WO2001012609A1 - Pyrazoloanthrone and derivatives thereof as jnk inhibitors and their compositions - Google Patents

Pyrazoloanthrone and derivatives thereof as jnk inhibitors and their compositions Download PDF

Info

Publication number
WO2001012609A1
WO2001012609A1 PCT/US2000/022727 US0022727W WO0112609A1 WO 2001012609 A1 WO2001012609 A1 WO 2001012609A1 US 0022727 W US0022727 W US 0022727W WO 0112609 A1 WO0112609 A1 WO 0112609A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
alkyl
jnk
pyrazol
aryl
Prior art date
Application number
PCT/US2000/022727
Other languages
French (fr)
Inventor
Brydon L. Bennett
Shripad S. Bhagwat
Anthony M. Manning
Brion W. Murray
Eoin C. O'leary
Yoshitaka Satoh
Original Assignee
Signal Pharmaceuticals, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Signal Pharmaceuticals, Inc. filed Critical Signal Pharmaceuticals, Inc.
Priority to CA002383268A priority Critical patent/CA2383268A1/en
Priority to IL14818700A priority patent/IL148187A0/en
Priority to EP00955709A priority patent/EP1218347A1/en
Priority to NZ517578A priority patent/NZ517578A/en
Priority to JP2001517507A priority patent/JP2003532626A/en
Priority to AU67863/00A priority patent/AU780306B2/en
Publication of WO2001012609A1 publication Critical patent/WO2001012609A1/en
Priority to HK02109283.5A priority patent/HK1053107A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D231/00Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings
    • C07D231/54Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings condensed with carbocyclic rings or ring systems
    • 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
    • A61P1/16Drugs 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
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • A61P21/02Muscle relaxants, e.g. for tetanus or cramps
    • 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
    • A61P25/08Antiepileptics; Anticonvulsants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • A61P25/16Anti-Parkinson drugs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • 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
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/24Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom with substituted hydrocarbon radicals attached to ring carbon atoms
    • C07D213/36Radicals substituted by singly-bound nitrogen atoms
    • C07D213/38Radicals substituted by singly-bound nitrogen atoms having only hydrogen or hydrocarbon radicals attached to the substituent nitrogen atom
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D295/00Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms
    • C07D295/04Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms with substituted hydrocarbon radicals attached to ring nitrogen atoms
    • C07D295/12Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms with substituted hydrocarbon radicals attached to ring nitrogen atoms substituted by singly or doubly bound nitrogen atoms
    • C07D295/125Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms with substituted hydrocarbon radicals attached to ring nitrogen atoms substituted by singly or doubly bound nitrogen atoms with the ring nitrogen atoms and the substituent nitrogen atoms attached to the same carbon chain, which is not interrupted by carbocyclic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D333/00Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom
    • C07D333/02Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom not condensed with other rings
    • C07D333/04Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom not condensed with other rings not substituted on the ring sulphur atom
    • C07D333/26Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom not condensed with other rings not substituted on the ring sulphur atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D333/38Carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D409/00Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms
    • C07D409/02Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings
    • C07D409/12Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/04Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings directly linked by a ring-member-to-ring-member bond

Definitions

  • TECHNICAL FIELD This invention is generally directed to pyrazoloanthrone and derivatives thereof which have utility over a wide range of indications, including activity as Jun N- terminal kinase inhibitors, and related compositions and methods.
  • JNK Jun N-terminal kinase pathway
  • c-jun and ATF2 Whitmarsh A.J., and Davis R.J. J. Mol. Med. 74:589-607, 1996).
  • bZIP basic leucine zipper
  • JNK binds to the N-terminal region of c-jun and ATF-2 and phosphorylates two sites within the activation domain of each transcription factor (Hibi M., Lin A., Smeal T., Minden A., Karin M. Genes Dev. 7:2135-2148. 1993; Mohit A.A., Martin M.H., and Miller CA. Neuron 14:67-75, 199)].
  • Three JNK enzymes have been identified as products of distinct genes (Hibi et al, supra; Mohit et al., supra).
  • Ten different isoforms of JNK have been identified. These represent alternatively spliced forms of three different genes: JNK1, JNK2 and JNK3.
  • JNK1 and 2 are ubiquitously expressed in human tissues, whereas JNK3 is selectively expressed in the brain, heart and testis (Dong, C, Yang, D., Wysk, M., Whitmarsh, A., Davis, R., Flavell, R. Science 270:1-4, 1998).
  • Gene transcripts are alternatively spliced to produce four- JNK 1 isoforms, four-JNK2 isoforms and two-JNK3 isoforms.
  • JNK1 and 2 are expressed widely in mammalian tissues, whereas JNK3 is expressed almost exclusively in the brain. Selectivity of JNK signaling is achieved via specific interactions of JNK pathway components and by use of scaffold proteins that selectively bind multiple components of the signaling cascade.
  • JIP-1 JNK-interacting protein- 1 selectively binds the MAPK module, MLK -> JNKK1 - JNK.12, 13 It has no binding affinity for a variety of other MAPK cascade enzymes. Different scaffold proteins are likely to exist for other MAPK signaling cascades to preserve substrate specificity. JNKs are activated by dual phosphorylation on Thr-183 and Tyr-185.
  • JNKK1 also known as MKK 4
  • JNKK2 MKK7
  • JNKK2 JNKK2
  • JNKK2 JNKK2
  • JNKK2 JNKK2
  • JNKK1 can also phosphorylate and activate p38.
  • JNKK1 and JNKK2 are widely expressed in mammalian tissues.
  • JNKK1 and JNKK2 are activated by the MAPKKK enzymes, MEKK1 and 2 (Lange-Carter C.A., Pleiman CM., Gardner A.M., Blumer K.J., and Johnson G.L. Science 260:315-319, 1993; Yan M., Dai J.C. Deak J.C., Kyriakis J.M., Zon L.I., Woodgett J.R., and Templeton D.J. Nature 372:798-781, 1994). Both MEKK1 and MEKK2 are widely expressed in mammalian tissues.
  • Activation of the JNK pathway has been documented in a number of disease settings, providing the rationale for targeting this pathway for drug discovery.
  • molecular genetic approaches have validated the pathogenic role of this pathway in several diseases.
  • autoimmune and inflammatory diseases arise from the over-activation of the immune system.
  • Activated immune cells express many genes encoding inflammatory molecules, including cytokines, growth factors, cell surface receptors, cell adhesion molecules and degradative enzymes. Many of these genes are regulated by the JNK pathway, through activation of the transcription factors AP-1 and ATF-2, including TNF a, IL-2, E-selectin and matrix metalloproteinases such as collagenase-1 (Manning A.M. and Mercurio F.
  • Monocytes, tissue macrophages and tissue mast cells are key sources of TNF a production.
  • the JNK pathway regulates TNFa production in bacterial lipopolysaccharide-stimulated macrophages, and in mast cells stimulated through the FceRII receptor (Swantek J.L., Cobb M.H., Geppert T.D. Mol. Cell. Biol. 17:627 '4- 6282, 1997; Ishizuka, T., Tereda N., Gerwins, P.. Hamelmann E., Oshiba A., Fanger G.R.. Johnson G.L., and Gelfland E.W. Proc. Nat. Acad.
  • MMPs Matrix metalloproteinases
  • JNK pathway In human rheumatoid synoviocytes activated with TNFa, IL-1, or Fas ligand the JNK pathway is activated (Han Z., Boyle D.L., Aupperle K.R., Bennett B., Manning A.M., Firestein G.S. J. Pharm. Exp. Therap. 291 :1-7, 1999; Okamoto K., Fujisawa K., Hasunuma T., Kobata T., Sumida T., and Nishioka K. Arth & Rheum 40: 919-92615, 1997). Inhibition of JNK activation results in decreased AP-1 activation and collagenase-1 expression (Han et al., supra). The JNK pathway therefore regulates MMP expression in cells involved in rheumatoid arthritis.
  • T lymphocytes Inappropriate activation of T lymphocytes initiates and perpetuates many autoimmune diseases, including asthma, inflammatory bowel disease and multiple sclerosis.
  • the JNK pathway is activated in T cells by antigen stimulation and CD28 receptor co-stimulation and regulates production of the growth factor IL-2 and cellular proliferation (Su B., Jacinto E.. Hibi M.. Kallunki T., Karin M., Ben-Neriah Y. Cell 11:121-136, 1994; Faris M., Kokot N., Lee L., and Nel A.E. J. Biol. Chem. 271 :27366- 27373, 1996).
  • Peripheral T cells from mice genetically deficient in JNKK1 show decreased proliferation and IL-2 production after CD28 co-stimulation and PMA / Ca2+ ionophore activation, providing important validation for the role of the JNK pathway in these cells (Nishina H, Bachmann M., Oliveria-dos-Santos A.J., et al. J. Exp. Med. 186: 941-953, 1997). It is known that T cells activated by antigen receptor stimulation in the absence of accessory cell-derived co-stimulatory signals lose the capacity to synthesize IL-2. a state called clonal anergy. This is an important process by which auto-reactive T cell populations are eliminated from the peripheral circulation.
  • JNK 1 or JNK2 knockout mice develop normally and are phenotypically unremarkable.
  • Activated na ⁇ ve CD4+ T cells from these mice fail to produce IL-2 and do not proliferate well (Sabapathy, K, Hu, Y, Kallunki, T, Schreiber, M, David, J-P, Jochum, W, Wagner, E, Karin, M. Curr Biol 9: 116-125, 1999). It is possible to induce T cell differentiation in T cells from these mice, generating Thl cells (producers of IFN-g and TNF ⁇ ) and Th2 effector cells (producers of IL-4, IL-5, IL-6, IL-10 and IL-13) [22,23].
  • JNK1 and JNK2 are not have redundant functions in T cells and that they play different roles in the control of cell growth, differentiation and death.
  • the JNK pathway therefore, is an important point for regulation of T cell responses to antigen.
  • Cardiovascular disease accounts for nearly one quarter of total annual deaths worldwide.
  • Vascular disorders such as atherosclerosis and restenosis result from dysregulated growth of the vessel wall, restricting blood flow to vital organs.
  • the JNK pathway is activated by atherogenic stimuli and regulates local cytokine and growth factor production in vascular cells (Yang, DD, Conze, D, Whitmarsh, AJ, et al, Immunity, 9:575, 1998).
  • alterations in blood flow, hemodynamic forces and blood volume lead to JNK activation in vascular endothelium, leading to AP-1 activation and pro-atherosclerotic gene expression (Aspenstrom P., Lindberg U., and Hall A. Curr. Biol. 6:70-77, 1996).
  • Ischemia and ischemia coupled with reperfusion in the heart, kidney or brain results in cell death and scar formation, which can ultimately lead to congestive heart failure, renal failure or cerebral dysfunction.
  • reperfusion of previously ischemic donor organs results in acute leukocyte-mediated tissue injury and delay of graft function.
  • the JNK pathway is activated by ischemia and reperfusion (Li Y., Shyy J.. Li S., Lee J., Su B., Karin M., Chien S Mol. Cell. Biol. 16:5947-5954, 1996), leading to the activation of
  • JNK-responsive genes and leukcoyte-mediated tissue damage.
  • JNK activation can be either pro- or anti-apoptotic.
  • JNK activation is correlated with enhanced apoptosis in cardiac tissues following ischemia and reperfusion (Pombo CM, Bonventre JV, Avruch J, Woodgett JR, Kyriakis J.M, Force T.
  • Cancer is characterized by uncontrolled growth, proliferation and migration of cells. Cancer is the second leading cause of death with 500,000 deaths and an estimated 1.3 million new cases in the United States in 1996. The role of signal transduction pathways contributing to cell transformation and cancer is a generally accepted concept. The JNK pathway leading to AP-1 appears to play a critical role in cancer. Expression of c-jun is altered in early lung cancer and may mediate growth factor signaling in non-small cell lung cancer (Yin T., Sandhu G., Wolfgang CD., Burrier A., Webb R.L., Rigel D.F. Hai T.. and Whelan J. J. Biol. Chem. 272:19943- 19950, 1997).
  • the oncogene BCR-Abl associated with t(9,22) Philadelphia chromosome translocation of chronic myelogenous leukemia, activates JNK and leads to transformation of hematopoietic cells (Milne D.M., Campbell L.E., Campbell D.G., Meek D.W. J. Biol. Chem. 270:551 1-5518, 1995).
  • JIP-1 a naturally occurring JNK inhibitory protein, called JIP-1, blocks cellular transformation caused by BCR-Abl expression (Raitano A.B., Halpern J.R., Hambuch T.M., Sawyers C.L. Proc. Nat. Acad.
  • JNK inhibitors may block transformation and tumor cell growth. Accordingly, there is a need in the art for selective inhibitors of JNK, as well as for methods for preparation thereof, pharmaceutical compositions comprising such inhibitors, and methods of inhibiting JNK's and treating diseases in mammals which are responsive to JNK inhibition.
  • the present invention fulfills these needs, and provides further related advantages.
  • the present invention is directed to compounds having activity as selective inhibitors of JNK, as well as to compositions and methods related thereto.
  • JNK inhibitors may generally be classified as "pyrazoloanthrone derivatives" having the following structure (I):
  • R, and R 2 are as defined below, including pharmaceutically acceptable salts thereof.
  • the present invention is also directed to methods for treating a variety of conditions by administering an effective amount of a JNK inhibitor to an animal or subject in need thereof (referred to herein as a "patient"), typically a warm-blooded animal (including a human).
  • a patient typically a warm-blooded animal (including a human).
  • the compounds of this invention are preferably formulated as a pharmaceutical composition which contains an effective dosage amount of one or more JNK inhibitors in combination with one (or more) pharmaceutically acceptable carrier(s).
  • Conditions that may be treated by the compounds of this invention, or a pharmaceutical composition containing the same, include any condition which may benefit from administration of JNK inhibitors, and are particularly useful for the prevention and/or treatment of various diseases including (but not limited to) rheumatoid arthritis, rheumatoid spondylitis, osteoarthritis, gout, asthma, bronchitis, cystic fibrosis, inflammatory bowel disease, irritable bowel syndrome, mucous colitis, ulcerative colitis, Crohn's disease, gastritis, esophagitis, hepatitis, multiple sclerosis, atherosclerosis, restenosis following angioplasty, left ventricular hypertrophy, myocardial infarction, stroke, ischemic damages to heart, kidney, liver, and brain, transplant rejection, endotoxin shock, psoriasis, eczema, dermatitis, epilepsy, Alzheimer's disease, Huntington's disease, Amyotrophic laterial
  • Figure 1 illustrates the ability of a representative compound of this invention to inhibit IL-2 in Jurkat T-Cell.
  • Figure 2 illustrates the ability of a representative compound of this invention to inhibit TNF- in a mouse model of endotoxin shock.
  • Figure 3 illustrates the ability of a representative compound of this invention to inhibit leukocyte recruitment in rat model for inflamed lung.
  • Figure 4 illustrates the ability of a representative compound of this invention to inhibit paw swelling (Figure 4A), joint destruction (Figure 4B), transcription factor AP-1 activation (Figure 4C), and expression of MMP-13 ( Figure 4D) in a rat model for adjuvant arthritis.
  • Figure 5 illustrates the ability of a representative compound of this invention to reduce kainic acid-induced seizure response.
  • the present invention is directed to compounds which have activity as selective inhibitors of JNK, as well as to compositions and methods relating to the same.
  • the compounds of this invention have the following structure (I):
  • R, and R 2 are optional substituents that are the same or different and independently represent alkyl, halogen, nitro, trifluoromethyl, sulfonyl, carboxyl, alkoxycarbonyl, alkoxy, aryl, aryloxy, arylalkyloxy, arylalkyl, cycloalkylalkyloxy, cycloalkyloxy, alkoxyalkyl. alkoxyalkoxy, aminoalkoxy. mono- or di- alkylaminoalkoxy, or a group represented by formula (a), (b), (c) or (d):
  • R, and R 4 taken together represent alkylidene or a heteroatom-containing alkylidene, or R 3 and R 4 are the same or different and independently represent hydrogen, alkyl, cycloalkyl, aryl, arylalkyl, cycloalkylalkyl, aryloxyalkyl, alkoxyalkyl, alkoxyamino, or alkoxy(mono- or di-alkylamino); and
  • R 5 represents hydrogen, alkyl, cycloalkyl, aryl, arylalkyl, cycloalkylalkyl, alkoxy, amino, mono- or di-alkylamino, arylamino, arylalkylamino, cycloalkylamino, or cycloalkylalkylamino.
  • the terms used above having following meaning.
  • Alkyl means a straight chain or branched, saturated or unsaturated alkyl chain having from 1 to 8 carbon atoms, such as methyl, ethyl, n-propyl, iso- propyl, n-butyl, iso-butyl, tert-butyl, propylenyl, 1 -butenyl, propynyl, and the like.
  • Halogen means fluorine, chlorine, bromine or iodine.
  • Carboxyl means -COOH.
  • Alkoxy means -O-(alkyl), such as methoxy, ethoxy. n-propyloxy, iso- propyloxy. n-butyloxy, iso-butyloxy, and the like.
  • Alkoxyalkoxy means -0-(alkyl)-O-(alkyl), such as -OCH 2 CH 2 OCH 3 , and the like.
  • Alkoxyalkyl means -(alkyl)-O-(alkyl), such as -CH 2 OCH 3 , -CH 2 OCH 2 CH 3 , and the like.
  • Aryl means a carbocyclic or heterocyclic aromatic group containing from 5 to 10 ring atoms.
  • the ring atoms of a carbocyclic aryl group are all carbon atoms, and includes phenyl and naphthyl.
  • the ring atoms of a heterocyclic aryl group contains at least one heteroatom selected from nitrogen, oxygen and sulfur, and include pyridinyl, pyrimidinyl, furanyl, thienyl, imidazolyl, thiazolyl, pyrazolyl, pyridazinyl, pyrazinyl, triazinyl, tetrazolyl, and indolyl.
  • Aryloxy means -O-(aryl), such as -O-phenyl, -O-pyridinyl, and the like.
  • Arylalkyl means -(alkyl)-(aryl), such as benzyl (i.e., -CH 2 phenyl), -CH 2 -pyrindinyl, and the like.
  • Arylalkyloxy means -O-(alkyl)-(aryl), such as -O-benzyl, -O-CH 2 - pyridinyl, and the like.
  • Cycloalkyl means a cyclic alkyl having from 3 to 7 carbon atoms, such as cyclopropyl, cyclopentyl, cyclohexyl, and the like.
  • Cycloalkyloxy means -O-(cycloalkyl), such as -O-cyclohexyl, and the like. "Cycloalkylalkyloxy” means -O-(alkyl)-(cycloalkyl, such as
  • Alkylidene means the divalent radical -C n H 2n -, wherein n is an integer from 1 to 8, such as -CH 2 -, -CH 2 CH 2 -, -CH 2 -CH 2 -CH 2 -, -CH 2 CH 2 CH 2 CH 2 -, -CH 2 CH 2 CH 2 CH 2 -, and the like.
  • Heteroatom-containing alkylidene means an alkylidene wherein at least one carbon atom is replaced by a heteroatom selected from nitrogen, oxygen or sulfur, such as -CH 2 CH 2 OCH 2 CH 2 -, and the like.
  • Aminoalkoxy means -0-(alkyl)-NH 2 , such as -OCH 2 NH 2 , -OCH 2 CH 2 NH 2 , and the like.
  • “Mono- or di-alkylamino” means -NH(alkyl) or -N(alkyl)(alkyl), respectively, such as -NHCH 3 , -N(CH 3 ) 2 , and the like.
  • “Mono- or di-alkylaminoalkoxy” means -O-(alkyl)-NH(alkyl) or -O-(alkyl)-N(alkyl)(alkyl), respectively, such as -OCH 2 NHCH 3 , -OCH 2 CH 2 N(CH 3 ) 2 , and the like.
  • “Arylamino” means -NH(aryl), such as -NH-phenyl, -NH-pyridinyl. and the like.
  • Arylalkylamino means -NH-(alkyl)-(aryl), such as -NH-benzyl, -NHCH 2 -pyridinyl, and the like.
  • Alkylamino means -NH(alkyl). such as -NHCH 3 , -NHCH 2 CH 3 , and the like.
  • Cycloalkylamino means -NH-(cycloalkyl), such as -NH-cyclohexyl, and the like.
  • compounds of this invention have one of the following structures (III) or (IV):
  • compounds of this invention have one of the following structures (V), (VI) or (VII):
  • compositions of structure (I) are also within the scope of this invention.
  • the compound may generally be utilized as the free base.
  • the compounds may be used in the form of acid addition salts.
  • Acid addition salts of the free base amino compounds of the present invention may be prepared by methods well known in the art, and may be formed from organic and inorganic acids.
  • Suitable organic acids include maleic, fumaric, benzoic, ascorbic, succinic, methanesulfonic, acetic, oxalic, propionic, tartaric, salicylic, citric, gluconic, lactic, mandelic, cinnamic, aspartic, stearic, palmitic, glycolic, glutamic, and benzenesulfonic acids.
  • Suitable inorganic acids include hydrochloric, hydrobromic, sulfuric, phosphoric, and nitric acids.
  • the compounds of this invention may generally be made by organic synthesis techniques known to those skilled in the art, as well as by the following general techniques and by the procedures set forth in the Examples. To that end, the compounds of this invention may be made according to the following Reaction Schemes 1 through 7.
  • pyrazoloanthrones of this invention may be prepared by condensation of appropriate anthraquinones having a leaving group at the 1 -position (such as fluoro, chloro, bromo, iodo, nitro, methanesulfonyloxy, tosyloxy or phenoxy) with hydrazine in a suitable solvent (such as pyridine, dimethylformamide, methylene chloride, chloroform, or dioxane).
  • a suitable solvent such as pyridine, dimethylformamide, methylene chloride, chloroform, or dioxane.
  • Suitable anthraquinone starting materials are commercially available from a variety of sources with the R, and/or R 2 groups at various positions on the anthraquinone ring.
  • the following reaction schemes depict synthesis of 5- and/or 7-substituted pyrazoloanthrones.
  • pyrazoloanthrones substituted at other positions may be made in a similar manner from the appropriately substituted pyrazoloanthrone starting material.
  • pyrazoloanthrones with 5-amino substituents may be prepared by condensation of 5-chloropyrazoloanthrone with mono- or disubstituted amines at 0 to 250°C for 1 to 16 hours, either in the absence or the presence of a solvent.
  • solvents are pyridine, dimethylformamide, dimethylsulfoxide, dichloroethane, chloroform, tetrahydrofuran, dioxane, diglyme, or triglyme in the presence of excess amount of the amine, or in the presence of an acid quenching agent such as triethylamine, diisopropylefhylamine, sodium bicarbonate, potassium carbonate, or sodium hydroxide.
  • pyrazoloanthrones with 7-amino substituents may be prepared by condensation of 7-chloropyrazoloanthrone with mono- or disubstituted amines at 0 to 250°C for 1 to 16 hours either in the absence or the presence of a solvent.
  • solvents are pyridine, dimethylformamide, dimethylsulfoxide, dichloroethane, chloroform, tetrahydrofuran, dioxane, diglyme. or triglyme in the presence of excess amount of the amine, or in the presence of an acid quenching agent such as triethylamine, diisopropylethylamine, sodium bicarbonate, potassium carbonate, or sodium hydroxide.
  • pyrazoloanthrones with 5-acyl- or sulfonylamino substituents may be prepared by condensation of 5-amino-2-(2- methoxyethoxymethyl)pyrazoloanthrone with acid chlorides and sulfonyl chlorides followed by the deprotection.
  • Condensation of 5-amino-2-(2- methoxyethoxymethyl)pyrazoloanthrone with appropriate acid chlorides R ⁇ COCl or sulfonyl chlorides is carried out in the presence of an acid quenching agent such as triethylamine, diisopropylethylamine, sodium bicarbonate, potassium carbonate, or sodium hydroxide at -20 to 50°C for 0.5 to 16 hours in solvents such as methylene chloride, chloroform, tetrahydrofuran, dioxane, dimefhylformamide, and ethyl acetate.
  • an acid quenching agent such as triethylamine, diisopropylethylamine, sodium bicarbonate, potassium carbonate, or sodium hydroxide at -20 to 50°C for 0.5 to 16 hours in solvents such as methylene chloride, chloroform, tetrahydrofuran, dioxane, dimefhylformamide, and
  • the deprotection step may be performed by the treatment of the product mentioned above with an acid such as trifluoroacetic acid, aqueous hydrochloric acid, aqueous hydrobromic acid, or aqueous sulfuric acid.
  • an acid such as trifluoroacetic acid, aqueous hydrochloric acid, aqueous hydrobromic acid, or aqueous sulfuric acid.
  • the starting material may be prepared in two steps.
  • the 2-position of 5- nitropyrazoloanthrone may be protected by a protective group such as methoxymethyl (MOM), methoxyethoxymethyl (MEM), 2-trimethylsilylethoxymethyl (SEM), or 4- methoxybenzyl (PMB) with an aid of a base such as triethylamine, diisopropylethylamine, pyridine, sodium hexamethyldisilazide, potassium hexame hyldisilazide, or lithium diisopropylamide.
  • a protective group such as methoxymethyl (MOM), methoxyethoxymethyl (MEM), 2-trimethylsilylethoxymethyl (SEM), or 4- methoxybenzyl (PMB) with an aid of a base such as triethylamine, diisopropylethylamine, pyridine, sodium hexamethyldisilazide, potassium hexame hyldisilazide, or
  • the reaction is typically carried out at -40 to 60°C for 1 to 16 hours in a solvent such as methylene chloride, chloroform, tetrahydrofuran, dioxane, or dimethoxy ethane.
  • a solvent such as methylene chloride, chloroform, tetrahydrofuran, dioxane, or dimethoxy ethane.
  • MEM group is preferred.
  • N-Protected 5-nitropyrazoloanthorone is then reduced to its 5-amino derivative by a variety of reducing agents such as Sn or Fe metal in acidic media such as acetic acid or aqueous hydrochloric acid.
  • the reaction is typically run at 20 to 160°C for 1 to 16 hours.
  • the same transformation can be carried out by hydrogenation in the presence of a transition-metal catalyst such as palladium, platinum, rhodium, or iridium with or without a support such as charcoal in a solvent such as ethanol, ethyl acetate, tetrahydrofuran, dioxane, or dimethoxyethane at 1 to 20 atmospheres of hydrogen at 20 to 60°C for 1 to 16 hours.
  • a transition-metal catalyst such as palladium, platinum, rhodium, or iridium with or without a support such as charcoal
  • a solvent such as ethanol, ethyl acetate, tetrahydro
  • pyrazoloanthrones with 7-acyl- or sulfonylamino substituents may be prepared by condensation of 7-amino-2-(2- methoxyethoxymethyl)pyrazoloanthrone with acid chlorides and sulfonyl chlorides followed by the deprotection.
  • Condensation of 7-amino-2-(2- methoxyethoxymethyl)pyrazoloanthrone with appropriate acid chlorides R 6 COCl or sulfonyl chlorides is carried out in the presence of an acid quenching agent such as triethylamine, diisopropylethylamine, sodium bicarbonate, potassium carbonate, or sodium hydroxide at -20 to 50°C for 0.5 to 16 hours in solvents such as methylene chloride, chloroform, tetrahydrofuran, dioxane, dimethylformamide, or ethyl acetate.
  • an acid quenching agent such as triethylamine, diisopropylethylamine, sodium bicarbonate, potassium carbonate, or sodium hydroxide at -20 to 50°C for 0.5 to 16 hours in solvents such as methylene chloride, chloroform, tetrahydrofuran, dioxane, dimethylformamide, or ethyl acetate
  • the deprotection step may be performed by the treatment of the product mentioned above with an acid such as trifluoroacetic acid, aqueous hydrochloric acid, aqueous hydrobromic acid, or aqueous sulfuric acid.
  • an acid such as trifluoroacetic acid, aqueous hydrochloric acid, aqueous hydrobromic acid, or aqueous sulfuric acid.
  • the starting material is prepared in two steps.
  • the 2-position of 7- nitropyrazoloanthrone is protected by a protective group such as methoxymethyl (MOM), methoxyethoxymethyl (MEM), 2-trimethylsilylethoxymethyl (SEM), or 4- methoxybenzyl (PMB) with an aid of a base such as triethylamine, diisopropylethylamine, pyridine, sodium hexamefhyldisilazide, potassium hexamethyldisilazide, or lithium diisopropylamide.
  • a protective group such as methoxymethyl (MOM), methoxyethoxymethyl (MEM), 2-trimethylsilylethoxymethyl (SEM), or 4- methoxybenzyl (PMB) with an aid of a base such as triethylamine, diisopropylethylamine, pyridine, sodium hexamefhyldisilazide, potassium hexamethyldisilazide, or
  • the reaction is typically carried out at -40 to 60°C for 1 to 16 hours in a solvent such as methylene chloride, chloroform, tetrahydrofuran, dioxane. or dimethoxy ethane.
  • a solvent such as methylene chloride, chloroform, tetrahydrofuran, dioxane. or dimethoxy ethane.
  • MEM group is preferred.
  • N-Protected 7-nitropyrazoloanthorone is then reduced to its 7-amino derivative by a variety of reducing agents such as Sn or Fe metal in acidic media such as acetic acid or aqueous hydrochloric acid.
  • the reaction is typically run at 20 to 160°C for 1 to 16 hours.
  • the same transformation can be carried out by hydrogenation in the presence of a transition-metal catalyst such as palladium, platinum, rhodium, or iridium with or without a support such as charcoal in a solvent such as ethanol, ethyl acetate, tetrahydrofuran, dioxane, or dimethoxyethane at 1 to 20 atmospheres of hydrogen at 20 to 60°C for 1 to 16 hours.
  • a transition-metal catalyst such as palladium, platinum, rhodium, or iridium with or without a support such as charcoal in a solvent such as ethanol, ethyl acetate, tetrahydr
  • pyrazoloanthrones with 5-alkoxy substituents may be prepared by condensation of 5-hydroxy-2-(2-methoxyethoxymethyl)- pyrazoloanthrone with alkyl halides and sulfonates R 7 -X followed by the deprotection.
  • the leaving group X chloride, bromide, iodide, methanesulfonate. tosylate, benzenesulfonate, or triflate can be used.
  • Condensation of 5-hydroxy-2-(2- methoxyethoxymethyl)pyrazoloanthrone with appropriate alkyl halides and sulfonates is carried out in the presence of an acid quenching agent such as triethylamine, diisopropylethylamine, sodium bicarbonate, potassium carbonate, or sodium hydroxide at -20 to 50°C for 0.5 to 16 hours in solvents such as methylene chloride, chloroform, tetrahydrofuran, dioxane, dimethylformamide, or ethyl acetate.
  • the deprotection step is performed by the treatment of the product mentioned above with an acid such as trifluoroacetic acid, aqueous hydrochloric acid, aqueous hydrobromic acid, or aqueous sulfuric acid.
  • the starting material is prepared in two steps.
  • the 2-position of 5- benzyloxypyrazoloanthrone is protected by a protective group such as methoxymethyl (MOM), methoxyethoxymethyl (MEM), 2-trimethylsilylethoxymethyl (SEM), or 4- methoxybenzyl (PMB) with an aid of a base such as triethylamine, diisopropylethylamine, pyridine, sodium hexamethyldisilazide, potassium hexamethyldisilazide, or lithium diisopropylamide.
  • a protective group such as methoxymethyl (MOM), methoxyethoxymethyl (MEM), 2-trimethylsilylethoxymethyl (SEM), or 4- methoxybenzyl (PMB) with an aid of a base such as triethylamine, diisopropylethylamine, pyridine, sodium hexamethyldisilazide, potassium hexamethyldisil
  • the reaction is typically carried out at -40 to 60°C for 1 to 16 hours in a solvent such as methylene chloride, chloroform, tetrahydrofuran, dioxane, or dimethoxy ethane.
  • a solvent such as methylene chloride, chloroform, tetrahydrofuran, dioxane, or dimethoxy ethane.
  • MEM group is preferred.
  • N-Protected 5-benzyloxypyrazoloanthorone is then reduced to its 5- hydroxy derivative by hydrogenation in the presence of a transition-metal catalyst, such as palladium platinum, rhodium, or iridium with or without a support such as charcoal in a solvent such as ethanol, ethyl acetate, tetrahydrofuran, dioxane, or dimethoxyethane at 1 to 20 atmospheres of hydrogen at 20 to 60°C for 1 to 16 hours.
  • a transition-metal catalyst such as palladium platinum, rhodium, or iridium with or without a support such as charcoal
  • a solvent such as ethanol, ethyl acetate, tetrahydrofuran, dioxane, or dimethoxyethane
  • pyrazoloanthrones with 5-alkoxy substituents may be prepared by condensation of 7-hydroxy-2-(2-methoxyethoxymethyl)- pyrazoloanthrone with alkyl halides and sulfonates R 7 -X followed by the deprotection.
  • the leaving group X chloride, bromide, iodide, methanesulfonate, tosylate, benzenesulfonate, or triflate can be used.
  • Condensation of 7-hydroxy-2-(2- methoxyethoxymethyl)pyrazoloanthrone with appropriate alkyl halides and sulfonates is carried out in the presence of an acid quenching agent such as triethylamine, diisopropylethylamine, sodium bicarbonate, potassium carbonate, or sodium hydroxide at -20 to 50°C for 0.5 to 16 hours in solvents such as methylene chloride, chloroform, tetrahydrofuran, dioxane, dimethylformamide, or ethyl acetate.
  • the deprotection step is performed by the treatment of the product mentioned above with an acid such as trifluoroacetic acid, aqueous hydrochloric acid, aqueous hydrobromic acid, or aqueous sulfuric acid.
  • the starting material is prepared in two steps.
  • the 2-position of 7- benzyloxypyrazoloanthrone is protected by a protective group such as methoxymethyl (MOM), methoxyethoxymethyl (MEM), 2-trimethylsilylethoxymethyl (SEM), or 4- methoxybenzyl (PMB) with an aid of a base such as triethylamine, diisopropylethylamine, pyridine.
  • a base such as triethylamine, diisopropylethylamine, pyridine.
  • 4-(N, N-dimethylamino)pyridine (DMAP) can be used as a catalyst when a tertiary amine is used as a base.
  • the reaction is typically carried out at -40 to 60°C for 1 to 16 hours in a solvent such as methylene chloride, chloroform, tetrahydrofuran, dioxane, or dimethoxy ethane.
  • a solvent such as methylene chloride, chloroform, tetrahydrofuran, dioxane, or dimethoxy ethane.
  • MEM group is preferred.
  • N-Protected 7-benzyloxypyrazoloanthorone is then reduced to its 7- hydroxy derivative by hydrogenation in the presence of a transition-metal catalyst, such as palladium platinum, rhodium, or iridium with or without a support such as charcoal in a solvent such as ethanol, ethyl acetate, tetrahydrofuran, dioxane, or dimethoxyethane at 1 to 20 atmospheres of hydrogen at 20 to 60°C for 1 to 16 hours.
  • a transition-metal catalyst such as palladium platinum, rhodium, or iridium with or without a support such as charcoal
  • a solvent such as ethanol, ethyl acetate, tetrahydrofuran, dioxane, or dimethoxyethane
  • the procedure using hydrogenation with palladium on charcoal as the catalyst is preferred.
  • Compounds of structures (V), (VI) and (VII) may be made by the same procedures
  • compositions containing one or more compounds of this invention are disclosed.
  • a compound of structure (I) is preferably formulated as a pharmaceutical composition.
  • Pharmaceutical compositions of the present invention comprise a compound of this invention and a pharmaceutically acceptable carrier, wherein the compound is present in the composition in an amount which is effective to treat the condition of interest.
  • the pharmaceutical compositions of the present invention include a compound of structure (I) in an amount from 0.1 mg to 250 mg per dosage depending upon the route of administration, and more typically from 1 mg to 60 mg. Appropriate concentrations and dosages can be readily determined by one skilled in the art.
  • Pharmaceutically acceptable carriers are familiar to those skilled in the art.
  • acceptable carriers include saline and sterile water, and may optionally include antioxidants, buffers, bacteriostats and other common additives.
  • the compositions can also be formulated as pills, capsules, granules, or tablets which contain, in addition to a compound of this invention, diluents, dispersing and surface active agents, binders, and lubricants.
  • diluents such as those disclosed in Remington 's Pharmaceutical Sciences, Gennaro, Ed., Mack Publishing Co., Easton, PA 1990.
  • the present invention provides a method for treating a variety of conditions by administering an effective amount of a JNK inhibitor to a patient in need thereof.
  • Conditions that may be treated by the compounds of this invention, or a pharmaceutical composition containing the same, include any condition which is responsive to JNK inhibition, and thereby benefit from administration of a JNK inhibitor.
  • Representative conditions in this regard include (but not limited to) rheumatoid arthritis, rheumatoid spondylitis, osteoarthritis, gout, asthma, bronchitis, cystic fibrosis, inflammatory bowel disease, irritable bowel syndrome, mucous colitis, ulcerative colitis, Crohn ' s disease, gastritis, esophagitis, hepatitis, multiple sclerosis, atherosclerosis, restenosis following angioplasty, left ventricular hypertrophy, myocardial infarction, stroke, ischemic damage to the heart, kidney, liver, or brain, transplant rejection (such as kidney, liver, heart, lung, and the like), endotoxin shock, psoriasis, eczema, dermatitis, epilepsy, Alzheimer's disease, Parkinson's disease, Huntington's disease, amyotrophic laterial sclerosis, peripheral neuropathies, spinal cord damage, and cancer.
  • the methods of this invention include systemic administration of a compound of this invention, preferably in the form of a pharmaceutical composition.
  • systemic administration encompasses both oral and parenteral methods of administration.
  • suitable pharmaceutical compositions include powders, granules, pills, tablets, and capsules as well as liquids, syrups, suspensions, and emulsions. These compositions may also include flavorants, preservatives, suspending, thickening and emulsifying agents, and other pharmaceutically acceptable additives.
  • the compounds of the present invention can be prepared in aqueous injection solutions which may contain buffers, antioxidants, bacteriostats. and other additives commonly employed in such solutions. The following examples are offered by way of illustration, not limitation.
  • purification of the same may be achieved by first derivatizing Compound 1 to a more soluble intermediate, such as the corresponding acetate, recrystallizing the intermediate, and then converting the intermediate to yield purified Compound 1 in good yield. More specifically, to solution of the pyrazoloanthrone (9.67 g, 43.9 mmol) in acetic acid (700 mL) is added acetic anhydride (12.4 mL, 132 mmol). The solution is heated to 80°C for 5 hours and then cooled to room temperature. After 16 hours, the reaction is cooled to 0°C for 2 hours. The reaction is then filtered to give the N-acetylpyrazoloanthrone intermediate.
  • This compound may be made in the same manner from 1,4- dichloroanthraquinone (commercial product).
  • This compound may be made in the same manner from 1,5- dichloroanthraquinone (commercial product).
  • This compound may be made from 1 ,4-dinitroanthraquinone (Krapcho, A. P.; Avery, K. L., Jr. J. Org. Chem. 55, 5562-4, 1990).
  • This compound may be made in the same manner from 1,5- dichloroanthraquinone (commercial product).
  • This compound may be made in the same manner from l-nitro-4- benzyloxy anthraquinone.
  • This starting material may be prepared as follows. Benzyl bromide is added to 1 -nitro-4-hydroxyanthraquinone (Aldrich) and potassium carbonate in dimethylformamide, and the mixture is stirred for 16 hours. Water is added and the mixture is extracted with ethyl acetate (x2). The combined organic layer is washed sequentially with sodium bicarbonate solution, water, IN hydrochloric acid, and brine, dried, and evaporated. The residue is chromatographed on silica gel to afford l-nitro-4- benzyloxy anthraquinone . 27
  • This compound may be made in the same manner from l-nitro-5- benzyloxyanthraquinone. which starting material may prepared as disclosed in German Patent No. DE 2254199 to Reubke, Hohmann and Bien.
  • This compound may be made in the same manner from 4-acetylamino-l- chloroanthraquinone.
  • This starting material may be prepared as follows. 4-Amino-l- chloroanthraquinone is taken in pyridine and treated with acetic anhydride. The mixture is stirred for 1 hour, and poured onto water. The solids are collected by filtration, washed with water, and dried in vacuo to give 4-acetylamino-l- chloroanthraquinone as a colorless solid.
  • This compound may be made in the same manner using piperidine as the amine.
  • This compound may be made in the same manner using mo ⁇ holine as the amine.
  • This compound may be made in the same manner using benzylamine as the amine.
  • This compound may be made in the same manner using 4- pyridylmethylamine as the amine.
  • This compound may be made in the same manner using 2-(l- piperidyl)ethylamine as the amine.
  • This compound may be made in the same manner using piperidine as the amine.
  • This compound may be made in the same manner using mo ⁇ holine as the amine.
  • This compound may be made in the same manner using benzylamine as the amine.
  • This compound may be made in the same manner using 4- pyridylmethylamine as the amine.
  • This compound may be made in the same manner using 2-(l- piperidyl)ethylamine as the amine.
  • Benzoyl chloride is added to a solution of 2-(methoxyethoxymethyl)-5- aminoanthra[l,9cd]pyrazol-6-(2H)one and triethylamine in methylene chloride at 0°C
  • the mixture is stirred for 16 hours, quenched with water, and extracted with ethyl acetate (x2).
  • the combined organic layer is washed with sodium bicarbonate solution, and brine, dried and evaporated.
  • the crude reaction mixture is then taken in aqueous 6N hydrochloric acid, and heated at 80°C for 4 hours. After cooling, the mixture is extracted with ethyl acetate (x2), washed with brine, dried, and evaporated.
  • the reside is chromatographed on silica gel to furnish the desired amide as a yellow solid.
  • the starting material is prepared as follows. Sodium hexamethyldisilazide is added to a cooled (0°C) solution of 5-nitroanthra[l ,9cd]pyrazol- 6(2H)-one (Example 1-D) in tetrahydrofuran, and the mixture is stirred for 30 minutes at 0°C MEM-chloride is added, and the mixture is stirred for 16 hours at room temperature. Water is added and the mixture is extracted with ethyl acetate (x2). The combined organic layer is washed with aqueous sodium bicarbonate solution, water, IN hydrochloric acid, and brine, dried and evaporated. The residue is chromatographed on silica gel to give 2-MEM-5-nitroanthra[l ,9cd]pyrazol-6(2H)-one as an oil.
  • This compound may be made in the same manner using isonicotinoyl chloride as the acid chloride
  • This compound may be made in the same manner using nicotinoyl chloride as the acid chloride.
  • This compound may be made in the same manner using 2- thiophenecarboxylic acid as the acid chloride.
  • This compound may be made in the same manner using isopentanoyl chloride as the acid chloride.
  • This compound may be made in the same manner using methanesulfonyl chloride as the sulfonyl chloride.
  • G. 5-(3-Benzenesulfonylamino)anthra 1.9cd]pyrazol-6(2H)-one
  • This compound may be made in the same manner using benzenesulfonyl chloride as the sulfonyl chloride.
  • Benzoyl chloride is added to a solution of 2-(methoxyethoxymethyl)-7- aminoanthra[l,9cd]pyrazol-6-(2H)one and triethylamine in methylene chloride at 0°C.
  • the mixture is stirred for 16 hours, quenched with water, and extracted with ethyl acetate (x2).
  • the combined organic layer is washed with sodium bicarbonate solution, and brine, dried and evaporated.
  • the crude reaction mixture is then taken in aqueous 6N hydrochloric acid, and heated at 80°C for 4 hours. After cooling, the mixture is extracted with ethyl acetate (x2), washed with brine, dried, and evaporated.
  • the reside is chromatographed on silica gel to furnish the desired amide as a yellow solid.
  • the starting material is prepared as follows. Sodium hexamethyldisilazide is added to a cooled (0°C) solution of 7-nitroanthra[l,9cd]pyrazol- 6(2H)-one (Example 1-E) in tetrahydrofuran, and the mixture is stirred for 30 minutes at 0°C. MEM-chloride is added, and the mixture is stirred for 16 hours at room temperature. Water is added and the mixture is extracted with ethyl acetate (x2). The combined organic layer is washed with aqueous sodium bicarbonate solution, water, IN hydrochloric acid, and brine, dried and evaporated. The residue is chromatographed on silica gel to give 2-MEM-7-nitroanthra[l ,9cd]pyrazol-6(2H)-one as an oil.
  • This compound may be made in the same manner using isonicotinoyl chloride as the acid chloride.
  • This compound may be made in the same manner using nicotinoyl chloride as the acid chloride.
  • This compound may be made in the same manner using 2- thiophenecarboxylic acid chloride as the acid chloride.
  • This compound may be made in the same manner using isopentanoyl chloride as the acid chloride.
  • This compound may be made in the same manner using methanesulfonyl chloride as the sulfonyl chloride.
  • This compound may be made in the same manner using benzenesulfonyl chloride as the sulfonyl chloride.
  • Isopentyl bromide is added to a mixture of 3-(2- methoxyethoxymethyl)5-hydroxyanthra[ 1 ,9cd]pyrazol-6(2H)-one and potassium carbonate in dimethylformamide at room temperature. After stirring the mixture for sixteen hours, water is added, and the mixture was extracted with ethyl acetate (x2). The combined organic layer is washed with aqueous sodium bicarbonate, water, IN hydrochloric acid, and brine, dried and evaporated. The reside is taken in 6N hydrochloric acid and heated at 80°C for 4 hours. After cooling, the mixture is extracted with ethyl acetate (x2), and the combined organic layer is washed with brine, dried, and evaporated. The residue is purified by column chromatography to afford the title compound as yellow solid.
  • the starting material is prepared as follows. Sodium hexamethyldisilazide is added to a cooled (0°C) solution of 5- benzyloxyanthra[l,9cd]pyrazol-6(2H)-one (Example 1-F) in tetrahydrofuran, and the mixture is stirred for 30 minutes at 0°C MEM-chloride is added, and the mixture is stirred for 16 hours at room temperature. Water is added and the mixture is extracted with ethyl acetate (x2). The combined organic layer is washed with aqueous sodium bicarbonate solution, water, IN hydrochloric acid, and brine, dried and evaporated. The residue is chromatographed on silica gel to give 2-MEM-5- benzyloxyanthra[l,9cd]pyrazol-6(2H)-one as an oil.
  • This compound may be made in the same manner using chloromethyl-4- pyridine as the alkyl halide.
  • This compound may be made in the same manner using chloromethyl-3 - pyridine as the alkyl halide.
  • This compound may be made in the same manner using 2 -methoxy ethyl bromide as the alkyl halide.
  • This compound may be made in the same manner using 2- dimethylaminoethyl chloride as the alkyl halide.
  • This compound may be made in the same manner using chloromethyl-4- pyridine as the alkyl halide.
  • This compound may be made in the same manner using chloromethyl-3 - pyridine as the alkyl halide.
  • This compound may be made in the same manner using 2-methoxyethyl bromide as the alkyl halide.
  • the compounds of this invention may be assayed for their activity accordingly to the following procedures.
  • JNK Assay To 10 ⁇ L of the test compound in 20% DMSO/80% dilution buffer consisting of 20 mM HEPES (pH 7.6). 0.1 mM EDTA, 2.5 mM magnesium chloride, 0.004% Triton xlOO, 2 ⁇ g/mL leupeptin, 20 mM ⁇ -glycerolphosphate, 0.1 mM sodium vanadate, and 2 mM DTT in water is added 30 ⁇ L of 50-200 ng His6-JNK1, JNK2 or JNK3 in the same dilution buffer. The mixture is preincubated for 30 minutes at room temperature.
  • IC 50 values are calculated as the concentration of the test compound at which the c-Jun phosphorylation is reduced to 50% of the control value.
  • Preferred compounds of the present invention have an IC 50 value ranging 0.01 - 10 ⁇ M in this assay.
  • a preferred compound of this invention is Compound 1 , which has an IC 50 according to this assay of 0.11 ⁇ M for JNK1 and JNK2, and 0.15 ⁇ M for JNK3.
  • Compound 1 was also assayed for its inhibitory activity against the following protein kinases by techniques known to those skilled in this field (see, e.g., Protein Phosphorylation, Sefton & Hunter, Eds., Academic Press, pp. 97-367, 1998):
  • EGF-TK 10,000 nM
  • Jurkat T cells (clone E6-1) are purchased from the American Tissue
  • Non-fasted mice are acclimatized for at least 7 days.
  • Groups of 4 to 6 female BALB/c or CD-I mice (8-10 weeks of age from Charles River laboratories) are pretreated with test compound, either by intravenous injection or by oral gavage 15 - 180 minutes prior to the injection of 0.5 mg/kg Bacto LPS from E. coli 055:B5 (Difco Labs).
  • Bacto LPS from E. coli 055:B5 (Difco Labs).
  • a terminal bleed is performed via abdominal vena cava and blood is allowed to clot at room temperature for 30 minutes in Microtainer serum separator tubes. After separation by centrifugation.
  • ELIZA is performed on thawed, diluted samples (1 :10 to 1 :20) using a Mouse TNF-alpha kit (Biosource International).
  • the ED 50 values are calculated as the dose of the test compound at which the TNF- ⁇ production is reduced to 50%> of the control value.
  • Preferred compounds of the present invention have an ED 50 value ranging 1 - 30 mg/kg in this assay.
  • Figure 2 illustrates the results of this experiment utilizing Compound 1 administered by intravenous injection (I.V.) at 15 and 30 mg/kg, as well as by per os (P.O.) at 7.5, 15 and 30 mg/kg. Vehicle alone (PEG-400, propylene glycol, cremophor EL.
  • Activation of AP-1 was determined by DNA binding activity in an electrophoretic mobility shift assay (EMSA) (Ausubel et al., Short Protocols in Molecular Biology, Second Edition. John Wiley & Sons Publisher, New York, 1992).
  • ESA electrophoretic mobility shift assay
  • Matrix metalloproteinase-13 expression was measured by nothern blot analysis of MMP-13 mRNA (Ausebel et al., supra) (see also Winter et al., Arthritis and Rheumatism P(3):394-404, 1966; Weichman et al., Pharmacological Methods in the Control of Inflammation, Chang and Lewis Eds., Alan R. Liss, Inc., Publ., New York, 1989).
  • Kainic Acid-Induced Seizure Response Compound 1 was administered to male CD rats at 10 mg/kg intravenously through a tail vein catheter. This was followed immediately by a 30 mg/kg subcutaneous injection. Vehicle controls received the same injection volumes of the PPCES vehicle alone. Thirty minutes later, animals were given a 1- mg/kg i.p. injection of kainic acid in normal saline solution. This dose of kainic acid has been previously reported to induce a seizure syndrome in rats (Maj et al., Eur. J. Pharm. 51

Landscapes

  • Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Chemical & Material Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Medicinal Chemistry (AREA)
  • General Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Neurology (AREA)
  • Biomedical Technology (AREA)
  • Neurosurgery (AREA)
  • Immunology (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Pulmonology (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Rheumatology (AREA)
  • Pain & Pain Management (AREA)
  • Dermatology (AREA)
  • Psychology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Psychiatry (AREA)
  • Hospice & Palliative Care (AREA)
  • Transplantation (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Cardiology (AREA)
  • Vascular Medicine (AREA)
  • Urology & Nephrology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Plural Heterocyclic Compounds (AREA)

Abstract

Compounds having activity as selective inhibitors of JNK are disclosed. The compounds of this invention are pyrazoloanthrone and derivatives thereof having structure (I) wherein R1 and R2 are as defined herein. Such compounds have utility in the treatment of a wide range of conditions that are responsive to JNK inhibition. Thus, methods of treating such conditions are also disclosed, as are pharmaceutical compositions containing one or more compounds of the above compounds.

Description

PYRAZOLOANTHRONE AND DERIVATIVES THEREOF AS JNK INHIBITORS AND THEIR COMPOSITIONS
TECHNICAL FIELD This invention is generally directed to pyrazoloanthrone and derivatives thereof which have utility over a wide range of indications, including activity as Jun N- terminal kinase inhibitors, and related compositions and methods.
BACKGROUND OF THE INVENTION The Jun N-terminal kinase (JNK) pathway is activated by exposure of cells to environmental stress or by treatment of cells with pro-inflammatory cytokines. Targets of the JNK pathway include the transcription factors c-jun and ATF2 (Whitmarsh A.J., and Davis R.J. J. Mol. Med. 74:589-607, 1996). These transcription factors are members of the basic leucine zipper (bZIP) group that bind as homo- and hetero-dimeric complexes to AP-1 and AP-1 -like sites in the promoters of many genes (Karin M., Liu Z.G. and Zandi E. Curr Opin Cell Biol 9:240-246, 1997). JNK binds to the N-terminal region of c-jun and ATF-2 and phosphorylates two sites within the activation domain of each transcription factor (Hibi M., Lin A., Smeal T., Minden A., Karin M. Genes Dev. 7:2135-2148. 1993; Mohit A.A., Martin M.H., and Miller CA. Neuron 14:67-75, 199)]. Three JNK enzymes have been identified as products of distinct genes (Hibi et al, supra; Mohit et al., supra). Ten different isoforms of JNK have been identified. These represent alternatively spliced forms of three different genes: JNK1, JNK2 and JNK3. JNK1 and 2 are ubiquitously expressed in human tissues, whereas JNK3 is selectively expressed in the brain, heart and testis (Dong, C, Yang, D., Wysk, M., Whitmarsh, A., Davis, R., Flavell, R. Science 270:1-4, 1998). Gene transcripts are alternatively spliced to produce four- JNK 1 isoforms, four-JNK2 isoforms and two-JNK3 isoforms. JNK1 and 2 are expressed widely in mammalian tissues, whereas JNK3 is expressed almost exclusively in the brain. Selectivity of JNK signaling is achieved via specific interactions of JNK pathway components and by use of scaffold proteins that selectively bind multiple components of the signaling cascade. JIP-1 (JNK-interacting protein- 1) selectively binds the MAPK module, MLK -> JNKK1 - JNK.12, 13 It has no binding affinity for a variety of other MAPK cascade enzymes. Different scaffold proteins are likely to exist for other MAPK signaling cascades to preserve substrate specificity. JNKs are activated by dual phosphorylation on Thr-183 and Tyr-185.
JNKK1 (also known as MKK 4) and JNKK2 (MKK7), two MAPKK level enzymes, can mediate JNK activation in cells (Lin A., Minden A., Martinetto H, Claret F.-Z., Lange-Carter C, Mercurio F., Johnson G.L., and Karin M. Science 268:286-289, 1995; Tournier C, Whitmarsh A.J., Cavanagh J., Barrett T., and Davis R.J. Proc. Nat. Acad. Sci. USA 94:7337-7342, 1997). JNKK2 specifically phosphorylates JNK. whereas JNKK1 can also phosphorylate and activate p38. Both JNKK1 and JNKK2 are widely expressed in mammalian tissues. JNKK1 and JNKK2 are activated by the MAPKKK enzymes, MEKK1 and 2 (Lange-Carter C.A., Pleiman CM., Gardner A.M., Blumer K.J., and Johnson G.L. Science 260:315-319, 1993; Yan M., Dai J.C. Deak J.C., Kyriakis J.M., Zon L.I., Woodgett J.R., and Templeton D.J. Nature 372:798-781, 1994). Both MEKK1 and MEKK2 are widely expressed in mammalian tissues.
Activation of the JNK pathway has been documented in a number of disease settings, providing the rationale for targeting this pathway for drug discovery. In addition, molecular genetic approaches have validated the pathogenic role of this pathway in several diseases. For example, autoimmune and inflammatory diseases arise from the over-activation of the immune system. Activated immune cells express many genes encoding inflammatory molecules, including cytokines, growth factors, cell surface receptors, cell adhesion molecules and degradative enzymes. Many of these genes are regulated by the JNK pathway, through activation of the transcription factors AP-1 and ATF-2, including TNF a, IL-2, E-selectin and matrix metalloproteinases such as collagenase-1 (Manning A.M. and Mercurio F. Exp Opin Invest Drugs 6: 555-567, 1997). Monocytes, tissue macrophages and tissue mast cells are key sources of TNF a production. The JNK pathway regulates TNFa production in bacterial lipopolysaccharide-stimulated macrophages, and in mast cells stimulated through the FceRII receptor (Swantek J.L., Cobb M.H., Geppert T.D. Mol. Cell. Biol. 17:627 '4- 6282, 1997; Ishizuka, T., Tereda N., Gerwins, P.. Hamelmann E., Oshiba A., Fanger G.R.. Johnson G.L., and Gelfland E.W. Proc. Nat. Acad. Sci. USA 94:6358-6363, 1997). Inhibition of JNK activation effectively modulates TNFa secretion from these cells. The JNK pathway therefore regulates production of this key pro-inflammatory cytokine. Matrix metalloproteinases (MMPs) promote cartilage and bone erosion in rheumatoid arthritis, and generalized tissue destruction in other autoimmune diseases. Inducible expression of MMPs, including MMP-3 and MMP-9, type II and IV collagenases, are regulated via activation of the JNK pathway and AP-1 (Gum, R., Wang, H, Lengyel, E., Juarez. J., and Boyd, D. Oncogene 14: 1481-1493, 1997). In human rheumatoid synoviocytes activated with TNFa, IL-1, or Fas ligand the JNK pathway is activated (Han Z., Boyle D.L., Aupperle K.R., Bennett B., Manning A.M., Firestein G.S. J. Pharm. Exp. Therap. 291 :1-7, 1999; Okamoto K., Fujisawa K., Hasunuma T., Kobata T., Sumida T., and Nishioka K. Arth & Rheum 40: 919-92615, 1997). Inhibition of JNK activation results in decreased AP-1 activation and collagenase-1 expression (Han et al., supra). The JNK pathway therefore regulates MMP expression in cells involved in rheumatoid arthritis.
Inappropriate activation of T lymphocytes initiates and perpetuates many autoimmune diseases, including asthma, inflammatory bowel disease and multiple sclerosis. The JNK pathway is activated in T cells by antigen stimulation and CD28 receptor co-stimulation and regulates production of the growth factor IL-2 and cellular proliferation (Su B., Jacinto E.. Hibi M.. Kallunki T., Karin M., Ben-Neriah Y. Cell 11:121-136, 1994; Faris M., Kokot N., Lee L., and Nel A.E. J. Biol. Chem. 271 :27366- 27373, 1996). Peripheral T cells from mice genetically deficient in JNKK1 show decreased proliferation and IL-2 production after CD28 co-stimulation and PMA / Ca2+ ionophore activation, providing important validation for the role of the JNK pathway in these cells (Nishina H, Bachmann M., Oliveria-dos-Santos A.J., et al. J. Exp. Med. 186: 941-953, 1997). It is known that T cells activated by antigen receptor stimulation in the absence of accessory cell-derived co-stimulatory signals lose the capacity to synthesize IL-2. a state called clonal anergy. This is an important process by which auto-reactive T cell populations are eliminated from the peripheral circulation. Of note, anergic T cells fail to activate the JNK pathway in response to CD3- and CD28-receptor co- stimulation, even though expression of the JNK enzymes is unchanged (Li W., Whaley CD., Mondino A., and Mueller D.L. Science 271 : 1272-1276, 1996). Recently, the examination of JNK-deficient mice revealed that the JNK pathway plays a key role in T cell activation and differentiation to T helper 1 and 2 cell types. JNK 1 or JNK2 knockout mice develop normally and are phenotypically unremarkable. Activated naϊve CD4+ T cells from these mice fail to produce IL-2 and do not proliferate well (Sabapathy, K, Hu, Y, Kallunki, T, Schreiber, M, David, J-P, Jochum, W, Wagner, E, Karin, M. Curr Biol 9: 116-125, 1999). It is possible to induce T cell differentiation in T cells from these mice, generating Thl cells (producers of IFN-g and TNFβ) and Th2 effector cells (producers of IL-4, IL-5, IL-6, IL-10 and IL-13) [22,23]. Deletion of either JNK1 or JNK2 in mice resulted in a selective defect in the ability of Thl effector cells to express IFNg. This suggests that JNK1 and JNK2 do not have redundant functions in T cells and that they play different roles in the control of cell growth, differentiation and death. The JNK pathway therefore, is an important point for regulation of T cell responses to antigen.
Cardiovascular disease (CVD) accounts for nearly one quarter of total annual deaths worldwide. Vascular disorders such as atherosclerosis and restenosis result from dysregulated growth of the vessel wall, restricting blood flow to vital organs. The JNK pathway is activated by atherogenic stimuli and regulates local cytokine and growth factor production in vascular cells (Yang, DD, Conze, D, Whitmarsh, AJ, et al, Immunity, 9:575, 1998). In addition, alterations in blood flow, hemodynamic forces and blood volume lead to JNK activation in vascular endothelium, leading to AP-1 activation and pro-atherosclerotic gene expression (Aspenstrom P., Lindberg U., and Hall A. Curr. Biol. 6:70-77, 1996). Ischemia and ischemia coupled with reperfusion in the heart, kidney or brain results in cell death and scar formation, which can ultimately lead to congestive heart failure, renal failure or cerebral dysfunction. In organ transplantation, reperfusion of previously ischemic donor organs results in acute leukocyte-mediated tissue injury and delay of graft function. The JNK pathway is activated by ischemia and reperfusion (Li Y., Shyy J.. Li S., Lee J., Su B., Karin M., Chien S Mol. Cell. Biol. 16:5947-5954, 1996), leading to the activation of
JNK-responsive genes and leukcoyte-mediated tissue damage. In a number of different settings JNK activation can be either pro- or anti-apoptotic. JNK activation is correlated with enhanced apoptosis in cardiac tissues following ischemia and reperfusion (Pombo CM, Bonventre JV, Avruch J, Woodgett JR, Kyriakis J.M, Force T.
J. Biol. Chem. 269:26546-26551, 1994).
Cancer is characterized by uncontrolled growth, proliferation and migration of cells. Cancer is the second leading cause of death with 500,000 deaths and an estimated 1.3 million new cases in the United States in 1996. The role of signal transduction pathways contributing to cell transformation and cancer is a generally accepted concept. The JNK pathway leading to AP-1 appears to play a critical role in cancer. Expression of c-jun is altered in early lung cancer and may mediate growth factor signaling in non-small cell lung cancer (Yin T., Sandhu G., Wolfgang CD., Burrier A., Webb R.L., Rigel D.F. Hai T.. and Whelan J. J. Biol. Chem. 272:19943- 19950, 1997). Indeed, over-expression of c-jun in cells results in transformation, and blocking c-jun activity inhibits MCF-7 colony formation (Szabo E., Riffe M., Steinberg S.M., Birrer M.J., Linnoila R.I. Cancer Res. 56:305-315, 1996). DNA-damaging agents, ionizing radiation and tumor necrosis factor activate the JNK pathway. In addition to regulating c-jun production and activity, JNK activation can regulate phosphorylation of p53, and thus can modulate cell cycle progression (Chen T.K., Smith L.M., Gebhardt D.K., Birrer M.J., Brown P.H. Mol. Carcinogenesis 15:215-226, 1996). The oncogene BCR-Abl, associated with t(9,22) Philadelphia chromosome translocation of chronic myelogenous leukemia, activates JNK and leads to transformation of hematopoietic cells (Milne D.M., Campbell L.E., Campbell D.G., Meek D.W. J. Biol. Chem. 270:551 1-5518, 1995). Selective inhibition of JNK activation by a naturally occurring JNK inhibitory protein, called JIP-1, blocks cellular transformation caused by BCR-Abl expression (Raitano A.B., Halpern J.R., Hambuch T.M., Sawyers C.L. Proc. Nat. Acad. Sci USA 92:11746-1 1750, 1995). Thus, JNK inhibitors may block transformation and tumor cell growth. Accordingly, there is a need in the art for selective inhibitors of JNK, as well as for methods for preparation thereof, pharmaceutical compositions comprising such inhibitors, and methods of inhibiting JNK's and treating diseases in mammals which are responsive to JNK inhibition. The present invention fulfills these needs, and provides further related advantages.
SUMMARY OF THE INVENTION
In brief, the present invention is directed to compounds having activity as selective inhibitors of JNK, as well as to compositions and methods related thereto.
The compounds of the present invention (also referred to herein as "JNK inhibitors") may generally be classified as "pyrazoloanthrone derivatives" having the following structure (I):
Figure imgf000008_0001
wherein R, and R2 are as defined below, including pharmaceutically acceptable salts thereof.
The present invention is also directed to methods for treating a variety of conditions by administering an effective amount of a JNK inhibitor to an animal or subject in need thereof (referred to herein as a "patient"), typically a warm-blooded animal (including a human). Prior to administration, the compounds of this invention are preferably formulated as a pharmaceutical composition which contains an effective dosage amount of one or more JNK inhibitors in combination with one (or more) pharmaceutically acceptable carrier(s). Conditions that may be treated by the compounds of this invention, or a pharmaceutical composition containing the same, include any condition which may benefit from administration of JNK inhibitors, and are particularly useful for the prevention and/or treatment of various diseases including (but not limited to) rheumatoid arthritis, rheumatoid spondylitis, osteoarthritis, gout, asthma, bronchitis, cystic fibrosis, inflammatory bowel disease, irritable bowel syndrome, mucous colitis, ulcerative colitis, Crohn's disease, gastritis, esophagitis, hepatitis, multiple sclerosis, atherosclerosis, restenosis following angioplasty, left ventricular hypertrophy, myocardial infarction, stroke, ischemic damages to heart, kidney, liver, and brain, transplant rejection, endotoxin shock, psoriasis, eczema, dermatitis, epilepsy, Alzheimer's disease, Huntington's disease, Amyotrophic laterial sclerosis, peripheral neuropathies, spinal cord damage, Parkinson's disease, and cancer.
These and other aspects of this invention will be apparent upon reference to the following detailed description. To that end, certain patent and other documents are cited herein to more specifically set forth various aspects of this invention. Each of these documents are hereby incorporated by reference in their entirety.
BRIEF DESCRIPTION OF THE DRAWINGS
Figure 1 illustrates the ability of a representative compound of this invention to inhibit IL-2 in Jurkat T-Cell.
Figure 2 illustrates the ability of a representative compound of this invention to inhibit TNF- in a mouse model of endotoxin shock.
Figure 3 illustrates the ability of a representative compound of this invention to inhibit leukocyte recruitment in rat model for inflamed lung. Figure 4 illustrates the ability of a representative compound of this invention to inhibit paw swelling (Figure 4A), joint destruction (Figure 4B), transcription factor AP-1 activation (Figure 4C), and expression of MMP-13 (Figure 4D) in a rat model for adjuvant arthritis.
Figure 5 illustrates the ability of a representative compound of this invention to reduce kainic acid-induced seizure response.
DETAILED DESCRIPTION OF THE INVENTION
As mentioned above, the present invention is directed to compounds which have activity as selective inhibitors of JNK, as well as to compositions and methods relating to the same. The compounds of this invention have the following structure (I):
Figure imgf000010_0001
and pharmaceutically acceptable salts thereof, wherein:
R, and R2 are optional substituents that are the same or different and independently represent alkyl, halogen, nitro, trifluoromethyl, sulfonyl, carboxyl, alkoxycarbonyl, alkoxy, aryl, aryloxy, arylalkyloxy, arylalkyl, cycloalkylalkyloxy, cycloalkyloxy, alkoxyalkyl. alkoxyalkoxy, aminoalkoxy. mono- or di- alkylaminoalkoxy, or a group represented by formula (a), (b), (c) or (d):
Figure imgf000010_0002
(a) (b) (c) ( )
R, and R4 taken together represent alkylidene or a heteroatom-containing alkylidene, or R3 and R4 are the same or different and independently represent hydrogen, alkyl, cycloalkyl, aryl, arylalkyl, cycloalkylalkyl, aryloxyalkyl, alkoxyalkyl, alkoxyamino, or alkoxy(mono- or di-alkylamino); and
R5 represents hydrogen, alkyl, cycloalkyl, aryl, arylalkyl, cycloalkylalkyl, alkoxy, amino, mono- or di-alkylamino, arylamino, arylalkylamino, cycloalkylamino, or cycloalkylalkylamino. As used herein, the terms used above having following meaning.
"Alkyl" means a straight chain or branched, saturated or unsaturated alkyl chain having from 1 to 8 carbon atoms, such as methyl, ethyl, n-propyl, iso- propyl, n-butyl, iso-butyl, tert-butyl, propylenyl, 1 -butenyl, propynyl, and the like.
"Halogen" means fluorine, chlorine, bromine or iodine.
"Trifluoromethyl'' means -CF3.
"Sulfonyl" means -SO3H;
"Carboxyl" means -COOH. "Alkoxy" means -O-(alkyl), such as methoxy, ethoxy. n-propyloxy, iso- propyloxy. n-butyloxy, iso-butyloxy, and the like.
"Alkoxyalkoxy" means -0-(alkyl)-O-(alkyl), such as -OCH2CH2OCH3, and the like.
"Alkoxycarbonyl" means -C(=O)O-(alkyl), such as -C(=O)OCH3, -C(=O)OCH2CH„ and the like.
"Alkoxyalkyl" means -(alkyl)-O-(alkyl), such as -CH2OCH3, -CH2OCH2CH3, and the like.
"Aryl" means a carbocyclic or heterocyclic aromatic group containing from 5 to 10 ring atoms. The ring atoms of a carbocyclic aryl group are all carbon atoms, and includes phenyl and naphthyl. The ring atoms of a heterocyclic aryl group contains at least one heteroatom selected from nitrogen, oxygen and sulfur, and include pyridinyl, pyrimidinyl, furanyl, thienyl, imidazolyl, thiazolyl, pyrazolyl, pyridazinyl, pyrazinyl, triazinyl, tetrazolyl, and indolyl.
"Aryloxy" means -O-(aryl), such as -O-phenyl, -O-pyridinyl, and the like.
"Arylalkyl" means -(alkyl)-(aryl), such as benzyl (i.e., -CH2phenyl), -CH2-pyrindinyl, and the like.
"Arylalkyloxy" means -O-(alkyl)-(aryl), such as -O-benzyl, -O-CH2- pyridinyl, and the like. "Cycloalkyl" means a cyclic alkyl having from 3 to 7 carbon atoms, such as cyclopropyl, cyclopentyl, cyclohexyl, and the like.
"Cycloalkyloxy" means -O-(cycloalkyl), such as -O-cyclohexyl, and the like. "Cycloalkylalkyloxy" means -O-(alkyl)-(cycloalkyl, such as
-OCHcyclohexyl, and the like.
"Alkylidene" means the divalent radical -CnH2n-, wherein n is an integer from 1 to 8, such as -CH2-, -CH2CH2-, -CH2-CH2-CH2-, -CH2CH2CH2CH2-, -CH2CH2CH2CH2CH2-, and the like. "Heteroatom-containing alkylidene" means an alkylidene wherein at least one carbon atom is replaced by a heteroatom selected from nitrogen, oxygen or sulfur, such as -CH2CH2OCH2CH2-, and the like.
"Aminoalkoxy" means -0-(alkyl)-NH2, such as -OCH2NH2, -OCH2CH2NH2, and the like. "Mono- or di-alkylamino" means -NH(alkyl) or -N(alkyl)(alkyl), respectively, such as -NHCH3, -N(CH3)2, and the like.
"Mono- or di-alkylaminoalkoxy" means -O-(alkyl)-NH(alkyl) or -O-(alkyl)-N(alkyl)(alkyl), respectively, such as -OCH2NHCH3, -OCH2CH2N(CH3)2, and the like. "Arylamino"means -NH(aryl), such as -NH-phenyl, -NH-pyridinyl. and the like.
"Arylalkylamino" means -NH-(alkyl)-(aryl), such as -NH-benzyl, -NHCH2-pyridinyl, and the like.
"Alkylamino" means -NH(alkyl). such as -NHCH3, -NHCH2CH3, and the like.
"Cycloalkylamino" means -NH-(cycloalkyl), such as -NH-cyclohexyl, and the like.
"Cycloalkylalkylamino" -NH-(alkyl)-(cycloalkyl), such as
-NHCH2-cyclohexyl, and the like. In the embodiment wherein R, and R2 are not present, compounds of this invention have the following structure (II) (also referred to herein as "Compound 1"):
Figure imgf000013_0001
This compound is commercially available from Pfaltz-Bauer (Conn., U.S.).
In the embodiment wherein only one of R, and R2 is present, compounds of this invention have one of the following structures (III) or (IV):
Figure imgf000013_0002
In the embodiment wherein both R, and R2 are present, compounds of this invention have one of the following structures (V), (VI) or (VII):
Figure imgf000013_0003
Pharmaceutically acceptable salts of compounds of structure (I) are also within the scope of this invention. To this end, the compound may generally be utilized as the free base. Alternatively, the compounds may be used in the form of acid addition salts. Acid addition salts of the free base amino compounds of the present invention may be prepared by methods well known in the art, and may be formed from organic and inorganic acids. Suitable organic acids include maleic, fumaric, benzoic, ascorbic, succinic, methanesulfonic, acetic, oxalic, propionic, tartaric, salicylic, citric, gluconic, lactic, mandelic, cinnamic, aspartic, stearic, palmitic, glycolic, glutamic, and benzenesulfonic acids. Suitable inorganic acids include hydrochloric, hydrobromic, sulfuric, phosphoric, and nitric acids. Thus, the term "pharmaceutically acceptable salt" of a compound of structure (I) is intended to encompass any and all acceptable salt forms.
The compounds of this invention may generally be made by organic synthesis techniques known to those skilled in the art, as well as by the following general techniques and by the procedures set forth in the Examples. To that end, the compounds of this invention may be made according to the following Reaction Schemes 1 through 7.
Reaction Scheme 1
Figure imgf000014_0001
In Reaction Scheme 1, pyrazoloanthrones of this invention may be prepared by condensation of appropriate anthraquinones having a leaving group at the 1 -position (such as fluoro, chloro, bromo, iodo, nitro, methanesulfonyloxy, tosyloxy or phenoxy) with hydrazine in a suitable solvent (such as pyridine, dimethylformamide, methylene chloride, chloroform, or dioxane). The reaction is carried out at temperatures ranging 0°C to 200°C for 1 to 16 hours. Suitable anthraquinone starting materials are commercially available from a variety of sources with the R, and/or R2 groups at various positions on the anthraquinone ring. For purpose of illustration, the following reaction schemes depict synthesis of 5- and/or 7-substituted pyrazoloanthrones. One skilled in the art will recognize that pyrazoloanthrones substituted at other positions may be made in a similar manner from the appropriately substituted pyrazoloanthrone starting material.
Reaction Scheme 2
Figure imgf000015_0001
In Reaction Scheme 2, pyrazoloanthrones with 5-amino substituents may be prepared by condensation of 5-chloropyrazoloanthrone with mono- or disubstituted amines at 0 to 250°C for 1 to 16 hours, either in the absence or the presence of a solvent. Typically solvents are pyridine, dimethylformamide, dimethylsulfoxide, dichloroethane, chloroform, tetrahydrofuran, dioxane, diglyme, or triglyme in the presence of excess amount of the amine, or in the presence of an acid quenching agent such as triethylamine, diisopropylefhylamine, sodium bicarbonate, potassium carbonate, or sodium hydroxide. Reaction Scheme 3
Figure imgf000016_0001
In Reaction Scheme 3, pyrazoloanthrones with 7-amino substituents may be prepared by condensation of 7-chloropyrazoloanthrone with mono- or disubstituted amines at 0 to 250°C for 1 to 16 hours either in the absence or the presence of a solvent. Typically solvents are pyridine, dimethylformamide, dimethylsulfoxide, dichloroethane, chloroform, tetrahydrofuran, dioxane, diglyme. or triglyme in the presence of excess amount of the amine, or in the presence of an acid quenching agent such as triethylamine, diisopropylethylamine, sodium bicarbonate, potassium carbonate, or sodium hydroxide.
Reaction Scheme 4
Figure imgf000017_0001
Y = C, n : = 1
Y = S, n = 2
In Reaction Scheme 4, pyrazoloanthrones with 5-acyl- or sulfonylamino substituents may be prepared by condensation of 5-amino-2-(2- methoxyethoxymethyl)pyrazoloanthrone with acid chlorides and sulfonyl chlorides followed by the deprotection. Condensation of 5-amino-2-(2- methoxyethoxymethyl)pyrazoloanthrone with appropriate acid chlorides R^COCl or sulfonyl chlorides
Figure imgf000017_0002
is carried out in the presence of an acid quenching agent such as triethylamine, diisopropylethylamine, sodium bicarbonate, potassium carbonate, or sodium hydroxide at -20 to 50°C for 0.5 to 16 hours in solvents such as methylene chloride, chloroform, tetrahydrofuran, dioxane, dimefhylformamide, and ethyl acetate. The deprotection step may be performed by the treatment of the product mentioned above with an acid such as trifluoroacetic acid, aqueous hydrochloric acid, aqueous hydrobromic acid, or aqueous sulfuric acid. The starting material may be prepared in two steps. The 2-position of 5- nitropyrazoloanthrone may be protected by a protective group such as methoxymethyl (MOM), methoxyethoxymethyl (MEM), 2-trimethylsilylethoxymethyl (SEM), or 4- methoxybenzyl (PMB) with an aid of a base such as triethylamine, diisopropylethylamine, pyridine, sodium hexamethyldisilazide, potassium hexame hyldisilazide, or lithium diisopropylamide. 4-(N, N-dimethylamino)pyridine (DMAP) may be used as a catalyst when a tertiary amine is used as a base. The reaction is typically carried out at -40 to 60°C for 1 to 16 hours in a solvent such as methylene chloride, chloroform, tetrahydrofuran, dioxane, or dimethoxy ethane. As the nitrogen protective group, MEM group is preferred.
N-Protected 5-nitropyrazoloanthorone is then reduced to its 5-amino derivative by a variety of reducing agents such as Sn or Fe metal in acidic media such as acetic acid or aqueous hydrochloric acid. The reaction is typically run at 20 to 160°C for 1 to 16 hours. The same transformation can be carried out by hydrogenation in the presence of a transition-metal catalyst such as palladium, platinum, rhodium, or iridium with or without a support such as charcoal in a solvent such as ethanol, ethyl acetate, tetrahydrofuran, dioxane, or dimethoxyethane at 1 to 20 atmospheres of hydrogen at 20 to 60°C for 1 to 16 hours. The procedure using hydrogenation with palladium on charcoal as the catalyst is preferred.
Reaction Scheme 5
Figure imgf000019_0001
Y = C, n = = 1
Y = S, n = 2
In Reaction Scheme 5, pyrazoloanthrones with 7-acyl- or sulfonylamino substituents may be prepared by condensation of 7-amino-2-(2- methoxyethoxymethyl)pyrazoloanthrone with acid chlorides and sulfonyl chlorides followed by the deprotection. Condensation of 7-amino-2-(2- methoxyethoxymethyl)pyrazoloanthrone with appropriate acid chlorides R6COCl or sulfonyl chlorides
Figure imgf000019_0002
is carried out in the presence of an acid quenching agent such as triethylamine, diisopropylethylamine, sodium bicarbonate, potassium carbonate, or sodium hydroxide at -20 to 50°C for 0.5 to 16 hours in solvents such as methylene chloride, chloroform, tetrahydrofuran, dioxane, dimethylformamide, or ethyl acetate. The deprotection step may be performed by the treatment of the product mentioned above with an acid such as trifluoroacetic acid, aqueous hydrochloric acid, aqueous hydrobromic acid, or aqueous sulfuric acid. The starting material is prepared in two steps. The 2-position of 7- nitropyrazoloanthrone is protected by a protective group such as methoxymethyl (MOM), methoxyethoxymethyl (MEM), 2-trimethylsilylethoxymethyl (SEM), or 4- methoxybenzyl (PMB) with an aid of a base such as triethylamine, diisopropylethylamine, pyridine, sodium hexamefhyldisilazide, potassium hexamethyldisilazide, or lithium diisopropylamide. 4-(N, N-dimethylamino)pyridine (DMAP) can be used as a catalyst when a tertiary amine is used as a base. The reaction is typically carried out at -40 to 60°C for 1 to 16 hours in a solvent such as methylene chloride, chloroform, tetrahydrofuran, dioxane. or dimethoxy ethane. As the nitrogen protective group, MEM group is preferred.
N-Protected 7-nitropyrazoloanthorone is then reduced to its 7-amino derivative by a variety of reducing agents such as Sn or Fe metal in acidic media such as acetic acid or aqueous hydrochloric acid. The reaction is typically run at 20 to 160°C for 1 to 16 hours. The same transformation can be carried out by hydrogenation in the presence of a transition-metal catalyst such as palladium, platinum, rhodium, or iridium with or without a support such as charcoal in a solvent such as ethanol, ethyl acetate, tetrahydrofuran, dioxane, or dimethoxyethane at 1 to 20 atmospheres of hydrogen at 20 to 60°C for 1 to 16 hours. The procedure using hydrogenation with palladium on charcoal as the catalyst is preferred.
Reaction Scheme 6
Figure imgf000021_0001
1) R7-X 2) H+
Figure imgf000021_0002
In Reaction Scheme 6, pyrazoloanthrones with 5-alkoxy substituents may be prepared by condensation of 5-hydroxy-2-(2-methoxyethoxymethyl)- pyrazoloanthrone with alkyl halides and sulfonates R7-X followed by the deprotection. As the leaving group X, chloride, bromide, iodide, methanesulfonate. tosylate, benzenesulfonate, or triflate can be used. Condensation of 5-hydroxy-2-(2- methoxyethoxymethyl)pyrazoloanthrone with appropriate alkyl halides and sulfonates is carried out in the presence of an acid quenching agent such as triethylamine, diisopropylethylamine, sodium bicarbonate, potassium carbonate, or sodium hydroxide at -20 to 50°C for 0.5 to 16 hours in solvents such as methylene chloride, chloroform, tetrahydrofuran, dioxane, dimethylformamide, or ethyl acetate. The deprotection step is performed by the treatment of the product mentioned above with an acid such as trifluoroacetic acid, aqueous hydrochloric acid, aqueous hydrobromic acid, or aqueous sulfuric acid.
The starting material is prepared in two steps. The 2-position of 5- benzyloxypyrazoloanthrone is protected by a protective group such as methoxymethyl (MOM), methoxyethoxymethyl (MEM), 2-trimethylsilylethoxymethyl (SEM), or 4- methoxybenzyl (PMB) with an aid of a base such as triethylamine, diisopropylethylamine, pyridine, sodium hexamethyldisilazide, potassium hexamethyldisilazide, or lithium diisopropylamide. 4-(N, N-dimethylamino)pyridine (DMAP) can be used as a catalyst when a tertiary amine is used as a base. The reaction is typically carried out at -40 to 60°C for 1 to 16 hours in a solvent such as methylene chloride, chloroform, tetrahydrofuran, dioxane, or dimethoxy ethane. As the nitrogen protective group, MEM group is preferred.
N-Protected 5-benzyloxypyrazoloanthorone is then reduced to its 5- hydroxy derivative by hydrogenation in the presence of a transition-metal catalyst, such as palladium platinum, rhodium, or iridium with or without a support such as charcoal in a solvent such as ethanol, ethyl acetate, tetrahydrofuran, dioxane, or dimethoxyethane at 1 to 20 atmospheres of hydrogen at 20 to 60°C for 1 to 16 hours. The procedure using hydrogenation with palladium on charcoal as the catalyst is preferred.
Reaction Scheme 7
Figure imgf000022_0001
1) R7-X
2) H+
Figure imgf000022_0002
In Reaction Scheme 7, pyrazoloanthrones with 5-alkoxy substituents may be prepared by condensation of 7-hydroxy-2-(2-methoxyethoxymethyl)- pyrazoloanthrone with alkyl halides and sulfonates R7-X followed by the deprotection. As the leaving group X, chloride, bromide, iodide, methanesulfonate, tosylate, benzenesulfonate, or triflate can be used. Condensation of 7-hydroxy-2-(2- methoxyethoxymethyl)pyrazoloanthrone with appropriate alkyl halides and sulfonates is carried out in the presence of an acid quenching agent such as triethylamine, diisopropylethylamine, sodium bicarbonate, potassium carbonate, or sodium hydroxide at -20 to 50°C for 0.5 to 16 hours in solvents such as methylene chloride, chloroform, tetrahydrofuran, dioxane, dimethylformamide, or ethyl acetate. The deprotection step is performed by the treatment of the product mentioned above with an acid such as trifluoroacetic acid, aqueous hydrochloric acid, aqueous hydrobromic acid, or aqueous sulfuric acid.
The starting material is prepared in two steps. The 2-position of 7- benzyloxypyrazoloanthrone is protected by a protective group such as methoxymethyl (MOM), methoxyethoxymethyl (MEM), 2-trimethylsilylethoxymethyl (SEM), or 4- methoxybenzyl (PMB) with an aid of a base such as triethylamine, diisopropylethylamine, pyridine. sodium hexamethyldisilazide, potassium hexamethyldisilazide, or lithium diisopropylamide. 4-(N, N-dimethylamino)pyridine (DMAP) can be used as a catalyst when a tertiary amine is used as a base. The reaction is typically carried out at -40 to 60°C for 1 to 16 hours in a solvent such as methylene chloride, chloroform, tetrahydrofuran, dioxane, or dimethoxy ethane. As the nitrogen protective group, MEM group is preferred.
N-Protected 7-benzyloxypyrazoloanthorone is then reduced to its 7- hydroxy derivative by hydrogenation in the presence of a transition-metal catalyst, such as palladium platinum, rhodium, or iridium with or without a support such as charcoal in a solvent such as ethanol, ethyl acetate, tetrahydrofuran, dioxane, or dimethoxyethane at 1 to 20 atmospheres of hydrogen at 20 to 60°C for 1 to 16 hours. The procedure using hydrogenation with palladium on charcoal as the catalyst is preferred. Compounds of structures (V), (VI) and (VII) may be made by the same procedures as outlined above by utilizing starting materials having multiple reactive sites at the corresponding positions to the desired product.
In another embodiment of the invention, pharmaceutical compositions containing one or more compounds of this invention are disclosed. For purpose of administration, a compound of structure (I) is preferably formulated as a pharmaceutical composition. Pharmaceutical compositions of the present invention comprise a compound of this invention and a pharmaceutically acceptable carrier, wherein the compound is present in the composition in an amount which is effective to treat the condition of interest. Preferably, the pharmaceutical compositions of the present invention include a compound of structure (I) in an amount from 0.1 mg to 250 mg per dosage depending upon the route of administration, and more typically from 1 mg to 60 mg. Appropriate concentrations and dosages can be readily determined by one skilled in the art. Pharmaceutically acceptable carriers are familiar to those skilled in the art. For compositions formulated as liquid solutions, acceptable carriers include saline and sterile water, and may optionally include antioxidants, buffers, bacteriostats and other common additives. The compositions can also be formulated as pills, capsules, granules, or tablets which contain, in addition to a compound of this invention, diluents, dispersing and surface active agents, binders, and lubricants. One skilled in this art may further formulate the compounds of this invention in an appropriate manner, and in accordance with accepted practices, such as those disclosed in Remington 's Pharmaceutical Sciences, Gennaro, Ed., Mack Publishing Co., Easton, PA 1990.
In another embodiment, the present invention provides a method for treating a variety of conditions by administering an effective amount of a JNK inhibitor to a patient in need thereof. Conditions that may be treated by the compounds of this invention, or a pharmaceutical composition containing the same, include any condition which is responsive to JNK inhibition, and thereby benefit from administration of a JNK inhibitor. Representative conditions in this regard include (but not limited to) rheumatoid arthritis, rheumatoid spondylitis, osteoarthritis, gout, asthma, bronchitis, cystic fibrosis, inflammatory bowel disease, irritable bowel syndrome, mucous colitis, ulcerative colitis, Crohn's disease, gastritis, esophagitis, hepatitis, multiple sclerosis, atherosclerosis, restenosis following angioplasty, left ventricular hypertrophy, myocardial infarction, stroke, ischemic damage to the heart, kidney, liver, or brain, transplant rejection (such as kidney, liver, heart, lung, and the like), endotoxin shock, psoriasis, eczema, dermatitis, epilepsy, Alzheimer's disease, Parkinson's disease, Huntington's disease, amyotrophic laterial sclerosis, peripheral neuropathies, spinal cord damage, and cancer.
The methods of this invention include systemic administration of a compound of this invention, preferably in the form of a pharmaceutical composition. As used herein, systemic administration encompasses both oral and parenteral methods of administration. For oral administration, suitable pharmaceutical compositions include powders, granules, pills, tablets, and capsules as well as liquids, syrups, suspensions, and emulsions. These compositions may also include flavorants, preservatives, suspending, thickening and emulsifying agents, and other pharmaceutically acceptable additives. For parental administration, the compounds of the present invention can be prepared in aqueous injection solutions which may contain buffers, antioxidants, bacteriostats. and other additives commonly employed in such solutions. The following examples are offered by way of illustration, not limitation.
EXAMPLES
EXAMPLE 1
Synthesis of Representative Compounds
Figure imgf000025_0001
X = Cl, NO2 A. AnthralT.9cdlpyrazol-6(2H)-one ("Compound 1")
Figure imgf000026_0001
Anhydrous hydrazine is added to a solution of 2-chloroanthraquinone (Aldrich) in 10 mL pyridine, and the mixture heated at 100°C for 16 hours. The mixture is cooled and the solvent is evaporated in vacuo. The residue is taken in hot 6N HC1, and the solid is collected by filtration. Flash chromatography of the crude material on silica gel affords anthra[l,9cd]pyrazol-6(2H)-one ("Compound 1") as yellow solids.
Due to limited solubility of Compound 1 , purification of the same may be achieved by first derivatizing Compound 1 to a more soluble intermediate, such as the corresponding acetate, recrystallizing the intermediate, and then converting the intermediate to yield purified Compound 1 in good yield. More specifically, to solution of the pyrazoloanthrone (9.67 g, 43.9 mmol) in acetic acid (700 mL) is added acetic anhydride (12.4 mL, 132 mmol). The solution is heated to 80°C for 5 hours and then cooled to room temperature. After 16 hours, the reaction is cooled to 0°C for 2 hours. The reaction is then filtered to give the N-acetylpyrazoloanthrone intermediate. This intermediate is recrystallized in acetic acid to give the pure intermediate (5.96 g, 52%). Η NMR (CDCL3) δ 10.6 (br s, IH), 8.46 (d, IH), 8.33 (d, IH), 8.26 (d, IH), 8.08 (d, IH), 7.96-7.87 (m, 2H), 7.78 (t, IH), 2.83 (s, 3H); ES-MS (m/z) 263 [M+1T\ To a solution of the pure intermediate (5.96 g, 23 mmol) in methanol (600 mL) is added ammonium hydroxide (60 mL). The reaction is stirred at room temperature for 16 hours and then filtered and dried in a vacuum oven. A second crop of crystals is recovered to give a total of 4.8 g of Compound 1 at greater than 98%> purity. ES-MS (m/z) 221 [M + l]+. B. 5-Chloroanthraπ.9cd]pyrazol-6(2H)-one
Figure imgf000027_0001
This compound may be made in the same manner from 1,4- dichloroanthraquinone (commercial product).
C. 7-Chloroanthra 1 ,9cd]pyrazol-6(2H)-one
Figure imgf000027_0002
This compound may be made in the same manner from 1,5- dichloroanthraquinone (commercial product).
D. 5-Nitroanthra[ 1.9cd1pyrazol-6(2H)-one
Figure imgf000027_0003
This compound may be made from 1 ,4-dinitroanthraquinone (Krapcho, A. P.; Avery, K. L., Jr. J. Org. Chem. 55, 5562-4, 1990). E. 7-Nitroanthra 1.9cd]pyrazol-6(2H)-one
Figure imgf000028_0001
This compound may be made in the same manner from 1,5- dichloroanthraquinone (commercial product).
F. 5-Benzyloxyanthra|T,9cd]pyrazol-6f2H)-one
Figure imgf000028_0002
This compound may be made in the same manner from l-nitro-4- benzyloxy anthraquinone. This starting material may be prepared as follows. Benzyl bromide is added to 1 -nitro-4-hydroxyanthraquinone (Aldrich) and potassium carbonate in dimethylformamide, and the mixture is stirred for 16 hours. Water is added and the mixture is extracted with ethyl acetate (x2). The combined organic layer is washed sequentially with sodium bicarbonate solution, water, IN hydrochloric acid, and brine, dried, and evaporated. The residue is chromatographed on silica gel to afford l-nitro-4- benzyloxy anthraquinone . 27
G. 7-Benzyloxyanthra|T,9cd1pyrazol-6(2H)-one
Figure imgf000029_0001
This compound may be made in the same manner from l-nitro-5- benzyloxyanthraquinone. which starting material may prepared as disclosed in German Patent No. DE 2254199 to Reubke, Hohmann and Bien.
H. 5-(Acetylamino)anthra[l,9cd]pyrazol-6(2H)-one
Figure imgf000029_0002
This compound may be made in the same manner from 4-acetylamino-l- chloroanthraquinone. This starting material may be prepared as follows. 4-Amino-l- chloroanthraquinone is taken in pyridine and treated with acetic anhydride. The mixture is stirred for 1 hour, and poured onto water. The solids are collected by filtration, washed with water, and dried in vacuo to give 4-acetylamino-l- chloroanthraquinone as a colorless solid.
EXAMPLE 2 Synthesis of Representative Compounds
Figure imgf000030_0001
A. 5-(Dimethylamino)anthra[1.9cd]pyrazol-6(2H)-one
Figure imgf000030_0002
A mixture of 5-chloroanthra[l,9cd]pyrazol-6(2H)-one (Example 1-B) and dimethylamine in pyridine is heated at 100 °C for 16 hours. The mixture is cooled and evaporated. The residue is chromatographed on silica gel to give the desired compound as yellow solids.
B. 5-(l-Piperidinyl)anthra[l,9cd pyrazol-6(2H)-one
Figure imgf000030_0003
This compound may be made in the same manner using piperidine as the amine.
C. 5-(l-Moφholinyl)anthra[1.9cd1pyrazol-6(2H)-one
Figure imgf000031_0001
This compound may be made in the same manner using moφholine as the amine.
D. 5-(Benzylamino)anthraπ .9cd]pyrazol-6(2H)-one
Figure imgf000031_0002
This compound may be made in the same manner using benzylamine as the amine.
E. 5- { (4-Pyridylmethyl)lamino I anthra[ 1.9cd]pyrazol-6(2H)-one
Figure imgf000032_0001
This compound may be made in the same manner using 4- pyridylmethylamine as the amine.
F. 5-{2-(l-Piperidinyl)ethylamino}anthra|"1.9cd]pyrazol-6(2H)-one
Figure imgf000032_0002
This compound may be made in the same manner using 2-(l- piperidyl)ethylamine as the amine.
EXAMPLE 3 Synthesis of Representative Compounds
Figure imgf000032_0003
A. 7-(Dimethylamino)anthra|T,9cd1pyrazol-6f2H)-one
Figure imgf000033_0001
A mixture of 6-chloroanthra[1.9cd]pyrazol-6(2H)-one (Example 1-C) and dimethylamine in pyridine is heated at 100°C for 16 hours. The mixture is cooled and evaporated. The residue is chromatographed on silica gel to give the desired compound as yellow solids.
B. 5-(l-Piperidinyl)anthra|T.9cd]pyrazol-6(2H)-one
Figure imgf000033_0002
This compound may be made in the same manner using piperidine as the amine.
C. 5-( 1 -Moφholinyl)anthra 1.9cd]pyrazol-6(2H)-one
Figure imgf000034_0001
This compound may be made in the same manner using moφholine as the amine.
D. 5-(Benzylamino)anthrari.9cd]pyrazol-6(2H)-one
Figure imgf000034_0002
This compound may be made in the same manner using benzylamine as the amine.
E. 5-{ (4-Pyridylmethyl)lamino ) anthra[" 1 ,9cd]pyrazol-6(2H)-one
Figure imgf000034_0003
This compound may be made in the same manner using 4- pyridylmethylamine as the amine.
F. 5-{2-(l-Piperidinyl)ethylamino}anthra[1.9cd]pyrazol-6(2H)-one
Figure imgf000035_0001
This compound may be made in the same manner using 2-(l- piperidyl)ethylamine as the amine.
EXAMPLE 4 Synthesis of Representative Compounds
Figure imgf000035_0002
Figure imgf000035_0003
Y = C, n = 1
Y = S, n = 2 A. 5-(Benzoylamino)anthra[T.9cd1pyrazol-6(2H)-one
Figure imgf000036_0001
Benzoyl chloride is added to a solution of 2-(methoxyethoxymethyl)-5- aminoanthra[l,9cd]pyrazol-6-(2H)one and triethylamine in methylene chloride at 0°C The mixture is stirred for 16 hours, quenched with water, and extracted with ethyl acetate (x2). The combined organic layer is washed with sodium bicarbonate solution, and brine, dried and evaporated. The crude reaction mixture is then taken in aqueous 6N hydrochloric acid, and heated at 80°C for 4 hours. After cooling, the mixture is extracted with ethyl acetate (x2), washed with brine, dried, and evaporated. The reside is chromatographed on silica gel to furnish the desired amide as a yellow solid.
The starting material is prepared as follows. Sodium hexamethyldisilazide is added to a cooled (0°C) solution of 5-nitroanthra[l ,9cd]pyrazol- 6(2H)-one (Example 1-D) in tetrahydrofuran, and the mixture is stirred for 30 minutes at 0°C MEM-chloride is added, and the mixture is stirred for 16 hours at room temperature. Water is added and the mixture is extracted with ethyl acetate (x2). The combined organic layer is washed with aqueous sodium bicarbonate solution, water, IN hydrochloric acid, and brine, dried and evaporated. The residue is chromatographed on silica gel to give 2-MEM-5-nitroanthra[l ,9cd]pyrazol-6(2H)-one as an oil.
Palladium(10%) on charcoal and 2-MEM-5-nitroanthra[l,9cd]pyrazol- 6(2H)-one in ethanol is placed under 1 -atm of hydrogen, and the mixture was stirred for 6 hours. The catalyst is filtered off over celite. and the filtrate is evaporated to dryness to give 2-(methoxyethoxymethyl)-5-aminoanthra[l,9cd]pyrazol-6-(2H)one, which is used without further purification. B. 5-(Isonicotinylamino)anthraπ.9cd]pyrazol-6(2H)-one
Figure imgf000037_0001
This compound may be made in the same manner using isonicotinoyl chloride as the acid chloride
C. 5-(Nicotinylamino)anthraπ.9cd]pyrazol-6(2H)-one
Figure imgf000037_0002
This compound may be made in the same manner using nicotinoyl chloride as the acid chloride.
D. 5-(2-Thiophenecarbonylamino)anthra l,9cd]pyrazol-6(2H)-one
Figure imgf000037_0003
This compound may be made in the same manner using 2- thiophenecarboxylic acid as the acid chloride.
E. 5-(3-Methylbutyrylamino)anthrari,9cd]pyrazol-6(2H)-one
Figure imgf000038_0001
This compound may be made in the same manner using isopentanoyl chloride as the acid chloride.
F. 5-(3-Methanesulfonylamino)anthra[1.9cd]pyrazol-6(2H)-one
This compound may be made in the same manner using methanesulfonyl chloride as the sulfonyl chloride. G. 5-(3-Benzenesulfonylamino)anthra 1.9cd]pyrazol-6(2H)-one
Figure imgf000039_0001
This compound may be made in the same manner using benzenesulfonyl chloride as the sulfonyl chloride.
EXAMPLE 5 Synthesis of Representative Compounds
Figure imgf000039_0002
Figure imgf000039_0003
Y = C, n = 1
Y = S, n = 2 A. 7-(Benzoylamino anthra[1.9cd]pyrazol-6(2H)-one
Figure imgf000040_0001
Benzoyl chloride is added to a solution of 2-(methoxyethoxymethyl)-7- aminoanthra[l,9cd]pyrazol-6-(2H)one and triethylamine in methylene chloride at 0°C. The mixture is stirred for 16 hours, quenched with water, and extracted with ethyl acetate (x2). The combined organic layer is washed with sodium bicarbonate solution, and brine, dried and evaporated. The crude reaction mixture is then taken in aqueous 6N hydrochloric acid, and heated at 80°C for 4 hours. After cooling, the mixture is extracted with ethyl acetate (x2), washed with brine, dried, and evaporated. The reside is chromatographed on silica gel to furnish the desired amide as a yellow solid.
The starting material is prepared as follows. Sodium hexamethyldisilazide is added to a cooled (0°C) solution of 7-nitroanthra[l,9cd]pyrazol- 6(2H)-one (Example 1-E) in tetrahydrofuran, and the mixture is stirred for 30 minutes at 0°C. MEM-chloride is added, and the mixture is stirred for 16 hours at room temperature. Water is added and the mixture is extracted with ethyl acetate (x2). The combined organic layer is washed with aqueous sodium bicarbonate solution, water, IN hydrochloric acid, and brine, dried and evaporated. The residue is chromatographed on silica gel to give 2-MEM-7-nitroanthra[l ,9cd]pyrazol-6(2H)-one as an oil.
Palladium(10%) on charcoal and 2-MEM-5-nitroanthra[l,9cd]pyrazol- 6(2H)-one in ethanol is placed under 1 -atm of hydrogen, and the mixture was stirred for 6 hours. The catalyst is filtered off over celite. and the filtrate is evaporated to dryness to give 2-(methoxyethoxymethyl)-7-aminoanthra[l,9cd]pyrazol-6-(2H)one, which is used without further purification. B. 7-(Isonicotinylamino)anthra[1.9cd]pyrazol-6(2H)-one
Figure imgf000041_0001
This compound may be made in the same manner using isonicotinoyl chloride as the acid chloride.
C. 7-(Nicotinylamino)anthra 1.9cd1pyrazol-6(2H)-one
Figure imgf000041_0002
This compound may be made in the same manner using nicotinoyl chloride as the acid chloride.
D. 5-(2-Thiophenecarbonylamino)anthra[1.9cd]pyrazol-6(2H)-one
Figure imgf000041_0003
This compound may be made in the same manner using 2- thiophenecarboxylic acid chloride as the acid chloride.
E. 7-(3-Methylbutyrylamino)anthra 1.9cd]pyrazol-6(2H -one
Figure imgf000042_0001
This compound may be made in the same manner using isopentanoyl chloride as the acid chloride.
F. 7-f3-Methanesulfonylamino)anthra[T.9cd]pyrazol-6(2H)-one
Figure imgf000042_0002
This compound may be made in the same manner using methanesulfonyl chloride as the sulfonyl chloride.
G. 7-f3-Benzenesulfonylamino)anthra|T.9cd]pyrazol-6(2H -one
Figure imgf000043_0001
This compound may be made in the same manner using benzenesulfonyl chloride as the sulfonyl chloride.
EXAMPLE 6 Synthesis of Representative Compounds
Figure imgf000043_0002
A. 5-(3-Methylbutyloxy)anthra[1.9cd]pyrazol-6(2H)-one
Figure imgf000043_0003
Isopentyl bromide is added to a mixture of 3-(2- methoxyethoxymethyl)5-hydroxyanthra[ 1 ,9cd]pyrazol-6(2H)-one and potassium carbonate in dimethylformamide at room temperature. After stirring the mixture for sixteen hours, water is added, and the mixture was extracted with ethyl acetate (x2). The combined organic layer is washed with aqueous sodium bicarbonate, water, IN hydrochloric acid, and brine, dried and evaporated. The reside is taken in 6N hydrochloric acid and heated at 80°C for 4 hours. After cooling, the mixture is extracted with ethyl acetate (x2), and the combined organic layer is washed with brine, dried, and evaporated. The residue is purified by column chromatography to afford the title compound as yellow solid.
The starting material is prepared as follows. Sodium hexamethyldisilazide is added to a cooled (0°C) solution of 5- benzyloxyanthra[l,9cd]pyrazol-6(2H)-one (Example 1-F) in tetrahydrofuran, and the mixture is stirred for 30 minutes at 0°C MEM-chloride is added, and the mixture is stirred for 16 hours at room temperature. Water is added and the mixture is extracted with ethyl acetate (x2). The combined organic layer is washed with aqueous sodium bicarbonate solution, water, IN hydrochloric acid, and brine, dried and evaporated. The residue is chromatographed on silica gel to give 2-MEM-5- benzyloxyanthra[l,9cd]pyrazol-6(2H)-one as an oil.
Palladium(10%) on charcoal and 2-MEM-5- benzyloxyanthra[l ,9cd]pyrazol-6(2H)-one in ethanol is placed under 1-atm of hydrogen, and the mixture stirred for 6 hours. The catalyst is filtered off over celite, and the filtrate is evaporated to dryness to give 2-(2-methoxyefhoxymethyl)-5- hydroxyanthra[l,9cd]pyrazol-6-(2H)one, which is used without further purification.
B. 5-(4-Pyridylmethoxy)anthra[T.9cd1pyrazol-6(2H -one
Figure imgf000044_0001
This compound may be made in the same manner using chloromethyl-4- pyridine as the alkyl halide.
C. 5-(3-Pyridylmethoxy)anthra|T.9cd1pyrazol-6(2H)-one
Figure imgf000045_0001
This compound may be made in the same manner using chloromethyl-3 - pyridine as the alkyl halide.
D. 5-(2-Methoxyethoxy)anthra[1.9cdlpyrazol-6(2H)-one
Figure imgf000045_0002
This compound may be made in the same manner using 2 -methoxy ethyl bromide as the alkyl halide.
E. 5-(2-Dimethylaminoethoxy)anthra|T.9cd1pyrazol-6(2H)-one
Figure imgf000045_0003
This compound may be made in the same manner using 2- dimethylaminoethyl chloride as the alkyl halide.
EXAMPLE 7 Synthesis of Representative Compounds
Figure imgf000046_0001
A. 7-(3-Methylbutyloxy)anthra[l,9cd]pyrazol-6(2H)-one
Figure imgf000046_0002
Isopentyl bromide is added to a mixture of 3-(2-methoxyethoxymethyl)-
7-hydroxyanthra[l,9cd]pyrazol-6(2H)-one and potassium carbonate in dimethylformamide at room temperature. After stirring the mixture for sixteen hours, water is added, and the mixture was extracted with ethyl acetate (x2). The combined organic layer is washed with aqueous sodium bicarbonate, water, IN hydrochloric acid, and brine, dried and evaporated. The reside is taken in 6N hydrochloric acid and heated at 80°C for 4 hours. After cooling, the mixture is extracted with ethyl acetate (x2), and the combined organic layer is washed with brine, dried, and evaporated. The residue is purified by column chromatography to afford the title compound as yellow solid. The starting material is prepared as follows. Sodium hexamethyldisilazide is added to a cooled (0°C) solution of 7- benzyloxyanthra[l,9cd]pyrazol-6(2H)-one (Example 1-F) in tetrahydrofuran, and the mixture is stirred for 30 minutes at 0°C. MEM-chloride is added, and the mixture is stirred for 16 hours at room temperature. Water is added and the mixture is extracted with ethyl acetate (x2). The combined organic layer is washed with aqueous sodium bicarbonate solution, water, IN hydrochloric acid, and brine, dried and evaporated. The residue is chromatographed on silica gel to give 2-MEM-7- benzyloxyanthra[l,9cd]pyrazol-6(2H)-one as an oil. Palladium(10%) on charcoal and 2-MEM-7- benzyloxyanthra[l,9cd]pyrazol-6(2H)-one in ethanol is placed under 1-atm of hydrogen, and the mixture was stirred for 6 h. The catalyst is filtered off over celite, and the filtrate is evaporated to dryness to give 2-(2-methoxyethoxymethyl)-7- hydroxyanthra[l ,9cd]pyrazol-6-(2H)one, which is used without further purification.
B. 7-(4-Pyridylmethoxy)anthraf 1 ,9cd]pyrazol-6(2H)-one
Figure imgf000047_0001
This compound may be made in the same manner using chloromethyl-4- pyridine as the alkyl halide. C. 7-(3-Pyridylmethoxy)anthra|T ,9cd]pyrazol-6(2H)-one
Figure imgf000048_0001
This compound may be made in the same manner using chloromethyl-3 - pyridine as the alkyl halide.
D. 7-(2-Methoxyethoxy)anthra|T,9cd]pyrazol-6(2H)-one
Figure imgf000048_0002
This compound may be made in the same manner using 2-methoxyethyl bromide as the alkyl halide.
E. 7-(2-Dimethylaminoethoxy)anthraπ,9cd]pyrazol-6(2H)-one
Figure imgf000048_0003
This compound may be made in the same manner using 2- dimethylaminoethyl chloride as the alkyl halide. EXAMPLE 8 Activity of Representative Compound
The compounds of this invention may be assayed for their activity accordingly to the following procedures.
JNK Assay To 10 μL of the test compound in 20% DMSO/80% dilution buffer consisting of 20 mM HEPES (pH 7.6). 0.1 mM EDTA, 2.5 mM magnesium chloride, 0.004% Triton xlOO, 2 μg/mL leupeptin, 20 mM β-glycerolphosphate, 0.1 mM sodium vanadate, and 2 mM DTT in water is added 30 μL of 50-200 ng His6-JNK1, JNK2 or JNK3 in the same dilution buffer. The mixture is preincubated for 30 minutes at room temperature. Sixty microliter of 10 μg GST-c-Jun(l-79) in assay buffer consisting of 20 mM HEPES (pH 7.6), 50 mM sodium chloride, 0.1 mM EDTA, 24 mM magnesium chloride, 1 mM DTT, 25 mM PNPP, 0.05% Triton xlOO, 11 μM ATP, and 0.5 μCi γ- 32P ATP in water is added and the reaction is allowed to proceed for 1 hour at room temperature. The c-Jun phosphorylation is terminated by addition of 150 μL of 12.5% trichloroacetic acid. After 30 minutes, the precipitate is harvested onto a filter plate, diluted with 50 μL of the scintillation fluid and quantified by a counter. The IC50 values are calculated as the concentration of the test compound at which the c-Jun phosphorylation is reduced to 50% of the control value. Preferred compounds of the present invention have an IC50 value ranging 0.01 - 10 μM in this assay. To this end, a preferred compound of this invention is Compound 1 , which has an IC50 according to this assay of 0.11 μM for JNK1 and JNK2, and 0.15 μM for JNK3.
Selectivity For JNK 48
Compound 1 was also assayed for its inhibitory activity against the following protein kinases by techniques known to those skilled in this field (see, e.g., Protein Phosphorylation, Sefton & Hunter, Eds., Academic Press, pp. 97-367, 1998):
Enzyme IC50 p38-2 >30,000 nM
ERK1 >30,000 nM
MEKK1 >30,000 nM
IKK1 >30,000 nM
IKK2 >30,000 nM
PKA >30,000 nM
PKC > 10,000 nM
EGF-TK > 10,000 nM
Jurkat T-cell 11-2 Production Assay
Jurkat T cells (clone E6-1) are purchased from the American Tissue
Culture Collection and maintained in growth media consisting of RPMI 1640 medium containing 2 mM L-glutamine (Mediatech), with 10% fetal bovine serum (Hyclone) and penicillin/streptomycin. All cells are cultured at 37°C in 95%) air and 5% CO2. Cells are plated at a density of 0.2 x 106 cells per well in 200 μL of media. Compound stock
(20 mM) is diluted in growth media and added to each well as a 1 Ox concentrated solution in a volume of 25 μL, mixed, and allowed to pre-incubate with cells for 30 minutes. The compound vehicle (dimethyl sulfoxide) is maintained at a final concentration of 0.5%> in all samples. After 30 minutes the cells are activated with
PMA (phorbol myristate acetate; final concentration 50 ng/mL) and PHA
(phytohemagglutinin; final concentration 2 μg/mL). PMA and PHA are added as a lOx concentrated solution made up in growth media and added in a volume of 25 μL per well. Cell plates are cultured for 10 hours. Cells are pelleted by centrifugation and the media removed and stored at -20 °C Media aliquots are analyzed by sandwich ELISA for the presence of IL-2 as per the manufacturers instructions (Endogen). The IC50 values are calculated as the concentration of the test compound at which the 11-2 production was reduced to 50%) of the control value. Preferred compounds of the present invention have an IC50 value ranging 0.1 - 30 μM in this assay. Figure 1 presents the dose dependent inhibition of IL-2 in Jarkat T-Cells by Compound 1 according to this procedure, with a resulting IC50 of 5 μM
Mouse in vivo LPS-Induced TNF-α Production Assay
Non-fasted mice are acclimatized for at least 7 days. Groups of 4 to 6 female BALB/c or CD-I mice (8-10 weeks of age from Charles River laboratories) are pretreated with test compound, either by intravenous injection or by oral gavage 15 - 180 minutes prior to the injection of 0.5 mg/kg Bacto LPS from E. coli 055:B5 (Difco Labs). Ninety minutes after LPS challenge, a terminal bleed is performed via abdominal vena cava and blood is allowed to clot at room temperature for 30 minutes in Microtainer serum separator tubes. After separation by centrifugation. the serum is stored frozen at -80°C ELIZA is performed on thawed, diluted samples (1 :10 to 1 :20) using a Mouse TNF-alpha kit (Biosource International). The ED50 values are calculated as the dose of the test compound at which the TNF-α production is reduced to 50%> of the control value. Preferred compounds of the present invention have an ED50 value ranging 1 - 30 mg/kg in this assay. Figure 2 illustrates the results of this experiment utilizing Compound 1 administered by intravenous injection (I.V.) at 15 and 30 mg/kg, as well as by per os (P.O.) at 7.5, 15 and 30 mg/kg. Vehicle alone (PEG-400, propylene glycol, cremophor EL. and ethanol in normal saline, "PPCES") and dexamethasone-21 acetate ("DEX") (1 mg/kg P.O.) were run as controls (n = 6, * = p 0.01). Compound 1 was administered 15 minutes pre-LPS challenge, and bleed occurred 90 minutes post LPS.
Inhibition of Leukocyte Recruitment in Rat Inflamed Lung
Aerosol administration of ovalbumun in Brown Norway Rats previously sensitized by injection of ovalbumin (OA) results in an allergic airway inflammation marked by the generation of an eosinophil- and T-lymphocyte-rich leukocytic 50
infiltration in the lungs (see Richards et al., Am. J. Physiol, 271:2 Pt 1, L267-76, 1996). Compound 1 was administered by subcutaneous injection at a dose of 30 mg/kg, b.i.d. for 3 days prior to ovalbumin challenge by aerosol. Cells counts were obtained from samples of broncho-alveolar lavage, the results of which are illustrated in Figure 3 (V = PPCES vehicle).
Rat In Vivo Adjuvant Arthritis
Male Lewis rats were immunized with complete Freund's adjuvant on day 0 to induce an aggressive arthritis characterized by joint destruction and paw swelling. Compound 1 was administered subcutaneously once daily from day 8 to day 20. Paw swelling was determined be water displacement plethysmometry (see Figure 4A; * = pO.Ol). Radiographs were obtained of the right hind paw to assess bone changes using a semi-quantitative scoring system: demineralization (0-2+), calcaneal erosion (0-1+), and heterotropic bone formation (0-1+), with a maximum possible score = 6 (see Figure 4B). Activation of AP-1 (see Figure 4C) was determined by DNA binding activity in an electrophoretic mobility shift assay (EMSA) (Ausubel et al., Short Protocols in Molecular Biology, Second Edition. John Wiley & Sons Publisher, New York, 1992). Matrix metalloproteinase-13 expression (see Figure 4D) was measured by nothern blot analysis of MMP-13 mRNA (Ausebel et al., supra) (see also Winter et al., Arthritis and Rheumatism P(3):394-404, 1966; Weichman et al., Pharmacological Methods in the Control of Inflammation, Chang and Lewis Eds., Alan R. Liss, Inc., Publ., New York, 1989).
Kainic Acid-Induced Seizure Response Compound 1 was administered to male CD rats at 10 mg/kg intravenously through a tail vein catheter. This was followed immediately by a 30 mg/kg subcutaneous injection. Vehicle controls received the same injection volumes of the PPCES vehicle alone. Thirty minutes later, animals were given a 1- mg/kg i.p. injection of kainic acid in normal saline solution. This dose of kainic acid has been previously reported to induce a seizure syndrome in rats (Maj et al., Eur. J. Pharm. 51
J5°:27-32, 1992). Seizure behavior was monitored for 4 hours following kainic acid injection. As presented in Figure 5, behaviors were assessed based on the following cumulative scoring system: 1 pt. = arrest of motion; 2 pts. = myoclonic jerks of the head and neck (moderate); 3 pts. = unilateral or bilateral forelimb clonic activity; 4 pts. = whole body clonus; 5 pts. = clonic-tonic seizures; 6 pts. = status epilepticus (see also Mathis and Ungerer, Exp. Brain Res. 88:211-282, 1992; Rong et al., Proc. Natl. Acad. Sci. USA 96:9891-9902, 1999; Yang et al., Nature 389:865-810, 1997)
It will be appreciated that, although specific embodiments of the invention have been described herein for puφoses of illustration, various modifications may be made without departing from the spirit and scope of the invention. Accordingly, the invention is not limited except as by the appended claims.

Claims

1. A compound having the structure:
Figure imgf000054_0001
or a pharmaceutically acceptable salt thereof, wherein
R, and R2 are optional substituents that are the same or different and independently represent alkyl, halogen, nitro, trifluoromethyl, sulfonyl, carboxyl, alkoxycarbonyl, alkoxy, aryl, aryloxy, arylalkyloxy, arylalkyl, cycloalkylalkyloxy, cycloalkyloxy, alkoxyalkyl, alkoxyalkoxy, aminoalkoxy, mono- or di-alkylaminoalkoxy, or a group represented by formula (a), (b), (c) or (d):
Figure imgf000054_0002
(a) (b) (c) (
R3 and R4 taken together represent alkylidene or a heteroatom-containing alkylidene, or R3 and R4 are the same or different and independently represent hydrogen, alkyl, cycloalkyl, aryl, arylalkyl, cycloalkylalkyl, aryloxyalkyl, alkoxyalkyl, alkoxyamino, or alkoxy(mono- or di-alkylamino); and R5 represents hydrogen, alkyl, cycloalkyl, aryl, arylalkyl, cycloalkylalkyl, alkoxy, amino, mono- or di-alkylamino, arylamino, arylalkylamino, cycloalkylamino, or cycloalkylalkylamino ; and with the proviso that at least R, or R2 is present.
2. The compound of claim 1 wherein R, or R2 is present, and having one of the following structures:
Figure imgf000055_0001
3. The compound of claim 1 wherein both R, and R2 are present, and having one of the following structures:
Figure imgf000055_0002
4. The compound of claim 2 wherein R, and R2 are:
Figure imgf000055_0003
R4
5. The compound of claim 2 wherein R, and R2 are:
,R3
-NH— (alkyl)— N
R4
6. The compound of claim 2 wherein R, and R2 are:
Q
V -Rs
H
7. The compound of claim 2 wherein R, and R2 are:
Figure imgf000056_0001
8. A composition comprising the compound of claim 1 and a pharmaceutically acceptable carrier.
9. A method for treating a condition responsive to JNK inhibition, comprising administering to a patient in need thereof and effective amount of a compound having the structure:
Figure imgf000057_0001
or a pharmaceutically acceptable salt thereof, wherein
R, and R2 are optional substituents that are the same or different and independently represent alkyl, halogen, nitro, trifluoromethyl, sulfonyl, carboxyl, alkoxycarbonyl, alkoxy, aryl, aryloxy, arylalkyloxy, arylalkyl, cycloalkylalkyloxy, cycloalkyloxy, alkoxyalkyl, alkoxyalkoxy, aminoalkoxy, mono- or di-alkylaminoalkoxy, or a group represented by formula (a), (b), (c) or (d):
Figure imgf000057_0002
(a) (b) (c) (d)
R3 and R4 taken together represent alkylidene or a heteroatom-containing alkylidene, or R3 and R4 are the same or different and independently represent hydrogen, alkyl, cycloalkyl, aryl, arylalkyl, cycloalkylalkyl, aryloxyalkyl, alkoxyalkyl, alkoxyamino, or alkoxy(mono- or di-alkylamino); and R5 represents hydrogen, alkyl, cycloalkyl, aryl, arylalkyl, cycloalkylalkyl, alkoxy, amino, mono- or di-alkylamino, arylamino, arylalkylamino, cycloalkylamino, or cycloalkylalkylamino.
10. The method of claim 9 wherein the condition is cancer.
11. The method of claim 9 wherein the condition is rheumatoid arthritis, rheumatoid spondylitis. osteoarthritis, gout, asthma, bronchitis, cystic fibrosis, inflammatory bowel disease, irritable bowel syndrome, mucous colitis, ulcerative colitis, Crohn's disease, gastritis, esophagitis, hepatitis, multiple sclerosis, endotoxin shock, psoriasis, eczema, or dermatitis.
12. The method of claim 9 wherein the condition is atherosclerosis, restenosis following angioplasty, left ventricular hypertrophy, or myocardial infarction.
13. The method of claim 9 wherein the condition is stroke or ischemic damage to the heart, kidney, liver, or brain.
14. The method of claim 9 wherein the condition is transplant rejection.
15. The method of claim 9 wherein the condition is a central or peripheral neurological degenerative disorder.
16. The method of claim 15 wherein the central or peripheral neurological degenerative disorder is epilepsy, Alzheimer's disease, Parkinson's disease, Huntington's disease, amyotrophic laterial sclerosis, a peripheral neuropathy, or spinal cord damage.
17. The method of claim 9 wherein R, and R2 are not present, and having the following structure:
Figure imgf000059_0001
18. The method of claim 9 wherein R, or R2 is present, and having one of the following structures:
Figure imgf000059_0002
19. The method of claim 9 wherein both R, and R2 are present, and having one of the following structures:
Figure imgf000059_0003
20. The method of claim 18 wherein R, and R, are: R3
R4
21. The method of claim 18 wherein R, and R7 are:
.R3
-NH— (alkyl)— N
R4
22. The method of claim 18 wherein R, and R, are:
Q
V -R5
H
23. The method of claim 18 wherein R, and R2 are:
Figure imgf000060_0001
24. A composition comprising a compound having the structure:
Figure imgf000061_0001
or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier.
PCT/US2000/022727 1999-08-19 2000-08-19 Pyrazoloanthrone and derivatives thereof as jnk inhibitors and their compositions WO2001012609A1 (en)

Priority Applications (7)

Application Number Priority Date Filing Date Title
CA002383268A CA2383268A1 (en) 1999-08-19 2000-08-19 Pyrazoloanthrone and derivatives thereof as jnk inhibitors and their compositions
IL14818700A IL148187A0 (en) 1999-08-19 2000-08-19 Pyrazoloanthrone and derivatives thereof as jnk inhibitors and their compositions
EP00955709A EP1218347A1 (en) 1999-08-19 2000-08-19 Pyrazoloanthrone and derivatives thereof as jnk inhibitors and their compositions
NZ517578A NZ517578A (en) 1999-08-19 2000-08-19 Pyrazoloanthrone and derivatives thereof as JNK inhibitors and their compositions
JP2001517507A JP2003532626A (en) 1999-08-19 2000-08-19 Pyrazoloanthrones as JNK inhibitors, derivatives thereof and compositions thereof
AU67863/00A AU780306B2 (en) 1999-08-19 2000-08-19 Pyrazoloanthrone and derivatives thereof as JNK inhibitors and their compositions
HK02109283.5A HK1053107A1 (en) 1999-08-19 2002-12-23 Pyrazoloanthrone and derivatives thereof as jnk inhibitors and their compositions

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US37823499A 1999-08-19 1999-08-19
US09/378,234 1999-08-19

Publications (1)

Publication Number Publication Date
WO2001012609A1 true WO2001012609A1 (en) 2001-02-22

Family

ID=23492297

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2000/022727 WO2001012609A1 (en) 1999-08-19 2000-08-19 Pyrazoloanthrone and derivatives thereof as jnk inhibitors and their compositions

Country Status (10)

Country Link
EP (1) EP1218347A1 (en)
JP (1) JP2003532626A (en)
KR (2) KR100835700B1 (en)
CN (1) CN1304375C (en)
AU (1) AU780306B2 (en)
CA (1) CA2383268A1 (en)
HK (1) HK1053107A1 (en)
NZ (1) NZ517578A (en)
WO (1) WO2001012609A1 (en)
ZA (1) ZA200201312B (en)

Cited By (29)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002081475A1 (en) * 2001-04-06 2002-10-17 Eisai Co., Limited Jun kinase inhibitors
WO2002092065A2 (en) * 2001-05-16 2002-11-21 Cephalon, Inc. Novel methods for the treatment and prevention of pain using stress-activated protein kinase inhibitors
WO2003072550A1 (en) 2002-02-28 2003-09-04 Eisai Co., Ltd. Novel fused ring indazole compounds
WO2003084503A2 (en) * 2002-04-05 2003-10-16 Boehringer Ingelheim Pharma Gmbh & Co. Kg P38 kinase inhibitors for treating mucus hypersecretion_
WO2003102151A2 (en) * 2002-05-30 2003-12-11 Celgene Corporation Modulating cell differentiation and treating myeloprolifertive disorders with jnk/mkk inhibitors
WO2004039325A2 (en) * 2002-10-24 2004-05-13 Celgene Corporation Treatment of pain with jnk inhibitors
WO2004041191A2 (en) * 2002-10-31 2004-05-21 Celgene Corporation Methods for the treatment, prevention and management of macular degeneration
WO2004084891A1 (en) * 2003-03-24 2004-10-07 Signal Pharmaceuticals, Llc Methods for treating inflammatory conditions or inhibiting jnk
WO2004084901A1 (en) * 2003-03-24 2004-10-07 Signal Pharmaceuticals, Llc Methods for treating or preventing an inflammatory or metabolic condition by inhibiting jnk
WO2005074921A1 (en) * 2004-02-09 2005-08-18 University Of Zurich Treatment of atherosclerosis
EP1587440A2 (en) * 2002-12-31 2005-10-26 Celgene Corporation Drug-coated stents and methods of use therefor
US6982274B2 (en) 2001-04-16 2006-01-03 Eisai Co., Ltd. 1H-indazole compound
WO2006010628A1 (en) * 2004-07-29 2006-02-02 Creabilis Therapeutics S.P.A. Use of k-252a and kinase inhibitors for the prevention or treatment of hmgb1-associated pathologies
WO2006050045A2 (en) * 2004-10-29 2006-05-11 Alcon, Inc. Inhibitors of jun n-terminal kinases for treating glaucomatous retinopathy and ocular diseases
WO2006058007A2 (en) * 2004-11-23 2006-06-01 Celgene Corporation Jnk inhibitors for treatment of cns injury
EP1684690A2 (en) * 2003-11-06 2006-08-02 Celgene Corporation Methods of using and compositions comprising a jnk inhibitor for the treatment and management of asbestos-related diseases and disorders
US7119114B1 (en) 1999-08-19 2006-10-10 Signal Pharmaceuticals, Llc Pyrazoloanthrone and derivatives thereof as JNK inhibitors and compositions and methods related thereto
US7291630B2 (en) 2002-03-28 2007-11-06 Eisai Co., Ltd. Azaindoles as inhibitors of c-Jun N-terminal kinases
US7429609B2 (en) 2002-05-31 2008-09-30 Eisai R & D Management Co., Ltd. Pyrazole compound and medicinal composition containing the same
US7534800B2 (en) 2002-03-28 2009-05-19 Eisai R & D Development Co., Ltd. 7-azaindoles as inhibitors of c-Jun N-terminal kinases for the treatment of neurodegenerative disorders
US7612086B2 (en) 2003-05-16 2009-11-03 Eisai R & D Management Co. Ltd. JNK inhibitors
US7645769B2 (en) 2005-08-05 2010-01-12 Eisai R & D Management Co., Ltd. Inhibitors of c-Jun N-terminal kinases for the treatment of neurodegenerative disorders relating to apoptosis and/or inflammation
US7652137B2 (en) 2003-03-06 2010-01-26 Eisai R & D Management Co., Ltd. Synthesis of 5 substituted 7-azaindoles and 7-azaindolines
US7683078B2 (en) 2001-07-23 2010-03-23 Laboratoires Serono S.A. Arylsulfonamide derivatives as C-Jun-N-Terminal Kinases (JNK's) inhibitors
US7803824B2 (en) 2004-10-29 2010-09-28 Alcon, Inc. Use of inhibitors of Jun N-terminal kinases to treat glaucoma
US8501812B2 (en) 2005-03-29 2013-08-06 University Of Massachusetts Therapeutic methods for type I diabetes
US8592414B2 (en) 2005-07-15 2013-11-26 Merck Serono, S.A. JNK inhibitors for the treatment of endometriosis
US8658640B2 (en) 2005-07-15 2014-02-25 Merck Serono Sa JNK inhibitors for the treatment of endometriosis
US9598669B2 (en) 2005-12-29 2017-03-21 Anthrogenesis Corporation Composition for collecting placental stem cells and methods of using the composition

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR102484810B1 (en) 2016-05-25 2023-01-05 (주)아모레퍼시픽 Skin brightening composition

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR2024807A1 (en) * 1968-11-07 1970-09-04 Ciba Geigy
FR2167626A1 (en) * 1972-01-03 1973-08-24 Basf Ag
FR2336708A1 (en) * 1975-12-27 1977-07-22 Hoechst Ag PHOTOSENSITIVE COPY MATERIAL CONTAINING A SYNERGIC COMBINATION OF INITIATORS.
FR2401915A1 (en) * 1977-09-02 1979-03-30 Ciba Geigy Ag PROCESS FOR THE PREPARATION OF PYRAZOLANTHRONES CARRYING A SUBSTITUTE IN POSITION 3

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR2024807A1 (en) * 1968-11-07 1970-09-04 Ciba Geigy
FR2167626A1 (en) * 1972-01-03 1973-08-24 Basf Ag
FR2336708A1 (en) * 1975-12-27 1977-07-22 Hoechst Ag PHOTOSENSITIVE COPY MATERIAL CONTAINING A SYNERGIC COMBINATION OF INITIATORS.
FR2401915A1 (en) * 1977-09-02 1979-03-30 Ciba Geigy Ag PROCESS FOR THE PREPARATION OF PYRAZOLANTHRONES CARRYING A SUBSTITUTE IN POSITION 3

Non-Patent Citations (21)

* Cited by examiner, † Cited by third party
Title
DATABASE CHEMABS [online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; AMES, JAMES R. ET AL: "An integrated concept of amebicidal action: electron transfer and oxy radicals", XP002152988, retrieved from STN Database accession no. 107:190407 *
DATABASE CHEMABS [online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; ARIENT, JOSEF: "5,8-Dimethylpyrazolanthrone", XP002152991, retrieved from STN Database accession no. 79:6796 *
DATABASE CHEMABS [online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; GALUSHKO, A. M. ET AL: "Derivatives of pyrazoloanthrone. I. Reactivity of 3-aminopyrazoloanthrone", XP002152990, retrieved from STN Database accession no. 87:184421 *
DATABASE CHEMABS [online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; GVON, HAN IR ET AL: "Amino-imino tautomerism and intramolecular cyclization of 4,9-diamino-1,10-anthraquinone-1-tosylimines", XP002152986, retrieved from STN Database accession no. 122:160236 *
DATABASE CHEMABS [online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; HARTLEY, JOHN A. ET AL: "Characteristics of the interaction of anthrapyrazole anticancer agents with deoxyribonucleic acids: structural requirements for DNA binding, intercalation, and photosensitization", XP002152983, retrieved from STN Database accession no. 109:216 *
DATABASE CHEMABS [online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; IVANOVA, T. ET AL: "XPS investigation of electronic structure of pyrazolanthrone and its derivatives", XP002152985, retrieved from STN Database accession no. 127:176108 *
DATABASE CHEMABS [online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; JUDSON, IAN R.: "The anthrapyrazoles: a new class of compounds with clinical activity in breast cancer", XP002152992, retrieved from STN Database accession no. 118:139035 CA *
DATABASE CHEMABS [online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; SHOWALTER, H. D. HOLLIS ET AL: "5-[(Aminoalkyl)amino]-substituted anthra[1,9-cd]pyrazol-6(2H)-ones as novel anticancer agents. Synthesis and biological evaluation", XP002152993, retrieved from STN Database accession no. 100:85627 CA *
DATABASE CHEMABS [online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; SHOWALTER, H. D. HOLLIS ET AL: "Anthrapyrazole anticancer agents. Synthesis and structure-activity relationships against murine leukemias", XP002152984, retrieved from STN Database accession no. 106:32913 *
DATABASE CHEMABS [online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; SINGH, M. R. ET AL: "Reactions of 2,2'-ethylenebis(anthrapyrazolone)", XP002152989, retrieved from STN Database accession no. 89:112272 *
DATABASE CHEMABS [online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; SOKOLYUK, N. T. ET AL: "Synthesis and photochemical properties of peri-phenoxy derivatives of 6H-anthra[1,9-cd]-6-pyrazolone (pyrazole anthrone)", XP002152987, retrieved from STN Database accession no. 120:30709 *
DOKL. AKAD. NAUK (1994), 334(4), 465-8 *
FREE RADICAL BIOL. MED. (1987), 3(2), 85-96 *
INDIAN J. CHEM., SECT. B (1978), 16B(2), 100-2 *
J. MED. CHEM. (1984), 27(3), 253-5, 1984 *
J. MED. CHEM. (1987), 30(1), 121-31 *
KHIM. GETEROTSIKL. SOEDIN. (1977), (7), 956-61 *
MOL. PHARMACOL. (1988), 33(3), 265-71 *
POVERKHNOST (1997), (4-5), 193-201 *
SEMIN. ONCOL. (1992), 19(6), 687-94, 1992 *
ZH. ORG. KHIM. (1992), 28(10), 2193-200 *

Cited By (54)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7119114B1 (en) 1999-08-19 2006-10-10 Signal Pharmaceuticals, Llc Pyrazoloanthrone and derivatives thereof as JNK inhibitors and compositions and methods related thereto
US7335676B2 (en) 1999-08-19 2008-02-26 Signal Pharmaceuticals, Llc Methods for treating inflammatory conditions or inhibiting JNK
US7429599B2 (en) 2000-12-06 2008-09-30 Signal Pharmaceuticals, Llc Methods for treating or preventing an inflammatory or metabolic condition or inhibiting JNK
US7314940B2 (en) 2001-04-06 2008-01-01 Eisai Co., Ltd. Jun kinase inhibitors
WO2002081475A1 (en) * 2001-04-06 2002-10-17 Eisai Co., Limited Jun kinase inhibitors
US7776890B2 (en) 2001-04-16 2010-08-17 Eisai R&D Management Co., Ltd. 1H-indazole compounds
US7541376B2 (en) 2001-04-16 2009-06-02 Eisai R&D Management Co., Ltd. 1H-indazole compounds
US6982274B2 (en) 2001-04-16 2006-01-03 Eisai Co., Ltd. 1H-indazole compound
WO2002092065A3 (en) * 2001-05-16 2003-07-31 Cephalon Inc Novel methods for the treatment and prevention of pain using stress-activated protein kinase inhibitors
WO2002092065A2 (en) * 2001-05-16 2002-11-21 Cephalon, Inc. Novel methods for the treatment and prevention of pain using stress-activated protein kinase inhibitors
US7018999B2 (en) 2001-05-16 2006-03-28 Cephalon, Inc. Methods for the treatment and prevention of pain
US7683078B2 (en) 2001-07-23 2010-03-23 Laboratoires Serono S.A. Arylsulfonamide derivatives as C-Jun-N-Terminal Kinases (JNK's) inhibitors
EP1479670A1 (en) * 2002-02-28 2004-11-24 Eisai Co., Ltd. Novel fused ring indazole compounds
US7390797B2 (en) 2002-02-28 2008-06-24 Eisai Co., Ltd. Fused indazole compounds
WO2003072550A1 (en) 2002-02-28 2003-09-04 Eisai Co., Ltd. Novel fused ring indazole compounds
EP1479670A4 (en) * 2002-02-28 2006-03-08 Eisai Co Ltd Novel fused ring indazole compounds
US7291630B2 (en) 2002-03-28 2007-11-06 Eisai Co., Ltd. Azaindoles as inhibitors of c-Jun N-terminal kinases
US7534800B2 (en) 2002-03-28 2009-05-19 Eisai R & D Development Co., Ltd. 7-azaindoles as inhibitors of c-Jun N-terminal kinases for the treatment of neurodegenerative disorders
WO2003084503A2 (en) * 2002-04-05 2003-10-16 Boehringer Ingelheim Pharma Gmbh & Co. Kg P38 kinase inhibitors for treating mucus hypersecretion_
WO2003084503A3 (en) * 2002-04-05 2004-04-08 Boehringer Ingelheim Pharma P38 kinase inhibitors for treating mucus hypersecretion_
WO2003102151A3 (en) * 2002-05-30 2005-03-03 Celgene Corp Modulating cell differentiation and treating myeloprolifertive disorders with jnk/mkk inhibitors
WO2003102151A2 (en) * 2002-05-30 2003-12-11 Celgene Corporation Modulating cell differentiation and treating myeloprolifertive disorders with jnk/mkk inhibitors
US7429609B2 (en) 2002-05-31 2008-09-30 Eisai R & D Management Co., Ltd. Pyrazole compound and medicinal composition containing the same
WO2004039325A2 (en) * 2002-10-24 2004-05-13 Celgene Corporation Treatment of pain with jnk inhibitors
JP2006511495A (en) * 2002-10-24 2006-04-06 セルジーン・コーポレーション Method for using JNK inhibitor for treatment, prevention, management and / or amelioration of pain and composition containing the same
WO2004039325A3 (en) * 2002-10-24 2004-11-11 Celgene Corp Treatment of pain with jnk inhibitors
WO2004041191A2 (en) * 2002-10-31 2004-05-21 Celgene Corporation Methods for the treatment, prevention and management of macular degeneration
WO2004041191A3 (en) * 2002-10-31 2004-12-02 Celgene Corp Methods for the treatment, prevention and management of macular degeneration
EP1587440A2 (en) * 2002-12-31 2005-10-26 Celgene Corporation Drug-coated stents and methods of use therefor
JP2006512143A (en) * 2002-12-31 2006-04-13 セルジーン・コーポレーション Drug-coated stent and method of using the same
EP1587440A4 (en) * 2002-12-31 2006-08-02 Celgene Corp Drug-coated stents and methods of use therefor
US7652137B2 (en) 2003-03-06 2010-01-26 Eisai R & D Management Co., Ltd. Synthesis of 5 substituted 7-azaindoles and 7-azaindolines
JP2006521394A (en) * 2003-03-24 2006-09-21 シグナル ファーマシューティカルズ,エルエルシー Method of treating or preventing inflammation or metabolic symptoms by inhibiting JNK
WO2004084901A1 (en) * 2003-03-24 2004-10-07 Signal Pharmaceuticals, Llc Methods for treating or preventing an inflammatory or metabolic condition by inhibiting jnk
WO2004084891A1 (en) * 2003-03-24 2004-10-07 Signal Pharmaceuticals, Llc Methods for treating inflammatory conditions or inhibiting jnk
JP2006521395A (en) * 2003-03-24 2006-09-21 シグナル ファーマシューティカルズ,エルエルシー Method for treating inflammatory disease or method for inhibiting JNK
US7612086B2 (en) 2003-05-16 2009-11-03 Eisai R & D Management Co. Ltd. JNK inhibitors
EP1684690A2 (en) * 2003-11-06 2006-08-02 Celgene Corporation Methods of using and compositions comprising a jnk inhibitor for the treatment and management of asbestos-related diseases and disorders
EP1684690A4 (en) * 2003-11-06 2008-10-15 Celgene Corp Methods of using and compositions comprising a jnk inhibitor for the treatment and management of asbestos-related diseases and disorders
WO2005074921A1 (en) * 2004-02-09 2005-08-18 University Of Zurich Treatment of atherosclerosis
WO2006010628A1 (en) * 2004-07-29 2006-02-02 Creabilis Therapeutics S.P.A. Use of k-252a and kinase inhibitors for the prevention or treatment of hmgb1-associated pathologies
WO2006050045A3 (en) * 2004-10-29 2006-12-07 Alcon Inc Inhibitors of jun n-terminal kinases for treating glaucomatous retinopathy and ocular diseases
WO2006050045A2 (en) * 2004-10-29 2006-05-11 Alcon, Inc. Inhibitors of jun n-terminal kinases for treating glaucomatous retinopathy and ocular diseases
US7803824B2 (en) 2004-10-29 2010-09-28 Alcon, Inc. Use of inhibitors of Jun N-terminal kinases to treat glaucoma
EP2248521A1 (en) 2004-10-29 2010-11-10 Alcon, Inc. Inhibitors of jun n-terminal kinases for treating glaucomatous retinopathy and ocular diseases
WO2006058007A3 (en) * 2004-11-23 2006-08-10 Celgene Corp Jnk inhibitors for treatment of cns injury
WO2006058007A2 (en) * 2004-11-23 2006-06-01 Celgene Corporation Jnk inhibitors for treatment of cns injury
US8501812B2 (en) 2005-03-29 2013-08-06 University Of Massachusetts Therapeutic methods for type I diabetes
US8592414B2 (en) 2005-07-15 2013-11-26 Merck Serono, S.A. JNK inhibitors for the treatment of endometriosis
US8658640B2 (en) 2005-07-15 2014-02-25 Merck Serono Sa JNK inhibitors for the treatment of endometriosis
US7645769B2 (en) 2005-08-05 2010-01-12 Eisai R & D Management Co., Ltd. Inhibitors of c-Jun N-terminal kinases for the treatment of neurodegenerative disorders relating to apoptosis and/or inflammation
US9598669B2 (en) 2005-12-29 2017-03-21 Anthrogenesis Corporation Composition for collecting placental stem cells and methods of using the composition
US9725694B2 (en) 2005-12-29 2017-08-08 Anthrogenesis Corporation Composition for collecting and preserving placental stem cells and methods of using the composition
AU2007235111B2 (en) * 2006-03-31 2012-03-22 Alcon, Inc. Use of inhibitors of Jun N-terminal kinases to treat glaucoma

Also Published As

Publication number Publication date
KR20020038733A (en) 2002-05-23
AU780306B2 (en) 2005-03-17
KR20080028516A (en) 2008-03-31
CN1304375C (en) 2007-03-14
HK1053107A1 (en) 2003-10-10
EP1218347A1 (en) 2002-07-03
CA2383268A1 (en) 2001-02-22
CN1379763A (en) 2002-11-13
NZ517578A (en) 2004-02-27
JP2003532626A (en) 2003-11-05
ZA200201312B (en) 2003-02-17
KR100835700B1 (en) 2008-06-09
AU6786300A (en) 2001-03-13

Similar Documents

Publication Publication Date Title
AU780306B2 (en) Pyrazoloanthrone and derivatives thereof as JNK inhibitors and their compositions
AU2002220195B2 (en) Anilinopyrimidine derivatives as IKK inhibitors and compositions and methods related thereto
JP5739527B2 (en) N-cyclic-3- (cyclic carbonylaminomethyl) benzamide derivatives as Rho kinase inhibitors
US7335676B2 (en) Methods for treating inflammatory conditions or inhibiting JNK
CA2659696C (en) 6-1h-imidazo-quinazoline and quinolines derivatives, new potent analgesics and anti-inflammatory agents
US8227480B2 (en) Indazole derivative having spiro ring structure in side chain
US20030073732A1 (en) Isothiazoloanthrones, isoxazoloanthrones, isoindolanthrones and derivatives thereof as JNK inhibitors and compositions and methods related thereto
KR20060025131A (en) Methods for treating or preventing an inflammatory or metabolic condition by inhibiting jnk
EP1349840A2 (en) Anilinopyrimidine derivatives as jnk pathway inhibitors and compositions and methods related thereto
AU2002227214A1 (en) Anilinopyrimidine derivatives as JNK pathway inhibitors and compositions and methods related thereto
EP2128156A1 (en) Novel oxadiazole derivatives and thiadiazole derivatives having neovascularization inhibiting activity
WO2007027842A1 (en) Anilinopyrazole derivatives useful for the treatment of diabetes
WO2003051277A2 (en) Novel compounds
US7119114B1 (en) Pyrazoloanthrone and derivatives thereof as JNK inhibitors and compositions and methods related thereto
JP2008543968A (en) Novel pyrazolopyrimidinone derivatives
CA2446051A1 (en) Phthalazine derivatives with angiogenesis inhibiting activity
AU2002251936B2 (en) Anthrone derivatives and their use as ink inhibitors
Duffy et al. The Discovery of VX-745: A Novel and Selective p38r Kinase Inhibitor
AU2002251936A1 (en) Anthrone derivatives and their use as ink inhibitors

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

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

AL Designated countries for regional patents

Kind code of ref document: A1

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

121 Ep: the epo has been informed by wipo that ep was designated in this application
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
WWE Wipo information: entry into national phase

Ref document number: 2383268

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 148187

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: 2002/01312

Country of ref document: ZA

Ref document number: 200201312

Country of ref document: ZA

WWE Wipo information: entry into national phase

Ref document number: 1020027002168

Country of ref document: KR

WWE Wipo information: entry into national phase

Ref document number: 67863/00

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 517578

Country of ref document: NZ

WWE Wipo information: entry into national phase

Ref document number: 2000955709

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 008143870

Country of ref document: CN

WWP Wipo information: published in national office

Ref document number: 1020027002168

Country of ref document: KR

WWP Wipo information: published in national office

Ref document number: 2000955709

Country of ref document: EP

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

WWP Wipo information: published in national office

Ref document number: 517578

Country of ref document: NZ

WWG Wipo information: grant in national office

Ref document number: 517578

Country of ref document: NZ

WWG Wipo information: grant in national office

Ref document number: 67863/00

Country of ref document: AU