WO2007084455A1 - Hydantoin compounds for the treatment of inflammatory disorders - Google Patents

Hydantoin compounds for the treatment of inflammatory disorders Download PDF

Info

Publication number
WO2007084455A1
WO2007084455A1 PCT/US2007/001030 US2007001030W WO2007084455A1 WO 2007084455 A1 WO2007084455 A1 WO 2007084455A1 US 2007001030 W US2007001030 W US 2007001030W WO 2007084455 A1 WO2007084455 A1 WO 2007084455A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
alkyl
group
aryl
heteroaryl
Prior art date
Application number
PCT/US2007/001030
Other languages
French (fr)
Inventor
Brian J. Lavey
Joseph A. Kozlowski
Guowei Zhou
Ling Tong
Wensheng Yu
Michael K.C. Wong
Bandarpalle B. Shankar
Neng-Yang Shih
M. Arshad Siddiqui
Kristin E. Rosner
Chaoyang Dai
Janeta Popovici-Muller
Vinay M. Girijavallabhan
Dansu Li
Aneta M. Micula
Seong-Heon Kim
De-Yi Yang
Razia Rizvi
Original Assignee
Schering Corporation
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
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=38038495&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=WO2007084455(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Schering Corporation filed Critical Schering Corporation
Priority to EP07716637.9A priority Critical patent/EP1973900B1/en
Priority to CA002637198A priority patent/CA2637198A1/en
Priority to JP2008551312A priority patent/JP2009523797A/en
Priority to AU2007207711A priority patent/AU2007207711A1/en
Priority to BRPI0706598-1A priority patent/BRPI0706598A2/en
Publication of WO2007084455A1 publication Critical patent/WO2007084455A1/en
Priority to IL192691A priority patent/IL192691A0/en
Priority to NO20083568A priority patent/NO20083568L/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/02Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains two hetero rings
    • C07D491/04Ortho-condensed systems
    • C07D491/044Ortho-condensed systems with only one oxygen atom as ring hetero atom in the oxygen-containing ring
    • C07D491/048Ortho-condensed systems with only one oxygen atom as ring hetero atom in the oxygen-containing ring the oxygen-containing ring being five-membered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing three or more hetero rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41641,3-Diazoles
    • A61K31/41781,3-Diazoles not condensed 1,3-diazoles and containing further heterocyclic rings, e.g. pilocarpine, nitrofurantoin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/02Stomatological preparations, e.g. drugs for caries, aphtae, periodontitis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/14Prodigestives, e.g. acids, enzymes, appetite stimulants, antidyspeptics, tonics, antiflatulents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/02Nasal agents, e.g. decongestants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/08Bronchodilators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/02Drugs for dermatological disorders for treating wounds, ulcers, burns, scars, keloids, or the like
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • A61P19/10Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease for osteoporosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/02Drugs for disorders of the nervous system for peripheral neuropathies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/04Centrally acting analgesics, e.g. opioids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/30Drugs for disorders of the nervous system for treating abuse or dependence
    • A61P25/32Alcohol-abuse
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • A61P27/06Antiglaucoma agents or miotics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • A61P29/02Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID] without antiinflammatory effect
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/12Drugs for disorders of the metabolism for electrolyte homeostasis
    • A61P3/14Drugs for disorders of the metabolism for electrolyte homeostasis for calcium homeostasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P33/00Antiparasitic agents
    • A61P33/02Antiprotozoals, e.g. for leishmaniasis, trichomoniasis, toxoplasmosis
    • A61P33/06Antimalarials
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • 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
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • 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
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P39/00General protective or antinoxious agents
    • 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
    • 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
    • A61P7/02Antithrombotic agents; Anticoagulants; Platelet aggregation inhibitors
    • 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
    • A61P7/04Antihaemorrhagics; Procoagulants; Haemostatic agents; Antifibrinolytic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/02Non-specific cardiovascular stimulants, e.g. drugs for syncope, antihypotensives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/04Inotropic agents, i.e. stimulants of cardiac contraction; Drugs for heart failure
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/08Vasodilators for multiple indications
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/06Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/06Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/14Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing three or more hetero rings
    • 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
    • C07D409/14Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing three or more hetero rings
    • 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
    • C07D413/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing three or more hetero rings
    • 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
    • C07D417/14Heterocyclic 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 three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D473/00Heterocyclic compounds containing purine ring systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D513/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for in groups C07D463/00, C07D477/00 or C07D499/00 - C07D507/00
    • C07D513/02Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for in groups C07D463/00, C07D477/00 or C07D499/00 - C07D507/00 in which the condensed system contains two hetero rings
    • C07D513/04Ortho-condensed systems

Definitions

  • This invention relates generally to novel hydantoin derivatives that can inhibit matrix metalloproteinases (MIVIPs) 1 a disintegrin and metalloproteases (ADAMs) and/or tumor necrosis factor alpha - converting enzyme (TACE) and in so doing prevent the release of tumor necrosis factor alpha (TNF- ⁇ ), pharmaceutical compositions comprising such compounds, and methods of treatment using such compounds.
  • MIVIPs matrix metalloproteinases
  • ADAMs disintegrin and metalloproteases
  • TACE tumor necrosis factor alpha - converting enzyme
  • Osteo- and rheumatoid arthritis are destructive diseases of articular cartilage characterized by localized erosion of the cartilage surface. Findings have shown that articular cartilage from the femoral heads of patients with OA, for example, had a reduced incorporation of radiolabeled sulfate over controls, suggesting that there must be an enhanced rate of cartilage degradation in OA (Mankin et al. J. Bone Joint Surg. 52A (1970) 424-434).
  • MPs Metalloproteases
  • MMPs are a family of over 20 different enzymes that are involved in a variety of biological processes important in the uncontrolled breakdown of connective tissue, including proteoglycan and collagen, leading to resorption of the extracellular matrix. This is a feature of many pathological conditions, such as RA and OA, corneal, epidermal or gastric ulceration; tumor metastasis or invasion; periodontal disease and bone disease. Normally these catabolic enzymes are tightly regulated at the level of their synthesis as well as at their level of extracellular activity through the action of specific inhibitors, such as alpha-2-macroglobulins and TIMPs (tissue inhibitor of MPs) 1 which form inactive complexes with the MMP's.
  • specific inhibitors such as alpha-2-macroglobulins and TIMPs (tissue inhibitor of MPs) 1 which form inactive complexes with the MMP's.
  • Tumor necrosis factor alpha is a cell-associated cytokine that is processed from a 26 kDa precursor form to a 17 kd active form. See Black R.A. "Tumor necrosis factor-alpha converting enzyme” lnt J Biochem Cell Biol. 2002 Jan; 34(1 ):1 -5 and Moss ML, White JM, Lambert MH, Andrews RC 11 TACE and other ADAM proteases as targets for drug discovery" Drug Discov Today. 2001 Apr 1 ;6(8):417-426, each of which is incorporated by reference herein.
  • TNF- ⁇ has been shown to play a pivotal role in immune and inflammatory responses. Inappropriate or over-expression of TNF- ⁇ is a hallmark of a number of diseases, including RA, Crohn's disease, multiple sclerosis, psoriasis and sepsis. Inhibition of TNF- ⁇ production has been shown to be beneficial in many preclinical models of inflammatory disease, making inhibition of TNF- ⁇ production or signaling an appealing target for the development of novel anti-inflammatory drugs.
  • TNF- ⁇ is a primary mediator in humans and animals of inflammation, fever and acute phase responses, similar to those observed during acute infection and shock. Excess TNF- ⁇ has been shown to be lethal. Blocking the effects of TNF- ⁇ with specific antibodies can be beneficial in a variety of conditions, including autoimmune diseases such as RA (Feldman et al, Lancet, (1994) 344, 1105), non-insulin dependent diabetes mellitus (Lohmander L. S. et al., Arthritis Rheum. 36 (1993) 1214-22) and Crohn's disease (Macdonald T. et al., Clin. Exp. Immunol. 81 (1990) 301).
  • RA Paindman et al, Lancet, (1994) 344, 1105
  • non-insulin dependent diabetes mellitus Lihmander L. S. et al., Arthritis Rheum. 36 (1993) 1214-22
  • Crohn's disease Macdonald T. e
  • TNF- ⁇ Compounds that inhibit the production of TNF- ⁇ are therefore of therapeutic importance for the treatment of inflammatory disorders. Recently it has been shown that metalloproteases, such as TACE, are capable of converting TNF- ⁇ from its inactive to active form (Gearing et al Nature, 1994, 370, 555). Since excessive TNF- ⁇ production has been noted in several disease conditions also characterized by MMP-mediated tissue degradation, compounds which inhibit both MMPs and TNF- ⁇ production may also have a particular advantage in diseases where both mechanisms are involved.
  • TACE is a member of the ADAM family of type I membrane proteins and mediates the ectodomain shedding of various membrane-anchored signaling and adhesion proteins. TACE has become increasingly important in the study of several diseases, including inflammatory disease, because of its role in cleaving TNF- ⁇ from its "stalk" sequence and thus releasing the soluble form of the TNF- ⁇ protein (Black R.A. lnt J Biochem Cell Biol. 2002 34,1-5).
  • PCT Publications WO2004024698 and WO2004024715 disclose sulphonamide derivatives that are potential inhibitors of MMPs.
  • PCT Publications WO2004056766, WO2003053940 and WO2003053941 also describe potential inhibitors of TACE and MMPs.
  • PCT Publication WO2006/019768 refers to hydantoin derivatives that are TACE inhibitors.
  • TNF- ⁇ MMPs, ADAMs, TACE, and TNF- ⁇
  • the inhibition of TNF- ⁇ , TACE and or other MMPs can prevent the degradation of cartilage by these enzymes, thereby alleviating the pathological conditions of OA and RA as well as many other auto-immune diseases.
  • the present invention provides a novel class of compounds as inhibitors of TACE, the production of TNF- ⁇ , MMPs, ADAMs 1 aggrecanase, or any combination thereof, methods of preparing such compounds, pharmaceutical compositions comprising one or more such compounds, methods of preparing pharmaceutical formulations comprising one or more such compounds, and methods of treatment, prevention, inhibition or amelioration of one or more diseases associated with TACE 1 aggrecanaseTNF- ⁇ , MMPs, ADAMs or any combination thereof using such compounds or pharmaceutical compositions.
  • the present application discloses a compound, or pharmaceutically acceptable salts or solvates of said compound, said compound having the general structure shown in Formula (I):
  • ring A is selected from the group consisting of aryl and heteroaryl, each of which is substituted with -Y-R 1 and -Z-R 2 as shown;
  • X is selected from the group consisting of -S-.-O-, -S(O) 2 , -S(O)-,
  • T is absent or present, and if present, T is selected from the group consisting of alkyl, aryl, and heteroaryl, wherein when each of said T aryl and heteroaryl contains two radicals on adjacent carbon atoms, said radicals may optionally be taken together with the carbon atoms to which they are attached to form a five- to eight-membered aryl or heteroaryl ring, wherein each of the aforementioned T aryl, and heteroaryl, optionally with said five- to eight- membered aryl or heteroaryl is independently unsubstituted or substituted with one to four R 10 moieties which can be the same or different;
  • V is absent or present, and if present V is selected from the group consisting of alkyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, and N- oxides of said heterocyclyl and heteroaryl, wherein when each of said V cycloalkyl, heterocyclyl, aryl, heteroaryl, and N-oxides of said heterocycyl and heteroaryl contains two radicals on adjacent carbon atoms, said radicals may optionally be taken together with the carbon atoms to which they are attached to form a five- to eight-membered cycloalkyl, aryl, heterocyclyl or heteroaryl ring; wherein each of said V alkyl, alkynyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl heterocyclyl, optionally with said five- to eight-membered cycloalkyl, aryl, heterocyclyl, or heteroaryl is independently unsubstit
  • Y is selected from the group consisting of a covalent bond, -(C(R 4 ) 2 ) n -, -N(R 4 )-, -C(O)N(R 4 )-, -N(R 4 )C(O)-, -N(R 4 )C(O)N(R 4 )-, -S(O) 2 N(R 4 )-, -N(R 4 )- S(O) 2 , -O- -S-, -C(O)-, -S(O)-, and -S(O) 2 -;
  • Z is selected from the group consisting of a covalent bond, -(C(R 4 ) 2 ) n -,
  • R is selected from the group consisting of H, cyano, -C(O)OH, - C(O)O-alkyl, -C(O)NH 2 , -C(O)NH(alkyl), -C(O)N(alkyl) 2 , alkynyl, halogen, alkyl, cycloalkyl, haloalkyl, aryl, heteroaryl, and heterocyclyl, wherein when each of said cycloalkyl, heterocyclyl, aryl and heteroaryl contains two radicals on adjacent carbon atoms, said radicals may optionally be taken together with the carbon atoms to which they are attached to form a five- to eight- membered cycloalkyl, aryl, heterocyclyl or heteroaryl ring; wherein each of the R 1 alkyl, alkynyl, aryl, heteroaryl, and heterocyclyl, optionally with the five or six-membered cycloalkyl,
  • R is selected from the group consisting of H, cyano, -C(O)OH, - C(O)O-alkyl, -C(O)NH 2 , -C(O)NH(alkyl), -C(O)N(alky0 2l alkynyl, halogen, alkyl, cycloalkyl, haloalkyl, aryl, heteroaryl, and heterocyclyl, wherein when each of said cycloalkyl, heterocyclyl, aryl and heteroaryl contains two radicals on adjacent carbon atoms, said radicals may optionally be taken together with the carbon atoms to which they are attached to form a five- to eight- membered cycloalkyl, aryl, heterocyclyl or heteroaryl ring; wherein each of the R 2 alkyl, cycloalkyl, aryl, heteroaryl, and heterocyclyl, optionally with the five or six-membered cycloalkyl, ary
  • each R is the same or different and is independently selected from the group consisting of H, alkyl, cycloalkyl, haloalkyl, hydroxy, -alkylcycloalkyl, - alkyl-N(alkyl) 2 , heterocyclyl, aryl, and heteroaryl, wherein when each of said cycloalkyl, heterocyclyl, ayl, and heteroaryl contains two radicals on adjacent carbon atoms, said radicals may optionally be taken together with the carbon atoms to which they are attached to form a five- to eight-membered cycloalkyl, aryl, heterocyclyl or heteroaryl ring;
  • R is selected from the group consisting of hydrogen, cyano, nitro,
  • R is selected from the group consisting of cyano, nitro,
  • R is selected from the group consisting of cyano, nitro,
  • -C(R 4 ) N-OR 4 , -OR 4 , -SR 4 , -N(R 4 ) 2 .
  • T and V is present, and V is other than alkynyl; wherein said T or V is substituted with at least one R 10 moiety selected from the group consisting of cyano, -C(O)OR 4 , -C(O)R 4 , -C(O)N(R 4 ) 2 , - C(O)N(R 4 )C(O)R 4 , -C(OJN(R 4 JC(O)NR 4 , -SR 4 , -S(O) 2 R 4 , -N(R 4 J-C(O)OR 4 , - OC(O)N(R 4 ) 2 , -N(R 4 )C(O)N(R 4 ) 2 , -N(R 4 )-C(O)-R 4 , -S(O) 2 N(R 4 J 2 , -S(O) 2 N(R 4 )- C(O)-R 4 ,
  • each R 4 independently is selected from the group consisting of H, alkyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl, wherein when each of said R 4 cycloalkyl, heterocyclyl, aryl, and heteroaryl contains contains two radicals on adjacent carbon atoms, said radicals may optionally be taken together with the carbon atoms to which they are attached to form a five- to eight- membered aryl, heteocyclyl, heteroaryl or cycloalkyl ring; with the proviso that when R 10 is
  • V is other than piperidinyl, and when R 10 is cyano, the compound of Formula (I) is other than
  • each of -Y-R 1 and -Z-R 2 is independently selected from the group consisting of cyano, -(C(R 4 ) 2 )n-C(O)OH, -(C(R 4 ) 2 ) n -C(O)O-alkyl, -(C(R 4 ) 2 ) n - C(O)NH 2 , -(C(R 4 ) 2 )n-C(O)NH(alkyl), and ⁇ (C(R 4 ) 2 ) n -C(O)N(alkyl) 2 .
  • each R 4 independently is H or alkyl; and n is 1-3;
  • T is aryl or heteroaryl, each of which is optionally substituted with one to four independently selected R 10 moieties, and V is alkynyl which is optionally substituted with one or two independently selected R 10 moieties;
  • ring A is heteroaryl, and V is other than alkynyl.
  • the present application discloses a compound, or pharmaceutically acceptable salts or solvates of said compound, said compound having the general structure shown in Formula (II): or a pharmaceutically acceptable salt, solvate, ester or isomer thereof, wherein: the ring labeled A is selected from the group consisting of aryl and heteroaryl, each of which is substituted with -Y-R 1 and -Z-R 2 as shown;
  • X is selected from the group consisting of -S-,-O-, -C(R ) 2 - or -N(R )- ;
  • T is absent or present, and if present, T is selected from the group consisting of H (with U and V being absent), alkyl, alkenyl, alkynyl, aryl, heteroaryl, heterocyclyl, cycloalkyl, alkylaryl-, and arylalkyl-, said aryl, heteroaryl, heterocyclyl, cycloalkyl, alkylaryl-, and arylalkyl- being optionally fused with one or more moieties selected from the group consisting of aryl, heteroaryl, heterocyclyl, cycloalkyl, alkylaryl and arylalkyl, wherein each of any of the aforementioned alkyl, alkenyl, aryl, heteroaryl, heterocyclyl, cycloalkyl, alkylaryl and arylalkyl groups of T is unsubstituted or optionally independently substituted with one to four R 10 moieties which can be the same or different, each R 10 moiety being
  • U is absent or present, and if present U is selected from the group consisting of alkynyl, -C(O)-, -C(O)O-, and -C(O)NR 4 -;
  • V is absent or present, and if present V is selected from the group consisting of hydrogen, alkyl, aryl, heteroaryl, heterocyclyl, heterocyclylalkyl-, cycloalkyl, alkylaryl-, and arylalkyl-, said aryl, heteroaryl, heterocyclyl, heterocyclylalkyl-, cycloalkyl, alkylaryl- and arylalkyl- being optionally fused with one or more moieties selected from the group consisting of aryl, heteroaryl, heterocyclyl, cycloalkyl, alkylaryl and arylalkyl, wherein each of any of the aforementioned alkyl, aryl, heteroaryl, heterocyclyl and cycloalkyl is unsubstituted or optionally independently substituted with one to four R 10 moieties which can be the same or different, each R 10 moiety being independently selected from the group of R 10 moieties below;
  • Y is selected from the group consisting of a covalent bond, -(C(R 4 ) 2 ) n -, -N(R 4 )-, -C(O)N(R 4 )-, -N(R 4 )C(O)-, -N(R 4 )C(O)N(R 4 )-, -S(O) 2 N(R 4 )-, -N(R 4 )- S(O) 2 , -O-.-S-, -C(O)-, -S(O)-, and -S(O) 2 -;
  • Z is selected from the group consisting of a covalent bond, -(C(R 4 ) 2 )n-, -N(R 4 )-, -C(O)N(R 4 )-, -N(R 4 )C(O)-, -N(R 4 )C(O)N(R 4 )-, -
  • R is selected from the group consisting of H 1 -OR 4 , cyano, -C(O)OR 4 , -C(O)N(R 4 J 2 , halogen, alkyl, fluoroalkyl, aryl, heteroaryl, heterocyclyl, alkylaryl, alkylheteroaryl and arylalkyl, wherein each of the alkyl, fluoroalkyl, aryl, heteroaryl, heterocyclyl, alkylaryl, alkylheteroaryl and arylalkyl groups of
  • R is unsubstituted or optionally independently substituted with one to four R 20 moieties which can be the same or different, each R 20 moiety being independently selected from the group of R 20 moieties below, with the proviso that when Y is present and Y is N, S or O, then R 1 is not halogen or cyano;
  • R is selected from the group consisting of H 1 -OR 4 , cyano, -C(O)OR 4 , -C(O)N(R 4 J 2 , halogen, alkyl, fluoroalkyl, aryl, heteroaryl, heterocyclyl, alkylaryl, alkylheteroaryl and arylalkyl, wherein each of the alkyl, fluoroalkyl, aryl,
  • R 2 heteroaryl, heterocyclyl, alkylaryl, alkylheteroaryl and arylalkyl groups of R is unsubstituted or optionally independently substituted with one to four R 20 moieties which can be the same or different, each R 20 moiety being independently selected from the group of R 20 moieties below, with the proviso that when Z is present and Z is N, S or O, then R 2 is not halogen; each R 3 is the same of different and is independently selected from the group consisting of H, alkyl, and aryl;
  • each R is the same or different and is independently selected from the group consisting of H, alkyl, heterocyclyl, aryl, and heteroaryl; R is selected from the group consisting of cyano, -OR 4 , -SR 4 , -N(R ) 2 ,
  • R is selected from the group consisting of halogen, alkyl, fluoroalkyl, -N(R 4 ) 2 , and -C(O)N(R 4 ) 2 ;
  • R is selected from the group consisting of halogen, alkyl, fluoroalkyl, -N(R 4 J 2 , and -C(O)N(R 4 ) 2 .
  • the compounds of Formula I can be useful as inhibitors of TACE and may be useful in the treatment and prevention of diseases associated with TACE, TNF- ⁇ , MMPs, ADAMs or any combination thereof.
  • the present invention provides a novel class of inhibitors of TACE, aggrecanase, the production of TNF- ⁇ , MMPs, ADAMs or any combination thereof, pharmaceutical compositions containing one or more of the compounds, methods of preparing pharmaceutical formulations comprising one or more such compounds, and methods of treatment, prevention or amelioration of one or more of the symptoms of inflammation.
  • the present invention provides compounds which are represented by structural Formula (I) or (II) above or a pharmaceutically acceptable salt, solvate, ester or isomer thereof, wherein the various moieties are as described above.
  • the isomer referred to the in the preceding paragraph is a stereoisomer.
  • X is selected from the group consisting of -(C(R 3 )2)m- and -N(R 3 )-.
  • X is -(C(R 3 ) 2 )m, wherein m is 1 or 2.
  • X is -(C(R 3 )2)m. wherein m is 1.
  • R 3 is H.
  • At least one of T and V is present, and V is other than alkynyl; wherein said T or V is substituted with at least one R 10 moiety selected from the group consisting of cyano, -C(O)OR 4 , - C(O)R 4 , -C(O)N(R 4 ) 2 , -C(O)N(R 4 )C(O)R 4 , -C(O)N(R 4 )C(O)NR 4 , -SR 4 , - S(O) 2 R 4 , -N(R 4 )-C(O)OR 4 , -OC(O)N(R 4 ) 2 , -N(R 4 )C(O)N(R 4 ) 2 , -N(R 4 J-C(O)-R 4 , - S(O) 2 N(R 4 J 2 , -S(O) 2 N(R 4 J-C(O)-R 4 , -S(O)
  • V is other than piperidinyl, and when R 10 is cyano, the compound of Formula (I) is other than
  • At least one of T and V is present, and V is other than alkynyl; wherein said T or V is substituted with at least one R 10 moiety selected from the group consisting of cyano, -C(O)OR 4 , - C(O)R 4 , -C(O)N(R 4 ) 2 , -C(O)N(R 4 JC(O)R 4 , -C(O)N(R 4 )C(O)NR 4 , -SR 4 , - S(O) 2 R 4 , -N(R 4 )-C(O)OR 4 , -OC(O)N(R 4 ) 2 , -N(R 4 )C(O)N(R 4 ) 2 , -N(R 4 J-C(O)-R 4 , - S(O) 2 N(R 4 J 2 , -S(O) 2 N(R 4 J-C(O)-R 4 ,
  • V is other than piperidinyl, and when R 10 is cyano, the compound of Formula (I) is other than
  • At least one of T and V is present, and V is other than alkynyl; wherein said T or V is substituted with at least one R 10 moiety selected from the group consisting of cyano, -C(O)OR 4 , - C(O)R 4 , -C(O)N(R 4 ) 2 , -C(O)N(R 4 )C(O)R 4 , -C(O)N(R 4 )C(O)NR 4 , -SR 4 , - S(O) 2 R 4 , -N(R 4 )-C(O)OR 4 , -OC(O)N(R 4 ) 2 , -N(R 4 )C(O)N(R 4 ) 2 , -N(R 4 )-C(O)-R 4 , - S(O) 2 N(R 4 ) 2 , -S(O) 2 N(R4)-C(O)-R 4
  • T and V is present, and V is other than alkynyl; wherein said T or V is substituted with at least one R 10 moiety selected from the group consisting of cyano, -C(O)OR 4 , - C(O)R 4 , -C(O)N(R 4 J 2 , -C(O)N(R 4 JC(O)R 4 , -C(O)N(R 4 JC(O)NR 4 , -SR 4 , - S(O) 2 R 4 , -N(R 4 J-C(O)OR 4 , -OC(O)N(R 4 J 2 , -N(R 4 JC(O)N(R 4 J 2 , -N(R 4 J-C(O)-R 4 , - S(O) 2 N(R 4 ) 2 , -S(O) 2 N(R 4 J-C(O)-R 4 , -N(
  • T or V is substituted with at least one R 10 moiety that is -SR 4 .
  • At least one of T and V is present, and V is other than alkynyl; wherein said T or V is substituted with at least one R 10 moiety selected from the group consisting of cyano, -C(O)OR 4 , - C(O)R 4 , -C(O)N(R 4 J 2 , -C(O)N(R 4 JC(O)R 4 , -C(O)N(R 4 )C(O)NR 4 , -SR 4 , - S(O) 2 R 4 , -N(R 4 J-C(O)OR 4 , -OC(O)N(R 4 ) 2 , -N(R 4 )C(O)N(R 4 ) 2 , -N(R 4 )-C(O)-R 4 , - S(O) 2 N(R 4 ) 2 , -S(O) 2 N(R 4 J-C(O)-R 4 ,
  • T or V is substituted with at least one R 10 moiety that is -S(O) 2 R 4 .
  • At least one of T and V is present, and V is other than alkynyl; wherein said T or V is substituted with at least one R 10 moiety selected from the group consisting of cyano, -C(O)OR 4 , - C(O)R 4 , -C(O)N(R 4 J 2 , -C(O)N(R 4 )C(O)R 4 , -C(O)N(R 4 )C(O)NR 4 , -SR 4 , - S(O) 2 R 4 , -N(R 4 )-C(O)OR 4 , -OC(O)N(R 4 J 2 , -N(R 4 )C(O)N(R 4 ) 2 , -N(R 4 )- C(O)-R 4 , - S(O) 2 N(R 4 ) 2 , -S(O) 2 N(R 4 J-C(O)-R 4 , -S(O)
  • T and V is present, and V is other than alkynyl; wherein said T or V is substituted with at least one R 10 moiety selected from the group consisting of cyano, -C(O)OR 4 , - C(O)R 4 , -C(O)N(R 4 ) 2 , -C(O)N(R 4 )C(O)R 4 , -C(O)N(R 4 )C(O)NR 4 , -SR 4 , - S(O) 2 R 4 , -N(R 4 )-C(O)OR 4 , -OC(O)N(R 4 ) 2 , -N(R 4 )C(O)N(R 4 ) 2 , -N(R 4 )-C(O)-R 4 , - S(O) 2 N(R 4 ) 2 , -S(O) 2 N(R 4 J-C(O)-
  • ring A is selected from the group consisting of phenyl, thiophenyl, pyridyl, pyrimidyl, and
  • T and V are present, and V is other than alkynyl; wherein said T or V is substituted with at least one R 10 moiety selected from the group consisting of cyano, -C(O)OR 4 , - C(O)R 4 , -C(O)N(R 4 ) 2 , -C(O)N(R 4 )C(O)R 4 , -C(O)N(R 4 )C(O)NR 4 , -SR 4 , - S(O) 2 R 4 , -N(R 4 )-C(O)OR 4 , -OC(O)N(R 4 ) 2 .
  • each R 4 independently is selected from the group consisting of H, alkyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl, wherein when each of said R 4 cycloalkyl, heterocyclyl, aryl, and heteroaryl contains contains two radicals on adjacent carbon atoms, said radicals may optionally be taken together with the carbon atoms to which they are attached to form a five- to eight-member
  • ring A is phenyl
  • At least one of T and V is present, and V is other than alkynyl; wherein said T or V is substituted with at least one R 10 moiety selected from the group consisting of cyano, -C(O)OR 4 , - C(O)R 4 , -C(O)N(R 4 J 2 , -C(O)N(R 4 JC(O)R 4 , -C(O)N(R 4 )C(O)NR 4 , -SR 4 , - S(O) 2 R 4 , -N(R 4 J-C(O)OR 4 , -OC(O)N(R 4 ) 2 , -N(R 4 )C(O)N(R 4 ) 2 , -N(R 4 )-C(O)-R 4 , - S(O) 2 N(R 4 ) 2 , -S(O) 2 N(R4)-C(O)-R 4 ,
  • At least one of T and V is present, and V is other than alkynyl; wherein said T or V is substituted with at least one R 10 moiety selected from the group consisting of cyano, -C(O)OR 4 , - C(O)R 4 , -C(O)N(R 4 J 2 , -C(O)N(R 4 )C(O)R 4 , -C(O)N(R 4 )C(O)NR 4 , -SR 4 , - S(O) 2 R 4 , -N(R 4 J-C(O)OR 4 , -OC(O)N(R 4 ) 2 , -N(R 4 )C(O)N(R 4 ) 2 , -N(R 4 )-C(O)-R 4 , - S(O) 2 N(R 4 J 2 , -S(O) 2 N(R 4 J-C(O)-R 4 ,
  • T is selected from the group consisting Of-CH 2 -, phenyl,
  • R 10 moieties optionally substituted with one to four R 10 moieties such that the number of R 10 moieties per each T does not exceed four.
  • At least one of T and V is present, and V is other than alkynyl; wherein said T or V is substituted with at least one R 10 moiety selected from the group consisting of cyano, -C(O)OR 4 , - C(O)R 4 , -C(O)N(R 4 ) 2> -C(O)N(R 4 )C(O)R 4 , -C(O)N(R 4 JC(O)NR 4 , -SR 4 , - S(O) 2 R 4 , -N(R 4 )-C(O)OR 4 , -OC(O)N(R 4 ) 2 , -N(R 4 )C(O)N(R 4 ) 2 , -N(R 4 )-C(O)-R 4 , - S(O) 2 N(R 4 J 2 , -S(O) 2 N(R 4 J-C(O)-R 4 ,
  • U is absent or present, and if present is selected from the group consisting of -C(O)-, and -C(O)O-.
  • At least one of T and V is present, and V is other than alkynyl; wherein said T or V is substituted with at least one R 10 moiety selected from the group consisting of cyano, -C(O)OR 4 , - C(O)R 4 , -C(O)N(R 4 ) 2 , -C(O)N(R 4 )C(O)R 4 , -C(O)N(R 4 )C(O)NR 4 , -SR 4 , - S(O) 2 R 4 , -N(R 4 J-C(O)OR 4 , -OC(O)N(R 4 ) 2 , -N(R 4 )C(O)N(R 4 ) 2l -N(R 4 J-C(O)-R 4 , - S(O) 2 N(R 4 J 2 , -S(O) 2 N(R 4 J-C(O)-R 4 ,
  • V is absent or present, and if present is selected from the group consisting of aryl, and heteroaryl, wherein when each of said V aryl and heteroaryl contains two radicals on adjacent carbon atoms, said radicals may optionally be taken together with the carbon atoms to which they are attached to form a five- to eight-membered aryl or heteroaryl ring; wherein each of the aforementioned V aryl, and heteroaryl, optionally with said five- to eight- membered aryl or heteroaryl is independently unsubstituted or substituted with one to four R 10 moieties which can be the same or different.
  • At least one of T and V is present, and V is other than alkynyl; wherein said T or V is substituted with at least one R 10 moiety selected from the group consisting of cyano, -C(O)OR 4 , - C(O)R 4 , -C(O)N(R 4 J 2 , -C(O)N(R 4 JC(O)R 4 , -C(O)N(R 4 )C(O)NR 4 , -SR 4 , - S(O) 2 R 4 , -N(R 4 )-C(O)OR 4 , -OC(O)N(R 4 J 2 , -N(R 4 JC(O)N(R 4 J 2 , -N(R 4 J-C(O)-R 4 , - S(OJ 2 N(R 4 J 2 , -S(O) 2 N(R 4 J-C(O)-R 4 , -N
  • V is selected from the group consisting of phenyl, pyridyl, pyrazinyl, indazolyl,
  • R 10 moieties which can be the same or different.
  • At least one of T and V is present, and V is other than alkynyl; wherein said T or V is substituted with at least one R 10 moiety selected from the group consisting of cyano, -C(O)OR 4 , - C(O)R 4 , -C(O)N(R 4 J 2 , -C(O)N(R 4 )C(O)R 4 , -C(O)N(R 4 JC(O)NR 4 , -SR 4 , - S(O) 2 R 4 , -N(R 4 J-C(O)OR 4 , -OC(O)N(R 4 ) 2 , -N(R 4 )C(O)N(R 4 ) 2 , -N(R 4 )-C(O)-R 4 , - S(O) 2 N(R 4 J 2 , -S(O) 2 N(R 4 J-C(O)-R 4 ,
  • At least one of T and V is present, and V is other than alkynyl; wherein said T or V is substituted with at least one R 10 moiety selected from the group consisting of cyano, -C(O)OR 4 , - C(O)R 4 , -C(O)N(R 4 ) 2 , -C(O)N(R 4 )C(O)R 4 , -C(O)N(R 4 JC(O)NR 4 , -SR 4 , - S(O) 2 R 4 , -N(R 4 )-C(O)OR 4 , -OC(O)N(R 4 ) 2> -N(R 4 )C(O)N(R 4 ) 2 , -N(R 4 )-C(O)-R 4 , - S(O) 2 N(R 4 ) 2 , -S(O) 2 N(R 4 J-C(O)-R 4
  • At least one of T and V is present, and V is other than alkynyl; wherein said T or V is substituted with at least one R 10 moiety selected from the group consisting of cyano, -C(O)OR 4 , - C(O)R 4 , -C(O)N(R 4 J 2 , -C(O)N(R 4 )C(O)R 4 , -C(O)N(R 4 )C(O)NR 4 , -SR 4 , - S(O) 2 R 4 , -N(R 4 J-C(O)OR 4 , -OC(O)N(R 4 J 2 , -N(R 4 )C(O)N(R 4 ) 2 , -N(R 4 )-C(O)-R 4 , - S(O) 2 N(R 4 J 2 , -S(O) 2 N(R4)-C(O)-R 4 .
  • each R 4 independently is selected from the group consisting of H, alkyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl, wherein when each of said R 4 cycloalkyl, heterocyclyl, aryl, and heteroaryl contains contains two radicals on adjacent carbon atoms, said radicals may optionally be taken together with the carbon atoms to which they are attached to form a five- to eight-membered aryl, heteocyclyl, heteroaryl or cycloalkyl ring; with the proviso that when R 10 is -S(O) 2 R 4 , V is other than piperidinyl, and when R 10 is cyano, the compound of Formula (I) is other
  • R 1 and R 2 are independently selected form the group consisting of H and alkyl.
  • At least one of T and V is present, and V is other than alkynyl; wherein said T or V is substituted with at least one R 10 moiety selected from the group consisting of cyano, -C(O)OR 4 , - C(O)R 4 , -C(O)N(R 4 ) 2 , -C(O)N(R 4 )C(O)R 4 , -C(O)N(R 4 )C(O)NR 4 , -SR 4 , - S(O) 2 R 4 , -N(R 4 )-C(O)OR 4 , -OC(O)N(R 4 J 2 , -N(R 4 )C(O)N(R 4 ) 2 , -N(R 4 )-C(O)-R 4 , - S(O) 2 N(R 4 J 2 , -S(O) 2 N(R-O-C(O)-R 4 , -S(O)
  • At least one of T and V is present, and V is other than alkynyl; wherein said T or V is substituted with at least one R 10 moiety selected from the group consisting of cyano, -C(O)OR 4 , - C(O)R 4 , -C(O)N(R 4 J 2 , -C(O)N(R 4 JC(O)R 4 , -C(O)N(R 4 )C(O)NR 4 , -SR 4 , - S(O) 2 R 4 , -N(R 4 )-C(O)OR 4 , -OC(O)N(R 4 ) 2 , -N(R 4 )C(O)N(R 4 ) 2> -N(R 4 )-C(O)-R 4 , - S(O) 2 N(R 4 J 2 , -S(O) 2 N(R 4 KKO)-R 4 , -N
  • R 1 is methyl.
  • T and V is present, and V is other than alkynyl; wherein said T or V is substituted with at least one R 10 moiety selected from the group consisting of cyano, -C(O)OR 4 , - C(O)R 4 , -C(O)N(R 4 ) 2l -C(O)N(R 4 JC(O)R 4 , -C(O)N(R 4 )C(O)NR 4 , -SR 4 , - S(O) 2 R 4 , -N(R 4 )-C(O)OR 4 , -OC(O)N(R 4 J 2 , -N(R 4 )C(O)N(R 4 ) 2 , -N(R 4 )-C(O)-R 4 , - S(O) 2 N(R 4 J 2 , -S(O) 2 N(R 4 J-C
  • each of -Y-R 1 and -Z-R 2 is independently selected from the group consisting of cyano, -(C(R 4 )2)n-C(O)OH, -(C(R 4 ) 2 ) n -C(O)O-alkyl, -(C(R 4 ) 2 ) n - C(O)NH 2 , -(C(R 4 Js) n -C(O)N H(alkyl), and -(C(R 4 ) 2 )n-C(O)N(alkyl) 2 , wherein each R 4 independently is H or alkyl; and n is 1-3.
  • each of -Y-R 1 and -Z-R 2 is independently selected from the group consisting of cyano, -(C(R 4 ) 2 ) n -C(O)OH, -(C(R 4 ) 2 ) n -C(O)O-alkyl, -(C(R 4 ) 2 ) n - C(O)NH 2 , -(C(R 4 ) 2 ) n -C(O)NH(alkyl), and -(C(R 4 ) 2 ) n -C(O)N(alkyl) 2 , wherein each R 4 independently is H or alkyl; and n is 1-3; wherein ring A is selected from the group consisting of phenyl, thiophenyl, pyridyl, pyrimidyl, and
  • each of -Y-R 1 and -Z-R 2 is independently selected from the group consisting of cyano, -(C(R 4 ) 2 ) n -C(O)OH, -(C(R 4 ) 2 ) n -C(O)O-alkyl, -(C(R 4 ) 2 ) n - C(O)NH 2 , -(C(R 4 ) 2 ) n -C(O)NH(alkyl), and -(C(R 4 ) 2 ) n -C(O)N(alkyl) 2 , wherein each R 4 independently is H or alkyl; and n is 1-3; wherein said ring A is phenyl.
  • each of -Y-R 1 and -Z-R 2 is independently selected from the group consisting of cyano, -(C(R 4 ) 2 ) n -C(O)OH, -(C(R 4 ) 2 ) n -C(O)O-alkyl, -(C(R 4 ) 2 ) n - C(O)NH 2 , -(C(R 4 ) 2 ) n -C(O)NH(alkyl), and -(C(R 4 ) 2 ) n -C(O)N(alkyl) 2 , wherein each R 4 independently is H or alkyl; and n is 1-3; wherein T or V is aryl which is unsubstituted or substituted with one to four R 10 moieties.
  • each of -Y-R 1 and -Z-R 2 is independently selected from the group consisting of cyano, -(C(R 4 ) 2 ) n -C(O)OH, -(C(R 4 ) 2 ) n -C(O)O-alkyl, - ⁇ C(R 4 ) 2 ) n - C(O)NH 2 , -(C(R 4 ) 2 ) n -C(O)NH(alkyl) I and -(C(R 4 ) 2 )n-C(O)N(alkyl) 2 , wherein each R 4 independently is H or alkyl; and n is 1-3; wherein said T or V is phenyl which is unsubstituted or substituted with one to four R 10 moieties.
  • each of -Y-R 1 and -Z-R 2 is independently selected from the group consisting of cyano, -(C(R 4 ) 2 ) n -C(O)OH, -(C(R 4 ) 2 ) n -C(O)O-alkyl, ⁇ iC(R 4 ) 2 ) n - C(O)NH 2 , -(C(R 4 ) 2 )n-C(O)NH(alkyl), and -(C(R 4 ) 2 ) n -C(O)N(alkyl) 2> wherein each R 4 independently is H or alkyl; and n is 1-3; wherein R 10 is fluoro.
  • each of -Y-R 1 and -Z-R 2 is independently selected from the group consisting of cyano, -(C(R 4 ) 2 )n-C(O)OH, -(C(R 4 ) 2 ) n -C(O)O-alkyl, -(C(R 4 ) 2 ) n - C(O)NH 2 , -(C(R 4 ) 2 )n-C(O)NH(alkyl), and -(C(R 4 ) 2 )n-C(O)N(alkyl) 2 , wherein each R 4 independently is H or alkyl; and n is 1-3; wherein only one of T and V is present.
  • each of -Y-R 1 and -Z-R 2 is independently selected from the group consisting of cyano, -(C(R 4 ) 2 ) n -C(O)OH, -(C(R 4 ) 2 ) n -C(O)O-alkyl, -(C(R 4 Ja) n -C(O)NH 2 , - (C(R 4 ) 2 ) n -C(O)NH(alkyl), and -(C(R 4 ) 2 ) n -C(O)N(alkyl) 2 , wherein each R 4 independently is H or alkyl; and n is 1-3; wherein U is absent.
  • each of -Y-R 1 and -Z-R 2 is independently selected from the group consisting of cyano, -(C(R 4 ) 2 ) n -C(O)OH, -(C(R 4 ) 2 ) n -C(O)O-alkyl, -(C(R 4 Jz) n -C(O)NH 2 , - (C(R 4 ) 2 ) n -C(O)NH(alkyl), and -(C(R 4 ) 2 ) n -C(O)N(alkyl) 2 , wherein each R 4 independently is H or alkyl; and n is 1-3, wherein n is 1.
  • each of -Y-R 1 and -Z-R 2 is independently selected from the group consisting of cyano, -(C(R 4 ) 2 ) n -C(O)OH, -(C(R 4 ) 2 ) n -C(O)O-alkyl, -(C(R 4 ) 2 ) n -C(O)NH 2) - (C(R 4 ) 2 ) n -C(O)NH(alkyl), and -(C(R 4 ) 2 ) n -C(O)N(alkyl) 2 , wherein each R 4 independently is H or alkyl; and n is 1-3; wherein each of Y and Z is independently selected from the group consisting of a covalent bond and - CH 2 -, and each of R 1 and R 2 is independently selected from the group consisting of cyano, -C(O)OH or -C(
  • each of -Y-R 1 and -Z-R 2 is independently selected from the group consisting of cyano, -(C(R 4 ) 2 ) n -C(O)OH, - ⁇ C(R 4 ) 2 )n-C(O)O-alkyl, -(C(R 4 ⁇ ) n -C(O)NH 2 , - (C(R 4 ) 2 ) n -C(O)NH(alkyl), and -(C(R 4 ) 2 ) ⁇ -C(O)N(alkyl) 2 , wherein each R 4 independently is H or alkyl; and n is 1-3; wherein Y is a covalent bond, and R 1 is H.
  • each of -Y-R 1 and -Z-R 2 is independently selected from the group consisting of cyano, -(C(R 4 ) 2 ) n -C(O)OH, -(C(R 4 ) 2 ) n -C(O)O-alkyl, -(C(R 4 ) 2 ) n -C(O)NH 2> - (C(R 4 ) 2 ) n -C(O)NH(alkyl), and -(C(R 4 ) 2 ) n -C(O)N(alkyl) 2 , wherein each R 4 independently is H or alkyl; and n is 1-3; wherein Z is a covalent bond, and R 2 is cyano.
  • each of -Y-R 1 and -Z-R 2 is independently selected from the group consisting of cyano, -(C(R 4 ) 2 ) n -C(O)OH, -(C(R 4 ) 2 ) n -C(O)O-alkyl, -(C(R 4 ) 2 ) n -C(O)NH 2 , - (C(R 4 ) 2 )n-C(O)NH(alkyl), and -(C(R 4 ) 2 ) n -C(O)N(alkyl) 2] wherein each R 4 independently is H or alkyl; and n is 1-3; wherein Z is -CH 2 -, and R 2 is - C(O)OH or -C(O)NH 2 .
  • each of -Y-R 1 and -Z-R 2 is independently selected from the group consisting of cyano, -(C(R 4 ) 2 ) n -C(O)OH, -(C(R 4 ) 2 ) n -C(O)O-alkyl, -(C(R 4 ) 2 ) n -C(O)NH 2 , - (C(R 4 ) 2 )n-C(O)NH(alkyl), and -(C(R 4 ) 2 ) n -C(O)N(alkyl) 2 , wherein each R 4 independently is H or alkyl; and n is 1-3; wherein the compound of Formula (I) is selected from the group consisting of:
  • T is aryl or heteroaryl, each of which is optionally substituted with one to four independently selected R 10 moieties, and V is alkynyl which is optionally substituted with one or two independently selected R 10 moieties.
  • T is aryl or heteroaryl, each of which is optionally substituted with one to four independently selected R 10 moieties, and V is alkynyl which is optionally substituted with one or two independently selected R 10 moieties; wherein ring A is selected from the group consisting of phenyl, thiophenyl, pyridyl, pyrimidyl, and
  • T is aryl or heteroaryl, each of which is optionally substituted with one to four independently selected R 10 moieties, and V is alkynyl which is optionally substituted with one or two independently selected R 10 moieties; wherein said ring A is phenyl.
  • T is aryl or heteroaryl, each of which is optionally substituted with one to four independently selected R 10 moieties, and V is alkynyl which is optionally substituted with one or two independently selected R 10 moieties; wherein T is aryl, U is -O- or absent, and V is alkynyl which is unsubstituted or substituted with one or two R 10 moieties selected from the group consisting of -OR 4 , -N(R 4 ⁇ 1 and heteroaryl; wherein when said heteroaryl contains two radicals on adjacent carbon atoms, said radicals may optionally be taken together with the carbon atoms to which they are attached to form a five- to eight-membered aryl, heteocyclyl, heteroaryl or cycloalkyl ring; wherein each R 4 independently is H or alkyl, and said R 10 heteroaryl is optionally independently substituted with one to four R 30 moieties which can be the same or different
  • T is aryl or heteroaryl, each of which is optionally substituted with one to four independently selected R 10 moieties, and V is alkynyl which is optionally substituted with one or two independently selected R 10 moieties; wherein T is phenyl.
  • T is aryl or heteroaryl, each of which is optionally substituted with one to four independently selected R 10 moieties, and V is alkynyl which is optionally substituted with one or two independently selected R 10 moieties; wherein said V alkynyl is selected from the group consisting of -CH 2 -C ⁇ C-CH 3 , and R 10 substituted -CsC-, and -CH2-C ⁇ C-CH2 ⁇ .
  • T is aryl or heteroaryl, each of which is optionally substituted with one to four independently selected R 10 moieties, and V is alkynyl which is optionally substituted with one or two independently selected R 10 moieties; wherein said R 10 substituents are selected from the group consisting of -N(alkyl) 2 , -OH, -OCH 3 , and pyridyl.
  • T is aryl or heteroaryl, each of which is optionally substituted with one to four independently selected R 10 moieties, and V is alkynyl which is optionally substituted with one or two independently selected R 10 moieties; wherein the compound of Formula (I) is selected from the group consisting of: or a pha rmaceutically acceptable salt, solvate, or ester thereof.
  • At least one of T and V is present, ring A is heteroaryl, and V is other than alkynyl.
  • At least one of T and V is present, ring A is heteroaryl, and V is other than alkynyl; wherein ring A is selected from the group consisting of thiophenyl, pyridyl, pyrimidyl, and each of which is substituted with -Y-R 1 and -Z-R 2 as shown.
  • At least one of T and V is present, ring A is heteroaryl, and V is other than alkynyl; wherein T is selected from the group consisting of alkyl, and halo-substituted aryl.
  • At least one of T and V is present, ring A is heteroaryl, and V is other than alkynyl; wherein U and V are absent.
  • At least one of T and V is present, ring A is heteroaryl, and V is other than alkynyl; wherein Y is selected from the group consisting of a covalent bond and — O-, and Z is a covalent bond.
  • At least one of T and V is present, ring A is heteroaryl, and V is other than alkynyl; wherein R 1 is selected from the group consisting of H and -CH 3 ; and R 2 is H.
  • At least one of T and V is present, ring A is heteroaryl, and V is other than alkynyl; wherein Y is a covalent bond R 1 is H.
  • At least one of T and V is present, ring A is heteroaryl, and V is other than alkynyl; wherein Y is is -O- and R 1 is -CH 3 .
  • At least one of T and V is present, ring A is heteroaryl, and V is other than alkynyl; wherein Z is a covalent bond and R 2 is H.
  • At least one of T and V is present, ring A is heteroaryl, and V is other than alkynyl; wherein the compound of Formula (I) is selected from the group consisting of:
  • the compound of Formula (I) is selected from the group consisting of compounds listed in the table below (Table 1), or a pharmaceutically acceptable salt, solvate, ester or isomer thereof.
  • Table 1 This table also lists the mass spectroscopy data and the Ki rating for each compound.
  • Those compounds having a Ki value of less than 10 nM ( ⁇ 10 nM) are designated with letter "A”; those with a Ki value of from 10 to less than 100 nM (10 - ⁇ 100 nM) are designated with letter "B”; those with a Ki value of from 100 to 1000 nM are designated with letter "C”; and those with a Ki value of more than 1000 nM (>1000 nM) are designated with letter "D”.
  • the syntheis and characterization of these compounds is described hereinbelow in the "EXAMPLES” section of the present application.
  • the compound of Formula (I) is selected from the group consisting of:
  • Patient includes both human and animals.
  • “Mammal” means humans and other mammalian animals.
  • Alkyl means an aliphatic hydrocarbon group which may be straight or branched and comprising about 1 to about 20 carbon atoms in the chain. Preferred alkyl groups contain about 1 to about 12 carbon atoms in the chain. More preferred alkyl groups contain about 1 to about 6 carbon atoms in the chain. Branched means that one or more lower alkyl groups such as methyl, ethyl or propyl, are attached to a linear alkyl chain. "Lower alkyl” means a group having about 1 to about 6 carbon atoms in the chain which may be straight or branched.
  • the alkyl group may be substituted by one or more substituents which may be the same or different, each substituent being independently selected from the group consisting of halo, alkyl, aryl, cycloalkyl, cyano, hydroxy, alkoxy, alkylthio, amino, -NH(alkyl), -NH(cycloalkyl), -N(alkyl) 2 , carboxy and -C(O)O-alkyl.
  • substituents which may be the same or different, each substituent being independently selected from the group consisting of halo, alkyl, aryl, cycloalkyl, cyano, hydroxy, alkoxy, alkylthio, amino, -NH(alkyl), -NH(cycloalkyl), -N(alkyl) 2 , carboxy and -C(O)O-alkyl.
  • suitable alkyl groups include methyl, ethyl, n-propyl, isopropyl
  • Alkenyl means an aliphatic hydrocarbon group containing at least one carbon-carbon double bond and which may be straight or branched and comprising about 2 to about 15 carbon atoms in the chain.
  • Preferred alkenyl groups have about 2 to about 12 carbon atoms in the chain; and more preferably about 2 to about 6 carbon atoms in the chain.
  • Branched means that one or more lower alkyl groups such as methyl, ethyl or propyl, are attached to a linear alkenyl chain.
  • “Lower alkenyl” means about 2 to about 6 carbon atoms in the chain which may be straight or branched.
  • suitable alkenyl groups include ethenyl, propenyl, n-butenyl, 3-methylbut-2- enyl, n-pentenyl, octenyl and decenyl.
  • Alkynyl means an aliphatic hydrocarbon group containing at least one carbon-carbon triple bond and which may be straight or branched and comprising about 2 to about 15 carbon atoms in the chain.
  • Preferred alkynyl groups have about 2 to about 12 carbon atoms in the chain; and more preferably about 2 to about 4 carbon atoms in the chain.
  • Branched means that one or more lower alkyl groups such as methyl, ethyl or propyl, are attached to a linear alkynyl chain.
  • “Lower alkynyl” means about 2 to about 6 carbon atoms in the chain which may be straight or branched.
  • Non-limiting examples of suitable alkynyl groups include ethynyl, propynyl, 2-butynyl and 3- methylbutynyl.
  • substituted alkynyl means that the alkynyl group may be substituted by one or more substituents which may be the same or different, each substituent being independently selected from the group consisting of alkyl, aryl and cycloalkyl.
  • Aryl means an aromatic monocyclic or multicyclic ring system comprising about 6 to about 14 carbon atoms, preferably about 6 to about 10 carbon atoms.
  • the aryl group can be optionally substituted with one or more "ring system substituents" which may be the same or different, and are as defined herein.
  • suitable aryl groups include phenyl and naphthyl.
  • Heteroaryl means an aromatic monocyclic or multicyclic ring system comprising about 5 to about 14 ring atoms, preferably about 5 to about 10 ring atoms, in which one or more of the ring atoms is an element other than carbon, for example nitrogen, oxygen or sulfur, alone or in combination. Preferred heteroaryls contain about 5 to about 6 ring atoms.
  • the "heteroaryl” can be optionally substituted by one or more "ring system substituents" which may be the same or different, and are as defined herein.
  • the prefix aza, oxa or thia before the heteroaryl root name means that at least a nitrogen, oxygen or sulfur atom respectively, is present as a ring atom.
  • heteroaryl may also include a heteroaryl as defined above fused to an aryl as defined above.
  • suitable heteroaryls include pyridyl, pyrazinyl, furanyl, thienyl, pyrimidinyl, pyridone (including N- substituted pyridones), isoxazolyl, isothiazolyl, oxazolyl, thiazolyl, pyrazolyl, furazanyl, pyrrolyl, pyrazolyl, triazolyl, 1 ,2,4-thiadiazolyl, pyrazinyl, pyridazinyl, quinoxalinyl, phthalazinyl, oxindolyl, imidazo[1 ,2-a]pyridinyl, imidazo[2,1- b]thiazolyl, benzofura
  • Aralkyl or “arylalkyl” means an aryl-alkyl- group in which the aryl and alkyl are as previously described. Preferred aralkyls comprise a lower alkyl group. Non-limiting examples of suitable aralkyl groups include benzyl, 2- phenethyl and naphthalenylmethyl. The bond to the parent moiety is through the alkyl.
  • Alkylaryl means an alkyl-aryl- group in which the alkyl and aryl are as previously described. Preferred alkylaryls comprise a lower alkyl group. Non- limiting example of a suitable alkylaryl group is tolyl. The bond to the parent moiety is through the aryl.
  • Cycloalkyl means a non-aromatic mono- or multicyclic ring system comprising about 3 to about 10 carbon atoms, preferably about 5 to about 10 carbon atoms. Preferred cycloalkyl rings contain about 5 to about 7 ring atoms.
  • the cycloalkyl can be optionally substituted with one or more "ring system substituents" which may be the same or different, and are as defined above.
  • suitable monocyclic cycloalkyls include cyclopropyl, cyclopentyl, cyclohexyl, cycloheptyl and the like.
  • suitable multicyclic cycloalkyls include 1-decalinyl, norbornyl, adamantyl and the like.
  • Cycloalkenyl means a non-aromatic mono or multicyclic ring system comprising about 3 to about 10 carbon atoms, preferably about 5 to about 10 carbon atoms which contains at least one carbon-carbon double bond. Preferred cycloalkenyl rings contain about 5 to about 7 ring atoms.
  • the cycloalkenyl can be optionally substituted with one or more "ring system substituents" which may be the same or different, and are as defined above.
  • suitable monocyclic cycloalkenyls include cyclopentenyl, cyclohexenyl, cyclohepta-1,3-dienyl, and the like.
  • Non-limiting example of a suitable multicyclic cycloalkenyl is norbornylenyl.
  • Halogen means fluorine, chlorine, bromine, or iodine. Preferred are fluorine, chlorine and bromine.
  • Ring system substituent means a substituent attached to an aromatic or non-aromatic ring system which, for example, replaces an available hydrogen on the ring system.
  • Ring system substituents may be the same or different, each being independently selected from the group consisting of alkyl, alkenyl, alkynyl, aryl, heteroaryl, aralkyl, alkylaryl, heteroaralkyl, heteroarylalkenyl, heteroarylalkynyl, alkylheteroaryl, hydroxy, hydroxyalkyl, alkoxy, aryloxy, aralkoxy, acyl, aroyl, halo, nitro, cyano, carboxy, alkoxycarbonyl, aryloxycarbonyl, aralkoxycarbonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfo ⁇ yl, alkylthio, arylthio, heteroarylthio, aralkylthi
  • Ring system substituent may also mean a single moiety which simultaneously replaces two available hydrogens on two adjacent carbon atoms (one H on each carbon) on a ring system.
  • Examples of such moiety are methylene dioxy, ethylenedioxy, -C(CHs) 2 - and the like which form moieties such as, for example:
  • Heterocyclyl means a non-aromatic saturated monocyclic or multicyclic ring system comprising about 3 to about 10 ring atoms, preferably about 5 to about 10 ring atoms, in which one or more of the atoms in the ring system is an element other than carbon, for example nitrogen, oxygen or sulfur, alone or in combination. There are no adjacent oxygen and/or sulfur atoms present in the ring system.
  • Preferred heterocyclyls contain about 5 to about 6 ring atoms.
  • the prefix aza, oxa orthia before the heterocyclyl root name means that at least a nitrogen, oxygen or sulfur atom respectively is present as a ring atom.
  • Any -NH in a heterocyclyl ring may exist protected such as, for example, as an -N(Boc), -N(CBz) 1 -N(Tos) group and the like; such protections are also considered part of this invention.
  • the heterocyclyl can be optionally substituted by one or more "ring system substituents" which may be the same or different, and are as defined herein.
  • the nitrogen or sulfur atom of the heterocyclyl can be optionally oxidized to the corresponding N-oxide, S-oxide or S,S-dioxide.
  • heterocyclyl rings include piperidyl, pyrrolidinyl, piperazinyl, morpholinyl, thiomorpholinyl, thiazolidinyl, 1,4-dioxanyl, tetrahydrofuranyl, tetrahydrothiophenyl, lactam, lactone, and the like.
  • Heterocyclyl may also mean a single moiety (e.g., carbonyl) which simultaneously replaces two available hydrogens on the same carbon atom on a ring system. Example of such moiety is pyrrolidone:
  • Alkynylalkyl means an alkynyl-alkyl- group in which the alkynyl and alkyl are as previously described. Preferred alkynylalkyls contain a lower alkynyl and a lower alkyl group. The bond to the parent moiety is through the alkyl.
  • suitable alkynylalkyl groups include propargylrnethyl.
  • Heteroaralkyl means a heteroaryl-alkyl- group in which the heteroaryl and alkyl are as previously described. Preferred heteroaralkyls contain a lower alkyl group. Non-limiting examples of suitable aralkyl groups include pyridylmethyl, and quinolin-3-ylmethyl. The bond to the parent moiety is through the alkyl.
  • Hydroxyalkyl means a HO-alkyl- group in which alkyl is as previously defined. Preferred hydroxyalkyls contain lower alkyl. Non-limiting examples of suitable hydroxyalkyl groups include hydroxymethyl and 2-hydroxyethyl.
  • acyl means an H-C(O)-, alkyl-C(O)- or cycloalkyl-C(O)-, group in which the various groups are as previously described. The bond to the parent moiety is through the carbonyl. Preferred acyls contain a lower alky!. Non- limiting examples of suitable acyl groups include formyl, acetyl and propanoyl.
  • Aroyl means an aryl-C(O)- group in which the aryl group is as previously described. The bond to the parent moiety is through the carbonyl.
  • suitable groups include benzoyl and 1- naphthoyl.
  • Alkoxy means an alkyl-O- group in which the alkyl group is as previously described.
  • suitable alkoxy groups include methoxy, ethoxy, n-propoxy, isopropoxy and n-butoxy.
  • the bond to the parent moiety is through the ether oxygen.
  • Aryloxy means an aryl-O- group in which the aryl group is as previously described.
  • suitable aryloxy groups include phenoxy and naphthoxy.
  • the bond to the parent moiety is through the ether oxygen.
  • Alkyloxy means an aralkyl-O- group in which the aralkyl group is as previously described.
  • suitable aralkyloxy groups include benzyloxy and 1 - or 2-naphthalenemethoxy.
  • the bond to the parent moiety is through the ether oxygen.
  • Alkylthio means an alkyl-S- group in which the alkyl group is as previously described.
  • suitable alkylthio groups include methylthio and ethylthio.
  • the bond to the parent moiety is through the sulfur.
  • Arylthio means an aryl-S- group in which the aryl group is as previously described.
  • suitable arylthio groups include phenylthio and naphthylthio.
  • the bond to the parent moiety is through the sulfur.
  • Alkylthio means an aralkyl-S- group in which the aralkyl group is as previously described.
  • Non-limiting example of a suitable aralkylthio group is benzylthio.
  • the bond to the parent moiety is through the sulfur.
  • Alkoxycarbonyl means an alkyl-O-CO- group. Non-limiting examples of suitable alkoxycarbonyl groups include methoxycarbonyl and ethoxycarbonyl. The bond to the parent moiety is through the carbonyl.
  • Aryloxycarbonyl means an aryl-O-C(O)- group. Non-limiting examples of suitable aryloxycarbonyl groups include phenoxycarbonyl and naphthoxycarbonyl. The bond to the parent moiety is through the carbonyl.
  • Alkoxycarbonyl means an aralkyl-O-C(O)- group.
  • a suitable aralkoxycarbonyl group is benzyloxycarbonyl.
  • the bond to the parent moiety is through the carbonyl.
  • Alkylsulfonyl means an alkyl-S(O2)- group. Preferred groups are those in which the alkyl group is lower alkyl. The bond to the parent moiety is through the sulfonyl.
  • Arylsulfonyl means an aryl-S(C> 2 )- group. The bond to the parent moiety is through the sulfonyl.
  • substituted means that one or more hydrogens on the designated atom is replaced with a selection from the indicated group, provided that the designated atom's normal valency under the existing circumstances is not exceeded, and that the substitution results in a stable compound. Combinations of substituents and/or variables are permissible only if such combinations result in stable compounds.
  • stable compound' or “stable structure” is meant a compound that is sufficiently robust to survive isolation to a useful degree of purity from a reaction mixture, and formulation into an efficacious therapeutic agent.
  • isolated or “in isolated form” for a compound refers to the physical state of said compound after being isolated from a synthetic process or natural source or combination thereof.
  • purified or “in purified form” for a compound refers to the physical state of said compound after being obtained from a purification process or processes described herein or well known to the skilled artisan, in sufficient purity to be characterizable by standard analytical techniques described herein or well known to the skilled artisan.
  • protecting groups When a functional group in a compound is termed "protected", this means that the group is in modified form to preclude undesired side reactions at the protected site when the compound is subjected to a reaction. Suitable protecting groups will be recognized by those with ordinary skill in the art as well as by reference to standard textbooks such as, for example, T. W. Greene etal, Protective Groups in organic Synthesis (1991), Wiley, New York.
  • any variable e.g., aryl, heterocycle, R 2 , etc.
  • any variable e.g., aryl, heterocycle, R 2 , etc.
  • composition is intended to encompass a product comprising the specified ingredients in the specified amounts, as well as any product which results, directly or indirectly, from combination of the specified ingredients in the specified amounts.
  • Prodrugs and solvates of the compounds of the invention are also contemplated herein.
  • the term "prodrug”, as employed herein, denotes a compound that is a drug precursor which, upon administration to a subject, undergoes chemical conversion by metabolic or chemical processes to yield a compound of Formula I or a salt and/or solvate thereof.
  • a discussion of prodrugs is provided in T. Higuchi and V. Stella, Pro-drugs as Novel Delivery Systems (1987) 14 of the A.C.S. Symposium Series, and in Bioreversible Carriers in Drug Design, (1987) Edward B. Roche, ed., American Pharmaceutical Association and Pergamon Press, both of which are incorporated herein by reference thereto.
  • prodrug means a compound (e.g, a drug precursor) that is transformed in vivo to yield a compound of Formula (I) or a pharmaceutically acceptable salt, hydrate or solvate of the compound.
  • the transformation may occur by various mechanisms (e.g., by metabolic or chemical processes), such as, for example, through hydrolysis in blood.
  • a discussion of the use of prodrugs is provided by T. Higuchi and W. Stella, "Pro-drugs as Novel Delivery Systems," Vol. 14 of the A.C.S. Symposium Series, and in Bioreversible Carriers in Drug Design, ed. Edward B. Roche, American Pharmaceutical Association and Pergamon Press, 1987.
  • a prodrug can comprise an ester formed by the replacement of the hydrogen atom of the acid group with a group such as, for example, (Ci-C ⁇ JalkyI, (C2-Ci 2 )alkanoyloxymethyl, 1-(alkanoyloxy)ethyl having from 4 to 9 carbon atoms, 1-methyl-1-(alkanoyloxy)-ethyl having from 5 to 10 carbon atoms, alkoxycarbonyloxymethyl having from 3 to 6 carbon atoms, 1- (alkoxycarbonyloxy)ethyl having from 4 to 7 carbon atoms, 1-methyl-1- (alkoxycarbonyloxy)ethyl having from 5 to 8 carbon atoms, N- (alkoxycarbonyl)aminomethyl having from 3 to 9 carbon atoms, 1-(N- (alkoxycarbonyl)
  • a prodrug can be formed by the replacement of the hydrogen atom of the alcohol group with a group such as, for example, (C 1 - C 6 )alkanoyloxymethyl, 1 -((C 1 -C 6 )alkanoyloxy)ethyl, 1-methyl-1-((Cr C 6 )alkanoyloxy)ethyl, (Ci-Cejalkoxycarbonyloxymethyl, N-(Cr Cejalkoxycarbonylaminomethyl, succinoyl, (CrC 6 )alkanoyl, ⁇ -amino(Ci- C4)alkanyl, arylacyl and ⁇ -aminoacyl, or ⁇ -aminoacyl- ⁇ -aminoacyl, where each ⁇ -aminoacyl group is independently selected from the naturally occurring L- amino acids, P(O)(OH) 2 , -P(O)
  • a prodrug can be formed by the replacement of a hydrogen atom in the amine group with a group such as, for example, R-carbonyl, RO-carbonyl, NRR'- carbonyl where R and R 1 are each independently (Ci-Ci O )alkyl, (C 3 -C 7 ) cycloalkyl, benzyl, or R-carbonyl is a natural ⁇ -aminoacyl or natural ⁇ - aminoacyl, -C(OH)C(O)OY 1 wherein Y 1 is H, (CrC 6 )alkyl or benzyl, — C(OY 2 )Y 3 wherein Y 2 is (C 1 -C 4 ) alkyl and Y 3 is (d-QOalkyl, carboxy (d- C 6 )alkyl, amino(Ci-C 4 )alkyl or mono-N — or di-N,N-
  • Solvate means a physical association of a compound of this invention with one or more solvent molecules. This physical association involves varying degrees of ionic and covalent bonding, including hydrogen bonding. In certain instances the solvate will be capable of isolation, for example when one or more solvent molecules are incorporated in the crystal lattice of the crystalline solid.
  • Solvate encompasses both solution-phase and isolatable solvates. Non-limiting examples of suitable solvates include ethanolates, methanolates, and the like.
  • “Hydrate” is a solvate wherein the solvent molecule is H 2 O.
  • Effective amount or “therapeutically effective amount” is meant to describe an amount of compound or a composition of the present invention effective in inhibiting TACE, the production of TNF- ⁇ , MMPs, ADAMS or any combination thereof and thus producing the desired therapeutic, ameliorative, inhibitory or preventative effect.
  • the compounds of Formula I can form salts which are also within the scope of this invention.
  • Reference to a compound of Formula I herein is understood to include reference to salts thereof, unless otherwise indicated.
  • the term "salt(s)", as employed herein, denotes acidic salts formed with inorganic and/or organic acids, as well as basic salts formed with inorganic and/or organic bases.
  • zwitterions inner salts may be formed and are included within the term "salt(s)" as used herein.
  • Salts of the compounds of the Formula I may be formed, for example, by reacting a compound of Formula I with an amount of acid or base, such as an equivalent amount, in a medium such as one in which the salt precipitates or in an aqueous medium followed by lyophilization.
  • Exemplary acid addition salts include acetates, ascorbates, benzoates, benzenesulfonates, bisulfates, borates, butyrates, citrates, camphorates, camphorsulfonates, fumarates, hydrochlorides, hydrobromides, hydroiodides, lactates, maleates, methanesulfonates, naphthalenesulfonates, nitrates, oxalates, phosphates, propionates, salicylates, succinates, sulfates, tartarates, thiocyanates, toluenesulfonates (also known as tosylates,) and the like.
  • Exemplary basic salts include ammonium salts, alkali metal salts such as sodium, lithium, and potassium salts, alkaline earth metal salts such as calcium and magnesium salts, salts with organic bases (for example, organic amines) such as dicyclohexylamines, t-butyl amines, and salts with amino acids such as arginine, lysine and the like.
  • Basic nitrogen-containing groups may be quartemized with agents such as lower alkyl halides (e.g. methyl, ethyl, and butyl chlorides, bromides and iodides), dialkyl sulfates (e.g.
  • dimethyl, diethyl, and dibutyl sulfates dimethyl, diethyl, and dibutyl sulfates
  • long chain halides e.g. decyl, lauryl, and stearyl chlorides, bromides and iodides
  • aralkyl halides e.g. benzyl and phenethyl bromides
  • All stereoisomers for example, geometric isomers, optical isomers and the like
  • of the present compounds including those of the salts, solvates and prodrugs of the compounds as well as the salts and solvates of the prodrugs
  • those which may exist due to asymmetric carbons on various substituents including enantiomeric forms (which may exist even in the absence of asymmetric carbons), rotameric forms, atropisomers, and diastereomeric forms, are contemplated within the scope of this invention, as are positional isomers (such as, for example, 4-pyridyl and 3-pyridyl).
  • Individual stereoisomers of the compounds of the invention may, for example, be substantially free of other isomers, or may be admixed, for example, as racemates or with all other, or other selected, stereoisomers.
  • the chiral centers of the present invention can have the S or R configuration as defined by the IUPAC 1974 Recommendations.
  • the use of the terms "salt”, “solvate” "prodrug” and the like, is intended to equally apply to the salt, solvate and prodrug of enantiomers, stereoisomers, rotamers, tautomers, positional isomers, racemates or prodrugs of the inventive compounds.
  • the compounds according to the invention have pharmacological properties; in particular, the compounds of Formula I can be inhibitors of TACE, aggrecanase, TNF- ⁇ and/or MMP activity.
  • the invention provides a pharmaceutical composition comprising as an active ingredient at least one compound of formula (I). In another aspect, the invention provides a pharmaceutical composition of formula (I) additionally comprising at least one pharmaceutically acceptable carrier.
  • the invention provides a method of treating disorders associated with TACE, aggrecanase, TNF- ⁇ , MMPs, ADAMs or any combination thereof, said method comprising administering to a patient in need of such treatment an effective amount of at least one compound of formula (I).
  • the invention provides a use of a compound of formula (I) for the manufacture of a medicament to treat disorders associated with TACE, aggrecanase, TNF- ⁇ , MMPs, ADAMs or any combination thereof.
  • the compounds of Formula (I) can have anti-inflammatory activity and/or immunomodulatory activity and can be useful in the treatment of diseases including but not limited to septic shock, haemodynamic shock, sepsis syndrome, post ischaemic reperfusion injury, malaria, mycobacterial infection, meningitis, psoriasis, congestive heart failure, fibrotic diseases, cachexia, graft rejection, cancers such as cutaneous T-cell lymphoma, diseases involving angiogenesis, autoimmune diseases, skin inflammatory diseases, inflammatory bowel diseases such as Crohn's disease and colitis, OA and RA, ankylosing spondylitis, psoriatic arthritis, adult Still's disease, ureitis, Wegener's granulomatosis, Behcehe disease, Sjogren's syndrome, sarcoidosis, polymyositis, dermatomyositis, multiple sclerosis, sciatica, complex regional pain syndrome, radiation damage, hyperoxic alveolar injury, period
  • the invention provides a method of preparing a pharmaceutical composition for treating the disorders associated with TACE, aggrecanase, TNF- ⁇ , MMPs, ADAMs or any combination thereof, said method comprising bringing into intimate contact at least one compound of formula (I) and at least one pharmaceutically acceptable carrier.
  • the invention provides a compound of formula (I) exhibiting TACE, TNF- ⁇ , MMPs, ADAMs or any combination thereof inhibitory activity, including enantiomers, stereoisomers and tautomers of said compound, and pharmaceutically acceptable salts, solvates, or esters of said compound, said compound being selected from the compounds of structures listed in Table 1 set forth above.
  • the invention provides a pharmaceutical composition for treating disorders associated with TACE, aggrecanase, TNF- ⁇ , MMP, ADAM or any combination thereof in a subject comprising, administering to the subject in need of such treatment a therapeutically effective amount of at least one compound of formula (I) or a pharmaceutically acceptable salt, solvate, ester, or isomer thereof.
  • the invention provides a compound of formula (I) in purified form.
  • the invention provides a method of treating a condition or disease mediated by TACE, MMPs, TNF- ⁇ , aggrecanase, or any combination thereof in a subject comprising: administering to the subject in need of such treatment a therapeutically effective amount of at least one compound of formula (I) or a pharmaceutically acceptable salt, solvate, ester or isomer thereof.
  • the invention provides a method of treating a condition or disease selected from the group consisting of rheumatoid arthritis, osteoarthritis, periodontitis, gingivitis, corneal ulceration, solid tumor growth and tumor invasion by secondary metastases, neovascular glaucoma, inflammatory bowel disease, multiple sclerosis and psoriasis in a subject, comprising: administering to the subject in need of such treatment a therapeutically effective amount of at least one compound of formula (I) or a pharmaceutically acceptable salt, solvate, ester or isomer thereof.
  • a condition or disease selected from the group consisting of rheumatoid arthritis, osteoarthritis, periodontitis, gingivitis, corneal ulceration, solid tumor growth and tumor invasion by secondary metastases, neovascular glaucoma, inflammatory bowel disease, multiple sclerosis and psoriasis in a subject, comprising: administering to the subject in need of such treatment a therapeutically effective amount
  • the invention provides a method of treating a condition or disease selected from the group consisting of fever, cardiovascular conditions, hemorrhage, coagulation, cachexia, anorexia, alcoholism, acute phase response, acute infection, shock, graft versus host reaction, autoimmune disease and HIV infection in a subject comprising administering to the subject in need of such treatment a therapeutically effective amount of at least one compound of formula (I) or a pharmaceutically acceptable salt, solvate, ester, or isomer thereof.
  • the invention provides a method of treating a condition or disease selected from the group consisting of septic shock, haemodynamic shock, sepsis syndrome, post ischaemic reperfusion injury, malaria, mycobacterial infection, meningitis, psoriasis, congestive heart failure, fibrotic diseases, cachexia, graft rejection, cancers such as cutaneous T-cell lymphoma, diseases involving angiogenesis, autoimmune diseases, skin inflammatory diseases, inflammatory bowel diseases such as Crohn's disease and colitis, osteo and rheumatoid arthritis, ankylosing spondylitis, psoriatic arthritis, adult Still's disease, ureitis, Wegener's granulomatosis, Behcehe disease, Sjogren's syndrome, sarcoidosis, polymyositis, dermatomyositis, multiple sclerosis, sciatica, complex regional pain syndrome, radiation damage, hyperoxic alveolar injury, periodontal disease, HIV
  • the invention provides a method of treating a condition or disease associated with COPD, comprising: administering to the subject in need of such treatment a therapeutically effective amount of at least one compound of Formula (I) or a pharmaceutically acceptable salt, solvate, ester or isomer thereof.
  • the invention provides a method of treating a condition or disease associated with rheumatoid arthritis, comprising: administering to the subject in need of such treatment a therapeutically effective amount of at least one compound of formula (I) or a pharmaceutically acceptable salt, solvate, ester, or isomer thereof.
  • the invention provides a method of treating a condition or disease associated with Crohn's disease, comprising: administering to the subject in need of such treatment a therapeutically effective amount of at least one compound of formula (I) or a pharmaceutically acceptable salt, solvate, ester or isomer thereof.
  • the invention provides a method of treating a condition or disease associated with psoriasis, comprising: administering to the subject in need of such treatment a therapeutically effective amount of at least one compound of formula (I) or a pharmaceutically acceptable salt, solvate, ester, or isomer thereof.
  • the invention provides a method of treating a condition or disease associated with ankylosing spondylitis, comprising: administering to the subject in need of such treatment a therapeutically effective amount of at least one compound of formula (I) or a pharmaceutically acceptable salt, solvate, ester or isomer thereof.
  • the invention provides a method of treating a condition or disease associated with sciatica, comprising: administering to the subject in need of such treatment a therapeutically effective amount of at least one compound of formula (I) or a pharmaceutically acceptable salt, solvate, ester or isomer thereof.
  • the invention provides a method of treating a condition or disease associated with complex regional pain syndrome, comprising: administering to the subject in need of such treatment a therapeutically effective amount of at least one compound of formula (I) or a pharmaceutically acceptable salt, solvate, ester, or isomer thereof.
  • the invention provides a method of treating a condition or disease associated with psoriatic arthritis, comprising: administering to the subject in need of such treatment a therapeutically effective amount of at least one compound of formula (I), or a pharmaceutically acceptable salt, solvate, ester, or isomer thereof.
  • the invention provides a method of treating a condition or disease associated with multiple sclerosis, comprising: administering to the subject in need of such treatment a therapeutically effective amount of at least one compound of formula (I) or a pharmaceutically acceptable salt, solvate, ester or isomer thereof, in combination with a compound selected from the group consisting of AvonexD, Betaseron, Copaxone or other compounds indicated for the treatment of multiple sclerosis.
  • a compound of the present invention may be coadministered or used in combination with disease-modifying antirheumatic drugs (DMARDS) such as methotrexate, azathioprine, leflunomide, pencillinamine, gold salts, mycophenolate mofetil, cyclophosphamide and other similar drugs.
  • DARDS disease-modifying antirheumatic drugs
  • NSAIDs non-steroidal anti-inflammatory drugs
  • COX-2 selective (COX-2) inhibitors such as VioxxD and CelebrexD
  • immunosuppressives such as steroids, cyclosporin, Tacrolimus, rapamycin and the like
  • biological response modifiers BRMs
  • EnbrelD, RemicadeD, IL-1 antagonists anti-CD40, anti-CD28, IL-10, anti-adhesion molecules and the like
  • other anti-inflammatory agents such as p38 kinase inhibitors, PDE4 inhibitors, other chemically different TACE inhibitors, chemokine receptor antagonists, Thalidomide and other small molecule inhibitors of pro-inflammatory cytokine production.
  • a compound of the present invention may be co-administered or used in combination with an H1 antagonist for the treatment of seasonal allergic rhinitis and/or asthma.
  • H1 antagonists may be, for example, Claritin®, Clarinex®, Allegra®, or Zyrtec®.
  • the invention provides a method of treating a condition or disease mediated by TACE, MMPs, TNF- ⁇ , aggrecanase, or any combination thereof in a subject comprising: administering to the subject in need of such treatment a therapeutically effective amount of at least one compound of formula (I) or a pharmaceutically acceptable salt, solvate or isomer thereof in combination with a therapeutically effective amount of at least one medicament selected from the group consisting of disease modifying anti-rheumatic drugs (DMARDS), NSAIDs, COX-2 inhibitors, COX-1 inhibitors, immunosuppressives, biological response modifiers (BRMs), antiinflammatory agents and H1 antagonists.
  • DARDS disease modifying anti-rheumatic drugs
  • NSAIDs NSAIDs
  • COX-2 inhibitors COX-1 inhibitors
  • immunosuppressives biological response modifiers
  • BRMs biological response modifiers
  • the invention provides a method of treating a condition or disease selected from the group consisting of rheumatoid arthritis, osteoarthritis, periodontitis, gingivitis, corneal ulceration, solid tumor growth and tumor invasion by secondary metastases, neovascular glaucoma, inflammatory bowel disease, multiple sclerosis and psoriasis in a subject, comprising: administering to the subject in need of such treatment a therapeutically effective amount of at least one compound of Formula (I) or a pharmaceutically acceptable salt, solvate, ester, or isomer thereof in combination with a therapeutically effective amount of at least one medicament selected from the group consisting of DMARDS, NSAIDs, COX-2 inhibitors, COX-1 inhibitors, immunosuppressives, BRMs, anti-inflammatory agents and H1 antagonists.
  • a condition or disease selected from the group consisting of rheumatoid arthritis, osteoarthritis, periodontitis, gingivitis, corneal ulceration, solid tumor growth
  • the invention provides a method of treating a condition or disease selected from the group consisting of septic shock, haemodynamic shock, sepsis syndrome, post ischaemic reperfusion injury, malaria, mycobacterial infection, meningitis, psoriasis, congestive heart failure, fibrotic diseases, cachexia, graft rejection, cancers such as cutaneous T-cell lymphoma, diseases involving angiogenesis, autoimmune diseases, skin inflammatory diseases, inflammatory bowel diseases such as Crohn's disease and colitis, osteo and rheumatoid arthritis, ankylosing spondylitis, psoriatic arthritis, adult Still's disease, ureitis, Wegener's granulomatosis, Behcehe disease, Sjogren's syndrome, sarcoidosis, polymyositis, dermatomyositis, multiple sclerosis, sciatica, complex regional pain syndrome, radiation damage, hyperoxic alveolar injury, periodontal disease, HIV
  • the invention provides a method for treating RA comprising administering a compound of the formula I in combination with compound selected from the class consisting of a COX-2 inhibitor e.g. Celebrex® or Vioxx®; a COX-1 inhibitor e.g. Feldene®; an immunosuppressive e.g. methotrexate or cyclosporin; a steroid e.g. D- methasone; and anti-TNF- ⁇ compound, e.g. Enbrel® or Remicade®; a PDE IV inhibitor, or other classes of compounds indicated for the treatment of RA.
  • a COX-2 inhibitor e.g. Celebrex® or Vioxx®
  • COX-1 inhibitor e.g. Feldene®
  • an immunosuppressive e.g. methotrexate or cyclosporin
  • a steroid e.g. D- methasone
  • anti-TNF- ⁇ compound e.g. Enbrel® or Remicade®
  • the invention provides a method for treating multiple sclerosis comprising administering a compound of the formula (I) in combination with a compound selected from the group consisting of Avonex®, Betaseron, Copaxone or other compounds indicated for the treatment of multiple sclerosis.
  • TACE activity is determined by a kinetic assay measuring the rate of increase in fluorescent intensity generated by TACE catalyzed cleavage of an internally quenched peptide substrate (SPDL-3).
  • SPDL-3 internally quenched peptide substrate
  • the purified catalytic domain of recombinant human TACE (rhTACEc, Residue 215 to 477 with two mutation (S266A and N452Q) and a 6xHis tail) is used in the assay. It is purified from the baculovirus/Hi5 cells expression system using affinity chromatography.
  • the substrate SPDL-3 is an internally quenched peptide (MCA-Pro-Leu-Ala-Gln-Ala-Val-Arg-Ser-Ser-Ser-Dpa-Arg-NH2), with its sequence derived from the pro-TNFD cleavage site.
  • MCA is (7- Methoxycoumarin-4-yl)acetyl.
  • Dpa is N-3-(2,4-Dinitrophenyl)-L-2,3- diaminopropionyl.
  • a 50 /J assay mixture contains 20 mM HEPES, pH 7.3, 5 mM CaCI 2 , 100 DM ZnCI 2 , 2 % DMSO, 0.04% Methylcellulose, 30 ⁇ M SPDL-3, 7O pM rhTACEc and a test compound.
  • RhTACEc is pre-incubated with the testing compound for 90 min. at 25 0 C. Reaction is started by addition of the substrate. The fluorescent intensity (excitation at 320 nm, emission at 405 nm) was measured every 45 seconds for 30 min. using a fluorospectrometer (GEMINI XS, Molecular Devices). Rate of enzymatic reaction is shown as Units per second. Effect of a test compound is shown as % of TACE activity in the absence of the compound.
  • compositions containing the active ingredient may be in a form suitable for oral use, for example, as tablets, lozenges, aqueous or oily suspensions, dispersible powders or granules, emulsions, hard or soft capsules, or syrups or elixirs.
  • Compositions intended for oral use may be prepared according to any method known to the art for the manufacture of pharmaceutical compositions and such compositions may contain one or more agents selected from the group consisting of sweetening agents, flavoring agents, coloring agents and preserving agents in order to provide pharmaceutically elegant and palatable preparations. Tablets contain the active ingredient in admixture with non-toxic pharmaceutically acceptable excipients that are suitable for the manufacture of tablets.
  • excipients may be for example, inert diluents, such as calcium carbonate, sodium carbonate, lactose, calcium phosphate or sodium phosphate; granulating and disintegrating agents, for example, corn starch, or alginic acid; binding agents, for example starch, gelatin or acacia, and lubricating agents, for example magnesium stearate, stearic acid or talc.
  • the tablets may be uncoated or they may be coated by known techniques to delay disintegration and absorption in the gastrointestinal tract and thereby provide a sustained action over a longer period.
  • a time delay material such as glyceryl monostearate or glyceryl distearate may be employed. They may also be coated by the technique described in the U.S. Pat. Nos. 4,256,108; 4,166,452; and 4,265,874 to form osmotic therapeutic tablets for controlled release.
  • composition is also intended to encompass both the bulk composition and individual dosage units comprised of more than one (e.g., two) pharmaceutically active agents such as, for example, a compound of the present invention and an additional agent selected from the lists of the additional agents described herein, along with any pharmaceutically inactive excipients.
  • the bulk composition and each individual dosage unit can contain fixed amounts of the afore-said "more than one pharmaceutically active agents".
  • the bulk composition is material that has not yet been formed into individual dosage units.
  • An illustrative dosage unit is an oral dosage unit such as tablets, pills and the like.
  • the herein-described method of treating a patient by administering a pharmaceutical composition of the present invention is also intended to encompass the administration of the afore-said bulk composition and individual dosage units.
  • Formulations for oral use may also be presented as hard gelatin capsules wherein the active ingredients is mixed with an inert solid diluent, for example, calcium carbonate, calcium phosphate or kaolin, or a soft gelatin capsules where in the active ingredient is mixed with water or an oil medium, for example peanut oil, liquid paraffin or olive oil.
  • Aqueous suspensions contain the active material in admixture with excipients suitable for the manufacture of aqueous suspensions.
  • excipients are suspending agents, for example, sodium carboxymethylcellulose, methylcellulose, hydroxypropylmethyl-cellulose, sodium alginate, polyvinyl-pyrrolidone, gum tragacanth and gum acacia; dispersing or wetting agents may be a naturally-occurring phosphatide, for example, lecithin, or condensation products of an alkylene oxide with fatty acids, for example polyoxyethylene stearate, or condensation products of ethylene oxide with long chain aliphatic alcohols, for example, heptadecaethylene-oxycetanol, or condensation products of ethylene oxide with partial esters derived from fatty acids and a hexitol such as polyoxyethylene sorbitol monooleate, or condensation products of ethylene oxide with partial esters derived from fatty acids and hexitol anhydrides, for example, polyethylene sorbitan monooleate.
  • dispersing or wetting agents may be a naturally-occurring phosphatide, for example,
  • the aqueous suspensions may also contain one or more preservatives, for example, ethyl or n-propyl, p- hydroxybenzoate, one or more coloring agents, one or more flavoring agents, and one or more sweetening agents, such as sucrose, saccharin or aspartame.
  • preservatives for example, ethyl or n-propyl, p- hydroxybenzoate, one or more coloring agents, one or more flavoring agents, and one or more sweetening agents, such as sucrose, saccharin or aspartame.
  • Oily suspensions may be formulated by suspending the active ingredient in a vegetable oil, for example, arachis oil, olive oil, sesame oil or coconut oil, or in mineral oil such as liquid paraffin.
  • the oily suspensions may contain a thickening agent, for example, beeswax, hard paraffin or cetyl alcohol. Sweetening agents such as those set forth above, and flavoring agents may be added to provide a palatable oral preparation. These compositions may be preserved by the addition of an anti-oxidant such as ascorbic acid.
  • Dispersible powders and granules suitable for preparation of an aqueous suspension by the addition of water provide the active ingredient in admixture with a dispersing or wetting agent, suspending agent and one or more preservatives.
  • a dispersing or wetting agent e.g., sodium EDTA
  • suspending agent e.g., sodium EDTA
  • preservatives e.g., sodium EDTA, sodium sulfate, sodium bicarbonate
  • the pharmaceutical compositions of the invention may also be in the form of an oil-in-water emulsion.
  • the oily phase may be a vegetable oil, e.g., olive oil or arachis oil, or a mineral oil, e.g., liquid paraffin or mixtures of these.
  • Suitable emulsifying agents may be naturally-occurring phosphatides, e.g., soy beans, lecithin, and esters or partial esters derived from fatty acids and hexitol anhydrides, for example, sorbitan monooleate, and condensation products of the said partial esters with ethylene oxide, e.g., polyoxyethylene sorbitan monooleate.
  • the emulsions may also contain sweetening and flavoring agents.
  • Syrups and elixirs may be formulated with sweetening agents, for example, glycerol, propylene glycol, sorbitol or sucrose. Such formulations may also contain a demulcent, a preservative and flavoring and coloring agents.
  • sweetening agents for example, glycerol, propylene glycol, sorbitol or sucrose.
  • Such formulations may also contain a demulcent, a preservative and flavoring and coloring agents.
  • the pharmaceutical compositions may be in the form of a sterile injectable aqueous or oleagenous suspension.
  • This suspension may be formulated according to the known art using those suitable dispersing or wetting agents and suspending agents which have been mentioned above.
  • the sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally-acceptable diluent or solvent, e.g., as a solution in 1 ,3-butane diol.
  • the acceptable vehicles and solvents that may be employed are water, Ringer's solution and isotonic sodium chloride solution.
  • sterile fixed oils are conventionally employed as a solvent or suspending medium.
  • any bland fixed oil may be employed including synthetic mono- or diglycerides.
  • fatty acids such as oleic acid find use in the preparation of injectables.
  • compositions can be prepared by mixing the drug with a suitable non-irritating excipient which is solid at ordinary temperatures but liquid at the rectal temperature and will W 2
  • Such materials are cocoa butter and polyethylene glycols.
  • creams, ointments, jellies, solutions or suspensions, etc., containing the compounds of the invention are employed.
  • topical application shall include mouthwashes and gargles.
  • the compounds for the present invention can be administered in the intranasal form via topical use of suitable intranasal vehicles, or via transdermal routes, using those forms of transdermal skin patches well known to those of ordinary skill in the art.
  • the dosage administration will, of course, be continuous rather than intermittent throughout the dosage regimen.
  • Compounds of the present invention may also be delivered as a suppository employing bases such as cocoa butter, glycerinated gelatin, hydrogenated vegetable oils, mixtures of polyethylene glycols of various molecular weights and fatty acid esters of polyethylene glycol.
  • the dosage regimen utilizing the compounds of the present invention is selected in accordance with a variety of factors including type, species, weight, sex and medical condition of the patient; the severity of the condition to be treated; the route of administration; the renal and hepatic function of the patient; and the particular compound thereof employed.
  • a physician or veterinarian of ordinary skill can readily determine and prescribe the effective amount of the drug required to prevent, counter, arrest or reverse the progress of the condition.
  • Optimal precision in achieving concentration of drug within the range that yields efficacy without toxicity requires a regimen based on the kinetics of the drug's availability to target sites. This involves a consideration of the distribution, equilibrium, and elimination of a drug.
  • doses of the compound of Formula I useful in the method of the present invention range from 0.01 to 1000 mg per day. More preferably, dosages range from 0.1 to 1000 mg/day. Most preferably, dosages range from 0.1 to 500 mg/day.
  • the compositions are preferably provided in the form of tablets containing 0.01 to 1000 milligrams of the active ingredient, particularly 0.01, 0.05, 0.1 , 0.5, 1.0, 2.5, 5.0, 10.0, 15.0, 25.0, 50.0, 100 and 500 milligrams of the active ingredient for the symptomatic adjustment of the dosage to the patient to be treated.
  • An effective amount of the drug is ordinarily supplied at a dosage level of from about 0.0002 mg/kg to about 50 mg/kg of body weight per day. The range is more particularly from about 0.001 mg/kg to 1 mg/kg of body weight per day.
  • the active agent of the present invention may be administered in a single daily dose, or the total daily dosage may be administered in dividend doses of two, three or four time daily.
  • the amount of active ingredient that may be combined with the carrier materials to produce single dosage form will vary depending upon the host treated and the particular mode of administration.
  • the compounds of the invention may be produced by processes known to those skilled in the art and as shown in the following reaction schemes and in the preparations and examples described below.
  • step 1 Compound 1A (either commercially available, or prepared by a procedure similar to that described by Abdalla, G. M. and Sowell, J. W. Journal of Heterocyclic Chemistry, 1987, 24(2), 297-301 ) was treated with one equivalent of Di-tert-butyl dicarbonate in polar solvent, such as DMF, for 30 minutes to 12 hours. The solvent was removed and compound 1B could be used without further purification or purified by silica gel chromatography.
  • polar solvent such as DMF
  • step 2 compound 1B was reacted with potassium cyanide and ammonium carbonate in an alcohol and water solution, at 50 0 C to 90 0 C, for 5 hours to 48 hours. After cooling down, water was added and compound 1C could be collected by filtration.
  • step 3 compound 1C was stirred with 2 to 20 equivalents of hydrogen chloride in methanol for 5 to 48 hours. After ethyl ether was added, compound 1D could be collected by filtration.
  • step 3 compound 1C was stirred with 2 to 20 equivalents of hydrogen chloride in methanol for 5 to 48 hours. After ethyl ether was added, compound 1D could be collected by filtration.
  • Step 2 Compound 4C was converted to Compound 4D using a procedure similar to that described in Example 3.
  • Compound 5A was prepared using chemistry similar to that described in Examples 1 , 2, 3 and 4.
  • Compound 6A was prepared using chemistry similar to that described in Examples 1 , 2, 34, and 5.
  • Step 4 Compound 7C was dissolved in 18 ml_ of absolute ethanol and 8 ml_ of water. The solution was added to a thick walled glass pressure bottle and ammonium carbonate (2.21 g, 23.0 mniol) was added. The bottle was capped and the reaction mixture was stirred at rt for 15 min. Potassium cyanide was added, the bottle was recapped, and the reaction mixture was stirred at 70 0 C for 16 hr. The resulting mixture was poured into 250 mL of water and suction filtered to give 7D (1.86 g) as a white solid.
  • Example 8 The procedures described in Example 7 were used to prepare compounds 1 through 17 in Table 1.
  • Example 8 The procedures described in Example 7 were used to prepare compounds 1 through 17 in Table 1.
  • Compound 8A was prepared using chemistry similar to that described in Examples 1 , 2, 3, 4 and 5.
  • Racemic compound 9A was prepared using chemistry similar to that described in Examples 1 , 2, 3, 4 and 5. The enantiomers were resolved by Chiralcel OD column (Mobile phase: Hexane/2-propanol 3:1). The first peak was collected and concentrated to give compound 9A in its enantiomerically pure form.
  • Compound 11 A was prepared via procedure similar to those described in Examples 8 and 10.
  • the two isomers were separated using a chiral OD column. One gram of material was injected into the column and the two peaks were separated by using a solvent mixture of 85% hexanes/ethanol. The second isomer was the desired compound 309B (400 mg, 80%).
  • Part B Compound 321 (2.1 g, 11.1 mmol) was dissolved in toluene (30 mL) and methanol (30 ml) and cooled in an ice bath. TMS diazomethane (2M in hexanes, 11 mL) was added dropwise until yellow color persisted. The solvent was evaporated under reduced pressure to provide the desired product 322 with no need for purification (2.2 g, quant.).
  • the aryl ether compounds 82 to 90 were prepared from compound 8B using a procedure based on that described by E. Buck and 2. J. Song in Organic Synthesis VoI 82, p. 69, followed by a standard SEM deprotection sequence. An example is provided below.
  • the Schlenck tube was placed in a 100 0 C oil bath and heated to 150 0 C.
  • the reaction mixture was stirred for 23 h at 150 0 C.
  • the reaction mixture was allowed to cool to rt, then diluted with EtOAc and water.
  • Aqueous 1% EDTA was added and the layers were separated.
  • the organic layer was washed with 1% aq EDTA, water, and brine.
  • the resulting organic solution was dried with MgSO 4 , filtered, and concentrated to dryness. A brown solid was obtained.
  • the crude product was purified via sgc using a Biotage SiO 2 cartridge and a 1%-2.5% MeOH/CH 2 CI 2 gradient as the mobile phase. The major spot was collected as product, giving 0.04 g of compound 8C.
  • the reaction was stirred for 15 min. The ice bath was removed, and the reaction mixture was stirred for 3 h at rt. Acetic acid was added until the reaction mixture was weakly acidic. The reaction mixture was partially concentrated on the rotovap. EtOAc and water were added. The layers were separated. The organic layer was washed with water and brine, dried with MgSCU, filtered, and concentrated to dryness.
  • the crude product was purified via reverse phase chromatography using an lsco C-18 cartridge (43g). The mobile phase was a 15% to 80% CH 3 CN/H 2 O gradient with 0.1 % (volume) formic acid added to both components of the mobile phase. The main peak was isolated as product giving compound 8D.
  • StepL Compound 401 (100 mg, 0.33 mmol) was combined with compound 45A (80 mg, 0.4 mmol), Pd(PPh 3 J 2 CI 2 (8 mg, 0.012 mmol), CuI (17 mg, 0.1 mmol), diisopropylamine (0.08 mL, 0.58 mmol) in DMF (1 mL) and stirred at 85°C for 2 h.
  • the reaction mixture was purified on a Gilson reverse phase HPLC (0-40% acetonitrile in H 2 O with formic acid 0.1%) afforded the desired product 45B(18 mg, 13%).
  • Step 2 Compound 45B (20 mg, 0.23 mmol) was stirred in MeOH (5 mL) and HCI (1N, aq., cat.)- The reaction was stirred at rt for 2 h. Solvent was removed and the crude material was purified on a Gilson reverse phase HPLC (0-50% acetonitrile in H 2 O with 0.1% formic acid) afforded the desired product 45 (20 mg, 99%).
  • Compound 41 B was prepared from compound 41 A according to the procedures described in Example 300D part A and B.
  • the two isomers of 41 C were separated using a chiral AD column. One gram of material was injected into the column and the two peaks were separated by using a solvent mixture of 80% hexanes/2-propanol. The second isomer was the desired compound 41 D (400 mg, 80%).
  • Compound 37A was prepared using procedures described in Example 375.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Immunology (AREA)
  • Diabetes (AREA)
  • Hematology (AREA)
  • Cardiology (AREA)
  • Rheumatology (AREA)
  • Pulmonology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Oncology (AREA)
  • Neurology (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Neurosurgery (AREA)
  • Communicable Diseases (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Biomedical Technology (AREA)
  • Ophthalmology & Optometry (AREA)
  • Pain & Pain Management (AREA)
  • Virology (AREA)
  • Obesity (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Dermatology (AREA)
  • Endocrinology (AREA)
  • Urology & Nephrology (AREA)
  • Addiction (AREA)
  • Psychiatry (AREA)
  • Nutrition Science (AREA)

Abstract

This invention relates to compounds of the Formula (I), or a pharmaceutically acceptable salt, solvate or isomer thereof, which can be useful for the treatment of diseases or conditions mediated by MMPs, ADAMs, TACE, aggrecanase, TNF-α or combinations thereof.

Description

HYDANTOIN COMPOUNDS FOR THE TREATMENT OF INFLAMMATORY DISORDERS
BACKGROUND OF THE INVENTION Field of the Invention
This invention relates generally to novel hydantoin derivatives that can inhibit matrix metalloproteinases (MIVIPs)1 a disintegrin and metalloproteases (ADAMs) and/or tumor necrosis factor alpha - converting enzyme (TACE) and in so doing prevent the release of tumor necrosis factor alpha (TNF-α), pharmaceutical compositions comprising such compounds, and methods of treatment using such compounds.
Description
Osteo- and rheumatoid arthritis (OA and RA, respectively) are destructive diseases of articular cartilage characterized by localized erosion of the cartilage surface. Findings have shown that articular cartilage from the femoral heads of patients with OA, for example, had a reduced incorporation of radiolabeled sulfate over controls, suggesting that there must be an enhanced rate of cartilage degradation in OA (Mankin et al. J. Bone Joint Surg. 52A (1970) 424-434). There are four classes of protein degradative enzymes in mammalian cells: serine, cysteine, aspartic and metalloproteases. The available evidence supports the belief that it is the metalloproteases that are responsible for the degradation of the extracellular matrix of articullar cartilage in OA and RA. Increased activities of collagenases and stromelysin have been found in OA cartilage and the activity correlates with severity of the lesion (Mankin et al. Arthritis Rheum. 21 , 1978, 761-766, Woessner et al. Arthritis Rheum. 26, 1983, 63-68 and Ibid. 27, 1984, 305-312). In addition, aggrecanase (a newly identified metalloprotease) has been identified that provides the specific cleavage product of proteoglycan, found in RA and OA patients (Lohmander L S. et al. Arthritis Rheum. 36, 1993, 1214-22).
Metalloproteases (MPs) have been implicated as the key enzymes in the destruction of mammalian cartilage and bone. It can be expected that the pathogenesis of such diseases can be modified in a beneficial manner by the administration of MP inhibitors (see Wahl et al. Ann. Rep. Med. Chem. 25, 175-184, AP, San Diego, 1990).
MMPs are a family of over 20 different enzymes that are involved in a variety of biological processes important in the uncontrolled breakdown of connective tissue, including proteoglycan and collagen, leading to resorption of the extracellular matrix. This is a feature of many pathological conditions, such as RA and OA, corneal, epidermal or gastric ulceration; tumor metastasis or invasion; periodontal disease and bone disease. Normally these catabolic enzymes are tightly regulated at the level of their synthesis as well as at their level of extracellular activity through the action of specific inhibitors, such as alpha-2-macroglobulins and TIMPs (tissue inhibitor of MPs)1 which form inactive complexes with the MMP's.
Tumor necrosis factor alpha (TNF-α) is a cell-associated cytokine that is processed from a 26 kDa precursor form to a 17 kd active form. See Black R.A. "Tumor necrosis factor-alpha converting enzyme" lnt J Biochem Cell Biol. 2002 Jan; 34(1 ):1 -5 and Moss ML, White JM, Lambert MH, Andrews RC11TACE and other ADAM proteases as targets for drug discovery" Drug Discov Today. 2001 Apr 1 ;6(8):417-426, each of which is incorporated by reference herein.
TNF-α has been shown to play a pivotal role in immune and inflammatory responses. Inappropriate or over-expression of TNF-α is a hallmark of a number of diseases, including RA, Crohn's disease, multiple sclerosis, psoriasis and sepsis. Inhibition of TNF-α production has been shown to be beneficial in many preclinical models of inflammatory disease, making inhibition of TNF-α production or signaling an appealing target for the development of novel anti-inflammatory drugs.
TNF-α is a primary mediator in humans and animals of inflammation, fever and acute phase responses, similar to those observed during acute infection and shock. Excess TNF-α has been shown to be lethal. Blocking the effects of TNF-α with specific antibodies can be beneficial in a variety of conditions, including autoimmune diseases such as RA (Feldman et al, Lancet, (1994) 344, 1105), non-insulin dependent diabetes mellitus (Lohmander L. S. et al., Arthritis Rheum. 36 (1993) 1214-22) and Crohn's disease (Macdonald T. et al., Clin. Exp. Immunol. 81 (1990) 301).
Compounds that inhibit the production of TNF-α are therefore of therapeutic importance for the treatment of inflammatory disorders. Recently it has been shown that metalloproteases, such as TACE, are capable of converting TNF-α from its inactive to active form (Gearing et al Nature, 1994, 370, 555). Since excessive TNF-α production has been noted in several disease conditions also characterized by MMP-mediated tissue degradation, compounds which inhibit both MMPs and TNF-α production may also have a particular advantage in diseases where both mechanisms are involved.
One approach to inhibiting the harmful effects of TNF-α is to inhibit the enzyme, TACE before it can process TNF-α to its soluble form. TACE is a member of the ADAM family of type I membrane proteins and mediates the ectodomain shedding of various membrane-anchored signaling and adhesion proteins. TACE has become increasingly important in the study of several diseases, including inflammatory disease, because of its role in cleaving TNF- α from its "stalk" sequence and thus releasing the soluble form of the TNF-α protein (Black R.A. lnt J Biochem Cell Biol. 2002 34,1-5).
There are numerous patents and publications which disclose hydroxamate, sulphonamide, hydantoin, carboxylate and/or lactam based MMP inhibitors.
US 6,677,355 and US 6,534,491 (B2), describe compounds that are hydroxamic acid derivatives and MMP inhibitors.
US 6,495,565 discloses lactam derivatives that are potential inhibitors of MMPs and/or TNF-α.
PCT Publications WO2002/074750, WO2002/096426, WO20040067996, WO2004012663, WO200274750 and WO2004024721 disclose hydantoin derivatives that are potential inhibitors of MMPs.
PCT Publications WO2004024698 and WO2004024715 disclose sulphonamide derivatives that are potential inhibitors of MMPs. PCT Publications WO2004056766, WO2003053940 and WO2003053941 also describe potential inhibitors of TACE and MMPs.
PCT Publication WO2006/019768 refers to hydantoin derivatives that are TACE inhibitors.
There is a need in the art for inhibitors of MMPs, ADAMs, TACE, and TNF-α, which can be useful as anti-inflammatory compounds and cartilage protecting therapeutics. The inhibition of TNF-α, TACE and or other MMPs can prevent the degradation of cartilage by these enzymes, thereby alleviating the pathological conditions of OA and RA as well as many other auto-immune diseases.
SUMMARY OF THE INVENTION
In its many embodiments, the present invention provides a novel class of compounds as inhibitors of TACE, the production of TNF-α, MMPs, ADAMs1 aggrecanase, or any combination thereof, methods of preparing such compounds, pharmaceutical compositions comprising one or more such compounds, methods of preparing pharmaceutical formulations comprising one or more such compounds, and methods of treatment, prevention, inhibition or amelioration of one or more diseases associated with TACE1 aggrecanaseTNF-α, MMPs, ADAMs or any combination thereof using such compounds or pharmaceutical compositions.
In one embodiment, the present application discloses a compound, or pharmaceutically acceptable salts or solvates of said compound, said compound having the general structure shown in Formula (I):
Figure imgf000005_0001
or a pharmaceutically acceptable salt, solvate, or ester thereof, wherein: ring A is selected from the group consisting of aryl and heteroaryl, each of which is substituted with -Y-R1 and -Z-R2 as shown;
X is selected from the group consisting of -S-.-O-, -S(O)2, -S(O)-,
-(C(R3)2)πr and -N(R3)-:
T is absent or present, and if present, T is selected from the group consisting of alkyl, aryl, and heteroaryl, wherein when each of said T aryl and heteroaryl contains two radicals on adjacent carbon atoms, said radicals may optionally be taken together with the carbon atoms to which they are attached to form a five- to eight-membered aryl or heteroaryl ring, wherein each of the aforementioned T aryl, and heteroaryl, optionally with said five- to eight- membered aryl or heteroaryl is independently unsubstituted or substituted with one to four R10 moieties which can be the same or different;
U is absent or present or absent, and if present, U is selected from the group consisting of -O-, -0-C(O)NH-, -OC(O)N(alkyl)-, -C(O)-, -C(O)O-, -C(O)NH-, -C(O)N(alkyl)-, -C(=N-OH)-alkyl-, and -C(=N-O-alkyl)-alkyl-;
V is absent or present, and if present V is selected from the group consisting of alkyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, and N- oxides of said heterocyclyl and heteroaryl, wherein when each of said V cycloalkyl, heterocyclyl, aryl, heteroaryl, and N-oxides of said heterocycyl and heteroaryl contains two radicals on adjacent carbon atoms, said radicals may optionally be taken together with the carbon atoms to which they are attached to form a five- to eight-membered cycloalkyl, aryl, heterocyclyl or heteroaryl ring; wherein each of said V alkyl, alkynyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl heterocyclyl, optionally with said five- to eight-membered cycloalkyl, aryl, heterocyclyl, or heteroaryl is independently unsubstituted or substituted with one to four R10 moieties which can be the same or different;
Y is selected from the group consisting of a covalent bond, -(C(R4)2)n-, -N(R4)-, -C(O)N(R4)-, -N(R4)C(O)-, -N(R4)C(O)N(R4)-, -S(O)2N(R4)-, -N(R4)- S(O)2, -O- -S-, -C(O)-, -S(O)-, and -S(O)2-;
Z is selected from the group consisting of a covalent bond, -(C(R4)2)n-,
-N(R4)-, -C(O)N(R4)-, -N(R4)C(O)-, -N(R4)C(O)N(R4)-, -S(O)2N(R4)-, -N(R4)- S(O)2-, -O-.-S-, -C(O)-, -S(O)-, and -S(O)2-; m is 1 to 3; n is 1 to 3;
R is selected from the group consisting of H, cyano, -C(O)OH, - C(O)O-alkyl, -C(O)NH2, -C(O)NH(alkyl), -C(O)N(alkyl)2, alkynyl, halogen, alkyl, cycloalkyl, haloalkyl, aryl, heteroaryl, and heterocyclyl, wherein when each of said cycloalkyl, heterocyclyl, aryl and heteroaryl contains two radicals on adjacent carbon atoms, said radicals may optionally be taken together with the carbon atoms to which they are attached to form a five- to eight- membered cycloalkyl, aryl, heterocyclyl or heteroaryl ring; wherein each of the R1 alkyl, alkynyl, aryl, heteroaryl, and heterocyclyl, optionally with the five or six-membered cycloalkyl, aryl, heterocyclyl or heteroaryl ring is unsubstituted or optionally independently substituted with one to four R20 moieties which can be the same or different; with the proviso that when Y is -N(R4)-, -S -or -O-, then R1 is not halogen or cyano;
2
R is selected from the group consisting of H, cyano, -C(O)OH, - C(O)O-alkyl, -C(O)NH2, -C(O)NH(alkyl), -C(O)N(alky02l alkynyl, halogen, alkyl, cycloalkyl, haloalkyl, aryl, heteroaryl, and heterocyclyl, wherein when each of said cycloalkyl, heterocyclyl, aryl and heteroaryl contains two radicals on adjacent carbon atoms, said radicals may optionally be taken together with the carbon atoms to which they are attached to form a five- to eight- membered cycloalkyl, aryl, heterocyclyl or heteroaryl ring; wherein each of the R2 alkyl, cycloalkyl, aryl, heteroaryl, and heterocyclyl, optionally with the five or six-membered cycloalkyl, aryl, heterocyclyl or heteroaryl ring is unsubstituted or optionally independently substituted with one to four R20 moieties which can be the same or different; with the proviso that when Y is - N(R4)-, -S -or -O-, then R2 is not halogen or cyano; each R3 is the same of different and is independently selected from the group consisting of H, alkyl, and aryl;
4 each R is the same or different and is independently selected from the group consisting of H, alkyl, cycloalkyl, haloalkyl, hydroxy, -alkylcycloalkyl, - alkyl-N(alkyl)2, heterocyclyl, aryl, and heteroaryl, wherein when each of said cycloalkyl, heterocyclyl, ayl, and heteroaryl contains two radicals on adjacent carbon atoms, said radicals may optionally be taken together with the carbon atoms to which they are attached to form a five- to eight-membered cycloalkyl, aryl, heterocyclyl or heteroaryl ring;
R is selected from the group consisting of hydrogen, cyano, nitro,
-C(R4J=N-OR4, -OR4, -SR4, -N(R4J2, -S(O)R4, -S(O)2R4, -N(R4)S(O)2R4, -N(R4)- C(O)-R4, -N(R4)-C(O)-N(R4)2 , -N(R4J-C(O)-OR4, -OC(O)N(R4)2, -C(O)N(R4)- S(O)2R4, -S(O)2N(R4J-C(O)-R4, -C(O)N(R4)C(O)R4, -C(O)N(R4JC(O)NR4, - S(O)2N(R4J2, -N(R4)-C(=NR4)-N(R4)2, -N(R4)-C(=N-CN)-N(R4)2, -haloalkoxy, - C(O)OR4, -C(O)R4, -C(O)N(R4)2r halogen, alkyl, haloalkyl, aryl, heteroaryl, heterocyclyl, and cycloalkyl, wherein each of the R10 alkyl, aryl, heteroaryl, heterocyclyl, and cycloalkyl is unsubstituted or optionally independently substituted with one to four R30 moieties which can be the same or different; or wherein two R10 moieties, when attached to the same or adjacent carbon atoms may optionally be taken together with the carbon atom(s) to which they are attached to form a cycloalkyl, cycloalkenyl, heterocyclyl, aryl, or heteroaryl ring;
20
R is selected from the group consisting of cyano, nitro,
-C(R4J=N-OR4, -OR4, -SR4, -N(R4J2, -S(O)R4, -S(O)2R4, -N(R4)S(O)2R4, -N(R4)- C(O)-R4, -N(R4)-C(O)-N(R4)2 , -N(R4J-C(O)-OR4, -OC(O)N(R4J2, -C(O)N(R4J- S(O)2R4, -S(O)2N(R-O-C(O)-R4, -C(O)N(R4)C(O)R4, -C(O)N(R4JC(O)NR4, - S(O)2N(R4J2, -N(R4)-C(=NR4)-N(R4)2, -N(R4)-C(=N-CN)-N(R4)2, -haloalkoxy, - C(O)OR4, -C(O)R4, -C(O)N(R4)2, halogen, alkyl, haloalkyl, aryl, heteroaryl, heterocyclyl, and cycloalkyl; wherein when each of said R20 aryl, heteroaryl, heterocyclyl and cycloalkyl contains two radicals on adjacent carbon atoms, said radicals may optionally be taken together with the carbon atoms to which they are attached to form a five- to eight-membered cycloalkyl, aryl, heterocyclyl or heteroaryl ring; wherein each of said R20 alkyl, aryl, heteroaryl, heterocyclyl, and cycloalkyl, optionally with said five- to eight-membered cycloalkyl, aryl, heterocyclyl or heteroaryl ring is unsubstituted or substituted with one to four moieties selected independently from the group consisting of alkyl, halo, haloalkyl, hydroxy, alkoxy, haloalkoxy, hydroxyalkyl, cyano, nitro, - NH2, -NH(alkyl), and -N(alkyl)2; or when two R20 moieties when attached to the same or adjacent carbon atoms may optionally be taken together with the carbon atom(s) to which they are attached to form a cycloalkyl, cycloalkenyl, heterocyclyl, aryl, or heteroaryl ring;
R is selected from the group consisting of cyano, nitro,
-C(R4)=N-OR4, -OR4, -SR4, -N(R4)2. -S(O)R4, -S(O)2R4, -N(R4)S(O)2R4, -N(R4)- C(O)-R4, -N(R4)-C(O)-N(R4)2 , -N(R4J-C(O)-OR4, -OC(O)N(R4J2, -C(O)N(R4)- S(O)2R4, -S(O)2N(R4)-C(O)-R4, -C(O)N(R4)C(O)R4, -C(O)N(R4)C(O)NR4, - S(O)2N(R4J2, -N(R4)-C(=NR4)-N(R4)2, -N(R4)-C(=N-CN)-N(R4)2, -haloalkoxy, - C(O)OR4, -C(O)R4, -C(O)N(R4J2, halogen, alkyl, haloalkyl, aryl, heteroaryl, heterocyclyl, and cycloalkyl; wherein when each of said R30 aryl, heteroaryl, heterocyclyl and cycloalkyl contains two radicals on adjacent carbon atoms, said radicals may optionally be taken together with the carbon atoms to which they are attached to form a five- to eight-membered cycloalkyl, aryl, heterocyclyl or heteroaryl ring; wherein each of said R30 alkyl, aryl, heteroaryl, heterocyclyl, and cycloalkyl, optionally with said five- to eight-membered cycloalkyl, aryl, heterocyclyl or heteroaryl ring is unsubstituted or substituted with one to four moieties selected independently from the group consisting of alkyl, halo, haloalkyl, hydroxy, alkoxy, haloalkoxy, cyano, nitro, -NH2, - NH(alkyl), and -N(alkyl)2; or when two R30 moieties when attached to the same or adjacent carbon atoms may optionally be taken together with the carbon atom(s) to which they are attached to form a cycloalkyl, cycloalkenyl, heterocyclyl, aryl, or heteroaryl ring; with the proviso that at least one of T, U, and V must be present; and further that at least one of conditions (1 ) — (5) below are satisfied:
(1 ) at least one of T and V is present, and V is other than alkynyl; wherein said T or V is substituted with at least one R10 moiety selected from the group consisting of cyano, -C(O)OR4, -C(O)R4, -C(O)N(R4)2, - C(O)N(R4)C(O)R4, -C(OJN(R4JC(O)NR4, -SR4, -S(O)2R4, -N(R4J-C(O)OR4, - OC(O)N(R4)2, -N(R4)C(O)N(R4)2, -N(R4)-C(O)-R4, -S(O)2N(R4J2, -S(O)2N(R4)- C(O)-R4,
-N(R4)-C(=NR4)-N(R4)2, -N(R4)-C(=N-CN)-N(R4)2, and -C(R4)=N-OR4, wherein each R4 independently is selected from the group consisting of H, alkyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl, wherein when each of said R4 cycloalkyl, heterocyclyl, aryl, and heteroaryl contains contains two radicals on adjacent carbon atoms, said radicals may optionally be taken together with the carbon atoms to which they are attached to form a five- to eight- membered aryl, heteocyclyl, heteroaryl or cycloalkyl ring; with the proviso that when R10 is
-S(O)2R4, V is other than piperidinyl, and when R10 is cyano, the compound of Formula (I) is other than
Figure imgf000010_0001
(2) U is present and is selected from the group consisting of -O- C(O)NH-, -OC(O)N(alkyl)-, -C(O)-, -C(O)O-, -C(O)NH-, -C(O)N(alkyl)-, - C(=N-OH)-alkyl-, and -C(=N-O-alkyl)-alkyl-;
(3) each of -Y-R1 and -Z-R2 is independently selected from the group consisting of cyano, -(C(R4)2)n-C(O)OH, -(C(R4)2)n-C(O)O-alkyl, -(C(R4)2)n- C(O)NH2, -(C(R4)2)n-C(O)NH(alkyl), and ~(C(R4)2)n-C(O)N(alkyl)2. wherein each R4 independently is H or alkyl; and n is 1-3;
(4) T is aryl or heteroaryl, each of which is optionally substituted with one to four independently selected R10 moieties, and V is alkynyl which is optionally substituted with one or two independently selected R10 moieties; and
(5) ring A is heteroaryl, and V is other than alkynyl.
In another embodiment, the present application discloses a compound, or pharmaceutically acceptable salts or solvates of said compound, said compound having the general structure shown in Formula (II):
Figure imgf000011_0001
or a pharmaceutically acceptable salt, solvate, ester or isomer thereof, wherein: the ring labeled A is selected from the group consisting of aryl and heteroaryl, each of which is substituted with -Y-R1 and -Z-R2 as shown;
3 3
X is selected from the group consisting of -S-,-O-, -C(R )2- or -N(R )-;
T is absent or present, and if present, T is selected from the group consisting of H (with U and V being absent), alkyl, alkenyl, alkynyl, aryl, heteroaryl, heterocyclyl, cycloalkyl, alkylaryl-, and arylalkyl-, said aryl, heteroaryl, heterocyclyl, cycloalkyl, alkylaryl-, and arylalkyl- being optionally fused with one or more moieties selected from the group consisting of aryl, heteroaryl, heterocyclyl, cycloalkyl, alkylaryl and arylalkyl, wherein each of any of the aforementioned alkyl, alkenyl, aryl, heteroaryl, heterocyclyl, cycloalkyl, alkylaryl and arylalkyl groups of T is unsubstituted or optionally independently substituted with one to four R10 moieties which can be the same or different, each R10 moiety being independently selected from the group of R10 moieties below;
U is absent or present, and if present U is selected from the group consisting of alkynyl, -C(O)-, -C(O)O-, and -C(O)NR4-;
V is absent or present, and if present V is selected from the group consisting of hydrogen, alkyl, aryl, heteroaryl, heterocyclyl, heterocyclylalkyl-, cycloalkyl, alkylaryl-, and arylalkyl-, said aryl, heteroaryl, heterocyclyl, heterocyclylalkyl-, cycloalkyl, alkylaryl- and arylalkyl- being optionally fused with one or more moieties selected from the group consisting of aryl, heteroaryl, heterocyclyl, cycloalkyl, alkylaryl and arylalkyl, wherein each of any of the aforementioned alkyl, aryl, heteroaryl, heterocyclyl and cycloalkyl is unsubstituted or optionally independently substituted with one to four R10 moieties which can be the same or different, each R10 moiety being independently selected from the group of R10 moieties below;
Y is selected from the group consisting of a covalent bond, -(C(R4)2)n-, -N(R4)-, -C(O)N(R4)-, -N(R4)C(O)-, -N(R4)C(O)N(R4)-, -S(O)2N(R4)-, -N(R4)- S(O)2, -O-.-S-, -C(O)-, -S(O)-, and -S(O)2-;
Z is selected from the group consisting of a covalent bond, -(C(R4)2)n-, -N(R4)-, -C(O)N(R4)-, -N(R4)C(O)-, -N(R4)C(O)N(R4)-, -
S(O)2N(R4)-, -N(R4J-S(O)2-, -O-.-S-, -C(O)-, -S(O)-, and -S(O)2-; n is 1 to 3;
R is selected from the group consisting of H1 -OR4, cyano, -C(O)OR4, -C(O)N(R4J2, halogen, alkyl, fluoroalkyl, aryl, heteroaryl, heterocyclyl, alkylaryl, alkylheteroaryl and arylalkyl, wherein each of the alkyl, fluoroalkyl, aryl, heteroaryl, heterocyclyl, alkylaryl, alkylheteroaryl and arylalkyl groups of
R is unsubstituted or optionally independently substituted with one to four R20 moieties which can be the same or different, each R20 moiety being independently selected from the group of R20 moieties below, with the proviso that when Y is present and Y is N, S or O, then R1 is not halogen or cyano;
R is selected from the group consisting of H1 -OR4, cyano, -C(O)OR4, -C(O)N(R4J2, halogen, alkyl, fluoroalkyl, aryl, heteroaryl, heterocyclyl, alkylaryl, alkylheteroaryl and arylalkyl, wherein each of the alkyl, fluoroalkyl, aryl,
2 heteroaryl, heterocyclyl, alkylaryl, alkylheteroaryl and arylalkyl groups of R is unsubstituted or optionally independently substituted with one to four R20 moieties which can be the same or different, each R20 moiety being independently selected from the group of R20 moieties below, with the proviso that when Z is present and Z is N, S or O, then R2 is not halogen; each R3 is the same of different and is independently selected from the group consisting of H, alkyl, and aryl;
4 each R is the same or different and is independently selected from the group consisting of H, alkyl, heterocyclyl, aryl, and heteroaryl; R is selected from the group consisting of cyano, -OR4, -SR4, -N(R )2,
-S(O)R4-, -S(O)2R4-, -N(R4JS(O)2R4, -S(O)2N(R4J2, -O(fluoroalkyl), -C(O)OR4, -C(O)N(R4)2, halogen, alkyl, fluoroalkyl, aryl, heteroaryl, heterocyclyl, cycloalkyl, alkylaryl and arylalkyl, wherein each of the alkyl, fluoroalkyl, aryl, heteroaryl, heterocyclyl, cycloalkyl, alkylaryl and arylalkyl groups of R is unsubstituted or optionally independently substituted with one to four R30 moieties which can be the same or different, each R30 moiety being independently selected from the group of R30 moieties below;
R is selected from the group consisting of halogen, alkyl, fluoroalkyl, -N(R4)2, and -C(O)N(R4)2 ; and
30
R is selected from the group consisting of halogen, alkyl, fluoroalkyl, -N(R4J2, and -C(O)N(R4)2.
The compounds of Formula I can be useful as inhibitors of TACE and may be useful in the treatment and prevention of diseases associated with TACE, TNF-α, MMPs, ADAMs or any combination thereof.
DETAILED DESCRIPTION OF THE INVENTION
In its several embodiments, the present invention provides a novel class of inhibitors of TACE, aggrecanase, the production of TNF-α, MMPs, ADAMs or any combination thereof, pharmaceutical compositions containing one or more of the compounds, methods of preparing pharmaceutical formulations comprising one or more such compounds, and methods of treatment, prevention or amelioration of one or more of the symptoms of inflammation.
In one embodiment, the present invention provides compounds which are represented by structural Formula (I) or (II) above or a pharmaceutically acceptable salt, solvate, ester or isomer thereof, wherein the various moieties are as described above.
In another embodiment, the isomer referred to the in the preceding paragraph is a stereoisomer. In another embodiment, in formula (I), X is selected from the group consisting of -(C(R3)2)m- and -N(R3)-.
In another embodiment, in formula (I)1 X is -(C(R3)2)m, wherein m is 1 or 2.
In another embodiment, in formula (I), X is -(C(R3)2)m. wherein m is 1.
In another embodiment, in formula (I), R3 is H.
In another embodiment, in formula (I), at least one of T and V is present, and V is other than alkynyl; wherein said T or V is substituted with at least one R10 moiety selected from the group consisting of cyano, -C(O)OR4, - C(O)R4, -C(O)N(R4)2, -C(O)N(R4)C(O)R4, -C(O)N(R4)C(O)NR4, -SR4, - S(O)2R4, -N(R4)-C(O)OR4, -OC(O)N(R4)2, -N(R4)C(O)N(R4)2, -N(R4J-C(O)-R4, - S(O)2N(R4J2, -S(O)2N(R4J-C(O)-R4, -N(R4)-C(=NR4)-N(R4)2, -N(R4)-C(=N-CNJ- N(R4J2, and -C(R4J=N-OR4, wherein each R4 independently is selected from the group consisting of H, alkyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl, wherein when each of said R4 cycloalkyl, heterocyclyl, aryl, and heteroaryl contains contains two radicals on adjacent carbon atoms, said radicals may optionally be taken together with the carbon atoms to which they are attached to form a five- to eight-membered aryl, heteocyclyl, heteroaryl or cycloalkyl ring; with the proviso that when R10 is
-S(O)2R4, V is other than piperidinyl, and when R10 is cyano, the compound of Formula (I) is other than
Figure imgf000014_0001
In another embodiment in formula (I), at least one of T and V is present, and V is other than alkynyl; wherein said T or V is substituted with at least one R10 moiety selected from the group consisting of cyano, -C(O)OR4, - C(O)R4, -C(O)N(R4)2, -C(O)N(R4JC(O)R4, -C(O)N(R4)C(O)NR4, -SR4, - S(O)2R4, -N(R4)-C(O)OR4, -OC(O)N(R4)2, -N(R4)C(O)N(R4)2, -N(R4J-C(O)-R4, - S(O)2N(R4J2, -S(O)2N(R4J-C(O)-R4, -N(R4)-C(=NR4)-N(R4)2, -N(R4)-C(=N-CNJ- N(R4)2, and -C(R4J=N-OR4, wherein each R4 independently is selected from the group consisting of H, alkyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl, wherein when each of said R4 cycloalkyl, heterocyclyl, aryl, and heteroaryl contains contains two radicals on adjacent carbon atoms, said radicals may optionally be taken together with the carbon atoms to which they are attached to form a five- to eight-membered aryl, heteocyclyl, heteroaryl or cycloalkyl ring; with the proviso that when R10 is
-S(O)2R4, V is other than piperidinyl, and when R10 is cyano, the compound of Formula (I) is other than
Figure imgf000015_0001
wherein said T or V is substituted with at least one R10 moiety selected from the group consisting of cyano, - C(O)OR4, -C(O)R4, -C(O)N(R4)2, and -C(R4)=N-OR4.
In another embodiment in formula (I), at least one of T and V is present, and V is other than alkynyl; wherein said T or V is substituted with at least one R10 moiety selected from the group consisting of cyano, -C(O)OR4, - C(O)R4, -C(O)N(R4)2, -C(O)N(R4)C(O)R4, -C(O)N(R4)C(O)NR4, -SR4, - S(O)2R4, -N(R4)-C(O)OR4, -OC(O)N(R4)2, -N(R4)C(O)N(R4)2, -N(R4)-C(O)-R4, - S(O)2N(R4)2, -S(O)2N(R4)-C(O)-R4, -N(R4)-C(=NR4)-N(R4)2, -N(R4)-C(=N-CN)- N(R4)2, and -C(R4J=N-OR4, wherein each R4 independently is selected from the group consisting of H, alkyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl, wherein when each of said R4 cycloalkyl, heterocyclyl, aryl, and heteroaryl contains contains two radicals on adjacent carbon atoms, said radicals may optionally be taken together with the carbon atoms to which they are attached to form a five- to eight-membered aryl, heteocyclyl, heteroaryl or cycloalkyl ring; with the proviso that when R10 is -S(O)2R4, V is other than piperidinyl, and when R10 is cyano, the compound of Formula (I) is other than
Figure imgf000016_0001
wherein said T or V is substituted with at least one R10 moiety that is cyano.
In another embodiment, in formula (I), at least one of T and V is present, and V is other than alkynyl; wherein said T or V is substituted with at least one R10 moiety selected from the group consisting of cyano, -C(O)OR4, - C(O)R4, -C(O)N(R4J2, -C(O)N(R4JC(O)R4, -C(O)N(R4JC(O)NR4, -SR4, - S(O)2R4, -N(R4J-C(O)OR4, -OC(O)N(R4J2, -N(R4JC(O)N(R4J2, -N(R4J-C(O)-R4, - S(O)2N(R4)2, -S(O)2N(R4J-C(O)-R4, -N(R4)-C(=NR4)-N(R4)2, -N(R4)-C(=N-CN)- N(R4)2, and -C(R4)=N-OR4, wherein each R4 independently is selected from the group consisting of H, alkyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl, wherein when each of said R4 cycloalkyl, heterocyclyl, aryl, and heteroaryl contains contains two radicals on adjacent carbon atoms, said radicals may optionally be taken together with the carbon atoms to which they are attached to form a five- to eight-membered aryl, heteocyclyl, heteroaryl or cycloalkyl ring; with the proviso that when R10 is -S(O)2R4, V is other than piperidinyl, and when R10 is cyano, the compound of Formula (I) is other than
Figure imgf000016_0002
wherein said T or V is substituted with at least one R10 moiety that is -SR4.
In another embodiment, in formula (I), at least one of T and V is present, and V is other than alkynyl; wherein said T or V is substituted with at least one R10 moiety selected from the group consisting of cyano, -C(O)OR4, - C(O)R4, -C(O)N(R4J2, -C(O)N(R4JC(O)R4, -C(O)N(R4)C(O)NR4, -SR4, - S(O)2R4, -N(R4J-C(O)OR4, -OC(O)N(R4)2, -N(R4)C(O)N(R4)2, -N(R4)-C(O)-R4, - S(O)2N(R4)2, -S(O)2N(R4J-C(O)-R4, -N(R4)-C(=NR4)-N(R4)2, -N(R4)-C(=N-CN)- N(R4)2, and -C(R4)=N-OR4, wherein each R4 independently is selected from the group consisting of H, alkyl, cycloalkyf, heterocyclyl, aryl, and heteroaryl, wherein when each of said R4 cycloalkyl, heterocyclyl, aryl, and heteroaryl contains contains two radicals on adjacent carbon atoms, said radicals may optionally be taken together with the carbon atoms to which they are attached to form a five- to eight-membered aryl, heteocyclyl, heteroaryl or cycloalkyl ring; with the proviso that when R10 is -S(O)2R4, V is other than piperidinyl, and when R10 is cyano, the compound of Formula (I) is other than
Figure imgf000017_0001
wherein said T or V is substituted with at least one R10 moiety that is -S(O)2R4.
In another embodiment, in formula (I), at least one of T and V is present, and V is other than alkynyl; wherein said T or V is substituted with at least one R10 moiety selected from the group consisting of cyano, -C(O)OR4, - C(O)R4, -C(O)N(R4J2, -C(O)N(R4)C(O)R4, -C(O)N(R4)C(O)NR4, -SR4, - S(O)2R4, -N(R4)-C(O)OR4, -OC(O)N(R4J2, -N(R4)C(O)N(R4)2, -N(R4)- C(O)-R4, - S(O)2N(R4)2, -S(O)2N(R4J-C(O)-R4, -N(R4)-C(=NR4)-N(R4)2, -N(R4)-C(=N-CN)- N(R4J2, and -C(R4J=N-OR4, wherein each R4 independently is selected from the group consisting of H, alkyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl, wherein when each of said R4 cycloalkyl, heterocyclyl, aryl, and heteroaryl contains contains two radicals on adjacent carbon atoms, said radicals may optionally be taken together with the carbon atoms to which they are attached to form a five- to eight-membered aryl, heteocyclyl, heteroaryl or cycloalkyl ring; with the proviso that when R10 is -S(O)2R4, V is other than piperidinyl, and when R10 is cyano, the compound of Formula (I) is other than
Figure imgf000018_0001
wherein said T or V is substituted with at least one R10 moiety that -S(O)2N(R4)2.
In another embodiment, in formula (I), at least one of T and V is present, and V is other than alkynyl; wherein said T or V is substituted with at least one R10 moiety selected from the group consisting of cyano, -C(O)OR4, - C(O)R4, -C(O)N(R4)2, -C(O)N(R4)C(O)R4, -C(O)N(R4)C(O)NR4, -SR4, - S(O)2R4, -N(R4)-C(O)OR4, -OC(O)N(R4)2, -N(R4)C(O)N(R4)2, -N(R4)-C(O)-R4, - S(O)2N(R4)2, -S(O)2N(R4J-C(O)-R4, -N(R4)-C(=NR4)-N(R4)2, -N(R4)-C(=N-CN)- N(R4)2, and -C(R4)=N-OR4, wherein each R4 independently is selected from the group consisting of H, alkyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl, wherein when each of said R4 cycloalkyl, heterocyclyl, aryl, and heteroaryl contains contains two radicals on adjacent carbon atoms, said radicals may optionally be taken together with the carbon atoms to which they are attached to form a five- to eight-membered aryl, heteocyclyl, heteroaryl or cycloalkyl ring; with the proviso that when R10 is -S(O)2R4, V is other than piperidinyl, and when R10 is cyano, the compound of Formula (I) is other than
Figure imgf000018_0002
wherein ring A is selected from the group consisting of phenyl, thiophenyl, pyridyl, pyrimidyl, and
Figure imgf000018_0003
each of which is substituted with -Y-R1 and -Z-R2 as shown. In another embodiment, in Formula (I), at least one of T and V is present, and V is other than alkynyl; wherein said T or V is substituted with at least one R10 moiety selected from the group consisting of cyano, -C(O)OR4, - C(O)R4, -C(O)N(R4)2, -C(O)N(R4)C(O)R4, -C(O)N(R4)C(O)NR4, -SR4, - S(O)2R4, -N(R4)-C(O)OR4, -OC(O)N(R4)2. -N(R4)C(O)N(R4)2, -N(R4)-C(O)-R4, - S(O)2N(R4J2, -S(O)2N(R4J-C(O)-R4, -N(R4)-C(=NR4)-N(R4)2, -N(R4)-C(=N-CN)- N(R4)2, and -C(R4)=N-OR4, wherein each R4 independently is selected from the group consisting of H, alkyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl, wherein when each of said R4 cycloalkyl, heterocyclyl, aryl, and heteroaryl contains contains two radicals on adjacent carbon atoms, said radicals may optionally be taken together with the carbon atoms to which they are attached to form a five- to eight-membered aryl, heteocyclyl, heteroaryl or cycloalkyl ring; with the proviso that when R10 is -S(O)2R4, V is other than piperidinyl, and when R10 is cyano, the compound of Formula (I) is other than
Figure imgf000019_0001
wherein ring A is phenyl.
In another embodiment, in Formula (I), at least one of T and V is present, and V is other than alkynyl; wherein said T or V is substituted with at least one R10 moiety selected from the group consisting of cyano, -C(O)OR4, - C(O)R4, -C(O)N(R4J2, -C(O)N(R4JC(O)R4, -C(O)N(R4)C(O)NR4, -SR4, - S(O)2R4, -N(R4J-C(O)OR4, -OC(O)N(R4)2, -N(R4)C(O)N(R4)2, -N(R4)-C(O)-R4, - S(O)2N(R4)2, -S(O)2N(R4)-C(O)-R4, -N(R4)-C(=NR4)-N(R4)2, -N(R4)-C(=N-CN)- N(R4)2, and -C(R4)=N-OR4, wherein each R4 independently is selected from the group consisting of H, alkyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl, wherein when each of said R4 cycloalkyl, heterocyclyl, aryl, and heteroaryl contains contains two radicals on adjacent carbon atoms, said radicals may optionally be taken together with the carbon atoms to which they are attached to form a five- to eight-membered aryl, heteocyclyl, heteroaryl or cycloalkyl ring; with the proviso that when R10 is -S(O)2R4, V is other than piperidinyl, and when R10 is cyano, the compound of Formula (I) is other than
Figure imgf000020_0001
wherein T is selected from the group consisting of alkyl, aryl, heteroaryl, wherein when each of said T aryl and heteroaryl contains two radicals on adjacent carbon atoms, said radicals may optionally be taken together with the carbon atoms to which they are attached to form a five- to eight-mem bered aryl or heteroaryl ring; wherein each of the aforementioned T aryl, and heteroaryl, optionally with said five- to eight- membered aryl or heteroaryl is independently unsubstituted or substituted with one to four R10 moieties which can be the same or different.
In another embodiment, in Formula (I)1 at least one of T and V is present, and V is other than alkynyl; wherein said T or V is substituted with at least one R10 moiety selected from the group consisting of cyano, -C(O)OR4, - C(O)R4, -C(O)N(R4J2, -C(O)N(R4)C(O)R4, -C(O)N(R4)C(O)NR4, -SR4, - S(O)2R4, -N(R4J-C(O)OR4, -OC(O)N(R4)2, -N(R4)C(O)N(R4)2, -N(R4)-C(O)-R4, - S(O)2N(R4J2, -S(O)2N(R4J-C(O)-R4, -N(R4)-C(=NR4)-N(R4)2, -N(R4)-C(=N-CN)- N(R4)2, and -C(R4)=N-OR4, wherein each R4 independently is selected from the group consisting of H, alkyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl, wherein when each of said R4 cycloalkyl, heterocyclyl, aryl, and heteroaryl contains contains two radicals on adjacent carbon atoms, said radicals may optionally be taken together with the carbon atoms to which they are attached to form a five- to eight-membered aryl, heteocyclyl, heteroaryl or cycloalkyl ring; with the proviso that when R10 is -S(O)2R4, V is other than piperidinyl, and when R10 is cyano, the compound of Formula (I) is other than
Figure imgf000020_0002
wherein T is selected from the group consisting Of-CH2-, phenyl,
Figure imgf000021_0001
optionally substituted with one to four R10 moieties such that the number of R10 moieties per each T does not exceed four.
In another embodiment, in Formula (I), at least one of T and V is present, and V is other than alkynyl; wherein said T or V is substituted with at least one R10 moiety selected from the group consisting of cyano, -C(O)OR4, - C(O)R4, -C(O)N(R4)2> -C(O)N(R4)C(O)R4, -C(O)N(R4JC(O)NR4, -SR4, - S(O)2R4, -N(R4)-C(O)OR4, -OC(O)N(R4)2, -N(R4)C(O)N(R4)2, -N(R4)-C(O)-R4, - S(O)2N(R4J2, -S(O)2N(R4J-C(O)-R4, -N(R4)-C(=NR4)-N(R4)2, -N(R4)-C(=N-CN)- N(R4)2, and -C(R4)=N-OR4, wherein each R4 independently is selected from the group consisting of H, alkyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl, wherein when each of said R4 cycloalkyl, heterocyclyl, aryl, and heteroaryl contains contains two radicals on adjacent carbon atoms, said radicals may optionally be taken together with the carbon atoms to which they are attached to form a five- to eight-membered aryl, heteocyclyl, heteroaryl or cycloalkyl ring; with the proviso that when R10 is -S(O)2R4, V is other than piperidinyl, and when R10 is cyano, the compound of Formula (I) is other than
Figure imgf000021_0002
wherein U is absent or present, and if present is selected from the group consisting of -C(O)-, and -C(O)O-.
In another embodiment, in Formula (I), at least one of T and V is present, and V is other than alkynyl; wherein said T or V is substituted with at least one R10 moiety selected from the group consisting of cyano, -C(O)OR4, - C(O)R4, -C(O)N(R4)2, -C(O)N(R4)C(O)R4, -C(O)N(R4)C(O)NR4, -SR4, - S(O)2R4, -N(R4J-C(O)OR4, -OC(O)N(R4)2, -N(R4)C(O)N(R4)2l -N(R4J-C(O)-R4, - S(O)2N(R4J2, -S(O)2N(R4J-C(O)-R4, -N(R4)-C(=NR4)-N(R4)2, -N(R4)-C(=N-CNJ- N(R4)2, and -C(R4J=N-OR4, wherein each R4 independently is selected from the group consisting of H, alkyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl, wherein when each of said R4 cycloalkyl, heterocyclyl, aryl, and heteroaryl contains contains two radicals on adjacent carbon atoms, said radicals may optionally be taken together with the carbon atoms to which they are attached to form a five- to eight-membered aryl, heteocyclyl, heteroaryl or cycloalkyl ring; with the proviso that when R10 is -S(O)2R4, V is other than piperidinyl, and when R10 is cyano, the compound of Formula (I) is other than
Figure imgf000022_0001
wherein V is absent or present, and if present is selected from the group consisting of aryl, and heteroaryl, wherein when each of said V aryl and heteroaryl contains two radicals on adjacent carbon atoms, said radicals may optionally be taken together with the carbon atoms to which they are attached to form a five- to eight-membered aryl or heteroaryl ring; wherein each of the aforementioned V aryl, and heteroaryl, optionally with said five- to eight- membered aryl or heteroaryl is independently unsubstituted or substituted with one to four R10 moieties which can be the same or different.
In another embodiment, in Formula (I), at least one of T and V is present, and V is other than alkynyl; wherein said T or V is substituted with at least one R10 moiety selected from the group consisting of cyano, -C(O)OR4, - C(O)R4, -C(O)N(R4J2, -C(O)N(R4JC(O)R4, -C(O)N(R4)C(O)NR4, -SR4, - S(O)2R4, -N(R4)-C(O)OR4, -OC(O)N(R4J2, -N(R4JC(O)N(R4J2, -N(R4J-C(O)-R4, - S(OJ2N(R4J2, -S(O)2N(R4J-C(O)-R4, -N(R4)-C(=NR4)-N(R4)2, -N(R4)-C(=N-CNJ- N(R4)2, and -C(R4J=N-OR4, wherein each R4 independently is selected from the group consisting of H, alkyl, cycloalkyl, heterocycJyl, aryl, and heteroaryl, wherein when each of said R4 cycloalkyl, heterocyclyl, aryl, and heteroaryl contains contains two radicals on adjacent carbon atoms, said radicals may optionally be taken together with the carbon atoms to which they are attached to form a five- to eight-membered aryl, heteocyclyl, heteroaryl or cycloalkyl ring; with the proviso that when R10 is -S(O)2R4, V is other than piperidinyl, and when R10 is cyano, the compound of Formula (I) is other than
Figure imgf000023_0001
wherein V is selected from the group consisting of phenyl, pyridyl, pyrazinyl, indazolyl,
Figure imgf000023_0002
and
Figure imgf000023_0003
each of which is optionally substituted with one to four R10 moieties which can be the same or different.
In another embodiment, in Formula (I), at least one of T and V is present, and V is other than alkynyl; wherein said T or V is substituted with at least one R10 moiety selected from the group consisting of cyano, -C(O)OR4, - C(O)R4, -C(O)N(R4J2, -C(O)N(R4)C(O)R4, -C(O)N(R4JC(O)NR4, -SR4, - S(O)2R4, -N(R4J-C(O)OR4, -OC(O)N(R4)2, -N(R4)C(O)N(R4)2, -N(R4)-C(O)-R4, - S(O)2N(R4J2, -S(O)2N(R4J-C(O)-R4, -N(R4)-C(=NR4)-N(R4)2, -N(R4)-C(=N-CN)- N(R4)2j and -C(R4)=N-OR4, wherein each R4 independently is selected from the group consisting of H, alkyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl, wherein when each of said R4 cycloalkyl, heterocyclyl, aryl, and heteroaryl contains contains two radicals on adjacent carbon atoms, said radicals may optionally be taken together with the carbon atoms to which they are attached to form a five- to eight-membered aryl, heteocyclyl, heteroaryl or cycloalkyl ring; with the proviso that when R10 is -S(O)2R4, V is other than piperidinyl, and when R10 is cyano, the compound of Formula (I) is other than
Figure imgf000024_0001
wherein each of Y and Z is independently selected from the group consisting of a covalent bond and -O-
In another embodiment, in Formula (I), at least one of T and V is present, and V is other than alkynyl; wherein said T or V is substituted with at least one R10 moiety selected from the group consisting of cyano, -C(O)OR4, - C(O)R4, -C(O)N(R4)2, -C(O)N(R4)C(O)R4, -C(O)N(R4JC(O)NR4, -SR4, - S(O)2R4, -N(R4)-C(O)OR4, -OC(O)N(R4)2> -N(R4)C(O)N(R4)2, -N(R4)-C(O)-R4, - S(O)2N(R4)2, -S(O)2N(R4J-C(O)-R4, -N(R4)-C(=NR4)-N(R4)2, -N(R4)-C(=N-CN)- N(R4)2, and -C(R4J=N-OR4, wherein each R4 independently is selected from the group consisting of H, alkyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl, wherein when each of said R4 cycloalkyl, heterocyclyl, aryl, and heteroaryl contains contains two radicals on adjacent carbon atoms, said radicals may optionally be taken together with the carbon atoms to which they are attached to form a five- to eight-membered aryl, heteocyclyl, heteroaryl or cycloalkyl ring; with the proviso that when R10 is -S(O)2R4, V is other than piperidinyl, and when R10 is cyano, the compound of Formula (I) is other than
Figure imgf000024_0002
wherein Y is -O- and Z is a covalent bond.
In another embodiment, in Formula (I), at least one of T and V is present, and V is other than alkynyl; wherein said T or V is substituted with at least one R10 moiety selected from the group consisting of cyano, -C(O)OR4, - C(O)R4, -C(O)N(R4J2, -C(O)N(R4)C(O)R4, -C(O)N(R4)C(O)NR4, -SR4, - S(O)2R4, -N(R4J-C(O)OR4, -OC(O)N(R4J2, -N(R4)C(O)N(R4)2, -N(R4)-C(O)-R4, - S(O)2N(R4J2, -S(O)2N(R4)-C(O)-R4. -N(R4)-C(=NR4)-N(R4)2, -N(R4)-C(=N-CN)- N(R4J2, and -C(R4)=N-OR4, wherein each R4 independently is selected from the group consisting of H, alkyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl, wherein when each of said R4 cycloalkyl, heterocyclyl, aryl, and heteroaryl contains contains two radicals on adjacent carbon atoms, said radicals may optionally be taken together with the carbon atoms to which they are attached to form a five- to eight-membered aryl, heteocyclyl, heteroaryl or cycloalkyl ring; with the proviso that when R10 is -S(O)2R4, V is other than piperidinyl, and when R10 is cyano, the compound of Formula (I) is other than
Figure imgf000025_0001
wherein each of R1 and R2 is independently selected form the group consisting of H and alkyl.
In another embodiment, in Formula (I), at least one of T and V is present, and V is other than alkynyl; wherein said T or V is substituted with at least one R10 moiety selected from the group consisting of cyano, -C(O)OR4, - C(O)R4, -C(O)N(R4)2, -C(O)N(R4)C(O)R4, -C(O)N(R4)C(O)NR4, -SR4, - S(O)2R4, -N(R4)-C(O)OR4, -OC(O)N(R4J2, -N(R4)C(O)N(R4)2, -N(R4)-C(O)-R4, - S(O)2N(R4J2, -S(O)2N(R-O-C(O)-R4, -N(R4)-C(=NR4)-N(R4)2> -N(R4)-C(=N-CN)- N(R4J2, and -C(R4)=N-OR4, wherein each R4 independently is selected from the group consisting of H, alkyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl, wherein when each of said R4 cycloalkyl, heterocyclyl, aryl, and heteroaryl contains contains two radicals on adjacent carbon atoms, said radicals may optionally be taken together with the carbon atoms to which they are attached to form a five- to eight-membered aryl, heteocyclyl, heteroaryl or cycloalkyl ring; with the proviso that when R10 is -S(O)2R4, V is other than piperidinyl, and when R10 is cyano, the compound of Formula (I) is other than
Figure imgf000026_0001
wherein R1 is alkyl and R2 is H. In another embodiment, in Formula (I), at least one of T and V is present, and V is other than alkynyl; wherein said T or V is substituted with at least one R10 moiety selected from the group consisting of cyano, -C(O)OR4, - C(O)R4, -C(O)N(R4J2, -C(O)N(R4JC(O)R4, -C(O)N(R4)C(O)NR4, -SR4, - S(O)2R4, -N(R4)-C(O)OR4, -OC(O)N(R4)2, -N(R4)C(O)N(R4)2> -N(R4)-C(O)-R4, - S(O)2N(R4J2, -S(O)2N(R4KKO)-R4, -N(R4)-C(=NR4)-N(R4)2, -N(R4)-C(=N-CNJ- N(R4)2, and -C(R4)=N-OR4, wherein each R4 independently is selected from the group consisting of H, alkyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl, wherein when each of said R4 cycloalkyl, heterocyclyl, aryl, and heteroaryl contains contains two radicals on adjacent carbon atoms, said radicals may optionally be taken together with the carbon atoms to which they are attached to form a five- to eight-membered aryl, heteocyclyl, heteroaryl or cycloalkyl ring; with the proviso that when R10 is -S(O)2R4, V is other than piperidinyl, and when R10 is cyano, the compound of Formula (I) is other than
Figure imgf000026_0002
wherein R1 is methyl. In another embodiment, in Formula (I), at least one of T and V is present, and V is other than alkynyl; wherein said T or V is substituted with at least one R10 moiety selected from the group consisting of cyano, -C(O)OR4, - C(O)R4, -C(O)N(R4)2l -C(O)N(R4JC(O)R4, -C(O)N(R4)C(O)NR4, -SR4, - S(O)2R4, -N(R4)-C(O)OR4, -OC(O)N(R4J2, -N(R4)C(O)N(R4)2, -N(R4)-C(O)-R4, - S(O)2N(R4J2, -S(O)2N(R4J-C(O)-R4, -N(R4)-C(=NR4)-N(R4)2( -N(R4)-C(=N-CN)- N(R4)2, and -C(R4J=N-OR4, wherein each R4 independently is selected from the group consisting of H, alkyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl, wherein when each of said R4 cycloalkyl, heterocyclyl, aryl, and heteroaryl contains contains two radicals on adjacent carbon atoms, said radicals may optionally be taken together with the carbon atoms to which they are attached to form a five- to eight-membered aryl, heteocyclyl, heteroaryl or cycloalkyl ring; with the proviso that when R10 is -S(O)2R4, V is other than piperidinyl, and when R10 is cyano, the compound of Formula (I) is other than
Figure imgf000027_0001
wherein the compound of Formula (I) is selected from the group consisting of:
Compound
Structures ID
Figure imgf000027_0002
23
24
Figure imgf000027_0003
Figure imgf000028_0001
Figure imgf000029_0001
Figure imgf000030_0001
Figure imgf000031_0001
Figure imgf000032_0001
Figure imgf000033_0001
Figure imgf000034_0001
Figure imgf000035_0001
Figure imgf000036_0001
Figure imgf000037_0001
Figure imgf000038_0002
or a pharmaceutically acceptable salt, solvate or ester thereof.
In another embodiment, in Formula (I), U and V are present; and U is selected from the group consisting Of -O-C(O)NH-, -OC(O)N(alkyl)-, -C(O)-, C(O)O-, -C(O)NH-, -C(O)N(alkyl)-, -C(=N-OH)-alkyl-, and -C(=N-O-alkyl)- alkyk
In another embodiment, in Formula (I), U and V are present; and U is selected from the group consisting of -0-C(O)NH-, -OC(O)N(alkyl)-, -C(O)-, C(O)O-, -C(O)NH-, -C(O)N(alkyl)-, -C(=N-OH)-alkyl-, and -C(=N-O-alkyl)- alkyl-; wherein ring A is selected from the group consisting of phenyl, thiophenyl, pyridyl, pyrimidyl, and
Figure imgf000038_0001
each of which is substituted with -Y-R1 and -Z-R2 as shown. In another embodiment, in Formula (I), U and V are present; and U is selected from the group consisting of -0-C(O)NH-, -OC(O)N(alkyl)-, -C(O)-, - C(O)O-, -C(O)NH-, -C(O)N(alkyl)-, -C(=N-OH)-alkyl-, and -C(=N-O-alkyl)- alkyl-; wherein ring A is phenyl.
In another embodiment, in Formula (I), U and V are present; and U is selected from the group consisting Of -O-C(O)NH-, -OC(O)N(alkyl)-, -C(O)-, - C(O)O-, -C(O)NH-, -C(O)N(alkyl)-, -C(=N-OH)-alkyl-, and -C(=N-O-alkyl)- alkyl-; and wherein T is absent or present, and when present, is selected from the group consisting of alkyl, and aryl, each of which is uπsubstitued or substituted with one to four R10 moieties which can be the same or different.
In another embodiment, in Formula (I), U and V are present; and U is selected from the group consisting of -0-C(O)NH-, -OC(O)N(alkyl)-, -C(O)-, - C(O)O-, -C(O)NH-, -C(O)N(alkyl)-, -C(=N-OH)-alkyl-, and -C(=N-O-alkyl)- alkyl-; and wherein wherein T is absent or present, and when present is selected from the group consisting Of-CH2-, and phenyl.
In another embodiment, in Formula (I), U and V are present; and U is selected from the group consisting Of -O-C(O)NH-, -OC(O)N(alkyl)-, -C(O)-, - C(O)O-, -C(O)NH-, -C(O)N(alkyl)-, -C(=N-OH)-alkyl-, and -C(=N-O-alkyl)- alkyl-; and wherein V is selected from the group consisting of alkyl, heterocyclyl, and cycloalkyl, wherein when each of said V heterocyclyl or cycloalkyl contains two radicals on adjacent carbon atoms, said radicals may optionally be taken together with the carbon atoms to which they are attached to form a five- to eight-membered cycloalkyl, heterocyclyl, aryl or heteroaryl ring; wherein each of aforementioned V alkyl, heterocyclyl, and cycloalkyl, optionally with said five- to eight-membered cycloalky!, heterocyclyl, aryl or heteroaryl ring is is independently unsubstituted or substituted with one to four R10 moieties which can be the same or different.
In another embodiment, in Formula (I), U and V are present; and U is selected from the group consisting of -0-C(O)NH-, -OC(O)N(alkyl)-, -C(O)-, - C(O)O-, -C(O)NH-, -C(O)N(alkyl)-, -C(=N-OH)-alkyl-, and -C(=N-O-alkyl)- alkyl-; and wherein V is selected from the group consisting of methyl, ethyl, isopropyl, morpholinyl, cyclohexyl, piperidinyl optionally substituted with cyano or phenyl, -CH2- substituted with tetrahydrofuranyl and
Figure imgf000040_0001
-CH(CHa)- substituted with phenyl, piperaziπyl substituted wth methyl, pyrrolidinyl substituted wth -CH2-phenyl,
substituted with cyclopropyl, and
Figure imgf000040_0002
In another embodiment, in Formula (I), U and V are present; and U is selected from the group consisting of -0-C(O)NH-, -OC(O)N(alkyl)-, -C(O)-, -C(O)O-, -C(O)NH-, -C(O)N(alkyl)-, -C(=N-OH)-alkyl-, and -C(=N-O-alkyl)- alkyl-; and wherein each of Y and Z is independently selected from the group consisting of a covalent bond and -O-.
In another embodiment, in Formula (I), U and V are present; and U is selected from the group consisting of -0-C(O)NH-, -OC(O)N(alkyl)-, -C(O)-, - C(O)O-, -C(O)NH-, -C(O)N(alkyl)-, -C(=N-OH)-alkyl-, and -C(=N-O-alkyl)- alkyl-; and wherein Y is — O- and Z is a covalent bond.
In another embodiment, in Formula (I), U and V are present; and U is selected from the group consisting of -0-C(O)NH-, -OC(O)N(alkyl)-, -C(O)-, - C(O)O-, -C(O)NH-, -C(O)N(alkyl)-, -C(=N-OH)-alkyl-, and -C(=N-O-alkyl)- alkyl-; and wherein each of R1 and R2 is independently selected form the group consisting of H and alkyl.
In another embodiment, in Formula (I), U and V are present; and U is selected from the group consisting of -0-C(O)NH-, -OC(O)N(alkyl)-, -C(O)-, - C(O)O-, -C(O)NH-, -C(O)N(alkyl)-, -C(=N-OH)-alkyl-, and -C(=N-O-alkyl)- alkyl-; and wherein R1 is alkyl and R2 is H. In another embodiment, in Formula (I), U and V are present; and U is selected from the group consisting of -0-C(O)NH-, -OC(O)N(alkyl)-, -C(O)-, C(O)O-, -C(O)NH-, -C(O)N(alkyl)-, -C(=N-OH)-alkyl-, and -C(=N-O-alkyl)- alkyl-; and wherein R1 is methyl.
In another embodiment, in Formula (I), U and V are present; and U is selected from the group consisting Of -O-C(O)NH-, -OC(O)N(alkyl)-, -C(O)-, C(O)O-, -C(O)NH-, -C(O)N(alkyl)-, -C(=N-OH)-alkyl-, and -C(=N-O-alkyl)- alkyl-; and wherein the compound of Formula (I) is selected from the group consisting of:
Figure imgf000041_0001
Figure imgf000042_0001
Figure imgf000043_0001
or a pharmaceut
Figure imgf000044_0001
In another embodiment, in Formula (I), at least one T and V is present, and each of -Y-R1 and -Z-R2 is independently selected from the group consisting of cyano, -(C(R4)2)n-C(O)OH, -(C(R4)2)n-C(O)O-alkyl, -(C(R4)2)n- C(O)NH2, -(C(R4Js)n-C(O)N H(alkyl), and -(C(R4)2)n-C(O)N(alkyl)2, wherein each R4 independently is H or alkyl; and n is 1-3.
In another embodiment, in Formula (I), at least one T and V is present, and each of -Y-R1 and -Z-R2 is independently selected from the group consisting of cyano, -(C(R4)2)n-C(O)OH, -(C(R4)2)n-C(O)O-alkyl, -(C(R4)2)n- C(O)NH2, -(C(R4)2)n-C(O)NH(alkyl), and -(C(R4)2)n-C(O)N(alkyl)2, wherein each R4 independently is H or alkyl; and n is 1-3; wherein ring A is selected from the group consisting of phenyl, thiophenyl, pyridyl, pyrimidyl, and
Figure imgf000044_0002
as shown.
In another embodiment, in Formula (I), at least one T and V is present, and each of -Y-R1 and -Z-R2 is independently selected from the group consisting of cyano, -(C(R4)2)n-C(O)OH, -(C(R4)2)n-C(O)O-alkyl, -(C(R4)2)n- C(O)NH2, -(C(R4)2)n-C(O)NH(alkyl), and -(C(R4)2)n-C(O)N(alkyl)2, wherein each R4 independently is H or alkyl; and n is 1-3; wherein said ring A is phenyl.
In another embodiment, in Formula (I), at least one T and V is present, and each of -Y-R1 and -Z-R2 is independently selected from the group consisting of cyano, -(C(R4)2)n-C(O)OH, -(C(R4)2)n-C(O)O-alkyl, -(C(R4)2)n- C(O)NH2, -(C(R4)2)n-C(O)NH(alkyl), and -(C(R4)2)n-C(O)N(alkyl)2, wherein each R4 independently is H or alkyl; and n is 1-3; wherein T or V is aryl which is unsubstituted or substituted with one to four R10 moieties. In another embodiment, in Formula (I), at least one T and V is present, and each of -Y-R1 and -Z-R2 is independently selected from the group consisting of cyano, -(C(R4)2)n-C(O)OH, -(C(R4)2)n-C(O)O-alkyl, -<C(R4)2)n- C(O)NH2, -(C(R4)2)n-C(O)NH(alkyl)I and -(C(R4)2)n-C(O)N(alkyl)2, wherein each R4 independently is H or alkyl; and n is 1-3; wherein said T or V is phenyl which is unsubstituted or substituted with one to four R10 moieties.
In another embodiment, in Formula (I), at least one T and V is present, and each of -Y-R1 and -Z-R2 is independently selected from the group consisting of cyano, -(C(R4)2)n-C(O)OH, -(C(R4)2)n-C(O)O-alkyl, ~iC(R4)2)n- C(O)NH2, -(C(R4)2)n-C(O)NH(alkyl), and -(C(R4)2)n-C(O)N(alkyl)2> wherein each R4 independently is H or alkyl; and n is 1-3; wherein R10 is fluoro.
In another embodiment, in Formula (I), at least one T and V is present, and each of -Y-R1 and -Z-R2 is independently selected from the group consisting of cyano, -(C(R4)2)n-C(O)OH, -(C(R4)2)n-C(O)O-alkyl, -(C(R4)2)n- C(O)NH2, -(C(R4)2)n-C(O)NH(alkyl), and -(C(R4)2)n-C(O)N(alkyl)2, wherein each R4 independently is H or alkyl; and n is 1-3; wherein only one of T and V is present.
In another embodiment, in Formula (I), at least one T and V is present; each of -Y-R1 and -Z-R2 is independently selected from the group consisting of cyano, -(C(R4)2)n-C(O)OH, -(C(R4)2)n-C(O)O-alkyl, -(C(R4Ja)n-C(O)NH2, - (C(R4)2)n-C(O)NH(alkyl), and -(C(R4)2)n-C(O)N(alkyl)2, wherein each R4 independently is H or alkyl; and n is 1-3; wherein U is absent.
In another embodiment, in Formula (I), at least one T and V is present; each of -Y-R1 and -Z-R2 is independently selected from the group consisting of cyano, -(C(R4)2)n-C(O)OH, -(C(R4)2)n-C(O)O-alkyl, -(C(R4Jz)n-C(O)NH2, - (C(R4)2)n-C(O)NH(alkyl), and -(C(R4)2)n-C(O)N(alkyl)2, wherein each R4 independently is H or alkyl; and n is 1-3, wherein n is 1.
In another embodiment, in Formula (I), at least one T and V is present; each of -Y-R1 and -Z-R2 is independently selected from the group consisting of cyano, -(C(R4)2)n-C(O)OH, -(C(R4)2)n-C(O)O-alkyl, -(C(R4)2)n-C(O)NH2) - (C(R4)2)n-C(O)NH(alkyl), and -(C(R4)2)n-C(O)N(alkyl)2, wherein each R4 independently is H or alkyl; and n is 1-3; wherein each of Y and Z is independently selected from the group consisting of a covalent bond and - CH2-, and each of R1 and R2 is independently selected from the group consisting of cyano, -C(O)OH or -C(O)NH2.
In another embodiment, in Formula (I), at least one T and V is present, each of -Y-R1 and -Z-R2 is independently selected from the group consisting of cyano, -(C(R4)2)n-C(O)OH, -<C(R4)2)n-C(O)O-alkyl, -(C(R4^)n-C(O)NH2, - (C(R4)2)n-C(O)NH(alkyl), and -(C(R4)2)π-C(O)N(alkyl)2, wherein each R4 independently is H or alkyl; and n is 1-3; wherein Y is a covalent bond, and R1 is H.
In another embodiment, in Formula (I), at least one T and V is present; each of -Y-R1 and -Z-R2 is independently selected from the group consisting of cyano, -(C(R4)2)n-C(O)OH, -(C(R4)2)n-C(O)O-alkyl, -(C(R4)2)n-C(O)NH2> - (C(R4)2)n-C(O)NH(alkyl), and -(C(R4)2)n-C(O)N(alkyl)2, wherein each R4 independently is H or alkyl; and n is 1-3; wherein Z is a covalent bond, and R2 is cyano.
In another embodiment, in Formula (I), at least one T and V is present; each of -Y-R1 and -Z-R2 is independently selected from the group consisting of cyano, -(C(R4)2)n-C(O)OH, -(C(R4)2)n-C(O)O-alkyl, -(C(R4)2)n-C(O)NH2, - (C(R4)2)n-C(O)NH(alkyl), and -(C(R4)2)n-C(O)N(alkyl)2] wherein each R4 independently is H or alkyl; and n is 1-3; wherein Z is -CH2-, and R2 is - C(O)OH or -C(O)NH2.
In another embodiment, in Formula (I), at least one T and V is present; each of -Y-R1 and -Z-R2 is independently selected from the group consisting of cyano, -(C(R4)2)n-C(O)OH, -(C(R4)2)n-C(O)O-alkyl, -(C(R4)2)n-C(O)NH2, - (C(R4)2)n-C(O)NH(alkyl), and -(C(R4)2)n-C(O)N(alkyl)2, wherein each R4 independently is H or alkyl; and n is 1-3; wherein the compound of Formula (I) is selected from the group consisting of:
Figure imgf000046_0001
Figure imgf000047_0001
or a pharmaceutically acceptable salt or solvate thereof.
In another embodiment, in Formula (I), T is aryl or heteroaryl, each of which is optionally substituted with one to four independently selected R10 moieties, and V is alkynyl which is optionally substituted with one or two independently selected R10 moieties.
In another embodiment, in Formula (I), T is aryl or heteroaryl, each of which is optionally substituted with one to four independently selected R10 moieties, and V is alkynyl which is optionally substituted with one or two independently selected R10 moieties; wherein ring A is selected from the group consisting of phenyl, thiophenyl, pyridyl, pyrimidyl, and
Figure imgf000047_0002
each of which is substituted with -Y-R1 and -Z-R2 as shown.
In another embodiment, in Formula (I), T is aryl or heteroaryl, each of which is optionally substituted with one to four independently selected R10 moieties, and V is alkynyl which is optionally substituted with one or two independently selected R10 moieties; wherein said ring A is phenyl.
In another embodiment, in Formula (I), T is aryl or heteroaryl, each of which is optionally substituted with one to four independently selected R10 moieties, and V is alkynyl which is optionally substituted with one or two independently selected R10 moieties; wherein T is aryl, U is -O- or absent, and V is alkynyl which is unsubstituted or substituted with one or two R10 moieties selected from the group consisting of -OR4, -N(R4^1 and heteroaryl; wherein when said heteroaryl contains two radicals on adjacent carbon atoms, said radicals may optionally be taken together with the carbon atoms to which they are attached to form a five- to eight-membered aryl, heteocyclyl, heteroaryl or cycloalkyl ring; wherein each R4 independently is H or alkyl, and said R10 heteroaryl is optionally independently substituted with one to four R30 moieties which can be the same or different.
In another embodiment, in Formula (I)1 T is aryl or heteroaryl, each of which is optionally substituted with one to four independently selected R10 moieties, and V is alkynyl which is optionally substituted with one or two independently selected R10 moieties; wherein T is phenyl.
In another embodiment, in Formula (I), T is aryl or heteroaryl, each of which is optionally substituted with one to four independently selected R10 moieties, and V is alkynyl which is optionally substituted with one or two independently selected R10 moieties; wherein said V alkynyl is selected from the group consisting of -CH2-C≡C-CH3, and R10 substituted -CsC-, and -CH2-C≡C-CH2~.
In another embodiment, in Formula (I), T is aryl or heteroaryl, each of which is optionally substituted with one to four independently selected R10 moieties, and V is alkynyl which is optionally substituted with one or two independently selected R10 moieties; wherein said R10 substituents are selected from the group consisting of -N(alkyl)2, -OH, -OCH3, and pyridyl.
In another embodiment, in Formula (I), T is aryl or heteroaryl, each of which is optionally substituted with one to four independently selected R10 moieties, and V is alkynyl which is optionally substituted with one or two independently selected R10 moieties; wherein the compound of Formula (I) is selected from the group consisting of:
Figure imgf000049_0001
or a pha
Figure imgf000050_0001
rmaceutically acceptable salt, solvate, or ester thereof.
In another embodiment, in Formula (I), at least one of T and V is present, ring A is heteroaryl, and V is other than alkynyl.
In another embodiment, in Formula (I), at least one of T and V is present, ring A is heteroaryl, and V is other than alkynyl; wherein ring A is selected from the group consisting of thiophenyl, pyridyl, pyrimidyl, and
Figure imgf000051_0001
each of which is substituted with -Y-R1 and -Z-R2 as shown.
In another embodiment, in Formula (I), at least one of T and V is present, ring A is heteroaryl, and V is other than alkynyl; wherein T is selected from the group consisting of alkyl, and halo-substituted aryl.
In another embodiment, in Formula (I), at least one of T and V is present, ring A is heteroaryl, and V is other than alkynyl; wherein U and V are absent.
In another embodiment, in Formula (I), at least one of T and V is present, ring A is heteroaryl, and V is other than alkynyl; wherein Y is selected from the group consisting of a covalent bond and — O-, and Z is a covalent bond.
In another embodiment, in Formula (I), at least one of T and V is present, ring A is heteroaryl, and V is other than alkynyl; wherein R1 is selected from the group consisting of H and -CH3; and R2 is H.
In another embodiment, in Formula (I), at least one of T and V is present, ring A is heteroaryl, and V is other than alkynyl; wherein Y is a covalent bond R1 is H.
In another embodiment, in Formula (I), at least one of T and V is present, ring A is heteroaryl, and V is other than alkynyl; wherein Y is is -O- and R1 is -CH3.
In another embodiment, in Formula (I), at least one of T and V is present, ring A is heteroaryl, and V is other than alkynyl; wherein Z is a covalent bond and R2 is H.
In another embodiment, in Formula (I), at least one of T and V is present, ring A is heteroaryl, and V is other than alkynyl; wherein the compound of Formula (I) is selected from the group consisting of:
Figure imgf000051_0002
or a pha
Figure imgf000052_0001
rmaceutically acceptable salt or solvate thereof.
In another embodiment, the compound of Formula (I) is selected from the group consisting of compounds listed in the table below (Table 1), or a pharmaceutically acceptable salt, solvate, ester or isomer thereof. This table also lists the mass spectroscopy data and the Ki rating for each compound. Those compounds having a Ki value of less than 10 nM (<10 nM) are designated with letter "A"; those with a Ki value of from 10 to less than 100 nM (10 - <100 nM) are designated with letter "B"; those with a Ki value of from 100 to 1000 nM are designated with letter "C"; and those with a Ki value of more than 1000 nM (>1000 nM) are designated with letter "D". The syntheis and characterization of these compounds is described hereinbelow in the "EXAMPLES" section of the present application.
Table 1
Figure imgf000054_0001
Figure imgf000055_0001
Figure imgf000056_0001
Figure imgf000057_0001
Figure imgf000058_0001
Figure imgf000059_0001
Figure imgf000060_0001
Figure imgf000061_0001
Figure imgf000062_0001
Figure imgf000063_0001
Figure imgf000064_0001
Figure imgf000065_0001
Figure imgf000066_0001
Figure imgf000067_0001
Figure imgf000068_0001
Figure imgf000069_0001
Figure imgf000070_0001
US2007/001030
70
Figure imgf000071_0001
Figure imgf000072_0002
In another embodiment, the compound of Formula (I) is selected from the group consisting of:
Figure imgf000072_0001
Figure imgf000073_0001
or a pharmaceut
Figure imgf000074_0001
.
Specific TACE inhibitory activity (Ki values) of some representative compounds of the present invention are set forth below.
Figure imgf000075_0001
Figure imgf000076_0001
As used above, and throughout this disclosure, the following terms, unless otherwise indicated, shall be understood to have the following meanings:
"Patient" includes both human and animals.
"Mammal" means humans and other mammalian animals.
"Alkyl" means an aliphatic hydrocarbon group which may be straight or branched and comprising about 1 to about 20 carbon atoms in the chain. Preferred alkyl groups contain about 1 to about 12 carbon atoms in the chain. More preferred alkyl groups contain about 1 to about 6 carbon atoms in the chain. Branched means that one or more lower alkyl groups such as methyl, ethyl or propyl, are attached to a linear alkyl chain. "Lower alkyl" means a group having about 1 to about 6 carbon atoms in the chain which may be straight or branched. The alkyl group may be substituted by one or more substituents which may be the same or different, each substituent being independently selected from the group consisting of halo, alkyl, aryl, cycloalkyl, cyano, hydroxy, alkoxy, alkylthio, amino, -NH(alkyl), -NH(cycloalkyl), -N(alkyl)2, carboxy and -C(O)O-alkyl. Non-limiting examples of suitable alkyl groups include methyl, ethyl, n-propyl, isopropyl and t-butyl.
"Alkenyl" means an aliphatic hydrocarbon group containing at least one carbon-carbon double bond and which may be straight or branched and comprising about 2 to about 15 carbon atoms in the chain. Preferred alkenyl groups have about 2 to about 12 carbon atoms in the chain; and more preferably about 2 to about 6 carbon atoms in the chain. Branched means that one or more lower alkyl groups such as methyl, ethyl or propyl, are attached to a linear alkenyl chain. "Lower alkenyl" means about 2 to about 6 carbon atoms in the chain which may be straight or branched. Non-limiting examples of suitable alkenyl groups include ethenyl, propenyl, n-butenyl, 3-methylbut-2- enyl, n-pentenyl, octenyl and decenyl.
"Alkynyl" means an aliphatic hydrocarbon group containing at least one carbon-carbon triple bond and which may be straight or branched and comprising about 2 to about 15 carbon atoms in the chain. Preferred alkynyl groups have about 2 to about 12 carbon atoms in the chain; and more preferably about 2 to about 4 carbon atoms in the chain. Branched means that one or more lower alkyl groups such as methyl, ethyl or propyl, are attached to a linear alkynyl chain. "Lower alkynyl" means about 2 to about 6 carbon atoms in the chain which may be straight or branched. Non-limiting examples of suitable alkynyl groups include ethynyl, propynyl, 2-butynyl and 3- methylbutynyl. The term "substituted alkynyl" means that the alkynyl group may be substituted by one or more substituents which may be the same or different, each substituent being independently selected from the group consisting of alkyl, aryl and cycloalkyl.
"Aryl" means an aromatic monocyclic or multicyclic ring system comprising about 6 to about 14 carbon atoms, preferably about 6 to about 10 carbon atoms. The aryl group can be optionally substituted with one or more "ring system substituents" which may be the same or different, and are as defined herein. Non-limiting examples of suitable aryl groups include phenyl and naphthyl.
"Heteroaryl" means an aromatic monocyclic or multicyclic ring system comprising about 5 to about 14 ring atoms, preferably about 5 to about 10 ring atoms, in which one or more of the ring atoms is an element other than carbon, for example nitrogen, oxygen or sulfur, alone or in combination. Preferred heteroaryls contain about 5 to about 6 ring atoms. The "heteroaryl" can be optionally substituted by one or more "ring system substituents" which may be the same or different, and are as defined herein. The prefix aza, oxa or thia before the heteroaryl root name means that at least a nitrogen, oxygen or sulfur atom respectively, is present as a ring atom. A nitrogen atom of a heteroaryl can be optionally oxidized to the corresponding N-oxide. "Heteroaryl" may also include a heteroaryl as defined above fused to an aryl as defined above. Non-limiting examples of suitable heteroaryls include pyridyl, pyrazinyl, furanyl, thienyl, pyrimidinyl, pyridone (including N- substituted pyridones), isoxazolyl, isothiazolyl, oxazolyl, thiazolyl, pyrazolyl, furazanyl, pyrrolyl, pyrazolyl, triazolyl, 1 ,2,4-thiadiazolyl, pyrazinyl, pyridazinyl, quinoxalinyl, phthalazinyl, oxindolyl, imidazo[1 ,2-a]pyridinyl, imidazo[2,1- b]thiazolyl, benzofurazanyl, indolyl, azaindolyl, benzimidazolyl, benzothienyl, quinolinyl, imidazolyl, thienopyridyl, quinazolinyl, thienopyrimidyl, pyrrolopyridyl, imidazopyridyl, isoquinolinyl, benzoazaindolyl, 1 ,2,4-triazinyl, benzothiazolyl and the like. The term "heteroaryl" also refers to partially saturated heteroaryl moieties such as, for example, tetrahydroisoquinolyl, tetrahydroquinolyl and the like.
"Aralkyl" or "arylalkyl" means an aryl-alkyl- group in which the aryl and alkyl are as previously described. Preferred aralkyls comprise a lower alkyl group. Non-limiting examples of suitable aralkyl groups include benzyl, 2- phenethyl and naphthalenylmethyl. The bond to the parent moiety is through the alkyl.
"Alkylaryl" means an alkyl-aryl- group in which the alkyl and aryl are as previously described. Preferred alkylaryls comprise a lower alkyl group. Non- limiting example of a suitable alkylaryl group is tolyl. The bond to the parent moiety is through the aryl.
"Cycloalkyl" means a non-aromatic mono- or multicyclic ring system comprising about 3 to about 10 carbon atoms, preferably about 5 to about 10 carbon atoms. Preferred cycloalkyl rings contain about 5 to about 7 ring atoms. The cycloalkyl can be optionally substituted with one or more "ring system substituents" which may be the same or different, and are as defined above. Non-limiting examples of suitable monocyclic cycloalkyls include cyclopropyl, cyclopentyl, cyclohexyl, cycloheptyl and the like. Non-limiting examples of suitable multicyclic cycloalkyls include 1-decalinyl, norbornyl, adamantyl and the like.
"Cycloalkenyl" means a non-aromatic mono or multicyclic ring system comprising about 3 to about 10 carbon atoms, preferably about 5 to about 10 carbon atoms which contains at least one carbon-carbon double bond. Preferred cycloalkenyl rings contain about 5 to about 7 ring atoms. The cycloalkenyl can be optionally substituted with one or more "ring system substituents" which may be the same or different, and are as defined above. Non-limiting examples of suitable monocyclic cycloalkenyls include cyclopentenyl, cyclohexenyl, cyclohepta-1,3-dienyl, and the like. Non-limiting example of a suitable multicyclic cycloalkenyl is norbornylenyl.
"Halogen" (or "halo") means fluorine, chlorine, bromine, or iodine. Preferred are fluorine, chlorine and bromine.
"Ring system substituent" means a substituent attached to an aromatic or non-aromatic ring system which, for example, replaces an available hydrogen on the ring system. Ring system substituents may be the same or different, each being independently selected from the group consisting of alkyl, alkenyl, alkynyl, aryl, heteroaryl, aralkyl, alkylaryl, heteroaralkyl, heteroarylalkenyl, heteroarylalkynyl, alkylheteroaryl, hydroxy, hydroxyalkyl, alkoxy, aryloxy, aralkoxy, acyl, aroyl, halo, nitro, cyano, carboxy, alkoxycarbonyl, aryloxycarbonyl, aralkoxycarbonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfoπyl, alkylthio, arylthio, heteroarylthio, aralkylthio, heteroaralkylthio, cycloalkyl, heterocyclyl, -C(=N-CN)-NH2, -C(=NH)-NH2, - C(=NH)-NH(alkyl), G1G2N-, GiG2N-alkyh G1G2NC(O)-, G1G2NSO2- and - SO2NG1G2, wherein G1 and G2 can be the same or different and are independently selected from the group consisting of hydrogen, alkyl, aryl, cycloalkyl, and aralkyl. "Ring system substituent" may also mean a single moiety which simultaneously replaces two available hydrogens on two adjacent carbon atoms (one H on each carbon) on a ring system. Examples of such moiety are methylene dioxy, ethylenedioxy, -C(CHs)2- and the like which form moieties such as, for example:
Figure imgf000080_0001
"Heterocyclyl" means a non-aromatic saturated monocyclic or multicyclic ring system comprising about 3 to about 10 ring atoms, preferably about 5 to about 10 ring atoms, in which one or more of the atoms in the ring system is an element other than carbon, for example nitrogen, oxygen or sulfur, alone or in combination. There are no adjacent oxygen and/or sulfur atoms present in the ring system. Preferred heterocyclyls contain about 5 to about 6 ring atoms. The prefix aza, oxa orthia before the heterocyclyl root name means that at least a nitrogen, oxygen or sulfur atom respectively is present as a ring atom. Any -NH in a heterocyclyl ring may exist protected such as, for example, as an -N(Boc), -N(CBz)1 -N(Tos) group and the like; such protections are also considered part of this invention. The heterocyclyl can be optionally substituted by one or more "ring system substituents" which may be the same or different, and are as defined herein. The nitrogen or sulfur atom of the heterocyclyl can be optionally oxidized to the corresponding N-oxide, S-oxide or S,S-dioxide. Non-limiting examples of suitable monocyclic heterocyclyl rings include piperidyl, pyrrolidinyl, piperazinyl, morpholinyl, thiomorpholinyl, thiazolidinyl, 1,4-dioxanyl, tetrahydrofuranyl, tetrahydrothiophenyl, lactam, lactone, and the like. "Heterocyclyl" may also mean a single moiety (e.g., carbonyl) which simultaneously replaces two available hydrogens on the same carbon atom on a ring system. Example of such moiety is pyrrolidone:
Figure imgf000081_0001
It should be noted that tautomeric forms such as, for example, the moieties:
Figure imgf000081_0002
are considered equivalent in certain embodiments of this invention.
"Alkynylalkyl" means an alkynyl-alkyl- group in which the alkynyl and alkyl are as previously described. Preferred alkynylalkyls contain a lower alkynyl and a lower alkyl group. The bond to the parent moiety is through the alkyl. Non-limiting examples of suitable alkynylalkyl groups include propargylrnethyl.
"Heteroaralkyl" means a heteroaryl-alkyl- group in which the heteroaryl and alkyl are as previously described. Preferred heteroaralkyls contain a lower alkyl group. Non-limiting examples of suitable aralkyl groups include pyridylmethyl, and quinolin-3-ylmethyl. The bond to the parent moiety is through the alkyl.
"Hydroxyalkyl" means a HO-alkyl- group in which alkyl is as previously defined. Preferred hydroxyalkyls contain lower alkyl. Non-limiting examples of suitable hydroxyalkyl groups include hydroxymethyl and 2-hydroxyethyl.
"Acyl" means an H-C(O)-, alkyl-C(O)- or cycloalkyl-C(O)-, group in which the various groups are as previously described. The bond to the parent moiety is through the carbonyl. Preferred acyls contain a lower alky!. Non- limiting examples of suitable acyl groups include formyl, acetyl and propanoyl.
"Aroyl" means an aryl-C(O)- group in which the aryl group is as previously described. The bond to the parent moiety is through the carbonyl. Non-limiting examples of suitable groups include benzoyl and 1- naphthoyl.
"Alkoxy" means an alkyl-O- group in which the alkyl group is as previously described. Non-limiting examples of suitable alkoxy groups include methoxy, ethoxy, n-propoxy, isopropoxy and n-butoxy. The bond to the parent moiety is through the ether oxygen.
"Aryloxy" means an aryl-O- group in which the aryl group is as previously described. Non-limiting examples of suitable aryloxy groups include phenoxy and naphthoxy. The bond to the parent moiety is through the ether oxygen.
"Aralkyloxy" means an aralkyl-O- group in which the aralkyl group is as previously described. Non-limiting examples of suitable aralkyloxy groups include benzyloxy and 1 - or 2-naphthalenemethoxy. The bond to the parent moiety is through the ether oxygen.
"Alkylthio" means an alkyl-S- group in which the alkyl group is as previously described. Non-limiting examples of suitable alkylthio groups include methylthio and ethylthio. The bond to the parent moiety is through the sulfur.
"Arylthio" means an aryl-S- group in which the aryl group is as previously described. Non-limiting examples of suitable arylthio groups include phenylthio and naphthylthio. The bond to the parent moiety is through the sulfur.
"Aralkylthio" means an aralkyl-S- group in which the aralkyl group is as previously described. Non-limiting example of a suitable aralkylthio group is benzylthio. The bond to the parent moiety is through the sulfur.
"Alkoxycarbonyl" means an alkyl-O-CO- group. Non-limiting examples of suitable alkoxycarbonyl groups include methoxycarbonyl and ethoxycarbonyl. The bond to the parent moiety is through the carbonyl. "Aryloxycarbonyl" means an aryl-O-C(O)- group. Non-limiting examples of suitable aryloxycarbonyl groups include phenoxycarbonyl and naphthoxycarbonyl. The bond to the parent moiety is through the carbonyl.
"Aralkoxycarbonyl" means an aralkyl-O-C(O)- group. Non-limiting example of a suitable aralkoxycarbonyl group is benzyloxycarbonyl. The bond to the parent moiety is through the carbonyl.
"Alkylsulfonyl" means an alkyl-S(O2)- group. Preferred groups are those in which the alkyl group is lower alkyl. The bond to the parent moiety is through the sulfonyl.
"Arylsulfonyl" means an aryl-S(C>2)- group. The bond to the parent moiety is through the sulfonyl.
The term "substituted" means that one or more hydrogens on the designated atom is replaced with a selection from the indicated group, provided that the designated atom's normal valency under the existing circumstances is not exceeded, and that the substitution results in a stable compound. Combinations of substituents and/or variables are permissible only if such combinations result in stable compounds. By "stable compound' or "stable structure" is meant a compound that is sufficiently robust to survive isolation to a useful degree of purity from a reaction mixture, and formulation into an efficacious therapeutic agent.
The term "optionally substituted" means optional substitution with the specified groups, radicals or moieties.
The term "isolated" or "in isolated form" for a compound refers to the physical state of said compound after being isolated from a synthetic process or natural source or combination thereof. The term "purified" or "in purified form" for a compound refers to the physical state of said compound after being obtained from a purification process or processes described herein or well known to the skilled artisan, in sufficient purity to be characterizable by standard analytical techniques described herein or well known to the skilled artisan.
It should also be noted that any carbon as well as heteroatom with unsatisfied valences in the text, schemes, examples and Tables herein is assumed to have the sufficient number of hydrogen atom(s) to satisfy the valences.
When a functional group in a compound is termed "protected", this means that the group is in modified form to preclude undesired side reactions at the protected site when the compound is subjected to a reaction. Suitable protecting groups will be recognized by those with ordinary skill in the art as well as by reference to standard textbooks such as, for example, T. W. Greene etal, Protective Groups in organic Synthesis (1991), Wiley, New York.
When any variable (e.g., aryl, heterocycle, R2, etc.) occurs more than one time in any constituent or in Formula I1 its definition on each occurrence is independent of its definition at every other occurrence.
As used herein, the term "composition" is intended to encompass a product comprising the specified ingredients in the specified amounts, as well as any product which results, directly or indirectly, from combination of the specified ingredients in the specified amounts.
Prodrugs and solvates of the compounds of the invention are also contemplated herein. The term "prodrug", as employed herein, denotes a compound that is a drug precursor which, upon administration to a subject, undergoes chemical conversion by metabolic or chemical processes to yield a compound of Formula I or a salt and/or solvate thereof. A discussion of prodrugs is provided in T. Higuchi and V. Stella, Pro-drugs as Novel Delivery Systems (1987) 14 of the A.C.S. Symposium Series, and in Bioreversible Carriers in Drug Design, (1987) Edward B. Roche, ed., American Pharmaceutical Association and Pergamon Press, both of which are incorporated herein by reference thereto. The term "prodrug" means a compound (e.g, a drug precursor) that is transformed in vivo to yield a compound of Formula (I) or a pharmaceutically acceptable salt, hydrate or solvate of the compound. The transformation may occur by various mechanisms (e.g., by metabolic or chemical processes), such as, for example, through hydrolysis in blood. A discussion of the use of prodrugs is provided by T. Higuchi and W. Stella, "Pro-drugs as Novel Delivery Systems," Vol. 14 of the A.C.S. Symposium Series, and in Bioreversible Carriers in Drug Design, ed. Edward B. Roche, American Pharmaceutical Association and Pergamon Press, 1987.
For example, if a compound of Formula (I) or a pharmaceutically acceptable salt, hydrate or solvate of the compound contains a carboxylic acid functional group, a prodrug can comprise an ester formed by the replacement of the hydrogen atom of the acid group with a group such as, for example, (Ci-CβJalkyI, (C2-Ci2)alkanoyloxymethyl, 1-(alkanoyloxy)ethyl having from 4 to 9 carbon atoms, 1-methyl-1-(alkanoyloxy)-ethyl having from 5 to 10 carbon atoms, alkoxycarbonyloxymethyl having from 3 to 6 carbon atoms, 1- (alkoxycarbonyloxy)ethyl having from 4 to 7 carbon atoms, 1-methyl-1- (alkoxycarbonyloxy)ethyl having from 5 to 8 carbon atoms, N- (alkoxycarbonyl)aminomethyl having from 3 to 9 carbon atoms, 1-(N- (alkoxycarbonyl)amino)ethyl having from 4 to 10 carbon atoms, 3-phthalidyl, 4-crotonolactonyl, gamma-butyrolacton-4-yl, di-N,N-(C1-C2)alkylamino(C2- C3)alkyl (such as β-dimethylaminoethyl), carbamoyl-(Ci-C2)alkyl, N,N-di (Cr C2)alkylcarbamoyl-(C1 -C2)alkyl and piperidino-, pyrrolidino- or morpholino(C2- C-3)alkyl, and the like.
Similarly, if a compound of Formula (I) contains an alcohol functional group, a prodrug can be formed by the replacement of the hydrogen atom of the alcohol group with a group such as, for example, (C1- C6)alkanoyloxymethyl, 1 -((C1-C6)alkanoyloxy)ethyl, 1-methyl-1-((Cr C6)alkanoyloxy)ethyl, (Ci-Cejalkoxycarbonyloxymethyl, N-(Cr Cejalkoxycarbonylaminomethyl, succinoyl, (CrC6)alkanoyl, α-amino(Ci- C4)alkanyl, arylacyl and α-aminoacyl, or α-aminoacyl-α-aminoacyl, where each α-aminoacyl group is independently selected from the naturally occurring L- amino acids, P(O)(OH)2, -P(O)(O(Ci-C6)alkyl)2 or glycosyl (the radical resulting from the removal of a hydroxyl group of the hemiacetal form of a carbohydrate), and the like.
If a compound of Formula (I) incorporates an amine functional group, a prodrug can be formed by the replacement of a hydrogen atom in the amine group with a group such as, for example, R-carbonyl, RO-carbonyl, NRR'- carbonyl where R and R1 are each independently (Ci-CiO)alkyl, (C3-C7) cycloalkyl, benzyl, or R-carbonyl is a natural α-aminoacyl or natural α- aminoacyl, -C(OH)C(O)OY1 wherein Y1 is H, (CrC6)alkyl or benzyl, — C(OY2)Y3 wherein Y2 is (C1-C4) alkyl and Y3 is (d-QOalkyl, carboxy (d- C6)alkyl, amino(Ci-C4)alkyl or mono-N — or di-N,N-(Ci-C6)alkylaminoalkyl, — C(Y4JY5 wherein Y4 is H or methyl and Y5 is mono-N— or di-N,N-(d- Cβ)alkylamino morpholino, piperidin-1-yl or pyrrolidin-1-yl, and the like.
"Solvate" means a physical association of a compound of this invention with one or more solvent molecules. This physical association involves varying degrees of ionic and covalent bonding, including hydrogen bonding. In certain instances the solvate will be capable of isolation, for example when one or more solvent molecules are incorporated in the crystal lattice of the crystalline solid. "Solvate" encompasses both solution-phase and isolatable solvates. Non-limiting examples of suitable solvates include ethanolates, methanolates, and the like. "Hydrate" is a solvate wherein the solvent molecule is H2O.
"Effective amount" or "therapeutically effective amount" is meant to describe an amount of compound or a composition of the present invention effective in inhibiting TACE, the production of TNF-α, MMPs, ADAMS or any combination thereof and thus producing the desired therapeutic, ameliorative, inhibitory or preventative effect.
The compounds of Formula I can form salts which are also within the scope of this invention. Reference to a compound of Formula I herein is understood to include reference to salts thereof, unless otherwise indicated. The term "salt(s)", as employed herein, denotes acidic salts formed with inorganic and/or organic acids, as well as basic salts formed with inorganic and/or organic bases. In addition, when a compound of Formula I contains both a basic moiety, such as, but not limited to a pyridine or imidazole, and an acidic moiety, such as, but not limited to a carboxylic acid, zwitterions ("inner salts") may be formed and are included within the term "salt(s)" as used herein. Pharmaceutically acceptable (i.e., non-toxic, physiologically acceptable) salts are preferred, although other salts are also useful. Salts of the compounds of the Formula I may be formed, for example, by reacting a compound of Formula I with an amount of acid or base, such as an equivalent amount, in a medium such as one in which the salt precipitates or in an aqueous medium followed by lyophilization.
Exemplary acid addition salts include acetates, ascorbates, benzoates, benzenesulfonates, bisulfates, borates, butyrates, citrates, camphorates, camphorsulfonates, fumarates, hydrochlorides, hydrobromides, hydroiodides, lactates, maleates, methanesulfonates, naphthalenesulfonates, nitrates, oxalates, phosphates, propionates, salicylates, succinates, sulfates, tartarates, thiocyanates, toluenesulfonates (also known as tosylates,) and the like. Additionally, acids which are generally considered suitable for the formation of pharmaceutically useful salts from basic pharmaceutical compounds are discussed, for example, by P. Stahl etal, Camille G. (eds.) Handbook of Pharmaceutical Salts. Properties, Selection and Use. (2002) Zurich: Wiley- VCH; S. Berge et al, Journal of Pharmaceutical Sciences (1977) 66(1) 1-19; P. Gould, International J. of Pharmaceutics (1986) 33201-217; Anderson etal, The Practice of Medicinal Chemistry (1996), Academic Press, New York; and in The Orange Book (Food & Drug Administration, Washington, D.C. on their website). These disclosures are incorporated herein by reference thereto.
Exemplary basic salts include ammonium salts, alkali metal salts such as sodium, lithium, and potassium salts, alkaline earth metal salts such as calcium and magnesium salts, salts with organic bases (for example, organic amines) such as dicyclohexylamines, t-butyl amines, and salts with amino acids such as arginine, lysine and the like. Basic nitrogen-containing groups may be quartemized with agents such as lower alkyl halides (e.g. methyl, ethyl, and butyl chlorides, bromides and iodides), dialkyl sulfates (e.g. dimethyl, diethyl, and dibutyl sulfates), long chain halides (e.g. decyl, lauryl, and stearyl chlorides, bromides and iodides), aralkyl halides (e.g. benzyl and phenethyl bromides), and others.
All such acid salts and base salts are intended to be pharmaceutically acceptable salts within the scope of the invention and all acid and base salts are considered equivalent to the free forms of the corresponding compounds for purposes of the invention.
Compounds of Formula I, and salts, solvates and prodrugs thereof, may exist in their tautomeric form (for example, as an amide or imino ether). All such tautomeric forms are contemplated herein as part of the present invention.
All stereoisomers (for example, geometric isomers, optical isomers and the like) of the present compounds (including those of the salts, solvates and prodrugs of the compounds as well as the salts and solvates of the prodrugs), such as those which may exist due to asymmetric carbons on various substituents, including enantiomeric forms (which may exist even in the absence of asymmetric carbons), rotameric forms, atropisomers, and diastereomeric forms, are contemplated within the scope of this invention, as are positional isomers (such as, for example, 4-pyridyl and 3-pyridyl). Individual stereoisomers of the compounds of the invention may, for example, be substantially free of other isomers, or may be admixed, for example, as racemates or with all other, or other selected, stereoisomers. The chiral centers of the present invention can have the S or R configuration as defined by the IUPAC 1974 Recommendations. The use of the terms "salt", "solvate" "prodrug" and the like, is intended to equally apply to the salt, solvate and prodrug of enantiomers, stereoisomers, rotamers, tautomers, positional isomers, racemates or prodrugs of the inventive compounds.
Polymorphic forms of the compounds of Formula I1 and of the salts, solvates and prodrugs of the compounds of Formula I, are intended to be included in the present invention.
The compounds according to the invention have pharmacological properties; in particular, the compounds of Formula I can be inhibitors of TACE, aggrecanase, TNF-α and/or MMP activity.
In one aspect, the invention provides a pharmaceutical composition comprising as an active ingredient at least one compound of formula (I). In another aspect, the invention provides a pharmaceutical composition of formula (I) additionally comprising at least one pharmaceutically acceptable carrier.
In another aspect, the invention provides a method of treating disorders associated with TACE, aggrecanase, TNF-α, MMPs, ADAMs or any combination thereof, said method comprising administering to a patient in need of such treatment an effective amount of at least one compound of formula (I).
In another aspect, the invention provides a use of a compound of formula (I) for the manufacture of a medicament to treat disorders associated with TACE, aggrecanase, TNF-α, MMPs, ADAMs or any combination thereof.
The compounds of Formula (I) can have anti-inflammatory activity and/or immunomodulatory activity and can be useful in the treatment of diseases including but not limited to septic shock, haemodynamic shock, sepsis syndrome, post ischaemic reperfusion injury, malaria, mycobacterial infection, meningitis, psoriasis, congestive heart failure, fibrotic diseases, cachexia, graft rejection, cancers such as cutaneous T-cell lymphoma, diseases involving angiogenesis, autoimmune diseases, skin inflammatory diseases, inflammatory bowel diseases such as Crohn's disease and colitis, OA and RA, ankylosing spondylitis, psoriatic arthritis, adult Still's disease, ureitis, Wegener's granulomatosis, Behcehe disease, Sjogren's syndrome, sarcoidosis, polymyositis, dermatomyositis, multiple sclerosis, sciatica, complex regional pain syndrome, radiation damage, hyperoxic alveolar injury, periodontal disease, HIV, non-insulin dependent diabetes mellitus, systemic lupus erythematosus, glaucoma, sarcoidosis, idiopathic pulmonary fibrosis, bronchopulmonary dysplasia, retinal disease, scleroderma, osteoporosis, renal ischemia, myocardial infarction, cerebral stroke, cerebral ischemia, nephritis, hepatitis, glomerulonephritis, cryptogenic fibrosing aveolitis, psoriasis, transplant rejection, atopic dermatitis, vasculitis, allergy, seasonal allergic rhinitis, reversible airway obstruction, adult respiratory distress syndrome, asthma, chronic obstructive pulmonary disease (COPD) and/or bronchitis. It is contemplated that a compound of this invention may be useful in treating one or more of the diseases listed.
In another aspect, the invention provides a method of preparing a pharmaceutical composition for treating the disorders associated with TACE, aggrecanase, TNF-α, MMPs, ADAMs or any combination thereof, said method comprising bringing into intimate contact at least one compound of formula (I) and at least one pharmaceutically acceptable carrier.
In another aspect, the invention provides a compound of formula (I) exhibiting TACE, TNF-α, MMPs, ADAMs or any combination thereof inhibitory activity, including enantiomers, stereoisomers and tautomers of said compound, and pharmaceutically acceptable salts, solvates, or esters of said compound, said compound being selected from the compounds of structures listed in Table 1 set forth above.
In another aspect, the invention provides a pharmaceutical composition for treating disorders associated with TACE, aggrecanase, TNF-α, MMP, ADAM or any combination thereof in a subject comprising, administering to the subject in need of such treatment a therapeutically effective amount of at least one compound of formula (I) or a pharmaceutically acceptable salt, solvate, ester, or isomer thereof.
Jn another aspect, the invention provides a compound of formula (I) in purified form.
In another aspect, the invention provides a method of treating a condition or disease mediated by TACE, MMPs, TNF-α, aggrecanase, or any combination thereof in a subject comprising: administering to the subject in need of such treatment a therapeutically effective amount of at least one compound of formula (I) or a pharmaceutically acceptable salt, solvate, ester or isomer thereof.
In another aspect, the invention provides a method of treating a condition or disease selected from the group consisting of rheumatoid arthritis, osteoarthritis, periodontitis, gingivitis, corneal ulceration, solid tumor growth and tumor invasion by secondary metastases, neovascular glaucoma, inflammatory bowel disease, multiple sclerosis and psoriasis in a subject, comprising: administering to the subject in need of such treatment a therapeutically effective amount of at least one compound of formula (I) or a pharmaceutically acceptable salt, solvate, ester or isomer thereof.
In another aspect, the invention provides a method of treating a condition or disease selected from the group consisting of fever, cardiovascular conditions, hemorrhage, coagulation, cachexia, anorexia, alcoholism, acute phase response, acute infection, shock, graft versus host reaction, autoimmune disease and HIV infection in a subject comprising administering to the subject in need of such treatment a therapeutically effective amount of at least one compound of formula (I) or a pharmaceutically acceptable salt, solvate, ester, or isomer thereof.
In another aspect, the invention provides a method of treating a condition or disease selected from the group consisting of septic shock, haemodynamic shock, sepsis syndrome, post ischaemic reperfusion injury, malaria, mycobacterial infection, meningitis, psoriasis, congestive heart failure, fibrotic diseases, cachexia, graft rejection, cancers such as cutaneous T-cell lymphoma, diseases involving angiogenesis, autoimmune diseases, skin inflammatory diseases, inflammatory bowel diseases such as Crohn's disease and colitis, osteo and rheumatoid arthritis, ankylosing spondylitis, psoriatic arthritis, adult Still's disease, ureitis, Wegener's granulomatosis, Behcehe disease, Sjogren's syndrome, sarcoidosis, polymyositis, dermatomyositis, multiple sclerosis, sciatica, complex regional pain syndrome, radiation damage, hyperoxic alveolar injury, periodontal disease, HIV1 non- insulin dependent diabetes mellitus, systemic lupus erythematosus, glaucoma, sarcoidosis, idiopathic pulmonary fibrosis, bronchopulmonary dysplasia, retinal disease, scleroderma, osteoporosis, renal ischemia, myocardial infarction, cerebral stroke, cerebral ischemia, nephritis, hepatitis, glomerulonephritis, cryptogenic fibrosing aveolitis, psoriasis, transplant rejection, atopic dermatitis, vasculitis, allergy, seasonal allergic rhinitis, reversible airway obstruction, adult respiratory distress syndrome, asthma, chronic obstructive pulmonary disease (COPD) and bronchitis in a subject comprising administering to the subject in need of such treatment a therapeutically effective amount of at least one compound of formula (I) or a pharmaceutically acceptable salt, solvate, ester or isomer thereof.
In another aspect, the invention provides a method of treating a condition or disease associated with COPD, comprising: administering to the subject in need of such treatment a therapeutically effective amount of at least one compound of Formula (I) or a pharmaceutically acceptable salt, solvate, ester or isomer thereof.
In another aspect, the invention provides a method of treating a condition or disease associated with rheumatoid arthritis, comprising: administering to the subject in need of such treatment a therapeutically effective amount of at least one compound of formula (I) or a pharmaceutically acceptable salt, solvate, ester, or isomer thereof.
In another aspect, the invention provides a method of treating a condition or disease associated with Crohn's disease, comprising: administering to the subject in need of such treatment a therapeutically effective amount of at least one compound of formula (I) or a pharmaceutically acceptable salt, solvate, ester or isomer thereof.
In another aspect, the invention provides a method of treating a condition or disease associated with psoriasis, comprising: administering to the subject in need of such treatment a therapeutically effective amount of at least one compound of formula (I) or a pharmaceutically acceptable salt, solvate, ester, or isomer thereof.
In another aspect, the invention provides a method of treating a condition or disease associated with ankylosing spondylitis, comprising: administering to the subject in need of such treatment a therapeutically effective amount of at least one compound of formula (I) or a pharmaceutically acceptable salt, solvate, ester or isomer thereof.
In another aspect, the invention provides a method of treating a condition or disease associated with sciatica, comprising: administering to the subject in need of such treatment a therapeutically effective amount of at least one compound of formula (I) or a pharmaceutically acceptable salt, solvate, ester or isomer thereof. In another aspect, the invention provides a method of treating a condition or disease associated with complex regional pain syndrome, comprising: administering to the subject in need of such treatment a therapeutically effective amount of at least one compound of formula (I) or a pharmaceutically acceptable salt, solvate, ester, or isomer thereof.
In another aspect, the invention provides a method of treating a condition or disease associated with psoriatic arthritis, comprising: administering to the subject in need of such treatment a therapeutically effective amount of at least one compound of formula (I), or a pharmaceutically acceptable salt, solvate, ester, or isomer thereof.
In another aspect, the invention provides a method of treating a condition or disease associated with multiple sclerosis, comprising: administering to the subject in need of such treatment a therapeutically effective amount of at least one compound of formula (I) or a pharmaceutically acceptable salt, solvate, ester or isomer thereof, in combination with a compound selected from the group consisting of AvonexD, Betaseron, Copaxone or other compounds indicated for the treatment of multiple sclerosis.
Additionally, a compound of the present invention may be coadministered or used in combination with disease-modifying antirheumatic drugs (DMARDS) such as methotrexate, azathioprine, leflunomide, pencillinamine, gold salts, mycophenolate mofetil, cyclophosphamide and other similar drugs. They may also be co-administered with or used in combination with non-steroidal anti-inflammatory drugs (NSAIDs) such as piroxicam, naproxen, indomethacin, ibuprofen and the like; cycloxygenase-2 selective (COX-2) inhibitors such as VioxxD and CelebrexD; immunosuppressives such as steroids, cyclosporin, Tacrolimus, rapamycin and the like; biological response modifiers (BRMs) such as EnbrelD, RemicadeD, IL-1 antagonists, anti-CD40, anti-CD28, IL-10, anti-adhesion molecules and the like; and other anti-inflammatory agents such as p38 kinase inhibitors, PDE4 inhibitors, other chemically different TACE inhibitors, chemokine receptor antagonists, Thalidomide and other small molecule inhibitors of pro-inflammatory cytokine production.
Also, a compound of the present invention may be co-administered or used in combination with an H1 antagonist for the treatment of seasonal allergic rhinitis and/or asthma. Suitable H1 antagonists may be, for example, Claritin®, Clarinex®, Allegra®, or Zyrtec®.
In another aspect, the invention provides a method of treating a condition or disease mediated by TACE, MMPs, TNF-α, aggrecanase, or any combination thereof in a subject comprising: administering to the subject in need of such treatment a therapeutically effective amount of at least one compound of formula (I) or a pharmaceutically acceptable salt, solvate or isomer thereof in combination with a therapeutically effective amount of at least one medicament selected from the group consisting of disease modifying anti-rheumatic drugs (DMARDS), NSAIDs, COX-2 inhibitors, COX-1 inhibitors, immunosuppressives, biological response modifiers (BRMs), antiinflammatory agents and H1 antagonists.
In another aspect, the invention provides a method of treating a condition or disease selected from the group consisting of rheumatoid arthritis, osteoarthritis, periodontitis, gingivitis, corneal ulceration, solid tumor growth and tumor invasion by secondary metastases, neovascular glaucoma, inflammatory bowel disease, multiple sclerosis and psoriasis in a subject, comprising: administering to the subject in need of such treatment a therapeutically effective amount of at least one compound of Formula (I) or a pharmaceutically acceptable salt, solvate, ester, or isomer thereof in combination with a therapeutically effective amount of at least one medicament selected from the group consisting of DMARDS, NSAIDs, COX-2 inhibitors, COX-1 inhibitors, immunosuppressives, BRMs, anti-inflammatory agents and H1 antagonists.
In another aspect, the invention provides a method of treating a condition or disease selected from the group consisting of septic shock, haemodynamic shock, sepsis syndrome, post ischaemic reperfusion injury, malaria, mycobacterial infection, meningitis, psoriasis, congestive heart failure, fibrotic diseases, cachexia, graft rejection, cancers such as cutaneous T-cell lymphoma, diseases involving angiogenesis, autoimmune diseases, skin inflammatory diseases, inflammatory bowel diseases such as Crohn's disease and colitis, osteo and rheumatoid arthritis, ankylosing spondylitis, psoriatic arthritis, adult Still's disease, ureitis, Wegener's granulomatosis, Behcehe disease, Sjogren's syndrome, sarcoidosis, polymyositis, dermatomyositis, multiple sclerosis, sciatica, complex regional pain syndrome, radiation damage, hyperoxic alveolar injury, periodontal disease, HIV, non- insulin dependent diabetes mellitus, systemic lupus erythematosus, glaucoma, sarcoidosis, idiopathic pulmonary fibrosis, bronchopulmonary dysplasia, retinal disease, scleroderma, osteoporosis, renal ischemia, myocardial infarction, cerebral stroke, cerebral ischemia, nephritis, hepatitis, glomerulonephritis, cryptogenic fibrosing aveolitis, psoriasis, transplant rejection, atopic dermatitis, vasculitis, allergy, seasonal allergic rhinitis, reversible airway obstruction, adult respiratory distress syndrome, asthma, chronic obstructive pulmonary disease (COPD) and bronchitis in a subject comprising administering to the subject in need of such treatment a therapeutically effective amount of at least one compound of Formula (I), or a pharmaceutically acceptable salt, solvate, ester or isomer thereof in combination with a therapeutically effective amount of at least one medicament selected from the group consisting of DMARDS, NSAIDs, COX-2 inhibitors, COX-1 inhibitors, immunosuppressives, BRMs, anti-inflammatory agents and H1 antagonists.
In another aspect, the invention provides a method for treating RA comprising administering a compound of the formula I in combination with compound selected from the class consisting of a COX-2 inhibitor e.g. Celebrex® or Vioxx®; a COX-1 inhibitor e.g. Feldene®; an immunosuppressive e.g. methotrexate or cyclosporin; a steroid e.g. D- methasone; and anti-TNF-α compound, e.g. Enbrel® or Remicade®; a PDE IV inhibitor, or other classes of compounds indicated for the treatment of RA.
In another aspect, the invention provides a method for treating multiple sclerosis comprising administering a compound of the formula (I) in combination with a compound selected from the group consisting of Avonex®, Betaseron, Copaxone or other compounds indicated for the treatment of multiple sclerosis.
TACE activity is determined by a kinetic assay measuring the rate of increase in fluorescent intensity generated by TACE catalyzed cleavage of an internally quenched peptide substrate (SPDL-3). The purified catalytic domain of recombinant human TACE (rhTACEc, Residue 215 to 477 with two mutation (S266A and N452Q) and a 6xHis tail) is used in the assay. It is purified from the baculovirus/Hi5 cells expression system using affinity chromatography. The substrate SPDL-3 is an internally quenched peptide (MCA-Pro-Leu-Ala-Gln-Ala-Val-Arg-Ser-Ser-Ser-Dpa-Arg-NH2), with its sequence derived from the pro-TNFD cleavage site. MCA is (7- Methoxycoumarin-4-yl)acetyl. Dpa is N-3-(2,4-Dinitrophenyl)-L-2,3- diaminopropionyl.
A 50 /J assay mixture contains 20 mM HEPES, pH 7.3, 5 mM CaCI2, 100 DM ZnCI2, 2 % DMSO, 0.04% Methylcellulose, 30 μM SPDL-3, 7O pM rhTACEc and a test compound. RhTACEc is pre-incubated with the testing compound for 90 min. at 250C. Reaction is started by addition of the substrate. The fluorescent intensity (excitation at 320 nm, emission at 405 nm) was measured every 45 seconds for 30 min. using a fluorospectrometer (GEMINI XS, Molecular Devices). Rate of enzymatic reaction is shown as Units per second. Effect of a test compound is shown as % of TACE activity in the absence of the compound.
The pharmaceutical compositions containing the active ingredient may be in a form suitable for oral use, for example, as tablets, lozenges, aqueous or oily suspensions, dispersible powders or granules, emulsions, hard or soft capsules, or syrups or elixirs. Compositions intended for oral use may be prepared according to any method known to the art for the manufacture of pharmaceutical compositions and such compositions may contain one or more agents selected from the group consisting of sweetening agents, flavoring agents, coloring agents and preserving agents in order to provide pharmaceutically elegant and palatable preparations. Tablets contain the active ingredient in admixture with non-toxic pharmaceutically acceptable excipients that are suitable for the manufacture of tablets. These excipients may be for example, inert diluents, such as calcium carbonate, sodium carbonate, lactose, calcium phosphate or sodium phosphate; granulating and disintegrating agents, for example, corn starch, or alginic acid; binding agents, for example starch, gelatin or acacia, and lubricating agents, for example magnesium stearate, stearic acid or talc. The tablets may be uncoated or they may be coated by known techniques to delay disintegration and absorption in the gastrointestinal tract and thereby provide a sustained action over a longer period. For example, a time delay material such as glyceryl monostearate or glyceryl distearate may be employed. They may also be coated by the technique described in the U.S. Pat. Nos. 4,256,108; 4,166,452; and 4,265,874 to form osmotic therapeutic tablets for controlled release.
The term pharmaceutical composition is also intended to encompass both the bulk composition and individual dosage units comprised of more than one (e.g., two) pharmaceutically active agents such as, for example, a compound of the present invention and an additional agent selected from the lists of the additional agents described herein, along with any pharmaceutically inactive excipients. The bulk composition and each individual dosage unit can contain fixed amounts of the afore-said "more than one pharmaceutically active agents". The bulk composition is material that has not yet been formed into individual dosage units. An illustrative dosage unit is an oral dosage unit such as tablets, pills and the like. Similarly, the herein-described method of treating a patient by administering a pharmaceutical composition of the present invention is also intended to encompass the administration of the afore-said bulk composition and individual dosage units.
Formulations for oral use may also be presented as hard gelatin capsules wherein the active ingredients is mixed with an inert solid diluent, for example, calcium carbonate, calcium phosphate or kaolin, or a soft gelatin capsules where in the active ingredient is mixed with water or an oil medium, for example peanut oil, liquid paraffin or olive oil. Aqueous suspensions contain the active material in admixture with excipients suitable for the manufacture of aqueous suspensions. Such excipients are suspending agents, for example, sodium carboxymethylcellulose, methylcellulose, hydroxypropylmethyl-cellulose, sodium alginate, polyvinyl-pyrrolidone, gum tragacanth and gum acacia; dispersing or wetting agents may be a naturally-occurring phosphatide, for example, lecithin, or condensation products of an alkylene oxide with fatty acids, for example polyoxyethylene stearate, or condensation products of ethylene oxide with long chain aliphatic alcohols, for example, heptadecaethylene-oxycetanol, or condensation products of ethylene oxide with partial esters derived from fatty acids and a hexitol such as polyoxyethylene sorbitol monooleate, or condensation products of ethylene oxide with partial esters derived from fatty acids and hexitol anhydrides, for example, polyethylene sorbitan monooleate. The aqueous suspensions may also contain one or more preservatives, for example, ethyl or n-propyl, p- hydroxybenzoate, one or more coloring agents, one or more flavoring agents, and one or more sweetening agents, such as sucrose, saccharin or aspartame.
Oily suspensions may be formulated by suspending the active ingredient in a vegetable oil, for example, arachis oil, olive oil, sesame oil or coconut oil, or in mineral oil such as liquid paraffin. The oily suspensions may contain a thickening agent, for example, beeswax, hard paraffin or cetyl alcohol. Sweetening agents such as those set forth above, and flavoring agents may be added to provide a palatable oral preparation. These compositions may be preserved by the addition of an anti-oxidant such as ascorbic acid.
Dispersible powders and granules suitable for preparation of an aqueous suspension by the addition of water provide the active ingredient in admixture with a dispersing or wetting agent, suspending agent and one or more preservatives. Suitable dispersing or wetting agents and suspending agents are exemplified by those already mentioned above. Additional excipients, e.g., sweetening, flavoring and coloring agents, may also be present.
The pharmaceutical compositions of the invention may also be in the form of an oil-in-water emulsion. The oily phase may be a vegetable oil, e.g., olive oil or arachis oil, or a mineral oil, e.g., liquid paraffin or mixtures of these. Suitable emulsifying agents may be naturally-occurring phosphatides, e.g., soy beans, lecithin, and esters or partial esters derived from fatty acids and hexitol anhydrides, for example, sorbitan monooleate, and condensation products of the said partial esters with ethylene oxide, e.g., polyoxyethylene sorbitan monooleate. The emulsions may also contain sweetening and flavoring agents.
Syrups and elixirs may be formulated with sweetening agents, for example, glycerol, propylene glycol, sorbitol or sucrose. Such formulations may also contain a demulcent, a preservative and flavoring and coloring agents.
The pharmaceutical compositions may be in the form of a sterile injectable aqueous or oleagenous suspension. This suspension may be formulated according to the known art using those suitable dispersing or wetting agents and suspending agents which have been mentioned above. The sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally-acceptable diluent or solvent, e.g., as a solution in 1 ,3-butane diol. Among the acceptable vehicles and solvents that may be employed are water, Ringer's solution and isotonic sodium chloride solution. In addition, sterile fixed oils are conventionally employed as a solvent or suspending medium. For this purpose any bland fixed oil may be employed including synthetic mono- or diglycerides. In addition, fatty acids such as oleic acid find use in the preparation of injectables.
Compounds of the invention may also be administered in the form of suppositories for rectal administration of the drug. The compositions can be prepared by mixing the drug with a suitable non-irritating excipient which is solid at ordinary temperatures but liquid at the rectal temperature and will W 2
99
therefore melt in the rectum to release the drug. Such materials are cocoa butter and polyethylene glycols.
For topical use, creams, ointments, jellies, solutions or suspensions, etc., containing the compounds of the invention are employed. (For purposes of this application, topical application shall include mouthwashes and gargles.)
The compounds for the present invention can be administered in the intranasal form via topical use of suitable intranasal vehicles, or via transdermal routes, using those forms of transdermal skin patches well known to those of ordinary skill in the art. To be administered in the form of a transdermal delivery system, the dosage administration will, of course, be continuous rather than intermittent throughout the dosage regimen. Compounds of the present invention may also be delivered as a suppository employing bases such as cocoa butter, glycerinated gelatin, hydrogenated vegetable oils, mixtures of polyethylene glycols of various molecular weights and fatty acid esters of polyethylene glycol.
The dosage regimen utilizing the compounds of the present invention is selected in accordance with a variety of factors including type, species, weight, sex and medical condition of the patient; the severity of the condition to be treated; the route of administration; the renal and hepatic function of the patient; and the particular compound thereof employed. A physician or veterinarian of ordinary skill can readily determine and prescribe the effective amount of the drug required to prevent, counter, arrest or reverse the progress of the condition. Optimal precision in achieving concentration of drug within the range that yields efficacy without toxicity requires a regimen based on the kinetics of the drug's availability to target sites. This involves a consideration of the distribution, equilibrium, and elimination of a drug. Preferably, doses of the compound of Formula I useful in the method of the present invention range from 0.01 to 1000 mg per day. More preferably, dosages range from 0.1 to 1000 mg/day. Most preferably, dosages range from 0.1 to 500 mg/day. For oral administration, the compositions are preferably provided in the form of tablets containing 0.01 to 1000 milligrams of the active ingredient, particularly 0.01, 0.05, 0.1 , 0.5, 1.0, 2.5, 5.0, 10.0, 15.0, 25.0, 50.0, 100 and 500 milligrams of the active ingredient for the symptomatic adjustment of the dosage to the patient to be treated. An effective amount of the drug is ordinarily supplied at a dosage level of from about 0.0002 mg/kg to about 50 mg/kg of body weight per day. The range is more particularly from about 0.001 mg/kg to 1 mg/kg of body weight per day.
Advantageously, the active agent of the present invention may be administered in a single daily dose, or the total daily dosage may be administered in dividend doses of two, three or four time daily.
The amount of active ingredient that may be combined with the carrier materials to produce single dosage form will vary depending upon the host treated and the particular mode of administration.
It will be understood, however, that the specific dose level for any particular patient will depend upon a variety of factors including the age, body weight, general health, sex, diet, time of administration, route or administration, rate of excretion, drug combination and the severity of the particular disease undergoing therapy.
The compounds of the invention may be produced by processes known to those skilled in the art and as shown in the following reaction schemes and in the preparations and examples described below.
EXAMPLES
The following abbreviations may be used in the procedures and schemes below:
ACN Acetonitrile
AcOH Acetic acid
Aq Aqueous
BOC tert-Butoxycarbonyl
BOC2O BOC Anhydride
C degrees Celsius
CBZCI Benzyl chloroformate
DBU 1 ,8-Diazabicyclo[5.4.0]undec-7-ene
DCM Dichloromethane DEAD Diethyl azodicarboxylate
(DHQ)2PHAL Hydroquinine 1,4-phthalazinediyl diether
DIAD Diisopropylazodicarboxylate
DIPEA Diisopropylethylamine
DMA N, N-Dimethylacetamide
DMAP 4-Dimethylaminopyridine
DME Dimethoxyethane
DMF Dimethylformamide
DMPU 1 ,3-Dimethyl-3,4,5,6-tetrahydro-2(1 h)-pyrimidinone
DMSO Dimethyl sulfoxide
EDCI 1 -(S-DimethylaminopropyO-S-ethylcarbodiimide hydrochloride
El Electron impact eq Equivalents
EtOAc Ethyl acetate
EtOH Ethanol g grams h hours hr hours
1H proton
HATU N, N1N1, N'-Tetramethyl-O-(7-Azabenzotriazol-1 -yl)Uronium hexafluorophosphate
Hex hexanes
HOBT 1 -Hydroxybenzotriazole
HMPA Hexamethyl phosphoramide
HPLC High pressure liquid chromatography
HPLC/MS High pressure liquid chromatography/Mass spectroscopy
LC/MS Liquid chromatorgraphy/mass spectroscopy
LAH Lithium aluminum hydride
LDA Lithium diisopropylamide
M Molar mmol millimoles mCPBA /nefa-Chloroperoxybenzoic acid Me Methyl
MeCN Acetonitrile
MeOH Methanol min Minutes mg Milligrams
MHz Megahertz mL Milliliter
MPLC Medium Pressure Liquid Chromatography
NMR Nuclear Magnetic Resonance
MS Mass Spectroscopy
NBS N-Bromosuccinimide
NMM N-Methylmorpholine
NMP 1 -methyl-2-pyrrolidone
ON Overnight
PCC Pyridinium Chlorochromate
PTLC Preparative thin layer chromatography
PyBrOP Bromo-tris-pyrrolidino-phosphonium hexafluorophosphate
Pyr Pyridine
RT Room temperature
SEM 2-(Trimethylsilyl)-ethoxymethyl sgc Silica gel 60 chromatography tBOC tert-Butoxycarbonyl
TACE Tumor Necrosis Factor-alpha converting enzyme
TEA Triethylamine
TFA Trifluoroacetic acid
THF Tetrahydrofuran
TLC Thin layer chromatography
TR Retention time
X-PHOS 2-dicyclohexylphosphino-2\ 4\ 6'-tri-isopropyl-1 \ 1 -biphenyl
SYNTHETIC ROUTES AND EXAMPLES
Example 1 -
Figure imgf000104_0001
General procedures for Example 1:
In step 1 , Compound 1A (either commercially available, or prepared by a procedure similar to that described by Abdalla, G. M. and Sowell, J. W. Journal of Heterocyclic Chemistry, 1987, 24(2), 297-301 ) was treated with one equivalent of Di-tert-butyl dicarbonate in polar solvent, such as DMF, for 30 minutes to 12 hours. The solvent was removed and compound 1B could be used without further purification or purified by silica gel chromatography.
In step 2, compound 1B was reacted with potassium cyanide and ammonium carbonate in an alcohol and water solution, at 500C to 90 0C, for 5 hours to 48 hours. After cooling down, water was added and compound 1C could be collected by filtration.
In step 3, compound 1C was stirred with 2 to 20 equivalents of hydrogen chloride in methanol for 5 to 48 hours. After ethyl ether was added, compound 1D could be collected by filtration. Example 2
Figure imgf000104_0002
Step i
Compound 2A (Abdalla, G. M. and Sowell, J. W. Journal of Heterocyclic Chemistry, 1987, 24(2), 297-301 ) (Hydrochloride salt, 8.6Og, 45.4 mmol), triethyl amine (19.0 ml_, 136 mmol), and di-tert-butyl dicarbonate (11.9g, 54.4 mmol) were stirred in methylene chloride (100 mL) at 25 0C for 16 hours. Saturated aqueous NaHCC« 3 (150 mL) was added. The aqueous layer was extracted with CH2CI2 (100 ml.) twice. The organic phase was washed with brine (100 ml_) and dried over Na2SO4. The solvent was removed by rotary evaporator to give compound 2B which was used without further purification.
Step 2
Compound 2B (9.06 g, 35.8 mmol), KCN (3.49 g, 53.7 mmol), and (NHU)2CO3 (12.0 g, 125.2 mmol) were suspended in a mixture of EtOH (35 mL) and water (35 mL). The solution was stirred at 70 0C for three days. After cooling down, water (35 mL) was added. The solid was filtered and washed with water three times. The solid was dried under vacuum at 40 0C for 16 hours to give compound 2C (7.9 g, 68%).
Step 3
Compound 2C (4.0 g) was suspended in methanol (50 mL) and HCI (4M in dioxane, 20 mL) was added. The solution was stirred at 25 0C for 3 hours.
Ethyl ether (50 ml) was added. The solid was filtered, washed with ethyl ether twice, and dried under vacuum for 12 hours to give compound 2D (2.7 g,
84%).
Example 3
Figure imgf000105_0001
3C
Step 1
Compound 3A (prepared according to the procedure described by Wyrick, S. D. et al. Journal of Medicinal Chemistry, 1987, 30(70), 1798-806) (3.33 g, 18.5 mmol) was dissolved in dry benzene (40 mL). NBS (3.45 g, 19.4 mmol) and benzoyl peroxide (134 mg, 0.55 mmol) were added. The solution was stirred in a 75 0C oil bath for about 2 hours. After cooling down, the solid was filtered and washed with Et2O (150 ml_). The organic solution was then washed with water (50 mL) twice, dried over Na2SO4 or MgSO4, filtered, and concentrated by rotary evaporator. The crude product was dried under vacuum to give compound 3B which was used without further purification. 1H- NMR appeared to indicate that approximately 75% of this material was compound 3B.
Step 2
Compound 3B (4.62 mmol), Compound 3C (824 mg, 4.62 mmol), and K2CO3 (1.28 g, 9.24 mmol) were mixed in DMF (30 mL). The solution was stirred at room temperature for 20 hours. DMF (15 mL) was added and the solid was filtered and washed with DMF. All the DMF solution was combined and concentrated to 25 mL. The resulting solution was purified via reverse phase MPLC (CH3CN/water, 5% to 90%, containing 0.1%HCO2H) to give compound 3D (198 mg, 15%).
Example 4
Figure imgf000106_0001
Step i
Compound 4A (2Og, 81.61 mmol), 4B (13.36 mL, 97.93 mmol), Pd(dppf)CI2 (1.Og, 1.36 mmol), dioxane (350 mL), water (50 mL), and Cs2CO3 (22.5g, 163 mmol) were stirred at 110 0C (oil bath) under nitrogen for 16 hours. After cooling, the solid was removed by filtration. The solution was concentrated and purified by sgc (Hexane/EtOAc, 10:1) to give 4C (12.1g, 80%).
Step 2 Compound 4C was converted to Compound 4D using a procedure similar to that described in Example 3.
Example 5
Figure imgf000107_0001
Compound 5A was prepared using chemistry similar to that described in Examples 1 , 2, 3 and 4.
Stepi
Compound 5A (1.18 g, 3.36mmol) and pyridine hydrochloride (2.33 g, 20.17 mmol) were added into a 20 mL microwave reactor tube and reacted at 200 0C for 1 hour. After cooling down, the solid was dissolved in DMF and purified by reverse phase chromatography on a C-18 cartridge (CH3CN/water 5% to 90%, with 0.1 % HCO2H) to give compound 5B (0.87 g, 77%).
Step 2
Compound 5B (0.75 g, 2.22 mmol) was dissolved in DMF (12 mL). SEMCI (0.48 mL, 2.44 mmol) and DIPEA (0.775 mL, 4.44 mmol) were added and the solution was stirred at 25 0C for 4 hours. DMF was removed under vacuum and the product was purified by sgc (Hexane/EtOAc: 3:1 to 1 :1) to give compound 5C (0.81 g, 78%).
Example 6
Figure imgf000108_0001
Compound 6A was prepared using chemistry similar to that described in Examples 1 , 2, 34, and 5.
Step i
Compound 6A was resolved by Chiralcel OD column (Mobile phase: Hexane:2-propanol 4:1). The first peak was collected and concentrated to give compound 6B.
Step 2
Compound 6B (0.2 g, 0.36 mmol), allyl palladium chloride dimer (3 mg, 0.008 mmol), and DMF (3 mL) were added to a round bottomed flask and cycled between vacuum and nitrogen three times. Tri-tert-butyl phosphine ( 30 microliters of 10% solution in hexanes-Strem), piperidine (61 mg, 0.7 mmol), and 3-dimethylamino-1-propyne were added via syringe. The reaction was left stirring overnight at rt. The following morning, the reaction was stirred for 1 hr at 50 0C. The resulting material was diluted with EtOAc, washed with water, dried with MgSO4, and concentrated to dryness. The crude product was purified via flash silica gel chromatography using a gradient elution of 70% (5% Methanol in EtOAC) in hexanes to 100% (5% Methanol in EtOAc) giving 55 mg of compound 6C.
Step 3
Compound 6C (55 mg) was dissolved in 10 ml_ of 4 M HCI in dioxane (Aldrich) and 10 ml of methanol and the solution was added to a pressure tube. The tube was capped and heated to 900C. The reaction mixture was stirred at 90 0C for 4 hr, then allowed to cool to room temperature. The reaction mixture was concentrated to dryness. Methanol was added and the reaction mixture was reconcentrated to dryness. Methanol (5 ml) and triethylamine (1mL) were added and the reaction mixture was stirred at rt for 2 hr. The resulting solution was concentrated to dryness. The crude product was purified on a C-18 lsco cartridge using an (acetonitrile:water(+0.1% formic acid)) gradient as the mobile phase to give 6D.
Chemistry similar to the procedures described in Examples 6, 8, 1 , 2, 3, 4, and 5 was used to prepare compounds 18, 19, 20, 21, and 22 in Table 1.
Example 7
Figure imgf000109_0001
Step 1
Compound 4D (16.26 g, 62.77 mmol) was dissolved in 100 mL of DMF and glycine ethyl ester hydrochloride (9.68 g, 69.35 mmol) was added. Diisopropylethylamine (21 mL, 15.6 mmol) was added and the reaction mixture was placed in a 70 0C oil bath. The reaction mixture was stirred overnight at 700C. After 17 hr, the reaction mixture was allowed to cool to rt. EtOAc, water, and 5 mL of 1 M aq NaHSO4 were added and the layers were separated. The organic layer was washed with water and brine, dried with MgSO4, filtered and concentrated to an orange oil (14.8 g). The crude product was purified via flash silica gel chromatoghraphy using a 20% to 60% EtOAc: Hexanes gradient as the mobile phase to give 6.8 g of 7A as product.
Step 2
Compound 7A (6.31 g, 25.3 mmol) was dissolved in dioxane (88 mL). A 1.0 M aq soln of LiOH (28 mL, 28 mmol) was added followed by 10 mL of absolute ethanol. The reaction mixture was stirred at rt for 3 hr, then partially concentrated on the rotary evaporator. Dichloromethane and 1 M aq NaHSO4 were added and the layers were separated. The aq layer was extracted with CH2CI2. The combined organic layer was filtered, dried with MgSO4, filtered again, and concentrated to dryness giving 4.04 g of 7B.
Step 3
Compound 7B (2.02 g, 9.13 mmol) was suspended in 15 mL of THF. Carbonyl diimidazole was added in one portion. After 10 min, acetonitrile (10 mL) was also added. The reaction mixture was stirred at rt for 1 hr. Magnesium chloride and ethyl potassium malonate were added. The reaction mixture was left stirring overnight at room temperature under a drying tube. The reaction mixture was concentrated to near dryness. EtOAc and 1.0 M pH 5.5 sodium phosphate buffer were added. The layers were separated. The organic layer was washed with water and brine, dried with MgSO4, filtered, and concentrated to dryness. An off white solid was obtained. The crude product was purified via silica gel chromatoghraphy using a 50% to 100% EtOAc: Hexanes gradient as the mobile phase to give 1.87 g of 7C.
Step 4 Compound 7C was dissolved in 18 ml_ of absolute ethanol and 8 ml_ of water. The solution was added to a thick walled glass pressure bottle and ammonium carbonate (2.21 g, 23.0 mniol) was added. The bottle was capped and the reaction mixture was stirred at rt for 15 min. Potassium cyanide was added, the bottle was recapped, and the reaction mixture was stirred at 700C for 16 hr. The resulting mixture was poured into 250 mL of water and suction filtered to give 7D (1.86 g) as a white solid.
Step 5
Compound 7D (0.83 g, 2.29 mmol) was suspended in 9 mL of dioxane. Aq 1.0 M LiOH (4.6 mL, 4.6 mmol) was added, causing the material to dissolve. The reaction mixture was stirred at rt for 5.5 hr. Additional LiOH was added (1.0 mL. 1.0 mmol) and the reaction mixture was stirred for 1 hr. The reaction mixture was concentrated to near dryness. The resulting mixture was acidified with aq 1 M NaHSO4 causing a precipitate to form. The flask was placed in an ice water bath and stirred for 30 min. The resulting mixture was suction filtered to give 7E (0.70 g) as a white solid.
Step 6
Compound 7E (31 mg, 0.093 mmol) was dissolved in DMF (400 μL). Carbonyl diimidazole (18 mg, 0.11 mmole) was added and the reaction mixture was stirred at rt for 30 min. Pyrrolidine (20 μL) was added and the reaction mixture was stirred at rt for 5 hr. Aq 1 M NaHSO4 was added (7 mL) followed by EtOAc. The layers were separated. The organic layer was dried with MgSO4, filtered, and concentrated to dryness, the crude product was purified via reverse phase (C-18 lsco cartridge) chromatography using a 10% to 60% acetonitrilerwater + (0.1% formic acid) gradient as the mobile phase. A white solid was obtained as product giving compound 5 in Table 1, which is one embodiment of the class of compounds 7F.
The procedures described in Example 7 were used to prepare compounds 1 through 17 in Table 1. Example 8
Figure imgf000112_0001
Compound 8A was prepared using chemistry similar to that described in Examples 1 , 2, 3, 4 and 5.
Step l
Compound 8A was resolved by Chiralcel OD column (Mobile phase: Hexane:2-propanol 4:1). The first peak was collected and concentrated to give compound 8B.
Example 9:
Figure imgf000112_0002
9A 9B 9C
Racemic compound 9A was prepared using chemistry similar to that described in Examples 1 , 2, 3, 4 and 5. The enantiomers were resolved by Chiralcel OD column (Mobile phase: Hexane/2-propanol 3:1). The first peak was collected and concentrated to give compound 9A in its enantiomerically pure form.
Step l
To a dry flask was added compound 9A (1.5 g, 2.73 mmol) and 4-pyridyl boronic acid (670 mg, 5.50 mmol). The flask was subjected to vacuum and refilled with nitrogen three times. Pd(dppf)Cl2 (220 mg, 0.30 mmol) was added and followed by addition of CH3CN (20 ml_) and aq. K2CO3 (1 M, 15 ml_). The solution was stirred at 80 0C (oil bath) for 16 hours. After cooling down, CH3CN (100 ml_) was added and the solid was removed by filtration. The aqueous layer was separated and extracted with EtOAc (20 ml_) once. The organic solution was combined and concentrated. The product was purified by SGC (CH2CI2/MeOH/NH4OH: 20:1 :0.1 ) to give compound 9B.
Step 2
Compound 9B was dissolved in a mixture of methanol and HCl (4M in dioxane) (2:1, 30 ml_) and was stirred overnight in a sealed pressure flask at 90 0C (oil bath). After the solution was cooled, the solution was transferred, into a 250 mL round bottom flask. It was concentrated and dried under vacuum. The crude mixture was dissolved in methanol (50 mL) and Et3N (0.5 mL) was added and stirred overnight at 250C. The solvent was then removed and the product was purified by C18 reverse phase chromatography (CH3CN/water 5% to 90%, with addition of 0.1% HCO2H) to give compound 9C (815 mg).
Chemistry similar to that described in Examples 8 and 9 was used to prepare compound 24 in Table 1.
Figure imgf000113_0001
Step i
A mixture of compound 9A (0.3 g, 0.55 mmol), bis(pinacolato)diboroπ (10A; 170 mg, 0.65 mmol), potassium acetate (170 mg, 1.70 mmol), and [PdCI2(dppf)]CH2CI2 (50 mg, 0.05 mmol) in 1,4-dioxane (10 mL) was cycled between vacuum and argon three times. The reaction mixture was stirred at 1000C (oil bath) for 1.5 hours. After cooling down, the mixture was diluted in EtOAc (50 ml_) and filtered through a Celite pad. The filtrate was concentrated in vacuo and the residual material was purified by silica gel column chromatography (2% MeOH in CH2CI2) to afford compound 1OB (300 mg, 91% yield).
Step 2
A solution of compound 1OB (60 mg, 0.10 mmol), 3-bromoimidazo[1 ,2- a]pyridine (30 mg, 0.15 mmol), and [PdCI2(dppf)]CH2CI2 (8.2 mg, 0.01 mmol) in CH3CN (3 ml_) was treated with potassium carbonate (0.6 mL, 0.6 mmol, 1M in H2O). The mixture was subjected to vacuum and refilled with argon three times. The reaction mixture was stirred at 90 0C (oil bath) for 17 hours. After cooling, the mixture was diluted in EtOAc (20 mL) and filtered through a Celite pad. The filtrate was concentrated in vacuo and the residual material was purified by preparative TLC (10% MeOH in CH2CI2) to afford compound 10C (42 mg, 71% yield).
Figure imgf000114_0001
HA 23
Compound 11 A was prepared via procedure similar to those described in Examples 8 and 10.
Step i W 2
114
Compounds 11 A (53 mg) was dissolved in 2 ml. methanol in a 15 mL pressure tube. HCI (4M in dioxane, 1 mL) was added. The tube was sealed and put into an 800C oil bath for 16 h. After cooling down, the solution was transferred into a 100 mL flask and the solvent was removed. NH3 (7N in methanol, 3 mL) was added and the solution was transferred into a 15 mL pressure tube. The tube was sealed and put into a 70 0C oil bath for three hours. After cooling down, the solution was transferred into 100 mL flask and the solvent was removed. The product was purified by C18 reverse phase chromatography ( CH3CN/water, 5% to 90%, with 0.1% HCO2H) to give compound 23. Compound 23 was dissolved in methanol, and HCI (4M in dioxane, 0.5 mL) was added. The solution was stirred at 250C for 30 minutes. The solvent was removed and the product was suspended in water. The water was removed by lyophilizer to give compound 23 (22 mg) in Table 1.
Figure imgf000115_0001
Amine hydrochloride 2D (1.13g, 3.75 mmol) in DMF (30 mL) was treated with thiophenedialdehyde (0.3g, 1.9 mmol) and the mixture was stirred at ambient temperature for 1 h. The reaction was diluted with ethyl acetate (100 mL) and the organics were washed with water (3 X 50 mL) and brine (1 X 50 mL), dried over MgSO4 and concentrated to provide a crude oil. The crude product was subjected to silica-gel chromatography using CH2CI2-5% CH3OH/CH2CI2 as the gradient eluting solvent to yield 26 (0.08 g).
Figure imgf000116_0001
Amine hydrochloride 2D (2.7 g, 8 mmol) in DMF (30 ml_) was treated with bromoethylester 27A (3.6 g, 15 mmol) and diisopropylethylamine ( 2.6 ml_). The mixture was heated to 550C and stirred for 24 h. The reaction was diluted with ethyl acetate (200 ml_) and the organics were washed with water (3 X 50 mL) and brine (1 X 50 mL), dried over MgSO4 and concentrated to provide crude solid. The product 27 (1.8 g) was isolated by recrystallization (ethyl acetate : ether; 2:1). The product 27A was prepared from commercially available (Aldrich) ethyl nicitinate using the process described in step 1 example 3.
Compound #25 (Table 1 ) was prepared analogously using appropriate starting materials.
Example 300A
Figure imgf000116_0002
Part A:
Compound 300 (20.0 g, 81.61 mmol), trimethylboroxine (13.36 mL, 97.93 mmol), Pd(dppf)CI2 (1-0 g, 1.36 mmol), dioxane (350 mL), water (50 mL), and cesium carbonate (22.5 g, 163 mmol) were stirred at 110 0C (oil bath) under nitrogen for 16 hours. After cooling, the solid was removed by filtration. The solution was concentrated and purified by sgc (10:1 EtOAc/hexanes) to give 301 (12.1 g, 80%).
Part B:
Compound 301 (4.4 g, 24.2 mmol) was dissolved in carbon tetrachloride (80 mL) and N-bromosuccinimide (4.48 g, 24.2 mmol) and benzoyl peroxide (276 mg, 1.13 mmol) were added. The reaction mixture was stirred at reflux for 3 hours and then solids were filtered and washed with ether. The combined organic layers were washed with water, dried over sodium sulfate, and concentrated to provide the desired product 302 (6.1 g, 98%).
Part C:
Compound 302 (32.0 g, 124.0 mmol) was dissolved in 7 M ammonia in MeOH (150 mL) and stirred in a sealed pressure flask at 60 0C overnight. The reaction mixture was cooled and the solvent was removed under reduced pressure. The residue was suspended in ethyl acetate and stirred for 30 minutes. The solids were filtered and dissolved in methylene chloride. The methylene chloride was washed with water, dried over sodium sulfate, and concentrated to provide the desired product 303 (13.5 g, 67%).
Part D:
Compound 303 (2.2 g, 13.4 mmol) was dissolved in THF (250 mL) and DMPU (40 mL). Sodium t-butoxide (1.55 g, 16.13 mmol) was added and stirred for 5 hours. Chloromethylpivalate (3.0 mL, 20.1 mmol) was added dropwise and stirred overnight. The reaction was quenched with saturated ammonium chloride and extracted with ethyl acetate. The combined ethyl acetate layers were washed with water, brine, dried over sodium sulfate and concentrated. Purification by column chromatography (SiO2, 25% ethyl acetate/hexanes) afforded the desired product 304 (2.5 g, 67%).
Part E: Compound 304 (288 mg, 1.04 mmol) was dissolved in methylene chloride (5 ml_) and cooled in an ice bath. Bromotrimethylsilane (0.3 mL, 2.08 mmol) was added dropwise and stirred in the ice bath for 30 minutes followed by 2 hours at room temperature. The reaction mixture was concentrated and re- dissotved in methylene chloride (2 mL). Hexanes (8 mL) was added and the solids were filtered to provide the desired product 305 (218 mg, 83%).
Example 400:
Figure imgf000118_0001
Part A:
Glyoxylic acid monohydrate (20.0 g, 218 mmol) and methyl carbamate (16.3 g, 218 mmol) were dissolved in diethyl ether (200 mL) and stirred overnight. The solids were filtered to provide the desired product 306B (32.0 g, 98%).
Part B:
Compound 306B (32.0 g, 214 mmol) was dissolved in MeOH (200 mL) and cooled in an ice bath. Concentrated sulfuric acid (8 mL) was added dropwise and the reaction was stirred overnight. The reaction mixture was diluted with ethyl acetate and water. The organic layer was washed with brine, dried over sodium sulfate, and concentrated to provide compound 306C that was used without purification (27.0 g, 71 %).
Part C:
Compound 306C (27.0 g, 152 mmol) was dissolved in carbon tetrachloride (700 ml_). Phosphorus pentachloride (50 g, 240 mmol) was added and the suspension was stirred for 18 hours (solution became clear over time). The solvent was removed under reduced pressure and the residue was stirred in petroleum ether (500 mL) overnight. The solids were filtered to provide compound 307 with no need for purification (26.5 g, 96%). Trituration step was repeated if mass yield was too high.
Part D:
Compound 307 (15.0 g, 82.7 mmol) was dissolved in methylene chloride (140 mL) and cooled in an ice bath. Bis(trimethylsilyl)acetylene (15.0 g, 88.2 mmol) was added in methylene chloride (20 mL). Freshly crushed aluminum chloride (11.0 g, 82.7 mmol) was added in portions over 20 minutes. The reaction mixture was allowed to slowly warm to room temperature and stirred overnight. The reaction was cooled in an ice bath and slowly quenched with water. The organic layer was washed several times with water, dried over sodium sulfate, and concentrated. The residue was triturated/recrystallized from hexanes to provide the desired product 308 (14.8 g, 69%). HPLC-MS tR = 1.84 min (ELSD); mass calculated for formula Ci0Hi7NO4Si 243.09, observed LCMS m/z 244.1 (M+H).
Part E:
Compound 308 (24.0 g, 98.7 mmol) and compound 305 (25.1 g, 99.0 mmol) were dissolved in THF (300 mL) and cooled to -78°C. A 1M solution of LiHMDS (198 mL, 198 mmol) was added dropwise over 30 minutes and the reaction mixture was stirred for 2 hours. Saturated ammonium chloride solution was added slowly and the reaction was allowed to warm to room temperature. The aqueous layer was extracted with ethyl acetate. The combined organic layers were washed with water, brine, dried over sodium sulfate, and concentrated. Purification by column chromatography (Siθ2, 33% ethyl acetate/hexanes to 50% ethyl acetate/hexanes) afforded the desired product 309 (26.0 g, 63%). HPLC-MS tR = 1.90 min (UV254 nm); mass calculated for formula C2OH2BN2O6Si 418.15, observed LCMS m/z 419.2 (M+H).
Part F:
The two isomers were separated using a chiral OD column. One gram of material was injected into the column and the two peaks were separated by using a solvent mixture of 85% hexanes/ethanol. The second isomer was the desired compound 309B (400 mg, 80%).
Part G:
Compound 309B (8.0 g, 19.1 mmol) was dissolved in THF (250 mL) and cooled to O0C. Tetrabutylammonium fluoride (1 M in THF, 22.9 mL, 22.9 mmol) was added dropwise and the reaction was stirred for 1 hour at room temperature. The reaction mixture was diluted with ethyl acetate and water. The organic layer was washed with water, saturated sodium bicarbonate, brine, dried over sodium sulfate and concentrated to provide compound 400 (5.8 g, 88%). The product was used without purification.
Part H:
Compound 400 (75 mg, 0.22 mmol) was combined with 3-bromoquinoline (0.032 mL, 0.24 mmol), Pd(PPh3J2CI2 (3 mg, 0.0044 mmol), CuI (2 mg, 0.009 mmol), diisopropylamine (0.062 mL, 0.44 mmol) in DMF (1 mL) and stirred overnight at 80 0C. The reaction mixture was diluted with ethyl acetate and water. The organic layer was washed with water, brine, dried over sodium sulfate and concentrated. Purification by column chromatography (SiO2, 50% ethyl acetate/hexane to 80% ethyl acetate/hexane) afforded the desired product 400A (93 mg, 89%). HPLC-MS tR = 1.66 min (UV254 nm); mass calculated for formula C26H23N3O6 473.16, observed LCMS m/z 474.1 (M+H). Part i:
Compound 400A (77 mg, 0.16 mmol) was dissolved in 7 M ammonia solution (3 mL) and stirred in a sealed pressure tube at 90 0C overnight. The reaction mixture was coofed to room temperature and concentrated to afford compound 400B. HPLC-MS tR = 1.41 min (UV254 nm); mass calculated for formula C24H18N4O4426.13, observed LCMS m/z 427.0 (M+H).
Example 300E
Figure imgf000121_0001
2D
Part A:
Sodium pellets (3.6 g, 156 mmol) were dissolved in MeOH (100 mL) at 0 0C. Compound 320 (3.0 g, 15.6 mmol) was added and stirred at 1000C in a sealed pressure flask overnight. The reaction was cooled to room temperature and diluted with ethyl acetate and 1N HCI. The organic layer was dried over sodium sulfate and concentrated to provide the desired product 321 with no need for purification (2.1 g, 72%).
Part B: Compound 321 (2.1 g, 11.1 mmol) was dissolved in toluene (30 mL) and methanol (30 ml) and cooled in an ice bath. TMS diazomethane (2M in hexanes, 11 mL) was added dropwise until yellow color persisted. The solvent was evaporated under reduced pressure to provide the desired product 322 with no need for purification (2.2 g, quant.).
Part C:
Compound 322 (1.0 g, 5.0 mmol) was combined with Pd(P-tBu3)2 (128 mg, 0.25 mmol), Pd(dba)3 (250 mg, 0.25 mmol), trimethylboroxine (1.0 mL, 6.5 mmol), potassium phosphate monohydrate (3.69 g, 15 mmol) in dioxane (25 mL) and stirred at 90 0C overnight. The reaction mixture was filtered and the solvent was evaporated under reduced pressure. Purification by column chromatography (Siθ2, 10% ethyl acetate/hexanes) afforded the desired product 323 (0.500 g, 55%).
Part D:
Compound 323 (210 mg, 1.16 mmol) was dissolved in carbon tetrachloride (6 mL) and N-bromosuccinimide (228 mg, 1.28 mmol) and benzoyl peroxide (10 mg) were added. The reaction mixture was stirred at reflux overnight, cooled to room temperature, and filtered (solids were washed with ether). The combined organic layers was washed with water, dried over sodium sulfate, and concentrated to provide the desired product 324 (0.20 g, 67%).
Part E:
Compound 324 (75 mg, 0.29 mmol) and compound 2D (75 mg, 0.29 mmol) were dissolved in DMF (5 mL) and DIEA (0.15 mL, 0.87 mmol) and stirred at 70 0C overnight. The reaction mixture was diluted with ethyl acetate and water. The organic layer was washed with water, brine, dried over sodium sulfate, and concentrated. The residue was purified by reverse phase chromatography to provide the desired product 325 (24.1 mg, 22%). HPLC- MS tR = 1.269 min (UV254 nm); mass calculated for formula Ci8Hi5N4O4F 370.10, observed LCMS m/z 371.1 (M+H). Example 300F
Figure imgf000123_0001
Part A:
Compound 325 (140 mg, 0.378 mmol), chlorotrimethylsilane (226 mg, 1.89 mmol), and sodium iodide (283 mg, 1.89 mmol) were dissolved in acetonitrile (5 ml_) and stirred at reflux for 10 minutes. Water (0.3 ml_) was added and the reaction was refluxed for 3 hours. The reaction mixture was cooled and diluted with ethyl acetate and water. The organic layer was dried over sodium sulfate and concentrated. Purification by reverse phase chromatography provided the desired product 326 (7.1 mg, 5%). HPLC-MS tR = 0.855 min (UV254 nm); mass calculated for formula C17H13N4O4F 356.09, observed LCMS m/z 357.1 (M+H).
Example 82
Figure imgf000123_0002
The aryl ether compounds 82 to 90 were prepared from compound 8B using a procedure based on that described by E. Buck and 2. J. Song in Organic Synthesis VoI 82, p. 69, followed by a standard SEM deprotection sequence. An example is provided below.
Compound 8B (0.248 g, 0.442 mmol), 5-Fluoro-2-hydroxypyridine (128 mg, 1.13 mmol), cesium carbonate (374 mg, 1.14 mmol), and copper (I) chloride (48 mg, 0.48 mmol) were added to a 10 mL Schlenck tube equipped with a stir bar. The tube was capped with a septum and cycled between vacuum and N2 three times. N-methyl-2-pyrrolidinone (2mL) was added via syringe and the Schlenck tube was cycled between vacuum and N2 three times. 2,2,6,6,6-Tetramethyl heptane-3,5-dione (33 μL) was added via syringe. The Schlenck tube was placed in a 1000C oil bath and heated to 1500C. The reaction mixture was stirred for 23 h at 1500C. The reaction mixture was allowed to cool to rt, then diluted with EtOAc and water. Aqueous 1% EDTA was added and the layers were separated. The organic layer was washed with 1% aq EDTA, water, and brine. The resulting organic solution was dried with MgSO4, filtered, and concentrated to dryness. A brown solid was obtained. The crude product was purified via sgc using a Biotage SiO2 cartridge and a 1%-2.5% MeOH/CH2CI2 gradient as the mobile phase. The major spot was collected as product, giving 0.04 g of compound 8C.
Compound 8C (0.04g) was dissolved in (10 mL) anhydrous acetonitrile and concentrated to dryness on the rotovap. This step was repeated. The compound was redissolved in anhydrous acetonitrile (3 mL) and placed under N2. The flask was cooled in an ice water bath. BF3 etherate (90μL) was added, the ice bath was removed, and the reaction mixture was stirred at rt for 7 h. The reaction mixture was capped and stored in a 4°C freezer overnight. The reaction mixture was cooled in an ice-water bath. Diisopropylethylamine (1.5 mL) was added, followed by aq 3.0 M sodium hydroxide. The reaction was stirred for 15 min. The ice bath was removed, and the reaction mixture was stirred for 3 h at rt. Acetic acid was added until the reaction mixture was weakly acidic. The reaction mixture was partially concentrated on the rotovap. EtOAc and water were added. The layers were separated. The organic layer was washed with water and brine, dried with MgSCU, filtered, and concentrated to dryness. The crude product was purified via reverse phase chromatography using an lsco C-18 cartridge (43g). The mobile phase was a 15% to 80% CH3CN/H2O gradient with 0.1 % (volume) formic acid added to both components of the mobile phase. The main peak was isolated as product giving compound 8D.
Figure imgf000125_0001
Compound 8B (1.50 g, 2.68 mmol); pinocolatodiboron (816 mg, 3.21 mmol), potassium acetate (785 mg, 8.0 mmol), and palladium (II) dichloride(dppf) CH2CI2 complex (250 mg, 0.306 mmol) were added to a 100 ml_ Schlenck flask equipped with a stir bar. The flask was caped with a septum, then cycled between vacuum and nitrogen four times. Dioxane (20 mL, Aldrich anhydrous) was added via syringe. The flask was cycled between vacuum and nitrogen three times, then placed in an 85CC oil bath. The bath was heated to 1000C1 then stirred for 1.5 h. the reaction mixture was allowed to cool to RT and diluted with EtOAc (80 mL). The resulting mixture was filtered through Celite. The Celite was rinsed with additional EtOAc. The combined filtrate was concentrated to near dryness then redissolved in EtOAc. The organic solution was washed with 1.0 M aq pH 7 sodium phosphate buffer, water, and brine. After drying with MgSO4, the organic layer was concentrated to dryness. The crude product was purified via sgc using a 2%- 4% MeOH/ CH2CI2 gradient as the mobile phase. A brown solid was obtained (1.9 g). The solid was dissolved in dioxane (16 mL) and water (11 mL) was added. Sodium perborate (3.0 g, 19.5 mmol) was added and the reaction mixture was stirred at rt overnight. The reaction mixture was diluted with EtOAc and aq 1M NH4CI. The layers were separated. The organic layer was washed with water and brine, dried with MgSO4, filtered and concentrated to dryness, giving an off white solid (1.37 g). SGC using a gradient of 25% to 100% (5%Methanol in EtOAc)/Hexanes as the mobile phase gave 0.25 g of pure 93A and 0.62 g of impure 93A.
Compound 93A (0.70 g, 1.40 mmol) was dissolved in 50 ml_ of Aldrich 4N HCI in dioxane and 50 mL of methanol. The solution was added to a pressure tube equipped with a stir bar. The tube was capped, placed in an oil bath, and heated to 95 C. The reaction was stirred at 95°C for 4 h, then allowed to cool to rt. The reaction mixture was concentrated to dryness. Methanol was added and the reaction mixture was reconcentrated. Methanol (50 mL) was added, followed by triethylamine (5 mL). The reaction mixture was stirred at rt for 1 h, then concentrated to dryness. EtOAc and 1.0 M aq pH 5.5 sodium phosphate buffer were added. The layers were separated. The organic layer was washed with water and brine, dried with MgSO4, filtered, and concentrated to dryness. The crude product was purified via S1O2 chromatography. The mobile phase was a gradient of 10% to 100% of (100:10:1-CH2CI2:MeOH:concentrated NH4OH) in CH2CI2.. The main UV active peak was isolated as product giving 0.42 g of compound 93B as white solid.
Example 93
Figure imgf000126_0001
Compound 93A (0.05 g, 0.10 mmol) was dissolved in CH2CI2 (5mL). N, N- Dimethylcarbamyl chloride (18 μL) and DMAP (8mg) were added and the reaction mixture was stirred at rt overnight. The reaction mixture was diluted with CH2CI2 and washed with 1.0 M aqueous pH 7.0 sodium phosphate buffer, water, and brine. The organic layer was dried with MgSCM, filtered, and concentrated to dryness. The crude product was purified via sgc using a 0.5% to 5% MeOH/CH2CI2 gradient on a 40 g lsco SiO2 Cartridge. The major UV active peak was isolated as compound 93C.
Compound 93C was converted to compound 93 using SEM deprotection procedures similar to those described previously. Example 401 :
Figure imgf000127_0001
Part A:
Compound 309B (1.26 g, 3.0 mmol) in 7 M ammonia in methanol (20 ml_) was heated to 85 0C in a pressure bottle overnight. The reaction mixture was concentrated to afford 401 (900 mg, 100%) which was used without further purification. HPLC-MS tR = 1.00 min (UV254 nm); mass calculated for formula Ci5Hi3N3O4 299.09, observed LCMS m/z 300.1 (M+H).
Example 45
CuI, DIPA
Figure imgf000127_0002
Figure imgf000127_0003
StepL Compound 401 (100 mg, 0.33 mmol) was combined with compound 45A (80 mg, 0.4 mmol), Pd(PPh3J2CI2 (8 mg, 0.012 mmol), CuI (17 mg, 0.1 mmol), diisopropylamine (0.08 mL, 0.58 mmol) in DMF (1 mL) and stirred at 85°C for 2 h. The reaction mixture was purified on a Gilson reverse phase HPLC (0-40% acetonitrile in H2O with formic acid 0.1%) afforded the desired product 45B(18 mg, 13%).
Step 2. Compound 45B (20 mg, 0.23 mmol) was stirred in MeOH (5 mL) and HCI (1N, aq., cat.)- The reaction was stirred at rt for 2 h. Solvent was removed and the crude material was purified on a Gilson reverse phase HPLC (0-50% acetonitrile in H2O with 0.1% formic acid) afforded the desired product 45 (20 mg, 99%).
Example 48
Figure imgf000128_0001
48D 48
Step i
A mixture of 48A (161 mg, 0.86 mmol), SEMCI (0.17 mL, 0.94 mmol), and diisopropylethylamine (0.22 mL, 1.28 mmol) in CH2CI2 (3 mL) was stirred at 250C for 2 h. The mixture was added to an aqueous NaHCO3 solution and the organic layers were extracted with CH2CI2. The combined organic solution was washed with brine solution, dried (Na2SO4), and concentrated in vacuo. The residue was purified by SiO2 column chromatography (CH2CI2/hexane = 2:1). to afford 48B (200 mg, 74% yield).
Step 2
A mixture of 401 (100 mg, 0.33 mmol), 48B (165 mg, 0.52 mmol), Pd(PPh3J2CI2 (4.9 mg, 7 Dmol), CuI (1.9 mg, 10 Dmol), and diisopropylethylamine (0.17 ml_, 0.99 mmol) in DMF (1.5 ml) was purged with N2 and heated to 70 0C. After heating for 17 h, the mixture was cooled to 25 0C and purified by column chromatography on a reverse phase C-18 column (0.01%HCO2H in water/0.01% HCO2H in CH3CN) to afford 48C (78 mg, 44% yield).
Step 3
48C (78 mg, 0.14 mmol) was dissolved in MeOH (15 mL) and treated with 4 N HCI in dioxane (3 mL). The mixture was heated to 60 0C in a pressure vessel for 16 h and cooled to 25 0C. The mixture was neutralized with NH3-MeOH (7 N solution) and the resulting precipitate was filtered off. The filtrate was concentrated in vacuo and the residue was purified by preparative TLC (10% MeOH in CH2CI2) to afford 48D (25 mg, 40% yield).
Step 4
To a solution of 48D (12 mg, 0.028 mmol) in EtOH (4 mL) were added NH2OH HCI salt (10 mg, 0.14 mmol) and pyridine (34 mL, 0.42 mmol) at 250C. The mixture was heated to reflux for 16 h and concentrated in vacuo. The residue was purified by preparative TLC (10% MeOH in CH2CI2) to afford 48 (5 mg, 42% yield).
Example 47
Figure imgf000130_0001
Compound 47A (20 mg) was dissolved in absolute ethano "Bl (10 m 4L7). Lindlar catalyst was added (18 mg) and the reaction mixture was placed under balloon pressure of hydrogen gas. The reaction mixture was left stirring overnight at rt. The reaction mixture was concentrated to dryness. CH2CI2 was added and the resulting material was loaded onto a 1g SiO2 Sep-Pak. The product was eluted with 95:5 CH2CI2: MeOH. The filtrate was concentrated to give 457 as a clear oil. Compound 47A was prepared using Example 400.
Example 41 :
Figure imgf000130_0002
Part A:
Compound 41 B was prepared from compound 41 A according to the procedures described in Example 300D part A and B.
Part B:
To a mixture of 41b (7.87 g, 21.7 mmol) and diisopropylethylamine (7.5 mL, 43.4 mmol) in DMF (80 mL) was added 2-trimethylsilylethoxy methyl chloride (4.7 mL, 23.8 mmol). The mixture was stirred at room temperature overnight, diluted with water and extracted with ethyl acetate. The combined organic layers were washed with water, brine, dried over sodium sulfate and concentrated. The residue was purified by column chromatography (SiO2, 15% EtOAc/hexane to 30 % EtOAc/hexane) to afford 41 C as a white solid (10.2 g, 95%). HPLC-MS tR = 2.17 min (UV2S4 nm); mass calculated for formula C23H35N3O7Si 493.2, observed LCMS m/z 516.1 (M+Na).
Part C:
The two isomers of 41 C were separated using a chiral AD column. One gram of material was injected into the column and the two peaks were separated by using a solvent mixture of 80% hexanes/2-propanol. The second isomer was the desired compound 41 D (400 mg, 80%).
Part D:
Compound 41 was prepared from 41 D following the procedures described in Example 2 step 3, Example 300E part C and Example 8. HPLC-MS tR = 1.32 min (UV254 nm); mass calculated for formula C2iHigN3O6 409.13, observed LCMS m/z 410.2 (M+H).
Example 44
Figure imgf000131_0001
Part A:
Compound 44A (670 mg, 3.91 mmol) and iodine (2.10 g, 8.0 mmol) were dissolved in THF (20 mL) and 1 M sodium carbonate (20 mL) and stirred for 4 hours. The reaction was diluted with ethyl acetate and water. The organic layer was dried over sodium sulfate and concentrated (1.05 g mixture of unseparable mono and di-iodinated products). The residue was combined with methyl iodide (1.5 mL) and cesium carbonate (5 g) in DMF (20 mL) and stirred at 600C for 3 hours. The reaction was diluted with water and ethyl acetate. The organic layer was dried over sodium sulfate and concentrated. Column chromatography (2:1 hexanes/ethyl acetate) provided the mono- iodinated methyl ether (460 mg). The methyl ether was dissolved in methylene chloride (7 mL) and 1 M boron tribromide (7 ml_) and stirred at room temperature for 5 hours. The reaction was quenched slowly with water and extracted with ethyl acetate. The organic layer was dried over sodium sulfate and concentrated to provide the desired product (390 mg, 34%). 1H NMR (400 MHz, CDCI3) δ 8.25 (d, 1H)1 7.85-7.8 (m, 1 H), 7.1 (d, 1 H)1 3.0 (s, 3H).
Part B:
Compound 401 (130 mg, 0.43 mmol), compound 44B (130 mg, 0.43 mmol), Pd(PPh3J2CI2 (15 mg), CuI (8 mg), and triethylamine (0.4 mL) were dissolved in DMF (3 mL) and stirred at 80 0C under an inert atmosphere. The solvent was evaporated and the residue was purified by reverse phase chromatography to provide the desired product (112.3 mg, 55%). HPLC-MS tR = 1.2 min (UV2S4 nm); mass calculated for formula C22H19N3O7S 469.47, observed LCMS m/z 470.0 (M+H). 1H NMR (400 MHz, DMSOd6) δ 11.2 (s, 1 H), 9.0 (s, 1 H), 8.3 (m, 2H), 7.9 (s, 1H), 7.5 (d, 1H), 7.35 (s, 1 H), 7.2-7.1 (m, 2H), 4.5-4.3 (m, 4 H). 3.8 (S, 3H), 3.2 (s, 3H).
Example 46
Figure imgf000132_0001
Step 1
Compound 46A (80 mg, 0.29 mmol) was combined with compound 401 (100 mg, 0.33 mmol), Pd(PPh3J2CI2 (5 mg, 0.007 mmol), CuI (12 mg, 0.06 mmol), diisopropylamine (0.16 mL, 1.13 mmol) in DMF (1 mL) and stirred at 85 0C. The reaction mixture was neutralized with acetic acid and purified with Gilson reverse phase (0-40% acetonitrile in H2O with 0.1% formic acid) afforded the desired product 46B (3 mg, 3%) mass calculated for formula C2OH17N5O4 391.13, observed LCMS m/z 392.2. (M+H) and compound 46 (22 mg, 15%), mass calculated for formula C25H25N5O6 491.18, observed LCMS m/z 492.2. (M+H)
Example 49
Figure imgf000133_0001
49A 49B 49C
Figure imgf000133_0002
49
Step i
The commercially available 2,6-dibromo-3-hydroxypyridine (588 mg, 2.32 mmol) was dissolved in THF (6 mL) and the solution was treated with triethylamine (0.49 mL, 3.48 mmol) and triisopropylsilyl triflate (0.75 mL, 2.78 mmol) at 00C. The mixture was stirred at the temperature for 10 mim then added to an aqueous NaHCθ3 solution. The organic layers were extracted by CH2CI2 and the combined organic solution was washed with brine solution, dried (Na2SO4), and concentrated in vacuo to afford a crude 49A (1.11 g, quantitative) which was used without further purification.
Step 2
A solution of 49A (100 mg, 0.24 mmol) in toluene (1 mL) was treated with t- BuLi (1.7 M in pentane, 0.32 mL, 0.54 mmol) at -78 0C. After stirring for 0.5 h at the temperature, methyl disulfide (65 μL, 0.72 mmol) was added to the mixture slowly and the resulting mixture was stirred for 4.5 h at -780C to 25 0C. The mixture was quenched by MeOH (0.3 mL) and diluted in CH2CI2 followed by filtration through SiO2 pad. The clear filtrate was concentrated in vacuo to afford a mixture of 49B and its bromide regioisomer (~1 :1 , 62 mg) which was used without further purification.
Step 3
A mixture of 49B and its regioisomer (62 mg, ~0.16 mmol) was dissolved in THF and solution was treated with TBAF (1M in THF, 0.24 mL, 0.24 mmol) at 0 0C. The mixture was stirred at the temperature for 1 h and poured to a cold mixture of EtOAc and water. The organic layers were extracted by EtOAc and the combined organic solution was washed with brine solution, dried (Na2SO4), and concentrated in vacuo to afford a crude mixture of 49C and its bromide regioisomer (~1 :1 , 41 mg) which was used without further purification.
Step 4
A mixture of 401 (150 mg, 0.50 mmol), 49C (-50% purity, 460 mg, ~1 mmol), Pd(PPh3)2CI2 (7 mg, 10 μmol), CuI (1.9 mg, 10 μmol), and diisopropylethylamine (0.43 mL, 2.5 mmol) in DMF (3 ml) was purged with N2 and heated to 600C. After heating for 18 h, the mixture was cooled to 25 0C and purified by column chromatography on a reverse phase C-18 column (0.01 %HCO2H in water/0.01 % HCO2H in CH3CN) to afford a crude 49 which was further purified by preparative TLC (5% MeOH in CH2CI2) to afford pure 49 (27 mg, 12% yield).
Example 50
Figure imgf000135_0001
A solution of 49 (27 mg, 0.06 mmol) in CH2CI2 (3 mL) was treated with 3- chloroperbenzoic acid (ca 70% purity, 30 mg, 0.12 mmol) at 0 0C. The mixture was stirred at 25 0C for 1.5 h. The suspension was dissolved in 10% MeOH-CH2CI2 and treated with ion exchange resin (Amberlyst, A-21, weakly basic) followed by filtration. The filtrate was concentrated in vacuo and the residue was purified by preparative TLC (10% MeOH in CH2CI2) to afford 50 as a white solid (18 mg, 61 % yield).
Example 32:
Figure imgf000135_0002
Part A
To a solution of methyl 4-acetylbenzoate (1.9 g, 10.6 mmol) in acetic acid (10 mL) was added dropwise bromine (1.7 g, 21.3 mmol). The mixture was heated at 60 0C for 30 min, then stirred at room temperature for 1 hour, and poured into cold water (30 mL). The light yellow precipitate was collected, washed with water and dried (2.6 g, 96%). Part B
Compound 32A was treated with one equivalent of hexamethylene tetraamine in chloroform for about 1 hour. The product was collected by filtration and then treated with HCI in methanol for 2 hours. The solid was then collected by filtration to give compound 32B.
Part C
Compound 30 was prepared following the procedures described in Example 2 Steps 1 , 2, 3 and in Example 300E Part E: HPLC-MS tR = 1.36 min (UV254 πm); mass calculated for formula C21H19N3O6 409.1, observed LCMS m/z 410.1 (M+H).
Part D
Compound 30 (60 mg, 0.147 mmol) was heated in 5% KOH in MeOH (2 mL) at 60 0C overnight, cooled to room temperature and concentrated. The residue was dissolved in water (5 mL), acidified with cone. HCI and filtered. The solid was collected and dried to give compound 32 (23 mg, 40%): HPLC- MS tR = 1.04 min (UV254 nm); mass calculated for formula C20H17N3O6395.1 , observed LCMS m/z 396.1 (M+H).
Part E
Compound 30 (39 mg, 0.095 mmol) was heated in pyrrolidine (2 mL) at 60 0C overnight, cooled to room temperature and concentrated. The residue was purified by reverse phase chromatography to give 31: HPLC-MS tR = 1.19 min (UV254 nm); mass calculated for formula C24H24N4O5 448.2, observed LCMS m/z 449.2 (M+H).
Part F
A mixture of compound 32 (49 mg, 0.12 mmol), dimethylamine hydrochloride (20 mg, 0.25 mmol), HATU (61 mg, 0.16 mmol), DMAP (2 mg, 0.012 mmol) and diisopropylethylamine (0.065 mL, 0.37 mmol) was stirred in DMF (2 mL) at room temperature overnight. The mixture was diluted with ethyl acetate, washed with 0.1 N HCI, water and brine, dried over sodium sulfate and evaporated. The residue was purified by reverse phase chromatography to give 34: HPLC-MS tR = 1.09 min (UV254 nm); mass calculated for formula C22H22N4O5 422.2, observed LCMS m/z 423.1 (M+H).
Example 33:
Figure imgf000137_0001
Part A
Compound 33 was prepared from 33A following procedures described in Example 32 Part B and Part C: HPLC-MS tR = 1.08 min (UV254 nm); mass calculated for formula C20H19N3O6S 429.1 , observed LCMS m/z 430.0 (M+H).
Example 35:
Figure imgf000137_0002
Part A: A mixture of 5-cyanophthalide (5.0 g, 31.4 mmol) and 1 N of NaOH (31.4 ml_) was stirred at 1000C for 1 h. The solution was concentrated to dryness with azotropic distillation with toluene. The resulting white solid was dissolved in dry DMF (30 mL). Methyl iodide (5.88 mL, 94.2 mmol) was added slowly, and the reaction mixture was allowed to stir at room temperature for 2 h. It was then diluted with H2O and back extracted with EtOAc (30 mL x 4). EtOAc extracts were combined, washed with brine, dried over Na2SO4, and concentrated. Flash column chromatography on silica (EtOAc / hexane 40:60) gave compound 35A as a white solid (5.5 g, 91 %)
Part B:
To compound 35A (5.5 g, 28.77 mmol) in THF (60 mL) was added carbon tetrabromide (11.45 g, 34.52 mmol). The solution was cooled to 00C in an ice/water bath, and triphenylphosphine (9.05 g, 34.52 mmol) was added portionwise. The reaction mixture was stirred at room temperature for 3 h under argon. After removing the precipitate by filtration, the solution was concentrated. The residue was dissolved in EtOAc (100 mL), washed with 1N HCI, H2O, brine, dried over Na2SO4, and concentrated. Flash column chromatography on silica (EtOAc / hexane 20:80) gave compound 35B as a pale yellow solid (6.8 g, 93%).
Part C:
Compound 35 was prepared using previously described methods from 2D and 35B. HPLC-MS tR = 2.943 min (UV254nm), Mass calculated for formula Ci9Hi3FN4O3 364.1 , observed LCMS m/z 365.0 (M+H).
Example 37:
Figure imgf000139_0001
Compound 37A was prepared using procedures described in Example 375.
Part A:
Compound 37A (550 mg, 1.32 mmol) and Pd(J-Bu3P)2 (34 mg, 0.066 mmol, 5 mol%) in NMP (5 mL) were added with a 0.5 M solution of 2-tert- butoxy-2-oxoethylzinc chloride in THF (10.5 mL, 5.2 mmol) under argon. The reaction mixture was allowed to stir at 90 0C overnight. After cooling to room temperature, it was diluted with EtOAc, washed with water, brine, dried over Na2SO4, and concentrated. Column chromatography on silica gel (MeOH / DCM1 10:90) afforded 37B as a pale yellow solid (260 mg, 43%). HPLC-MS (5 min) tR = 1.69 min (UV254nm), Mass calculated for formula C19H13FN4O3 453.2, observed LCMS m/z 454.1 (M+H).
Part B:
Compound 37B (40 mg, 0.088 mmol) was treated with TFA in DCM at room temperature to afford 36 as a white solid (20 mg, 27%). HPLC-MS XR = 2.64min (UV254nm). Mass calculated for formula C2OH16FN3O5 397.1, observed LCMS m/z 398.0 (M+H).
Part C: Compound 36 (20 mg, 0.05 mmol) in DMF (1 ml_) was added with HOBt (14 mg, 0.1 mmol) and EDC (19 mg, 0.1 mmol). After stirring at room temperature for 10 min, NH4CI (20 mg, 0.15 mol) was added, followed by the addition of DIEA (0.026 mL). The reaction mixture was then stirred at room temperature overnight. It was diluted with EtOAc, washed with 1N HCI, saturated NaHCO3, and brine, dried over Na2SO4, and concentrated. Recrystallization in EtOAc gave 37 (7.4 mg, 37%) as a white solid. HPLC-MS tR = 2.64min (UV254nm), Mass calculated for formula C20H17FN4O4 396.1, observed LCMS m/z 397.1 (M+H).
NMR spectral data for the some of the above compounds are provided below:
Compound 50. 1H NMR (500 Hz, CD3OD) .δ 8.27 (d, 1 H, J = 8.6 Hz), 8.14 (d, 1H, J = 8.6 Hz), 7.45 (d, 1H, J = 8.3 Hz), 7.37 (s, 1H), 7.29 (d, 1H, J = 2.5 Hz), 7.20 (dd, 1 H, J = 8.3 Hz, 2.5 Hz), 4.56 (d, 1 H, J = 8.7 Hz), 4.53 (d, 1 H, J = 8.7 Hz), 4.50 (d, 1 H, J = 12.5 Hz), 4.46 (d, 1 H, J = 12.6 Hz), 3.87 (s, 3H), 3.30 (s, 3H).
Compound 98. 1H NMR (400 Hz DMSO-d6) δ (ppm): 11.24 (s, 1H), 8.97 (s, 1H), 8.80 (d, J = 2.01 Hz, 1H ), 8.57 (d, J = 2.46 Hz, 1H ), 8.17 (s, 1H), 7.60 (s, 1 H), 7.49 (d, J = 8.19 Hz, 1H), 7.24 (s, 1H), 7.17-7.14 (m, 2H), 4.46 - 4.25 (m, 4H), 3.79 (s, 3H).
Compound 68. 1H NMR (500 Hz, CD3OD) δ 8.42 (s, 1 H), 8.32 (d, 1H, J = 2.1 Hz), 7.79 (d, 2H, J = 8.5 Hz), 7.74 (d, 2H, J = 8.5 Hz), 7.42 (d, 1H, J = 8.4 Hz), 7.28-7.32 (m, 1 H), 7.19 (dd, 1H, J = 8.4, 2.5 Hz), 4.32-4.46 (m, 3H), 4.24 (d, 1 H, J = 14.2 Hz), 3.87 (s, 3H).
Compound 66. 1H NMR (500 Hz, CD3OD) δ 8.88 (d, 1 H, J = 5.0 Hz), 8.30 (s, 1H), 8.19 (d, 2H, J = 8.5 Hz), 7.88 (dr 2H, J = 8.2 Hz), 7.67 (dd, 1H, J = 5.1, 1.3 Hz), 7.41 (d, 1H1 J = 8.6 Hz), 7.31 (d, 1 H, J = 2.5 Hz), 7.19 (dd, 1H, J = 8.2, 2.5 Hz), 4.34-4.47 (m, 3H), 4.26 (d, 1 H, J = 14.7 Hz), 3.87 (s, 3H).
Compound 64. 1H NMR (500 Hz, CD3OD) δ 8.10 (d, 2H, J = 8.5 Hz), 7.99 (s, 1H), 7.84 (d, 2H1 J = 8.5 Hz), 7.56 (s, 1H), 7.37 (d, 1H, J = 8.2 Hz), 7.27 (d, 1H, J = 2.7 Hz), 7.15 (dd, 1H, J = 8.4, 2.5 Hz), 4.28-4.43 (m, 3H), 4.24 (d, 1H, J = 14.5 Hz), 3.84 (s, 3H)1 2.65 (s, 3H), 0.84-0.89 (m, 1H), 0.69-0.74 (m, 2H), 0.49-0.52 (m, 2H).
Compound 97. 1H NMR (500 MHz, MeOH-d4) δ 8.19 (s, 1H) , 7.83 (s,1H), 7.66-7.70 (m,1H), 7.55-7.58 (m,1H), 7.41-7.44 (m,1H), 7.29-7.32 (m,1H), 7.17-7.21 (m,1H) , 7.07 (s,1H), 4.35-4.55 (m,4H), 3.87 (s,3H).
Compound 85. 1H NMR (500 MHz, MeOH-d4) δ 7.70-7.75 (m, 2H) 7.65- 7.70 (m,1H), 7.41-7.56 (m, 3H), 7.28-7.31 (m, 1H), 7.17-7.23 (m, 2H), 7.08- 7.13 (m, 2H) , 4.29-4.47 (m, 3H), 4.15-4.24 (m, 1H), 3.87 (s, 3H).
Compound 84. 1H NMR (500 MHz, MeOH-d4) .δ 7.80-7.88 (m, 2H), 7.70- 7.78 (m, 2H) 7.38-7.46 (m, 1H), 7.26-7.33 (m, 1H), 7.16-7.24 (m, 1H), 7.08- 7.15 (m, 2H) , 6.98-7.06 (m, 2H)1 4.29-4.46 (m, 3H), 4.15-4.26 (m, 1H)1 3.87 (s, 3H), 2.93 (S, 3H).
Compound 43 (400 Hz DMSO-d6) δ 8.99 (s, 1H), 7.98 (dd, J = 14.8 Hz, 2 Hz, 2H), 7.90 (dd, J = 14.8 Hz, 2 Hz, 2H)1 7.47 (d, J = 8.4 Hz, 1H), 7.14 (m, 2H), 4.26 (m, 3H), 4.08 (m, 1H), 3.79 (s, 3H), 3.22 (s, 3H).
Compound 76. 1H NMR (500 MHz, DMSO-d6).δ 10.97 (s, 1H), 9.00 (d, J = 2.0 Hz, 1H), 8.39 (d, J = 8.5 Hz, 1H), 8.22 (d, J = 7.5 Hz, 1H), 8.08 (t, J = 7.5 Hz, 1H), 8.00 (d, J = 7.5 Hz, 1 H), 7.78 (d, J = 8.5 Hz, 1H), 7.75 (br. s, 1H), 7.48 (d, J = 8.5 Hz, 1H), 7.19 (d, J = 2.5 Hz, 1H), 7.16 (dd, J = 8.5, 2.5 Hz, 1H)1 4.34 (d, J = 17.5 Hz, 1H), 4.26 (d, J = 17.5 Hz, 1H), 4.24 (d, J = 14.5 Hz, 1H), 4.17 (d, J = 14.5 Hz, 1H), 3.81 (s, 3H).
It will be appreciated by those skilled in the art that changes could be made to the embodiments described above without departing from the broad inventive concept thereof. It is understood, therefore, that this invention is not limited to the particular embodiments disclosed, but it is intended to cover modifications that are within the spirit and scope of the invention, as defined by the appended claims.
Each document referred to herein is incorporated by reference in its entirety for all purposes.

Claims

THEREFORE, WE CLAIM:
1. A compound represented by Formula (I):
Figure imgf000143_0001
or a pharmaceutically acceptable salt, solvate, or ester thereof, wherein: ring A is selected from the group consisting of aryl and heteroaryl, each of which is substituted with -Y-R1 and -Z-R2 as shown;
X is selected from the group consisting of -S-.-O-, -S(O)2, -S(O)-,
-(C(R3)2)m- and -N(R3)-;
T is absent or present, and if present, T is selected from the group consisting of alkyl, aryl, and heteroaryl, wherein when each of said T aryl and heteroaryl contains two radicals on adjacent carbon atoms, said radicals may optionally be taken together with the carbon atoms to which they are attached to form a five- to eight-membered aryl or heteroaryl ring, wherein each of the aforementioned T aryl, and heteroaryl, optionally with said five- to eight- membered aryl or heteroaryl is independently unsubstituted or substituted with one to four R10 moieties which can be the same or different;
U is absent or present or absent, and if present, U is selected from the group consisting of -O-, -0-C(O)NH-, -OC(O)N(alkyl)-, -C(O)-, -C(O)O-, -C(O)NH-, -C(O)N(alkyl)-, -C(=N-OH)-alkyh and -C(=N-O-alkyl)-alkyl-;
V is absent or present, and if present V is selected from the group consisting of alkyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, and N- oxides of said heterocyclyl and heteroaryl, wherein when each of said V cycloalkyl, heterocyclyl, aryl, heteroaryl, and N-oxides of said heterocycyl and heteroaryl contains two radicals on adjacent carbon atoms, said radicals may optionally be taken together with the carbon atoms to which they are attached to form a five- to eight-membered cycloalkyl, aryl, heterocyclyl or heteroaryl ring; wherein each of said V alkyl, alkynyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl heterocyclyl, optionally with said five- to eight-mem be red cycloalkyl, aryl, heterocyclyl, or heteroaryl is independently unsubstituted or substituted with one to four R10 moieties which can be the same or different; Y is selected from the group consisting of a covalent bond, -(C(R4)2)n-,
-N(R4)-, -C(O)N(R4)-, -N(R4)C(O)-, -N(R4JC(O)N(R4)-, -S(O)2N(R4)-, -N(R4)- S(O)2, -O-.-S-, -C(O)-, -S(O)-, and -S(O)2-;
2 is selected from the group consisting of a covalent bond, -(C(R4)2)n-,
-N(R4)-, -C(O)N(R4)-, -N(R4)C(O)-, -N(R4)C(O)N(R4)-, -S(O)2N(R4)-, -N(R4)- S(O)2-, -O- -S-, -C(O)-, -S(O)-, and -S(O)2-; m is 1 to 3; n is 1 to 3;
R is selected from the group consisting of H, cyano, -C(O)OH, - C(O)O-alkyl, -C(O)NH2, -C(O)NH(alkyl), -C(O)N(alkyl)2, alkynyl, halogen, alkyl, cycloalkyl, haloalkyl, aryl, heteroaryl, and heterocyclyl, wherein when each of said cycloalkyl, heterocyclyl, aryl and heteroaryl contains two radicals on adjacent carbon atoms, said radicals may optionally be taken together with the carbon atoms to which they are attached to form a five- to eight- membered cycloalkyl, aryl, heterocyclyl or heteroaryl ring; wherein each of the R1 alkyl, alkynyl, aryl, heteroaryl, and heterocyclyl, optionally with the five or six-membered cycloalkyl, aryl, heterocyclyl or heteroaryl ring is unsubstituted or optionally independently substituted with one to four R20 moieties which can be the same or different; with the proviso that when Y is -N(R4)-, -S -or -O-, then R1 is not halogen or cyano;
R is selected from the group consisting of H, cyano, -C(O)OH, - C(O)O-alkyl, -C(O)NH2, -C(O)NH(alkyl), -C(O)N(alkyl)2, alkynyl, halogen, alkyl, cycloalkyl, haloalkyl, aryl, heteroaryl, and heterocyclyl; wherein when each of said cycloalkyl, heterocyclyl, aryl and heteroaryl contains two radicals on adjacent carbon atoms, said radicals may optionally be taken together with the carbon atoms to which they are attached to form a five- to eight- membered cycloalkyl, aryl, heterocyclyl or heteroaryl ring; wherein each of the R2 alkyl, cycloalkyl, aryl, heteroaryl, and heterocyclyl, optionally with the five or six-membered cycloalkyl, aryl, heterocyclyl or heteroaryl ring is unsubstituted or optionally independently substituted with one to four R20 moieties which can be the same or different; with the proviso that when Y is - N(R4)-, -S -or -O- , then R2 is not halogen or cyano; each R3 is the same of different and is independently selected from the group consisting of H, alkyl, and aryl;
4 each R is the same or different and is independently selected from the group consisting of H, alkyl, cycloalkyl, haloalkyl, hydroxy, -alkylcycloalkyl, - alkyl-N(alkyl)2, heterocyclyl, aryl, and heteroaryl, wherein when each of said cycloalkyl, heterocyclyl, ayl, and heteroaryl contains two radicals on adjacent carbon atoms, said radicals may optionally be taken together with the carbon atoms to which they are attached to form a five- to eight-membered cycloalkyl, aryl, heterocyclyl or heteroaryl ring;
10
R is selected from the group consisting of hydrogen, cyano, nitro,
-C(R4)=N-OR4, -OR4, -SR4, -N(R4)2, -S(O)R4, -S(O)2R4, -N(R4)S(O)2R4, -N(R4)- C(O)-R4, -N(R4)-C(O)-N(R4)2 , -N(R4)-C(O)-OR4, -OC(O)N(R4)2, -C(O)N(R4)- S(O)2R4, -S(O)2N(R4J-C(O)-R4, -C(O)N(R4)C(O)R4, -C(O)N(R4)C(O)NR4, - S(O)2N(R4)2, -N(R4)-C(=NR4)-N(R4)2, -N(R4)-C(=N-CN)-N(R4)2, -haloalkoxy, - C(O)OR4, -C(O)R4, -C(O)N(R4)2, halogen, alkyl, haloalkyl, aryl, heteroaryl, heterocyclyl, and cycloalkyl, wherein each of the R10 alkyl, aryl, heteroaryl, heterocyclyl, and cycloalkyl is unsubstituted or optionally independently substituted with one to four R30 moieties which can be the same or different; or wherein two R10 moieties, when attached to the same or adjacent carbon atoms may optionally be taken together with the carbon atom(s) to which they are attached to form a cycloalkyl, cycloalkenyl, heterocyclyl, aryl, or heteroaryl ring;
20
R is selected from the group consisting of cyano, nitro,
-C(R4)=N-OR4, -OR4, -SR4, -N(R4)2, -S(O)R4, -S(O)2R4, -N(R4)S(O)2R4, -N(R4)- C(O)-R4, -N(R4)-C(O)-N(R4)2 , -N(R4)-C(O)-OR4, -OC(O)N(R4)2, -C(O)N(R4)- S(O)2R4, -S(O)2N(R4)-C(O)-R4, -C(O)N(R4)C(O)R4, -C(O) N (R4) C (O) N R4, - S(O)2N(R4)2l -N(R4)-C(=NR4)-N(R4)2, -N(R4)-C(=N-CN)-N(R4)2, -haloalkoxy, - C(O)OR4, -C(O)R4, -C(O)N(R4J2, halogen, alkyl, haloalkyl, aryl, heteroaryl, heterocyclyl, and cycloalkyl; wherein when each of said R20 aryl, heteroaryl, heterocyclyl and cycloalkyl contains two radicals on adjacent carbon atoms, said radicals may optionally be taken together with the carbon atoms to which they are attached to form a five- to eight-membered cycloalkyl, aryl, heterocyclyl or heteroaryl ring; wherein each of said R20 alkyl, aryl, heteroaryl, heterocyclyl, and cycloalkyl, optionally with said five- to eight-membered cycloalkyl, aryl, heterocyclyl or heteroaryl ring is unsubstituted or substituted with one to four moieties selected independently from the group consisting of alkyl, halo, haloalkyl, hydroxy, alkoxy, haloalkoxy, hydroxyalkyl, cyano, nitro, - NH2, -NH(alkyl), and -N(alkyl)2; or when two R20 moieties when attached to the same or adjacent carbon atoms may optionally be taken together with the carbon atom(s) to which they are attached to form a cycloalkyl, cycloalkenyl, heterocyclyl, aryl, or heteroaryl ring;
30
R is selected from the group consisting of cyano, nitro,
-C(R4)=N-OR4, -OR4, -SR4, -N(R4J2, -S(O)R4, -S(O)2R4, -N (R4)S(O)2R4, -N(R4)- C(O)-R4, -N(R4)-C(O)-N(R4)2 , -N(R4)-C(O)-OR4, -OC(O)N(R4)2, -C(O)N(R4)- S(O)2R4, -S(O)2N(R-O-C(O)-R4, -C(O)N(R4)C(O)R4, -C(O)N(R4)C(O)NR4, - S(O)2N(R4J2, -N(R4)-C(=NR4)-N(R4)2, -N(R4)-C(=N-CN)-N(R4)2, -haloalkoxy, - C(O)OR4, -C(O)R4, -C(O)N(R4)2, halogen, alkyl, haloalkyl, aryl, heteroaryl, heterocyclyl, and cycloalkyl; wherein when each of said R30 aryl, heteroaryl, heterocyclyl and cycloalkyl contains two radicals on adjacent carbon atoms, said radicals may optionally be taken together with the carbon atoms to which they are attached to form a five- to eight-membered cycloalkyl, aryl, heterocyclyl or heteroaryl ring; wherein each of said R30 alkyl, aryl, heteroaryl, heterocyclyl, and cycloalkyl, optionally with said five- to eight-membered cycloalkyl, aryl, heterocyclyl or heteroaryl ring is unsubstituted or substituted with one to four moieties selected independently from the group consisting of alkyl, halo, haloalkyl, hydroxy, alkoxy, haloalkoxy, cyano, nitro, -NH2, - NH(alkyl), and -N(alkyl)2; or when two R30 moieties when attached to the same or adjacent carbon atoms may optionally be taken together with the carbon atom(s) to which they are attached to form a cycloalkyl, cycloalkenyl, heterocyclyl, aryl, or heteroaryl ring; with the proviso that at least one of T, U, and V must be present; and further that at least one of conditions (1) - (5) below are satisfied:
(1) at least one of T and V is present, and V is other than alkynyl; wherein said T or V is substituted with at least one R10 moiety selected from the group consisting of cyano, -C(O)OR4, -C(O)R4, -C(O)N(R4)2, - C(O)N(R4)C(O)R4, -C(O)N(R4)C(O)NR4, -SR4, -S(O)2R4, -N(R4)-C(O)OR4, - OC(O)N(R4)2, -N(R4)C(O)N(R4)2, -N(R4J-C(O)-R4, -S(O)2N(R4)2, -S(O)2N(R4)- C(O)-R4,
-N(R4)-C(=NR4)-N(R4)2, -N(R4)-C(=N-CN)-N(R4)2, and -C(R4)=N-OR4, wherein each R4 independently is selected from the group consisting of H, alkyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl, wherein when each of said R4 cycloalkyl, heterocyclyl, aryl, and heteroaryl contains contains two radicals on adjacent carbon atoms, said radicals may optionally be taken together with the carbon atoms to which they are attached to form a five- to eight- membered aryl, heteocyclyl, heteroaryl or cycloalkyl ring; with the proviso that when R10 is
-S(O)2R4, V is other than piperidinyl, and when R10 is cyano, the compound of Formula (I) is other than
Figure imgf000147_0001
(2) U and V are present; and U is selected from the group consisting of -0-C(O)NH-, -OC(O)N(alkyl)-, -C(O)-, -C(O)O-, -C(O)NH-, -C(O)N(alkyl)-( - C(=N-OH)-alkyl-, and -C(=N-O-alkyl)-alkyl-;
(3) at least one T and V is present, and each of -Y-R1 and -Z-R2 is independently selected from the group consisting of cyano, -(C(R4)2)n- C(O)OH, -(C(R4)2)n-C(O)O-alkyl, -(C(R4^)n-C(O)NH2, -(C(R4)2)n- C(O)NH(alkyl), and -(C(R4)2)n-C(O)N(alkyl)2, wherein each R4 independently is H or alkyl; and n is 1 -3;
(4) T is aryl or heteroaryl, each of which is optionally substituted with one to four independently selected R10 moieties, and V is alkynyl which is optionally substituted with one or two independently selected R10 moieties; and
(5) at least one of T and V is present, ring A is heteroaryl, and V is other than alkynyl.
2. The compound of claim 1 , wherein X is selected from the group consisting of -(C(R3)2)m- and -N(R3)-.
3. The compound of claim 2, wherein X is -(C(R3)2)m, wherein m is 1 or 2.
4. The compound of claim 3, wherein m is 1.
5. The compound of any of claims 1-4, wherein R3 is H.
6. The compound of claim 1 , wherein at least one of T and V is present, and V is other than alkynyl; wherein said T or V is substituted with at least one R10 moiety selected from the group consisting of cyano, -C(O)OR4, - C(O)R4, -C(O)N(R4)2, -C(O)N(R4)C(O)R4, -C(O)N(R4)C(O)NR4, -SR4, - S(O)2R4, -N(R4)-C(O)OR4, -OC(O)N(R4)2, -N(R4)C(O)N(R4)2, -N(R4)-C(O)-R4, - S(O)2N(R4J2, -S(O)2N (R4)-C(O)-R4, -N(R4)-C(=NR4)-N(R4)2, -N(R4)-C(=N-CN)- N(R4)2, and -C(R4)=N-OR4, wherein each R4 independently is selected from the group consisting of H, alkyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl, wherein when each of said R4 cycloalkyl, heterocyclyl, aryl, and heteroaryl contains contains two radicals on adjacent carbon atoms, said radicals may optionally be taken together with the carbon atoms to which they are attached to form a five- to eight-membered aryl, heteocyclyl, heteroaryl or cycloalkyl ring; with the proviso that when R10 is -S(O)2R4, V is other than piperidinyl, and when R10 is cyano, the compound of Formula (I) is other than
Figure imgf000149_0001
7. The compound of claim 6, wherein said T or V is substituted with at least one R10 moiety selected from the group consisting of cyano, - C(O)OR4, -C(O)R4, -C(O)N(R4)2, and -C(R4)=N-OR4.
8. The compound of claim 6, wherein said T or V is substituted with at least one R10 moiety that is cyano.
9. The compound of claim 6, wherein said T or V is substituted with at least one R10 moiety that is -SR4.
10. The compound of claim 6, wherein said T or V is substituted with at least one R10 moiety that is -S(O)2R4.
11. The compound of claim 6, wherein said T or V is substituted with at least one R10 moiety that is -S(O)2N(R4)2.
12. The compound of claim 6, wherein ring A is selected from the group consisting of phenyl, thiophenyl, pyridyl, pyrimidyl, and
Figure imgf000149_0002
each of which is substituted with -Y-R1 and -Z-R2 as shown.
13. The compound of claim 12, wherein said ring A is phenyl.
14. The compound of claim 6, wherein X is selected from the group consisting of -(C(R3)2)m- and -N(R3)-.
15. The compound of claim 14, wherein X is -(C(R3)2)mι wherein m is 1 or 2.
16. The compound of claim 15, wherein m is 1.
17. The compound of claim 14, wherein R3 is H.
18. The compound of claim 6, wherein T is selected from the group consisting of alkyl, aryl, heteroaryl, wherein when each of said T aryl and heteroaryl contains two radicals on adjacent carbon atoms, said radicals may optionally be taken together with the carbon atoms to which they are attached to form a five- to eight-membered aryl or heteroaryl ring; wherein each of the aforementioned T aryl, and heteroaryl, optionally with said five- to eight- membered aryl or heteroaryl is independently unsubstituted or substituted with one to four R10 moieties which can be the same or different.
19. The compound of claim 18, wherein T is selected from the group consisting of -CH2-, phenyl,
Figure imgf000150_0001
optionally substituted with one to four R moieties such that the number of R10 moieties per each T does not exceed four.
20. The compound of claim 6, wherein U is absent or present, and if present is selected from the group consisting of -C(O)-, and -C(O)O-.
21. The compound of claim 6, wherein V is absent or present, and if present is selected from the group consisting of aryl, and heteroaryl, wherein when each of said V aryl and heteroaryl contains two radicals on adjacent carbon atoms, said radicals may optionally be taken together with the carbon atoms to which they are attached to form a five- to eight-membered aryl or heteroaryl ring; wherein each of the aforementioned V aryl, and heteroaryl, optionally with said five- to eight-membered aryl or heteroaryl is independently unsubstituted or substituted with one to four R10 moieties which can be the same or different.
22. The compound of claim 21 , wherein V is selected from the group consisting of phenyl, pyridyl, pyrazinyl, indazolyl,
Figure imgf000150_0002
each of which is optionally substituted with one to four R10 moieties which can be the same or different.
23. The compound of claim 6, wherein each of Y and Z is independently selected from the group consisting of a covalent bond and -O-
24. The compound of claim 23, wherein Y is -O- and Z is a covalent bond.
25. The compound of claim 6, wherein each of R1 and R2 is independently selected form the group consisting of H and alkyl.
26. The compound of claim 25, wherein R1 is alkyl and R2 is H.
27. The compound of claim 26, wherein R1 is methyl.
28. The compound of claim 6, selected from the group consisting of:
Figure imgf000151_0001
Figure imgf000152_0001
Figure imgf000153_0001
Figure imgf000154_0001
Figure imgf000155_0001
Figure imgf000156_0001
Figure imgf000157_0001
Figure imgf000158_0001
Figure imgf000159_0001
Figure imgf000160_0001
Figure imgf000161_0001
Figure imgf000162_0001
or a pharmaceutically acceptable salt, solvate or ester thereof.
29. The compound of claim 1 , wherein U and V are present; and U is selected from the group consisting of -0-C(O)NH-, -OC(O)N(alkyl)-, -C(O)-, -C(O)O-, -C(O)NH-, -C(O)N(alkyl)-, -C(=N-OH)-alkyl-, and -C(=N-O-alkyl)- alkyl-.
30. The compound of claim 29, wherein ring A is selected from the group consisting of phenyl, thiophenyl, pyridyl, pyrimidyl, and
Figure imgf000162_0002
each of which is substituted with -Y-R1 and -Z-R2 as shown.
31. The compound of claim 30, wherein ring A is phenyl.
32. The compound of claim 29, wherein X is selected from the group consisting of -(C(R3)2)m- and -N(R3)-.
33. The compound of claim 32, wherein X is -(C(R3)2)m, wherein m is 1 or 2.
34. The compound of claim 33, wherein m is 1.
35. The compound of claim 32, wherein R3 is H.
36. The compound of claim 29, wherein T is absent or present, and when present, is selected from the group consisting of alkyl, and aryl, each of which is unsubstitued or substituted with one to four R10 moieties which can be the same or different.
37. The compound of claim 36, wherein T is absent or present, and when present is selected from the group consisting of -CH2-, and phenyl.
38. The compound of claim 29, wherein V is selected from the group consisting of alkyl, heterocyclyl, and cycloalkyl, wherein when each of said V heterocyclyl or cycloalkyl contains two radicals on adjacent carbon atoms, said radicals may optionally be taken together with the carbon atoms to which they are attached to form a five- to eight-membered cycloalkyl, heterocyclyl, aryl or heteroaryl ring; wherein each of aforementioned V alkyl, heterocyclyl, and cycloalkyl, optionally with said five- to eight-membered cycloalkyl, heterocyclyl, aryl or heteroaryl ring is is independently unsubstituted or substituted with one to four R10 moieties which can be the same or different.
39. The compound of claim 38, wherein V is selected from the group consisting of methyl, ethyl, isopropyl, morpholinyl, cyclohexyl, piperidinyl optionally substituted with cyano or phenyl, -CH2- substituted with tetrahydrofuranyl and
Figure imgf000163_0001
-CH(CH3)- substituted with phenyl, piperazinyl substituted wth methyl, pyrrolidinyl substituted wth -CH2-phenyl,
substituted with cyclopropyl, and
Figure imgf000163_0002
40. The compound of claim 29, wherein each of Y and Z is independently selected from the group consisting of a covalent bond and -O-
41. The compound of claim 40, wherein Y is -O- and Z is a covalent bond.
42. The compound of claim 29, wherein each of R1 and R2 is independently selected form the group consisting of H and alkyl.
43. The compound of claim 42, wherein R1 is alkyl and R2 is H.
44. The compound of claim 43, wherein R1 is methyl.
45. The compound of claim 29, selected from the group consisting of:
Figure imgf000164_0001
Figure imgf000165_0001
Figure imgf000166_0001
30
Figure imgf000167_0001
31
Figure imgf000167_0002
or a pharmaceutically acceptable salt, solvate or ester thereof.
46. The compound of claim 1 , wherein at least one T and V is present, and each of -Y-R1 and -Z-R2 is independently selected from the group consisting of cyano, -(C(R4)2)n-C(O)OH, -(C(R4)2)n-C(O)O-alkyl, - (C(R4)2)n-C(O)NH2) -(C(R4)2)n-C(O)NH(alkyl), and
-(C{R4)2)n-C(O)N(alkyl)2. wherein each R4 independently is H or atkyl; and n is 1-3.
47. The compound of claim 46, wherein ring A is selected from the group consisting of phenyl, thiophenyl, pyridyl, pyrimidyl, and
Figure imgf000167_0003
each of which is substituted with -Y-R1 and -Z-R2 as shown.
48. The compound of claim 47, wherein said ring A is phenyl.
49. The compound of claim 46, wherein X is selected from the group consisting of -(C(R3)2)m- and -N(R3)-.
50. The compound of claim 49, wherein X is -(C(R3)2)m, wherein m is 1 or 2.
51. The compound of claim 50, wherein m is 1.
52. The compound of claim 49, wherein R3 is H.
53. The compound of claim 46, wherein T or V is aryl which is unsubstituted or substituted with one to four R10 moieties.
54. The compound of claim 53, wherein said T or V is phenyl which is unsubstituted or substituted with one to four R10 moieties.
55. The compound of claim 54, wherein R10 is fluoro.
56. The compound of claim 55, wherein only one of T and V is present.
57. The compound of claim 46, wherein U is absent.
58. The compound of claim 46, wherein n is 1.
59. The compound of claim 46, wherein each of Y and Z is independently selected from the group consisting of a covalent bond and - CHz-, and each of R1 and R2 is independently selected from the group consisting of cyano, -C(O)OH or -C(O)NH2.
60. The compound of claim 55, wherein Y is a covalent bond, and R1 is H.
61. The compound of claim 55, wherein Z is a covalent bond, and R2 is cyano.
62. The compound of claim 55, wherein Z Is -CH2-, and R2 is - C(O)OH or -C(O)NH2.
63. The compound of claim 46, selected from the group consisting of:
Figure imgf000168_0001
Figure imgf000169_0001
or a pharmaceutically acceptable salt or solvate thereof.
64. The compound of claim 1 , wherein T is aryl or heteroaryl, each of which is optionally substituted with one to four independently selected R10 moieties, and V is alkynyl which is optionally substituted with one or two independently selected R10 moieties.
65. The compound of claim 64, wherein ring A is selected from the group consisting of phenyl, thiophenyl, pyridyl, pyrimidyl, and
Figure imgf000169_0002
each of which is substituted with -Y-R1 and -Z-R2 as shown.
66. The compound of claim 65, wherein said ring A is phenyl.
67. The compound of claim 64, wherein X is selected from the group consisting of -(C(R3)2)m- and -N(R3)-.
68. The compound of claim 67, wherein X is -(C(R3)2)m, wherein m is 1 or 2.
69. The compound of claim 68, wherein m is 1.
70. The compound of claim 66, wherein R3 is H.
71. The compound of claim 64, wherein T is aryl, U is -O- or absent, and V is alkynyl which is unsubstituted or substituted with one or two R10 moieties selected from the group consisting of -OR4, -N(R4)2, and heteroaryl; wherein when said heteroaryl contains two radicals on adjacent carbon atoms, said radicals may optionally be taken together with the carbon atoms to which they are attached to form a five- to eight-membered aryl, heteocyclyl, heteroaryl or cycloalkyl ring; wherein each R4 independently is H or alkyl, and said R10 heteroaryl is optionally independently substituted with one to four R30 moieties which can be the same or different.
72. The compound of claim 71 , wherein T is phenyl.
73. Th© compound of claim 71 , wherein said V alkynyl is selected from the group consisting of -CH2-C≡C-CH3l R10 substituted -C≡C- and R10 substituted -CH2-C≡C-CH2-.
74. The compound of claim 71 , wherein said R10 substituents are selected from the group consisting of -N(alkyl)2, -OH1 -OCH3, and pyridyl.
75. The compound of claim 64, selected from the group consisting of:
Figure imgf000170_0001
Figure imgf000171_0001
or a phar
Figure imgf000172_0001
76. The compound of claim 1 , wherein at least one of T and V is present, ring A is heteroaryl, and V is other than alkynyl.
77. The compound of claim 76, wherein ring A is selected from the group consisting of thiophenyl, pyridyl, pyrimidyl, and
Figure imgf000172_0002
each of which is substituted with -Y-R1 and -Z-R2 as shown.
78. The compound of claim 77, wherein X is selected from the group consisting of -(C(R3)2)m- and -N(R3)-.
79. The compound of claim 78, wherein X is -(C(R3)2)m, wherein m is 1 or 2.
80. The compound of claim 79, wherein m is 1.
81. The compound of claim 78, wherein R3 is H.
82. The compound of claim 76, wherein T is selected from the group consisting of alkyl, and halo-substituted aryl.
83. The compound of claim 76, wherein U and V are absent.
84. The compound of claim 76, wherein Y is selected from the group consisting of a covalent bond and -O-, and Z is a covalent bond.
85. The compound of claim 76, wherein R1 is selected from the group consisting of H and -CH3; and R2 is H.
86. The compound of claim 76, wherein Y is a covalent bond R1 is H.
87. The compound of claim 76, wherein Y is is -O-and R1 is -CH3.
88. The compound of claim 76, wherein Z is a covalent bond and R2 is H.
89. The compound of claim 76, selected from the group consisting of:
Figure imgf000173_0001
or a pharmaceutically acceptable salt or solvate thereof.
90. A compound selected from the group consisting of:
Figure imgf000174_0001
Figure imgf000175_0001
Figure imgf000176_0001
Figure imgf000177_0001
Figure imgf000178_0001
Figure imgf000179_0001
Figure imgf000180_0001
Figure imgf000181_0001
Figure imgf000182_0001
Figure imgf000183_0001
Figure imgf000184_0001
Figure imgf000185_0001
Figure imgf000186_0001
Figure imgf000187_0001
Figure imgf000188_0001
Figure imgf000189_0001
Figure imgf000190_0001
Figure imgf000191_0001
Figure imgf000192_0002
or a pharmaceutically acceptable salt, solvate, or ester thereof.
91. The compound of claim 90, selected from the group consisting of:
Figure imgf000192_0001
Figure imgf000193_0001
or a pharmaceut
Figure imgf000194_0001
92. A compound of claim 1 in purified form.
93. A pharmaceutical composition comprising the compound of claim 1 or a pharmaceutically acceptable salt, solvate or ester thereof, and at least one pharmaceutically acceptable carrier.
94. A method of treating disorders associated with TACE, TNF-α, MMPs, aggrecanase, ADAMs or any combination thereof, said method comprising administering to a patient in need of such treatment a therapeutically effective amount of at least one compound of claim 1 , or a pharmaceutically acceptable salt, solvate or ester thereof.
95. A method of treating disorders associated with TACE, TNF-α, aggrecanase, MMPs, ADAMs or any combination thereof, said method comprising administering to a patient in need of such treatment the pharmaceutical composition of claim 92.
96. A method of treating a condition or disease selected from the group consisting of rheumatoid arthritis, osteoarthritis, periodontitis, gingivitis, corneal ulceration, solid tumor growth and tumor invasion by secondary metastases, neovascular glaucoma, inflammatory bowel disease, multiple sclerosis and psoriasis in a subject, comprising: administering to the subject in need of such treatment a therapeutically effective amount of at least one compound of claim 1 , or a pharmaceutically acceptable salt, solvate or ester thereof.
97. A method of treating a condition or disease selected from the group consisting of fever, cardiovascular conditions, hemorrhage, coagulation, cachexia, anorexia, alcoholism, acute phase response, acute infection, shock, graft versus host reaction, autoimmune disease and HIV infection in a subject comprising administering to the subject in need of such treatment a therapeutically effective amount of at least one compound of claim 1 , or a pharmaceutically acceptable salt, solvate or ester thereof.
98. A method of treating a condition or disease selected from the group consisting of septic shock, haemodynamic shock, sepsis syndrome, post ischaemic reperfusion injury, malaria, mycobacterial infection, meningitis, psoriasis, congestive heart failure, fibrotic diseases, cachexia, graft rejection, cancers such as cutaneous T-cell lymphoma, diseases involving angiogenesis, autoimmune diseases, skin inflammatory diseases, inflammatory bowel diseases such as Crohn's disease and colitis, osteo and rheumatoid arthritis, ankylosing spondylitis, psoriatic arthritis, adult Still's disease, ureitis, Wegener's granulomatosis, Behcehe disease, Sjogren's syndrome, sarcoidosis, polymyositis, dermatomyositis, multiple sclerosis, sciatica, complex regional pain syndrome, radiation damage, hyperoxic alveolar injury, periodontal disease, HIV, non-insulin dependent diabetes mellitus, systemic lupus erythematosus, glaucoma, sarcoidosis, idiopathic pulmonary fibrosis, bronchopulmonary dysplasia, retinal disease, scleroderma, osteoporosis, renal ischemia, myocardial infarction, cerebral stroke, cerebral ischemia, nephritis, hepatitis, glomerulonephritis, cryptogenic fibrosing aveolitis, psoriasis, transplant rejection, atopic dermatitis, vasculitis, allergy, seasonal allergic rhinitis, reversible airway obstruction, adult respiratory distress syndrome, asthma, chronic obstructive pulmonary disease (COPD) and bronchitis in a subject comprising administering to the subject in need of such treatment a therapeutically effective amount of at least one compound of claim 1 , or a pharmaceutically acceptable salt, solvate or ester thereof.
99. A method of treating a condition or disease associated with COPD, comprising: administering to the subject in need of such treatment a therapeutically effective amount of at least one compound of claim 1 or a pharmaceutically acceptable salt, solvate or ester thereof.
100. A method of treating a condition or disease associated with rheumatoid arthritis, comprising: administering to the subject in need of such treatment a therapeutically effective amount of at least one compound of claim 1 or a pharmaceutically acceptable salt, solvate or ester thereof.
101. A method of treating a condition or disease associated with Crohn's disease, comprising: administering to the subject in need of such treatment a therapeutically effective amount of at least one compound of claim 1 or a pharmaceutically acceptable salt, solvate or ester thereof.
102. A method of treating a condition or disease associated with psoriasis, comprising: administering to the subject in need of such treatment a therapeutically effective amount of at least one compound of claim 1 or a pharmaceutically acceptable salt, solvate or ester thereof.
103. A method of treating a condition or disease associated with ankylosing spondylitis, comprising: administering to the subject in need of such treatment a therapeutically effective amount of at least one compound of claim 1 or a pharmaceutically acceptable salt, solvate or ester thereof.
104. A method of treating a condition or disease associated with sciatica, comprising: administering to the subject in need of such treatment a therapeutically effective amount of at least one compound of claim 1 or a pharmaceutically acceptable salt, solvate or ester thereof.
105. A method of treating a condition or disease associated with complex regional pain syndrome, comprising: administering to the subject in need of such treatment a therapeutically effective amount of at least one compound of claim 1 or a pharmaceutically acceptable salt, solvate or ester thereof.
106. A method of treating a condition or disease associated with psoriatic arthritis, comprising: administering to the subject in need of such treatment a therapeutically effective amount of at least one compound of claim 1 or a pharmaceutically acceptable salt, solvate or ester thereof.
107. A method of treating a condition or disease associated with multiple sclerosis, comprising: administering to the subject in need of such treatment a therapeutically effective amount of at least one compound of claim 1 or a pharmaceutically acceptable salt, solvate or isomer thereof, in combination with a compound selected from the group consisting of Avonex®, Betaseron, Copaxone or other compounds indicated for the treatment of multiple sclerosis.
108. The method of claim 96, further comprising administering to said subject a therapeutically effective amount of at least one medicament selected from the group consisting of disease modifying anti-rheumatic drugs (DMARDS), non-steroidal anti-inflammatory drugs (NSAIDs), cycloxygenase-2 selective (COX-2) inhibitors, COX-1 inhibitors, immunosuppressives, biological response modifiers (BRMs)1 anti-inflammatory agents and H1 antagonists.
109. A method of claim 97, further comprising administering to said subject a therapeutically effective amount of at least one medicament selected from the group consisting of DMARDS, NSAIDs, COX-2 inhibitors, COX-1 inhibitors, immunosuppressives, BRMs, anti-inflammatory agents and H1 antagonists.
110. A method of claim 98, further comprising administering to said subject a therapeutically effective amount of at least one medicament selected from the group consisting of DMARDS, NSAIDs, COX-2 inhibitors, COX-1 inhibitors, immunosuppressives, BRMs, anti-inflammatory agents and H1 antagonists.
PCT/US2007/001030 2006-01-17 2007-01-16 Hydantoin compounds for the treatment of inflammatory disorders WO2007084455A1 (en)

Priority Applications (7)

Application Number Priority Date Filing Date Title
EP07716637.9A EP1973900B1 (en) 2006-01-17 2007-01-16 Hydantoin compounds for the treatment of inflammatory disorders
CA002637198A CA2637198A1 (en) 2006-01-17 2007-01-16 Hydantoin compounds for the treatment of inflammatory disorders
JP2008551312A JP2009523797A (en) 2006-01-17 2007-01-16 Hydantoin compounds for the treatment of inflammatory disorders
AU2007207711A AU2007207711A1 (en) 2006-01-17 2007-01-16 Hydantoin compounds for the treatment of inflammatory disorders
BRPI0706598-1A BRPI0706598A2 (en) 2006-01-17 2007-01-16 compounds for the treatment of inflammatory disorders
IL192691A IL192691A0 (en) 2006-01-17 2008-07-08 Hydantoin compounds for the treatment of inflammatory disorders
NO20083568A NO20083568L (en) 2006-01-17 2008-08-15 Hydantoin compounds for the treatment of inflammatory disorders

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US75930006P 2006-01-17 2006-01-17
US60/759,300 2006-01-17

Publications (1)

Publication Number Publication Date
WO2007084455A1 true WO2007084455A1 (en) 2007-07-26

Family

ID=38038495

Family Applications (2)

Application Number Title Priority Date Filing Date
PCT/US2007/001030 WO2007084455A1 (en) 2006-01-17 2007-01-16 Hydantoin compounds for the treatment of inflammatory disorders
PCT/US2007/001025 WO2007084451A1 (en) 2006-01-17 2007-01-16 Hydantoin derivatives for the treatment of inflammatory disorders

Family Applications After (1)

Application Number Title Priority Date Filing Date
PCT/US2007/001025 WO2007084451A1 (en) 2006-01-17 2007-01-16 Hydantoin derivatives for the treatment of inflammatory disorders

Country Status (17)

Country Link
US (4) US7524842B2 (en)
EP (2) EP1973900B1 (en)
JP (2) JP2009523797A (en)
KR (2) KR20080085221A (en)
CN (2) CN101405286A (en)
AR (2) AR059036A1 (en)
AU (2) AU2007207707A1 (en)
BR (2) BRPI0706599A2 (en)
CA (2) CA2637196A1 (en)
EC (2) ECSP088625A (en)
IL (2) IL192690A0 (en)
NO (2) NO20083568L (en)
PE (2) PE20071240A1 (en)
RU (2) RU2008133383A (en)
TW (2) TW200736247A (en)
WO (2) WO2007084455A1 (en)
ZA (1) ZA200806072B (en)

Cited By (25)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007084415A2 (en) * 2006-01-17 2007-07-26 Schering Corporation Compounds for the treatment of inflammatory disorders
EP1848435A2 (en) * 2005-01-25 2007-10-31 Synta Pharmaceuticals Corporation Compounds for inflammation and immune-related uses
WO2008058449A1 (en) 2006-11-15 2008-05-22 Tianjin Hemay Bio-Tech Co. Ltd Pyrroline-2-one derivatives against cell releasing tumor necrosis factor, preparation methods and uses thereof
US7482370B2 (en) 2004-07-16 2009-01-27 Schering Corporation Compounds for the treatment of inflammatory disorders
US7488745B2 (en) 2004-07-16 2009-02-10 Schering Corporation Compounds for the treatment of inflammatory disorders
EP2083011A1 (en) * 2006-11-15 2009-07-29 Tian Jin Hemay Bio-Tech Co., Ltd. Pyrroline derivatives against cell releasing tumor necrosis factor, preparation methods and uses thereof
WO2010088368A2 (en) 2009-01-29 2010-08-05 Schering Corporation Imidazopyrazines as protein kinase inhibitors
US7772263B2 (en) 2006-01-17 2010-08-10 Schering Corporation Compounds for the treatment of inflammatory disorders
WO2012005229A1 (en) 2010-07-08 2012-01-12 科研製薬株式会社 N-hydroxyformamide derivative and pharmaceutical containing same
WO2013085017A1 (en) 2011-12-09 2013-06-13 科研製薬株式会社 Pyridone derivative and medicine containing same
WO2014196623A1 (en) 2013-06-07 2014-12-11 科研製薬株式会社 (+)-5-(3,4-difluorophenyl)-5-[(3-methyl-2-oxopyridin-1(2h)-yl)methyl]imidazolidine-2,4-dione and drug containing same
US9266886B2 (en) 2014-02-03 2016-02-23 Vitae Pharmaceuticals, Inc. Dihydropyrrolopyridine inhibitors of ROR-gamma
US9481674B1 (en) 2016-06-10 2016-11-01 Vitae Pharmaceuticals, Inc. Dihydropyrrolopyridine inhibitors of ROR-gamma
US9663515B2 (en) 2014-11-05 2017-05-30 Vitae Pharmaceuticals, Inc. Dihydropyrrolopyridine inhibitors of ROR-gamma
US9796710B2 (en) 2014-10-14 2017-10-24 Vitae Pharmaceuticals, Inc. Dihydropyrrolopyridine inhibitors of ROR-gamma
US9845308B2 (en) 2014-11-05 2017-12-19 Vitae Pharmaceuticals, Inc. Isoindoline inhibitors of ROR-gamma
KR20190026917A (en) * 2016-07-20 2019-03-13 노파르티스 아게 Aminopyridine derivatives and their use as selective ALK-2 inhibitors
US10301261B2 (en) 2015-08-05 2019-05-28 Vitae Pharmaceuticals, Llc Substituted indoles as modulators of ROR-gamma
US10829481B2 (en) 2016-01-29 2020-11-10 Vitae Pharmaceuticals, Llc Benzimidazole derivatives as modulators of ROR-gamma
US10913739B2 (en) 2017-07-24 2021-02-09 Vitae Pharmaceuticals, LLC (121374) Inhibitors of RORγ
US11008340B2 (en) 2015-11-20 2021-05-18 Vitae Pharmaceuticals, Llc Modulators of ROR-gamma
US11186573B2 (en) 2017-07-24 2021-11-30 Vitae Pharmaceuticals, Llc Inhibitors of ROR gamma
US11738026B2 (en) 2019-11-22 2023-08-29 Incyte Corporation Combination therapy comprising an ALK2 inhibitor and a JAK2 inhibitor
US11938134B2 (en) 2017-03-10 2024-03-26 Eikonizo Therapeutics, Inc. Metalloenzyme inhibitor compounds
KR102667331B1 (en) 2016-07-20 2024-05-21 노파르티스 아게 Aminopyridine derivatives and their use as selective alk-2 inhibitors

Families Citing this family (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7902352B2 (en) 2005-05-06 2011-03-08 Medtronic, Inc. Isolated nucleic acid duplex for reducing huntington gene expression
EP1948638B1 (en) * 2005-08-12 2011-08-03 Schering Corporation Compounds for the treatment of inflammatory disorders
CA2661605A1 (en) 2006-08-31 2008-03-06 Schering Corporation Hydantoin derivatives useful as antibacterial agents
TW201024303A (en) 2008-09-24 2010-07-01 Schering Corp Compounds for the treatment of inflammatory disorders
AR073307A1 (en) 2008-09-24 2010-10-28 Schering Corp COMPOUNDS FOR THE TREATMENT OF INFLAMMATORY DISORDERS
EP2356111A1 (en) * 2008-11-10 2011-08-17 Schering Corporation Compounds for the treatment of inflammatory disorders
WO2010054278A2 (en) 2008-11-10 2010-05-14 Schering Corporation Compounds for the treatment of inflammatory disorders
US8772276B2 (en) * 2011-05-03 2014-07-08 Merck Sharp & Dohme Corp. Alkyne benzotriazole derivatives
AR092971A1 (en) * 2012-10-26 2015-05-06 Lilly Co Eli AGRECANASA INHIBITORS
CN104803861B (en) * 2014-01-27 2017-05-24 上海博邦医药科技有限公司 Method for synthesizing tapentadol hydrochloride
US10183934B2 (en) 2015-02-02 2019-01-22 Forma Therapeutics, Inc. Bicyclic [4,6,0] hydroxamic acids as HDAC inhibitors
WO2016126725A1 (en) 2015-02-02 2016-08-11 Forma Therapeutics, Inc. 3-alkyl-4-amido-bicyclic [4,5,0] hydroxamic acids as hdac inhibitors
US10407408B2 (en) * 2015-07-09 2019-09-10 Mitsubishi Tanabe Pharma Corporation Imide derivatives and use thereof as medicine
CN105153048B (en) * 2015-07-31 2017-10-24 苏州大学 A kind of preparation method of 2,4 quinazoline diones class compound
WO2017218950A1 (en) 2016-06-17 2017-12-21 Forma Therapeutics, Inc. 2-spiro-5- and 6-hydroxamic acid indanes as hdac inhibitors
MX2018015872A (en) 2016-06-29 2019-04-22 Orion Corp Benzodioxane derivatives and their pharmaceutical use.
CN107311962A (en) * 2017-07-12 2017-11-03 安徽省诚联医药科技有限公司 The preparation method of the net intermediates of Yi Palie
CN107382679A (en) * 2017-07-12 2017-11-24 安徽省诚联医药科技有限公司 The preparation method of Dapagliflozin intermediate
CN107573311A (en) * 2017-08-09 2018-01-12 江苏工程职业技术学院 A kind of synthetic method of Dapagliflozin
CN107556309B (en) * 2017-09-11 2020-12-01 浙江永宁药业股份有限公司 Pharmaceutical application of polysubstituted tetrahydronaphthyridine compound and preparation method thereof
EP3822271A4 (en) 2018-07-03 2022-04-13 Jiangsu Hengrui Medicine Co., Ltd. Pyridopyrimidine derivative, preparation method therefor and medical use thereof
WO2021204185A1 (en) * 2020-04-10 2021-10-14 深圳信立泰药业股份有限公司 Benzo[d]azepine derivative as inhibitor of aggrecanase-2, preparation method therefor, and pharmaceutical use thereof
CN113754635A (en) * 2020-06-02 2021-12-07 成都康弘药业集团股份有限公司 Fused ring compound and preparation method and application thereof
IL298767A (en) 2020-06-16 2023-02-01 Incyte Corp Alk2 inhibitors for the treatment of anemia

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004024721A1 (en) * 2002-09-13 2004-03-25 Astrazeneca Ab Hydantoin derivatives und deren verwendung als tace inhibitoren
WO2006019768A1 (en) * 2004-07-16 2006-02-23 Schering Corporation Hydantoin derivatives for the treatment of inflammatory disorders

Family Cites Families (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6228869B1 (en) * 1996-10-16 2001-05-08 American Cyanamid Company Ortho-sulfonamido bicyclic hydroxamic acids as matrix metalloproteinase and TACE inhibitors
DE60008548T2 (en) * 1999-08-18 2004-08-05 Warner-Lambert Co. Llc HYDROXAMIC ACID DERIVATIVES AS MATRIX METALLOPROTEINASE INHIBITORS
ES2215893T3 (en) * 2000-03-17 2004-10-16 Bristol-Myers Squibb Pharma Company DERIVATIVES OF BETA-AMINO ACIDS AS INHIBITORS OF METALOPROTEASES OF MATRIX AND TNF-ALFA.
SK10932003A3 (en) 2001-03-15 2004-04-06 Astrazeneca Ab Metalloproteinase inhibitors
SE0100902D0 (en) 2001-03-15 2001-03-15 Astrazeneca Ab Compounds
ATE445400T1 (en) 2001-05-25 2009-10-15 Bristol Myers Squibb Co HYDANTION DERIVATIVES AS INHIBITORS OF MATRIX METALLOPROTEINASES
AU2002346729A1 (en) 2001-12-20 2003-07-09 Bristol-Myers Squibb Company Barbituric acid derivatives as inhibitors of tnf-$g(a) converting enzyme (tace) and/or matrix metalloproteinases
AU2002357312A1 (en) 2001-12-20 2003-07-09 Bristol-Myers Squibb Company Barbituric acid derivatives as inhibitors of tnf-alpha converting enzyme (tace) and/or matrix metalloproteinases
AU2003261319A1 (en) 2002-08-01 2004-02-23 Bristol-Myers Squibb Company Hydantoin derivatives as inhibitors of matrix metalloproteinases and/or tnf-alpha converting enzyme
GB0221250D0 (en) 2002-09-13 2002-10-23 Astrazeneca Ab Compounds
AU2003282920A1 (en) * 2002-10-04 2004-05-04 Bristol-Myers Squibb Company Hydantoin derivatives as inhibitors of matrix metalloproteinases and/or tnf-alpha converting enzyme (tace)
EP1581488B1 (en) 2002-12-19 2009-09-16 Vertex Pharmaceuticals Incorporated Inhibitors of tace
US7488745B2 (en) * 2004-07-16 2009-02-10 Schering Corporation Compounds for the treatment of inflammatory disorders
US7504424B2 (en) * 2004-07-16 2009-03-17 Schering Corporation Compounds for the treatment of inflammatory disorders
AR059036A1 (en) * 2006-01-17 2008-03-12 Schering Corp COMPOUNDS FOR THE TREATMENT OF INFLAMMATORY DISORDERS
CA2661605A1 (en) * 2006-08-31 2008-03-06 Schering Corporation Hydantoin derivatives useful as antibacterial agents

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004024721A1 (en) * 2002-09-13 2004-03-25 Astrazeneca Ab Hydantoin derivatives und deren verwendung als tace inhibitoren
WO2006019768A1 (en) * 2004-07-16 2006-02-23 Schering Corporation Hydantoin derivatives for the treatment of inflammatory disorders
US20060205797A1 (en) * 2004-07-16 2006-09-14 Schering Corporation Compounds for the treatment of inflammatory disorders

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
KNAGGS A R ET AL: "Biotransformation of Alosetron: Mechanism of Hydantoin Formation", TETRAHEDRON LETTERS, ELSEVIER, AMSTERDAM, NL, vol. 36, no. 3, 16 January 1995 (1995-01-16), pages 477 - 480, XP004028856, ISSN: 0040-4039 *

Cited By (55)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7879890B2 (en) 2004-07-16 2011-02-01 Schering Corporation Compounds for the treatment of inflammatory disorders
US7482370B2 (en) 2004-07-16 2009-01-27 Schering Corporation Compounds for the treatment of inflammatory disorders
US7488745B2 (en) 2004-07-16 2009-02-10 Schering Corporation Compounds for the treatment of inflammatory disorders
US7504424B2 (en) 2004-07-16 2009-03-17 Schering Corporation Compounds for the treatment of inflammatory disorders
US7683085B2 (en) 2004-07-16 2010-03-23 Schering Corporation Compounds for the treatment of inflammatory disorders
US7687527B2 (en) 2004-07-16 2010-03-30 Schering Corporation Compounds for the treatment of inflammatory disorders
EP1848435A2 (en) * 2005-01-25 2007-10-31 Synta Pharmaceuticals Corporation Compounds for inflammation and immune-related uses
US9090570B2 (en) 2005-01-25 2015-07-28 Synta Pharmaceuticals Corp. Compounds for inflammation and immune-related uses
US8518950B2 (en) 2005-01-25 2013-08-27 Synta Pharmaceuticals Corp. 2-amido pyrazines for inflammation and immune related uses
EP1848435A4 (en) * 2005-01-25 2010-09-29 Synta Pharmaceuticals Corp Compounds for inflammation and immune-related uses
WO2007084415A3 (en) * 2006-01-17 2007-10-18 Schering Corp Compounds for the treatment of inflammatory disorders
WO2007084415A2 (en) * 2006-01-17 2007-07-26 Schering Corporation Compounds for the treatment of inflammatory disorders
US7772263B2 (en) 2006-01-17 2010-08-10 Schering Corporation Compounds for the treatment of inflammatory disorders
EP2083011A1 (en) * 2006-11-15 2009-07-29 Tian Jin Hemay Bio-Tech Co., Ltd. Pyrroline derivatives against cell releasing tumor necrosis factor, preparation methods and uses thereof
EP2093228A1 (en) * 2006-11-15 2009-08-26 Tianjin Hemey Bio-Tech Co., Ltd. Pyrroline-2-one derivatives against cell releasing tumor necrosis factor, preparation methods and uses thereof
EP2093228A4 (en) * 2006-11-15 2009-11-11 Tianjin Hemey Bio Tech Co Ltd Pyrroline-2-one derivatives against cell releasing tumor necrosis factor, preparation methods and uses thereof
EP2083011A4 (en) * 2006-11-15 2011-03-09 Tian Jin Hemay Bio Tech Co Ltd Pyrroline derivatives against cell releasing tumor necrosis factor, preparation methods and uses thereof
WO2008058449A1 (en) 2006-11-15 2008-05-22 Tianjin Hemay Bio-Tech Co. Ltd Pyrroline-2-one derivatives against cell releasing tumor necrosis factor, preparation methods and uses thereof
WO2010088368A2 (en) 2009-01-29 2010-08-05 Schering Corporation Imidazopyrazines as protein kinase inhibitors
WO2012005229A1 (en) 2010-07-08 2012-01-12 科研製薬株式会社 N-hydroxyformamide derivative and pharmaceutical containing same
WO2013085016A1 (en) * 2011-12-09 2013-06-13 科研製薬株式会社 Pyridone derivative and medicine containing same
JPWO2013085016A1 (en) * 2011-12-09 2015-04-27 科研製薬株式会社 Pyridone derivative and pharmaceutical containing the same
US10000476B2 (en) 2011-12-09 2018-06-19 Kaken Pharmaceutical Co., Ltd. Pyridone derivative, pharmaceutical containing the same and methods of use thereof
WO2013085017A1 (en) 2011-12-09 2013-06-13 科研製薬株式会社 Pyridone derivative and medicine containing same
US9447072B2 (en) 2011-12-09 2016-09-20 Kaken Pharmaceutical Co., Ltd. Pyridone derivative and pharmaceutical containing same
WO2014196623A1 (en) 2013-06-07 2014-12-11 科研製薬株式会社 (+)-5-(3,4-difluorophenyl)-5-[(3-methyl-2-oxopyridin-1(2h)-yl)methyl]imidazolidine-2,4-dione and drug containing same
KR20160016834A (en) 2013-06-07 2016-02-15 가껭세이야꾸가부시기가이샤 (+)-5-(3,4-difluorophenyl)-5-[(3-methyl-2-oxopyridin-1(2h)-yl)methyl]imidazolidine-2,4-di-one and drug containing same
KR20210133326A (en) 2013-06-07 2021-11-05 가껭세이야꾸가부시기가이샤 (+)-5-(3,4-difluorophenyl)-5-[(3-methyl-2-oxopyridin-1(2h)-yl)methyl]imidazolidine-2,4-di-one and drug containing same
US9518041B2 (en) 2013-06-07 2016-12-13 Kaken Pharmaceutical Co., Ltd. (+)-5-(3,4-difluorophenyl)-5-[(3-methyl-2-oxopyridin-1(2H)-yl)methyl]imidazolidine-2,4-dione and drug containing same
US9266886B2 (en) 2014-02-03 2016-02-23 Vitae Pharmaceuticals, Inc. Dihydropyrrolopyridine inhibitors of ROR-gamma
US9624217B2 (en) 2014-02-03 2017-04-18 Vitae Pharmaceuticals, Inc. Dihydropyrrolopyridine inhibitors of ROR-gamma
US10047085B2 (en) 2014-02-03 2018-08-14 Vitae Pharmaceuticals, Inc. Dihydropyrrolopyridine inhibitors of ROR-gamma
US11535614B2 (en) 2014-02-03 2022-12-27 Vitae Pharmaceuticals, Llc Dihydropyrrolopyridine inhibitors of ROR-gamma
US10399976B2 (en) 2014-02-03 2019-09-03 Vitae Pharmaceuticals, Llc Dihydropyrrolopyridine inhibitors of ROR-gamma
US10807980B2 (en) 2014-02-03 2020-10-20 Vitae Pharmaceuticals, Llc Dihydropyrrolopyridine inhibitors of ROR-gamma
US9796710B2 (en) 2014-10-14 2017-10-24 Vitae Pharmaceuticals, Inc. Dihydropyrrolopyridine inhibitors of ROR-gamma
US10087184B2 (en) 2014-10-14 2018-10-02 Vitae Pharmaceuticals, Inc. Dihydropyrrolopyridine inhibitors of RORγ
US9663515B2 (en) 2014-11-05 2017-05-30 Vitae Pharmaceuticals, Inc. Dihydropyrrolopyridine inhibitors of ROR-gamma
US9845308B2 (en) 2014-11-05 2017-12-19 Vitae Pharmaceuticals, Inc. Isoindoline inhibitors of ROR-gamma
US11001583B2 (en) 2014-11-05 2021-05-11 Vitae Pharmaceuticals, Llc Dihydropyrrolopyridine inhibitors of ROR-gamma
US10301261B2 (en) 2015-08-05 2019-05-28 Vitae Pharmaceuticals, Llc Substituted indoles as modulators of ROR-gamma
US10829448B2 (en) 2015-08-05 2020-11-10 Vitae Pharmaceuticals, Llc Substituted benzoimidazoles as modulators of ROR-γ
US11008340B2 (en) 2015-11-20 2021-05-18 Vitae Pharmaceuticals, Llc Modulators of ROR-gamma
US10829481B2 (en) 2016-01-29 2020-11-10 Vitae Pharmaceuticals, Llc Benzimidazole derivatives as modulators of ROR-gamma
US9481674B1 (en) 2016-06-10 2016-11-01 Vitae Pharmaceuticals, Inc. Dihydropyrrolopyridine inhibitors of ROR-gamma
US10947218B2 (en) 2016-07-20 2021-03-16 Novartis Ag Aminopyridine derivatives and their use as selective ALK-2 inhibitors
KR102281550B1 (en) * 2016-07-20 2021-07-27 노파르티스 아게 Aminopyridine derivatives and their use as selective ALK-2 inhibitors
KR102667331B1 (en) 2016-07-20 2024-05-21 노파르티스 아게 Aminopyridine derivatives and their use as selective alk-2 inhibitors
KR20220143770A (en) * 2016-07-20 2022-10-25 노파르티스 아게 Aminopyridine derivatives and their use as selective alk-2 inhibitors
KR20190026917A (en) * 2016-07-20 2019-03-13 노파르티스 아게 Aminopyridine derivatives and their use as selective ALK-2 inhibitors
KR102559539B1 (en) 2016-07-20 2023-07-26 노파르티스 아게 Aminopyridine derivatives and their use as selective alk-2 inhibitors
US11938134B2 (en) 2017-03-10 2024-03-26 Eikonizo Therapeutics, Inc. Metalloenzyme inhibitor compounds
US10913739B2 (en) 2017-07-24 2021-02-09 Vitae Pharmaceuticals, LLC (121374) Inhibitors of RORγ
US11186573B2 (en) 2017-07-24 2021-11-30 Vitae Pharmaceuticals, Llc Inhibitors of ROR gamma
US11738026B2 (en) 2019-11-22 2023-08-29 Incyte Corporation Combination therapy comprising an ALK2 inhibitor and a JAK2 inhibitor

Also Published As

Publication number Publication date
ECSP088626A (en) 2008-08-29
US20100120838A1 (en) 2010-05-13
IL192690A0 (en) 2009-02-11
CA2637196A1 (en) 2007-07-26
PE20071241A1 (en) 2008-01-14
ZA200806072B (en) 2009-04-29
PE20071240A1 (en) 2008-01-14
BRPI0706599A2 (en) 2011-03-29
KR20080085221A (en) 2008-09-23
TW200738688A (en) 2007-10-16
EP1973901B1 (en) 2014-12-17
EP1973901A1 (en) 2008-10-01
RU2008133383A (en) 2010-02-27
AU2007207711A1 (en) 2007-07-26
US20070197564A1 (en) 2007-08-23
CN101405286A (en) 2009-04-08
RU2008133382A (en) 2010-02-27
US7772263B2 (en) 2010-08-10
US8178553B2 (en) 2012-05-15
US7683088B2 (en) 2010-03-23
WO2007084451A1 (en) 2007-07-26
IL192691A0 (en) 2009-02-11
JP2009523796A (en) 2009-06-25
US20090156586A1 (en) 2009-06-18
NO20083568L (en) 2008-10-16
CA2637198A1 (en) 2007-07-26
NO20083567L (en) 2008-10-16
AR059036A1 (en) 2008-03-12
ECSP088625A (en) 2008-08-29
CN101410390A (en) 2009-04-15
JP2009523797A (en) 2009-06-25
AU2007207707A1 (en) 2007-07-26
BRPI0706598A2 (en) 2011-04-05
TW200736247A (en) 2007-10-01
US20070265299A1 (en) 2007-11-15
AR059037A1 (en) 2008-03-12
US7524842B2 (en) 2009-04-28
EP1973900A1 (en) 2008-10-01
KR20080085222A (en) 2008-09-23
EP1973900B1 (en) 2017-04-05

Similar Documents

Publication Publication Date Title
US7683088B2 (en) Compounds for the treatment of inflammatory disorders
EP1778676B1 (en) Hydantoin derivatives for the treatment of inflammatory disorders
US7687527B2 (en) Compounds for the treatment of inflammatory disorders
EP2356111A1 (en) Compounds for the treatment of inflammatory disorders
EP2331528B1 (en) Compounds for the treatment of inflammatory disorders
EP2346857B1 (en) Compounds for the treatment of inflammatory disorders
US7879997B2 (en) Compounds for the treatment of inflammatory disorders
MX2008009297A (en) Hydantoin compounds for the treatment of inflammatory disorders
US20090239890A1 (en) Spiro substituted cyclopropane compounds for the treatment of inflammatory disorders
MX2008009295A (en) Hydantoin derivatives for the treatment of inflammatory disorders

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application
REEP Request for entry into the european phase

Ref document number: 2007716637

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2007716637

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 12008501595

Country of ref document: PH

WWE Wipo information: entry into national phase

Ref document number: 569663

Country of ref document: NZ

Ref document number: 192691

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: 2007207711

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 08072554

Country of ref document: CO

WWE Wipo information: entry into national phase

Ref document number: 2637198

Country of ref document: CA

Ref document number: 3640/CHENP/2008

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: 2008551312

Country of ref document: JP

WWE Wipo information: entry into national phase

Ref document number: MX/a/2008/009297

Country of ref document: MX

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2007207711

Country of ref document: AU

Date of ref document: 20070116

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 1020087019785

Country of ref document: KR

ENP Entry into the national phase

Ref document number: 2008133382

Country of ref document: RU

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 200780009127.8

Country of ref document: CN

ENP Entry into the national phase

Ref document number: PI0706598

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20080717