US20020058678A1 - Aromatic heterocyclic compounds as antiinflammatory agents - Google Patents

Aromatic heterocyclic compounds as antiinflammatory agents Download PDF

Info

Publication number
US20020058678A1
US20020058678A1 US09/879,776 US87977601A US2002058678A1 US 20020058678 A1 US20020058678 A1 US 20020058678A1 US 87977601 A US87977601 A US 87977601A US 2002058678 A1 US2002058678 A1 US 2002058678A1
Authority
US
United States
Prior art keywords
alkyl
urea
pyrazol
naphthalen
butyl
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US09/879,776
Inventor
Pier Cirillo
Thomas Gilmore
Eugene Hickey
John Regan
Lin-Hua Zhang
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Individual
Original Assignee
Individual
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Individual filed Critical Individual
Priority to US09/879,776 priority Critical patent/US20020058678A1/en
Publication of US20020058678A1 publication Critical patent/US20020058678A1/en
Priority to US10/165,372 priority patent/US6525046B1/en
Abandoned legal-status Critical Current

Links

Classifications

    • 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
    • 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
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • A61P19/10Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease for osteoporosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • 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
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D231/00Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings
    • C07D231/02Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings
    • C07D231/10Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members
    • C07D231/14Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D231/38Nitrogen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D231/00Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings
    • C07D231/02Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings
    • C07D231/10Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members
    • C07D231/14Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D231/38Nitrogen atoms
    • C07D231/40Acylated on said nitrogen atom
    • 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
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/02Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings
    • C07D405/12Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/12Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
    • C07D417/12Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings linked by a chain containing hetero atoms as chain links

Definitions

  • This invention relates to novel aromatic heterocyclic compounds of formula (I):
  • Ar 1 , Ar 2 , X, L and Q are defined below, which inhibit production of cytokines involved in inflammatory processes and are thus usefuil for treating diseases and pathological conditions involving inflammation such as chronic inflammatory disease.
  • This invention also relates to processes for preparing these compounds and to pharmaceutical compositions comprising these compounds.
  • TNF Tumor necrosis factor
  • IL-1 interleukin-1
  • Elevated levels of proinflammatory cytokines are also associated with a number of diseases of autoimmunity such as toxic shock syndrome, rheumatoid arthritis, osteoarthritis, diabetes and inflammatory bowel disease (Dinarello, C. A., et al., 1984, Rev. Infect. Disease 6:51).
  • diseases of autoimmunity such as toxic shock syndrome, rheumatoid arthritis, osteoarthritis, diabetes and inflammatory bowel disease.
  • chronic elevation of inflammation exacerbates or causes much of the pathophysiology observed.
  • rheumatoid synovial tissue becomes invaded with inflammatory cells that result in destruction to cartilage and bone (Koch, A. E., et al., 1995, J. Invest. Med. 43: 28-38).
  • TNF proinflammatory cytokines
  • IL-1 ⁇ proinflammatory cytokines
  • TNF ⁇ also referred to in its secreted cell-free form termed TNF ⁇
  • IL-1 ⁇ proinflammatory cytokines
  • Efficacy has been demonstrated with a monoclonal antibody directed against TNF ⁇ in a number of autoimmune diseases (Heath, P., “CDP571: An Engineered Human IgG4 Anti-TNF ⁇ Antibody” IBC Meeting on Cytokine Antagonists, Philadelphia, Pa., Apr. 24-5, 1997). These include the treatment of rheumatoid arthritis, Crohn's disease and ulcerative colitis (Rankin, E. C.
  • the monoclonal antibody is thought to function by binding to both soluble TNF ⁇ and to membrane bound TNF.
  • a soluble TNF ⁇ receptor has been engineered that interacts with TNF ⁇ . The approach is similar to that described above for the monoclonal antibodies directed against TNF ⁇ ; both agents bind to soluble TNF ⁇ , thus reducing its concentration.
  • Enbrel Immunex, Seattle, Wash.
  • Another version of the TNF ⁇ receptor, Ro 45-2081 has demonstrated efficacy in various animal models of allergic lung inflammation and acute lung injury.
  • Ro 45-2081 is a recombinant chimeric molecule constructed from the soluble 55 kDa human TNF receptor fused to the hinge region of the heavy chain IgG1 gene and expressed in eukaryotic cells (Renzetti, et al., 1997, Inflamm. Res. 46: S143). IL-1 has been implicated as an immunological effector molecule in a large number of disease processes. IL-1 receptor antagonist (IL-1ra) had been examined in human clinical trials. Efficacy has been demonstrated for the treatment of rheumatoid arthritis (Antril, Amgen). In a phase III human clinical trial IL-1ra reduced the mortality rate in patients with septic shock syndrome (Dinarello, 1995, Nutrution 11, 492).
  • Osteoarthritis is a slow progressive disease characterized by destruction of the articular cartilage.
  • IL-1 is detected in synovial fluid and in the cartilage matrix of osteoarthritic joints.
  • Antagonists of IL-1 have been shown to diminish the degradation of cartilage matrix components in a variety of experimental models of arthritis (Chevalier, 1997, Biomed Pharmacother. 51, 58).
  • Nitric oxide (NO) is a mediator of cardiovascular homeostasis, neurotransmission and immune function; recently it has been shown to have important effects in the modulation of bone remodeling.
  • Cytokines such as IL-1 and TNF are potent stimulators of NO production.
  • NO is an important regulatory molecule in bone with effects on cells of the osteoblast and osteoclast lineage (Evans, et al., 1996, J Bone Miner Res. 11, 300).
  • the promotion of beta-cell destruction leading to insulin dependent diabetes mellitis shows dependence on IL-1. Some of this damage may be mediated through other effectors such as prostaglandins and thromboxanes.
  • IL-1 can effect this process by controlling the level of both cyclooxygenase II and inducible nitric oxide synthetase expression (McDaniel et al., 1996, Proc Soc Exp Biol Med. 211, 24).
  • Inhibitors of cytokine production are expected to block inducible cyclooxygenase (COX-2) expression.
  • COX-2 expression has been shown to be increased by cytokines and it is believed to be the isoform of cyclooxygenase responsible for inflammation (M. K. O'Banion et al., Proc. Natl. Acad. Sci. U.S.A, 1992, 89, 4888.)
  • inhibitors of cytokines such as IL-1 would be expected to exhibit efficacy against those disorders currently treated with COX inhibitors such as the familiar NSAIDs. These disorders include acute and chronic pain as well as symptoms of inflammation and cardiovascular disease.
  • IBD active inflammatory bowel disease
  • IL-1ra showed a clear relationship to acute inflammatory events as well as to the different disease stages in the pathophysiology of HIV infection (Kreuzer, et al., 1997, Clin Exp Immunol. 109, 54).
  • IL-1 and TNF are both involved in periodontal disease. The destructive process associated with periodontal disease may be due to a disregulation of both IL-1 and TNF (Howells, 1995, Oral Dis. 1, 266).
  • TNF ⁇ and IL-1 ⁇ are also important mediators of septic shock and associated cardiopulmonary dysfunction, acute respiratory distress syndrome (ARDS) and multiple organ failure.
  • TNF ⁇ has also been implicated in cachexia and muscle degradation, associated with HIV infection (Lahdiverta et al., 1988, Amer. J. Med., 85, 289). Obesity is associated with an increase incidence of infection, diabetes and cardiovascular disease. Abnormalities in TNF ⁇ expression have been noted for each of the above conditions (Lrissada, et al., 1998, FASEB J. 12, 57). It has been proposed that elevated levels of TNF ⁇ are involved in other eating related disorders such as anorexia and bulimia nervosa.
  • Pathophysiological parallels are drawn between anorexia nervosa and cancer cachexia (Holden, et al., 1996, Med Hypotheses 47, 423).
  • An inhibitor of TNF ⁇ production, HU-211 was shown to improve the outcome of closed brain injury in an experimental model (Shohami, et al., 1997, J Neuroimmunol. 72, 169).
  • Atherosclerosis is known to have an inflammatory component and cytokines such as IL-1 and TNF have been suggested to promote the disease.
  • an IL-1 receptor antagonist was shown to inhibit fatty streak formation (Elhage et al., 1998, Circulation, 97, 242).
  • iNOS inducible nitric oxide synthetase
  • IL-1 has also been shown to induce uveitis in rats which could be inhibited with IL-1 blockers.
  • Cytokines including IL-1, TNF and GM-CSF have been shown to stimulate proliferation of acute myelogenous leukemia blasts (Bruserud, 1996, Leukemia Res. 20, 65).
  • IL-1 was shown to be essential for the development of both irritant and allergic contact dermatitis. Epicutaneous sensitization can be prevented by the administration of an anti- IL-1 monoclonal antibody before epicutaneous application of an allergen (Muller, et al., 1996, Am J Contact Dermat. 7, 177).
  • IL-1 knock out mice Data obtained from IL-1 knock out mice indicates the critical involvement in fever for this cytokine (Kluger et al., 1998, Clin Exp Pharmacol Physiol. 25, 141).
  • a variety of cytokines including TNF, IL-1, IL-6 and IL-8 initiate the acute-phase reaction which is stereotyped in fever, malaise, myalgia, headaches, cellular hypermetabolism and multiple endocrine and enzyme responses (Beisel, 1995, Am J Clin Nutr. 62, 813).
  • the production of these inflammatory cytokines rapidly follows trauma or pathogenic organism invasion.
  • IL-8 correlates with influx of neutrophils into sites of inflammation or injury. Blocking antibodies against IL-8 have demonstrated a role for IL-8 in the neutrophil associated tissue injury in acute inflammation (Harada et al., 1996, Molecular Medicine Today 2, 482).
  • an inhibitor of IL-8 production may be useful in the treatment of diseases mediated predominantly by neutrophils such as stroke and myocardial infarction, alone or following thrombolytic therapy, thermal injury, adult respiratory distress syndrome (ARDS), multiple organ injury secondary to trauma, acute glomerulonephritis, dermatoses with acute inflammatory components, acute purulent meningitis or other central nervous system disorders, hemodialysis, leukopherisis, granulocyte transfusion associated syndromes, and necrotizing entrerocolitis.
  • Rhinovirus triggers the production of various proinflammatory cytokines, predominantly IL-8, which results in symptomatic illnesses such as acute rhinitis (Winther et al., 1998, Am J Rhinol. 12, 17).
  • IL-8 diseases that are effected by IL-8 include myocardial ischemia and reperfusion, inflammatory bowel disease and many others.
  • IL-6 The proinflammatory cytokine IL-6 has been implicated with the acute phase response.
  • IL-6 is a growth factor in a number in oncological diseases including multiple myeloma and related plasma cell dyscrasias (Treon, et al., 1998, Current Opinion in Hematology 5: 42). It has also been shown to be an important mediator of inflammation within the central nervous system. Elevated levels of IL-6 are found in several neurological disorders including AIDS dememtia complex, Alzheimer's disease, multiple sclerosis, systemic lupus erythematosus, CNS trauma and viral and bacterial meningitis (Gruol, et al., 1997, Molecular Neurobiology 15: 307).
  • IL-6 also plays a significant role in osteoporosis. In murine models it has been shown to effect bone resorption and to induce osteoclast activity (Ershler et al., 1997, Development and Comparative Immunol. 21: 487). Marked cytokine differences, such as IL-6 levels, exist in vivo between osteoclasts of normal bone and bone from patients with Paget's disease (Mills, et al., 1997, Calcif Tissue Int. 61, 16). A number of cytokines have been shown to be involved in cancer cachexia.
  • IL-6 and IFN alpha as key factors in both symptom formation and in host defense.
  • Overexpression of IL-6 has been implicated in the pathology of a number of diseases including multiple myeloma, rheumatoid arthritis, Castleman's disease, psoriasis and post-menopausal osteoporosis (Simpson, et al., 1997, Protein Sci.
  • GM-CSF is another proinflammatory cytokine with relevance to a number of therapeutic diseases. It influences not only proliferation and differentiation of stem cells but also regulates several other cells involved in acute and chronic inflammation. Treatment with GM-CSF has been attempted in a number of disease states including burn-wound healing, skin-graft resolution as well as cytostatic and radiotherapy induced mucositis (Masucci, 1996, Medical Oncology 13: 149). GM-CSF also appears to play a role in the replication of human immunodeficiency virus (HIV) in cells of macrophage lineage with relevance to AIDS therapy (Crowe et al., 1997, Journal of Leukocyte Biology 62, 41). Bronchial asthma is characterised by an inflammatory process in lungs. Involved cytokines include GM-CSF amongst others (Lee, 1998, J R Coll Physicians Lond 32, 56).
  • HIV human immunodeficiency virus
  • Interferon ⁇ has been implicated in a number of diseases. It has been associated with increased collagen deposition that is a central histopathological feature of graft-versus-host disease (Parkman, 1998, Curr Opin Hematol. 5, 22). Following kidney transplantation, a patient was diagnosed with acute myelogenous leukemia. Retrospective analysis of peripheral blood cytokines revealed elevated levels of GM-CSF and IFN ⁇ . These elevated levels coincided with a rise in peripheral blood white cell count (Burke, et al., 1995, Leuk Lymphoma. 19, 173).
  • IFN ⁇ insulin-dependent diabetes
  • MS multiple sclerosis
  • AIDS dementia complex Alzheimer's disease
  • T-cells and macrophages are present in these lesions, mainly T-cells and macrophages. These cells produce large amounts of proinflammatory cytokines such as TNF, IL-1 and IFN ⁇ . These cytokines are thought to be involved in promoting apoptosis or programmed cell death of the surrounding vascular smooth muscle cells resulting in the atherosclerotic lesions (Geng, 1997, Heart Vessels Suppl 12, 76). Allergic subjects produce mRNA specific for IFN ⁇ following challenge with Vespula venom (Bonay, et al., 1997, Clin Exp Immunol. 109, 342).
  • cytokines including IFN ⁇ has been shown to increase following a delayed type hypersensitivity reaction thus indicating a role for IFN ⁇ in atopic dermatitis (Szepietowski, et al., 1997, Br J Dermatol. 137, 195). Histopathologic and immunohistologic studies were performed in cases of fatal cerebral malaria. Evidence for elevated IFN ⁇ amongst other cytokines was observed indicating a role in this disease (Udomsangpetch et al., 1997, Am J Trop Med Hyg. 57, 501). The importance of free radical species in the pathogenesis of various infectious diseases has been established.
  • the nitric oxide synthesis pathway is activated in response to infection with certain viruses via the induction of proinflammatory cytokines such as IFN ⁇ (Akaike, et al., 1998, Proc Soc Exp Biol Med. 217, 64).
  • proinflammatory cytokines such as IFN ⁇
  • Patients, chronically infected with hepatitis B virus (HBV) can develop cirrhosis and hepatocellular carcinoma.
  • Viral gene expression and replication in HBV transgenic mice can be suppressed by a post-transcriptional mechanism mediated by IFN ⁇ , TNF and IL-2 (Chisari, et al., 1995, Springer Semin Immunopathol. 17, 261).
  • IFN ⁇ can selectively inhibit cytokine induced bone resorption.
  • NO nitric oxide
  • NO is an important vasodilator and convincing evidence exists for its role in cardiovascular shock (Kilbourn, et al., 1997, Dis Mon. 43, 277).
  • IFN ⁇ is required for progression of chronic intestinal inflammation in such diseases as Crohn's disease and inflammatory bowel disease (IBD) presumably through the intermediacy of CD4 + lymphocytes probably of the TH1 phenotype (Sartor 1996, Aliment Pharmacol Ther. 10 Suppl 2, 43).
  • An elevated level of serum IgE is associated with various atopic diseases such as bronchial asthma and atopic dermatitis.
  • the level of IFN ⁇ was negatively correlated with serum IgE suggesting a role for IFN ⁇ in atopic patients (Teramoto et al., 1998, Clin Exp Allergy 28, 74).
  • WO 98/52558 discloses heteroaryl urea compounds which are indicated to be useful in treating cytokine mediated diseases.
  • U.S. Pat. No. 5,162,360 discloses N-substituted aryl-N′-heterocyclic substituted urea compounds which are described as being useful for treating hypercholesterolemia and atheroclerosis.
  • Ar 1 is a heterocyclic group selected from the group consisting of pyrrole, pyrrolidine, pyrazole, imidazole, oxazole, thiazole, furan and thiophene;
  • Ar 1 may be substituted by one or more R 1 , R 2 or R 3 ;
  • Ar 2 is:
  • L a linking group
  • one or more methylene groups are optionally independently replaced by O, N or S;
  • linking group is optionally substituted with 0-2 oxo groups and one or more C 1-4 branched or unbranched alkyl which may be substituted by one or more halogen atoms;
  • Q is selected from the group consisting of:
  • R 1 is selected from the group consisting of:
  • C 3-7 cycloalkyl selected from the group consisting of cyclopropyl, cyclobutyl, cyclopentanyl, cyclohexanyl, cycloheptanyl, bicyclopentanyl, bicyclohexanyl and bicycloheptanyl, which may optionally be partially or fully halogenated and which may optionally be substituted with one to three C 1-3 alkyl groups, or an analog of such cycloalkyl group wherein one to three ring methylene groups are replaced by groups independently selected from O, S, CHOH, >C ⁇ O, >C ⁇ S and NH;
  • R 2 is selected from the group consisting of:
  • a C 1-6 branched or unbranched alkyl which may optionally be partially or fully halogenated, acetyl, aroyl, C 1-4 branched or unbranched alkoxy, which may optionally be partially or fully halogenated, halogen, methoxycarbonyl and phenylsulfonyl;
  • R 3 is selected from the group consisting of:
  • a phenyl, naphthyl or heterocyclic group selected from the group consisting of pyridinyl, pyrimidinyl, pyrazinyl, pyridazinyl, pyrrolyl, imidazolyl, pyrazolyl, thienyl, furyl, tetrahydrofuryl, isoxazolyl, isothiazolyl, quinolinyl, isoquinolinyl, indolyl, benzimidazolyl, benzofuranyl, benzoxazolyl, benzisoxazolyl, benzpyrazolyl, benzothiofuranyl, cinnolinyl, pterindinyl, phthalazinyl, naphthypyridinyl, quinoxalinyl, quinazolinyl, purinyl and indazolyl; wherein such phenyl, naphthyl or heterocyclic group is optional
  • a fused aryl selected from the group consisting of benzocyclobutanyl, indanyl, indenyl, dihydronaphthyl, tetrahydronaphthyl, benzocycloheptanyl and benzocycloheptenyl, or a fused heterocyclyl selected from the group consisting of cyclopentenopyridine, cyclohexanopyridine, cyclopentanopyrimidine, cyclohexanopyrimidine, cyclopentanopyrazine, cyclohexanopyrazine, cyclopentanopyridazine, cyclohexanopyridazine, cyclopentanoquinoline, cyclohexanoquinoline, cyclopentanoisoquinoline, cyclohexanoisoquinoline, cyclopentanoindole, cyclohexanoindole, cyclopentano
  • cycloalkyl selected from the group consisting of cyclopentanyl, cyclohexanyl, cycloheptanyl, bicyclopentanyl, bicyclohexanyl and bicycloheptanyl, which the cycloalkyl may optionally be partially or fully halogenated and which may optionally be substituted with one to three C 1-3 alkyl groups;
  • R 1 and R 2 taken together may optionally form a fused phenyl or pyridinyl ring,
  • each R 8 , R 13 is independently selected from the group consisting of:
  • each R 4 , R 5 , R 6 , R 7 , R 9 , R 10 , R 11 , and R 12 is independently selected from the group consisting of:
  • a preferred subgeneric aspect of the invention comprises compounds of the formula(I) wherein Ar 2 is naphthyl, tetrahydronaphthyl, indanyl or indenyl.
  • a more preferred subgeneric aspect of the invention comprises compounds of the formula(I) wherein Ar 2 is naphthyl.
  • a yet more preferred subgeneric aspect of the invention comprises compounds of the formula (I), as described in the immediate previous paragraph, wherein:
  • Ar 1 is thiophene or pyrazole
  • Ar 2 is 1-naphthyl
  • L is C 1-6 saturated or unsaturated branched or unbranched carbon chain
  • one or more methylene groups are optionally independently replaced by O, N or S;
  • linking group is optionally substituted with 0-2 oxo groups and one or more C 1-4 branched or unbranched alkyl which may be substituted by one or more halogen atoms;
  • R 1 is selected from the group consisting of C 1-4 alkyl branched or unbranched, cyclopropyl and cyclohexyl which may optionally be partially or fully halogenated and which may optionally be substituted with one to three C 1-3 alkyl groups;
  • R 3 is selected from the group consisting of C 1-4 alkyl branched or unbranched, cyclopropyl, phenyl, pyridinyl each being optionally substituted as described above, alkoxycarbonylalkyl; C 1-6 alkyl branched or unbranched; cyclopropyl or cyclopentyl optionally substituted as described above.
  • a yet further preferred subgeneric aspect of the invention comprises compounds of the formula (I), as described in the immediate previous paragraph, wherein Ar 1 is pyrazole.
  • a still yet further preferred subgeneric aspect of previous the invention comprises compounds of the formula (I), as described in the immediate paragraph, wherein L is C 1-5 saturated carbon chain wherein one or more methylene groups are optionally independently replaced by O, N or S; and
  • linking group is optionally substituted with 0-2 oxo groups and one or more C 1-4 branched or unbranched alkyl which may be substituted by one or more halogen atoms;
  • L is propoxy, ethoxy, methoxy, methyl, propyl, C 3-5 acetylene or methylamino each being optionally substituted are described herein.
  • a more particularly preferred embodiment of L is ethoxy optionally substituted.
  • Preferred compounds of the formula(I) are:
  • Any compounds of this invention containing one or more asymmetric carbon atoms may occur as racemates and racemic mixtures, single enantiomers, diastereomeric mixtures and individual diastereomers. All such isomeric forms of these compounds are expressly included in the present invention.
  • Each stereogenic carbon may be in the R or S configuration, or a combination of configurations.
  • aroyl as used in the present specification shall be understood to mean “benzoyl” or “naphthoyl”.
  • the invention includes pharmaceutically acceptable derivatives of compounds of formula (I).
  • a “pharmaceutically acceptable derivative” refers to any pharmaceutically acceptable salt or ester of a compound of this invention, or any other compound which, upon administration to a patient, is capable of providing (directly or indirectly) a compound of this invention, a pharmacologically active metabolite or pharmacologically active residue thereof.
  • metabolite shall be understood to mean any of the compounds of the formula (I) which are capable of being hydroxylated or oxidized, enzymatically or chemically, as will be appreciated by those skilled in the art.
  • Nonlimiting examples of metabolites of the formula (I) are shown in the table below: Structure Name 1-[5-(2-hydroxy-1,1-dimethyl-ethyl)-2-p- tolyl-2H-pyrazol-3-yl]-3-[4-(2-morpholin- 4-yl-ethoxy)-naphthalen-1-yl]-urea 1-[5-tert-butyl-2-(3-hydroxy-4-methyl- phenyl)-2H-pyrazol-3-yl]-3-[4-(2- morpholin-4-yl-ethoxy)-naphthalen-1-yl]- urea 1-[5-tert-butyl-2-(4-hydroxymethyl- phenyl)-2H-pyrazol-3-yl]-3
  • Pharmaceutically acceptable salts of the compounds of this invention include those derived from pharmaceutically acceptable inorganic and organic acids and bases.
  • suitable acids include hydrochloric, hydrobromic, sulfuric, nitric, perchloric, fumaric, maleic, phosphoric, glycolic, lactic, salicylic, succinic, toluene-p-sulfuric, tartaric, acetic, citric, methanesulfonic, formic, benzoic, malonic, naphthalene-2-sulfuric and benzenesulfonic acids.
  • Salts derived from appropriate bases include alkali metal (e.g., sodium), alkaline earth metal (e.g., magnesium), ammonium and N-(C 1 -C 4 alkyl) 4 + salts.
  • the compounds of this invention include prodrugs of compounds of the formula (I).
  • Prodrugs include those compounds that, upon simple chemical transformation, are modified to produce a compound of formula (I).
  • Simple chemical transformations include hydrolysis, oxidation and reduction. Specifically, when a prodrug of this invention is administered to a patient, the prodrug may be transformed into a compound of formula (I), thereby imparting the desired pharmacological effect.
  • the compounds of the invention may be prepared by Method A, B, or C as illustrated in Scheme I, preferably method C.
  • Method A a mixture of an aminoheterocycle of formula II and an arylisocyanate of formula III is dissolved in a non-protic, anhydrous solvent such as THF, ether, toluene, dioxane or ethyl acetate.
  • a non-protic, anhydrous solvent such as THF, ether, toluene, dioxane or ethyl acetate.
  • the preferred solvent is THF.
  • the mixture is stirred at between 0-45° C., preferably at 25° C., for 2-24 hr, and the volatiles are removed.
  • Method B an aminoheterocycle of formula II is dissolved in a halogenated solvent, such as methylene chloride, chloroform or dichloroethane.
  • a halogenated solvent such as methylene chloride, chloroform or dichloroethane.
  • the preferred solvent is methylene chloride.
  • the mixture is diluted with aqueous alkali, such as sodium bicarbonate or potassium carbonate, cooled in an ice bath and phosgene is added.
  • the mixture is vigorously stirred for 5-30 min, with 10 min being preferable.
  • the organic layer is dried, with agents such as MgSO 4 or Na 2 SO 4 , and the volatiles removed to provide the corresponding isocyanate of formula II.
  • the isocyanate and arylamine IV are mixed in a non-protic, anhydrous solvent such as THF, ether, toluene, dioxane, methylene chloride or ethyl acetate.
  • the preferred solvent is THF.
  • the mixture is stirred at between 0-45° C., preferably at 25° C., for 2-24 hr, and the volatiles are removed. Purification of the residue by recrystallization or by silica gel chromatography, as above, provides the product of formula I.
  • an aminoheterocycle of formula II is dissolved in a halogenated solvent, such as methylene chloride, chloroform or dichloroethane.
  • a halogenated solvent such as methylene chloride, chloroform or dichloroethane.
  • the preferred solvent is methylene chloride.
  • a suitable base such as triethylamine may be added, followed by phenyl chloroformate.
  • the mixture is stirred at between 0-85° C., preferably at reflux temperature, for 2-24 hr, and the volatiles are removed providing carbamate V.
  • the carbamate and arylamine IV are mixed in a non-protic, anhydrous solvent such as THF, ether, toluene, dioxane, methylene chloride or ethyl acetate.
  • the preferred solvent is THF.
  • the mixture is stirred at between 0-110 C., preferably at reflux temperature, for 2-24 hr, and the volatiles are removed.
  • Desired aminopyrazoles of formula XIII can be prepared as described in Scheme II.
  • a hydrazine of formula VIII, bearing substituent R 3 may be prepared by Method D or E.
  • an aryl bromide of formula VI is dissolved in a non-protic, inert solvent, such as THF, 1,4-dioxane or diethyl ether, and cooled to low temperature under an inert atmosphere.
  • the preferred temperature for the solution is ⁇ 77° C.
  • a strong base dissolved in a non-protic, inert solvent, such as hexanes, THF or ether is added dropwise while maintaing a reaction temperature below 0° C. and preferrably below ⁇ 60° C.
  • the preferred bases are alkyl lithium reagents and the most preferred is sec-butyl lithium.
  • the reaction mixture is stirred for a period of time between thirty and ninety minutes or until all the starting aryl bromide has been consumed.
  • An excess of dialkyl azodicarboxylate is added while maintaining a reaction temperature below 0° C. and preferrably below ⁇ 60° C.
  • the preferred dialkyl azodicarboxylate is di-tert-butyl azodicarboxylate.
  • the reaction is stirred at cold temperatures and warmed to room temperature after 0.5 hr to 2 hr.
  • the reaction is quenched with the addition of water and the product extracted into a non-protic solvent, such as ethyl acetate, diethyl ether or chloroform.
  • a non-protic solvent such as ethyl acetate, diethyl ether or chloroform.
  • the organic layers are dried with agents such as MgSO 4 or Na 2 SO 4 and the volatiles removed.
  • the residue is dissolved in protic solvents, such as methanol or iso-propanol, cooled, preferably to 0-5° C. and treated with acid.
  • Preferred acids are hydrochloric, hydrobromic, sulfuric and trifluoroacetic. The most preferred is hydrochloric in gaseous form. After the addition of excess acid the mixture is heated at the reflux temperature of the solvent until all starting material has been consumed. After cooling the product aryl-hydrazine of formula VIII salt is filtered and dried.
  • an aryl amine bearing R 3 of formula VII is dissolved in a concentrated aqueous acid such as hydrochloric, hydrobromic or sulfuric and cooled to ice bath temperatures.
  • a concentrated aqueous acid such as hydrochloric, hydrobromic or sulfuric and cooled to ice bath temperatures.
  • the most preferred acid is hydrochloric with concentrations between 3-8N with the most preferred concentration of 6N.
  • a nitrosating reagent in water is added dropwise while maintaining a cold temperature. The preferred temperature is 0-5° C.
  • the preferred reagent is sodium nitrite.
  • the reaction is stirred between 10-90 min and a reducing agent is added while maintaing cold temperatures. The preferred temperature is 0-5° C.
  • Reducing agents include zinc, iron, samarium iodide and tin(II) chloride.
  • the most preferred agent is tin(II) chlroride dissolved in aqueous hydrochloride with a concentration of 3-8 N with a most preferred concentration of 6N.
  • the reaction is stirred between 0.5-3 hr and quenched with alkali to a pH between 12-14.
  • Alkali reagents include sodium hydroxide, potassium hydroxide, lithium hydroxide and calcium hydroxide.
  • the most preferred alkali reagent is potassium hydroxide.
  • the aqueous solution is extracted with a non-protic organic solvent, such as diethyl ether, chloroform, ethyl acetate and methylene chloride.
  • the organic layers are dried with agents such as MgSO 4 and Na 2 SO 4 and the volatiles removed to provide the aryl-hydrazine (VIII) which can be carried forward without further purification.
  • a ⁇ -ketonitrile bearing R 1 (XII) may be prepared by Method F or G.
  • a metal hydride such as sodium hydride, potassium hydride or lithium hydride
  • an anhydrous, inert, non-protic solvent such as diethyl ether, THF and dioxane
  • the most preferred metal hydride is sodium hydride and the most preferred solvent is THF at a temperature of 75° C.
  • An alkyl ester, preferably a methyl ester (IX), and acetonitrile is dissolved in an anhydrous, inert, non-protic solvent, such as diethyl ether, THF or dioxane and added dropwise to the metal hydride suspension.
  • the preferred solvent is THF.
  • the mixture is kept at elevated temperatures between 3-24 hours, cooled to room temperature and diluted with a non-protic solvent and aqueous acid.
  • the organic layer is washed with water and brine, dried, with agents such as MgSO 4 and Na 2 SO 4 , and the volatiles removed to provide the ⁇ -ketonitrile (XII) which could be used without further purification.
  • a solution of a strong base such as alkyl lithium reagents and metal amide reagents, such as n-butyl lithium, sec-butyl lithium, methyl lithium and lithium diisopropylamide, in an anhydrous, inert, non-protic solvent, such as diethyl ether, THF and dioxane, is cooled below 0° C.
  • a strong base such as alkyl lithium reagents and metal amide reagents, such as n-butyl lithium, sec-butyl lithium, methyl lithium and lithium diisopropylamide
  • an anhydrous, inert, non-protic solvent such as diethyl ether, THF and dioxane
  • the reaction is stirred between 10-45 min while warming to 0° C.
  • the solution of the dianion of cyanoacetic is cooled to temperatures below ⁇ 25° C. and preferrably at ⁇ 77° C.
  • an anhydrous, inert, non-protic solvent such as diethyl ether, THF and dioxane, and most preferrably THF.
  • the reaction mixture is warmed to 0° C. betweeen 10-30 min. and quenched with aqueous acid.
  • the product is extracted with an organic solvent, such as chloroform, ethyl acetate, ether and methylene chloride.
  • the combined organic extracts are dried, with agents such as MgSO 4 and Na 2 SO 4 , and the volatiles removed to provide the ⁇ -ketonitrile (XII) which could be used without further purification.
  • the desired aminopyrazole (XIII) may then be prepared by Method H or I.
  • aryl hydrazine VIII and ⁇ -ketonitrile XII are mixed in an organic solvent, such as toluene, ethanol, iso-propanol or t-butanol.
  • the preferred solvent is ethanol.
  • An acid, such as hydrochloric acid, p-toluene sulfonic acid or sulfuric acid, is added, The preferred acid is concentrated hydrochloric acid.
  • the mixture is heated to temperatures between 50-100° C., preferrably at 80° C., for 10-24 hr and cooled to room temperature.
  • the mixture is diluted with non-protic organic solvent, such as ethyl acetate, ether, chloroform and methylene chloride, and washed with aqueous alkali, such as sodium bicarbonate and potassium carbonate.
  • aqueous alkali such as sodium bicarbonate and potassium carbonate.
  • the organic layer is dried, with agents such as MgSO 4 and Na 2 SO 4 , and the volatiles removed to provide a residue which is purified by recrystallization or silica gel chromatography using hexanes and ethyl acetate as eluents.
  • the product-rich fractions are collected and the volatiles removed to provide the desired amonopyrazole (XIII).
  • aryl hydrazine VIII and ⁇ -ketonitrile XII are mixed in an organic solvent, such as toluene, ethanol, iso-propanol or t-butanol.
  • the preferred solvent is toluene.
  • the mixture is heated at reflux temperatures for 3-24 hrs with azeotropic removal of water and worked up as described above providing the aminopyrazole XIII.
  • a mixture of 1-aryl-5-alkyl-butane-1,4-dione (XIV) and a sulfating reagent, such as Lawesson's reagent or phosphorous (V) sulfide, and preferrably Lawesson's reagent, is dissolved in a non-protic, anhydrous solvent, such as toluene, THF and dioxane.
  • a non-protic, anhydrous solvent such as toluene, THF and dioxane.
  • the preferred solvent is toluene.
  • the mixture is heated at elevated temperatures and preferably at a solvent-refluxing temperature for 1-10 hr.
  • the volatiles are removed and the residue is purified by silica gel chromatography using hexanes and ethyl acetate as eluent.
  • the product-rich fractions are collected and the volatiles removed to provide the substituted thiophene XV.
  • a mixture of substituted thiophene XV is dissolved in a solvent such as acetic anhydride or acetic acid.
  • the preferred solvent is acetic anhydride.
  • the mixture is cooled to 0-30° C. and preferrably to ⁇ 10° C.
  • a solution of concentrated nitric acid in a solvent such as acetic anhydride or acetic acid, with the preferred solvent being acetic anhydride is added while cooling 0-30° C. and preferrably to ⁇ 10° C.
  • the mixture is stirred between 10-120 min, poured onto ice and extracted with a non-protic solvent such as diethyl ether, chloroform, ethyl acetate or methylene chloride.
  • the organic extracts are washed with aqueous alkali, dried with agents such as MgSO 4 and Na 2 SO 4 and the volatiles removed.
  • the residue is purified by silica gel chromatography using hexanes and ethyl acetate as eluents.
  • the product-rich fractions are collected and the volatiles removed to provide the 2-aryl-5-alkyl-3-nitrothiophene.
  • the 2-aryl-5-alkyl-3-nitrothiophene is reduced by metals, such as iron, tin and zinc or catalytic hydrogenation.
  • the preferred reduction occurs with iron in acetic acid at temperatures between 50-110° C. and preferrably at 100° C. for 5-30 min.
  • reaction After cooling to room temperature the reaction is diluted with water, neutralized with alkali, such as sodium hydroxide, potassium hydroxide, potassium carbonate or sodium bicarbonate, and extracted with a non-protic solvent such as diethyl ether, ethyl acetate or methylene chloride.
  • alkali such as sodium hydroxide, potassium hydroxide, potassium carbonate or sodium bicarbonate
  • non-protic solvent such as diethyl ether, ethyl acetate or methylene chloride.
  • organic extracts are dried with agents such as MgSO 4 and NA 2 SO 4 and the volatiles removed to provide the desired aminothiophene XVI.
  • Scheme IV outlines a general scheme for desired aminofurans as described by Stevenson et al. (J. Am. Chem. Soc., 1937, 59, 2525).
  • An ethyl aroylacetate (XVII) is dissolved in a non-protic solvent, such as ether or THF, and treated with a strong base, such as sodium, sodium ethoxide or sodium hydride, and the anion is reacted with a bromomethyl alkylketone (XVIII) at low temperatures, such as 0° C. After stirring the reaction until no starting material remains, it is poured onto cold water and extracted with a non-protic solvent. The combined extracts are dried with agents such as MgSO 4 or Na 2 SO 4 .
  • the diketo-ester (XIX) may be carried forward without further purification or purified by distillation or silica gel chromatography.
  • the diketo-ester in a protic solvent, such as ethanol is heated in the presence of a mineral acid, such as sulfuric or hydrochloric, for 5-10 hr. and extracted with a non-protic solvent. The combined extracts are dried with agents such as MgSO 4 or Na 2 SO 4 .
  • the furan-ester (XX) may be carried forward without further purification or purified by distillation or silica gel chromatography.
  • the furan-ester in a protic solvent, such as ethanol is treated with hydrazine hydrate and the mixture heated for 2-5 days.
  • the hydrazide is isloated as above and treated with hot formic acid and the resulting furan-amine (XXI) purified by distillation or silica gel chromatography.
  • the combined extracts are dried with agents such as MgSO 4 and Na 2 SO 4 . Removal of the volatiles provides the nitro-pyrrole which which may be carried forward without further purification or purified by recrystallization or silica gel chromatography.
  • the nitro-pyrrole is reduced to the amine with iron in acetic acid or by catalytic hydrogenation using palladium on activated carbon.
  • the aminopyrrole (XXVIII) may be carried forward without further purification or purified by recrystallization or silica gel chromatography.
  • Substituted 5-aminothiazoles may be prepared in a manner analogous to Gerwald et al., J. Prakt. Chem. 1973, 315, 539.
  • anhydrous solvent such as ethanol and methanol
  • a base such as triethylamine
  • the imine XXXIV is treated with acid, such as hydrchloric and toluenesulfonic acid, in water and an organic solvent. After the starting material is consumed the reaction is worked up and the product XXXV may be carried forward without further purification or purified by recrystallization or silica gel chromatography.
  • acid such as hydrchloric and toluenesulfonic acid
  • the combined extracts are dried with agents such as MgSO 4 and Na 2 SO 4 and the volatiles removed.
  • the nitrothiophene is reduced to the amine with iron in acetic acid or by catalytic hydrogenation using palladium on activated carbon.
  • the amino-thiophene may be carried forward without further purification or purified by recrystallization or silica gel chromatography.
  • 1,5-Disubstituted-3-aminopyrazoles may be prepared as shown in Scheme X, in a fashion analogous to the procedure described by Ege et al. (J. Het. Chem., 1982, 19, 1267). Potassium is added to anhydrous t-butanol and the mixture cooled to 5° C. Hydrazine XXXVIII is added, followed by cyanodibromoalkane XXXIX. The mixture is heated at refluxing temperatures for 3-10 hr. The mixture is cooled to room temperature and poured onto ice water. The product is extracted with an organic solvent. The combined extracts are dried with agents such as MgSO 4 or Na 2 SO 4 and the volatiles removed. The product XL may be carried forward without further purification or purified by recrystallization or silica gel chromatography.
  • XLVIII 1,4-disubstituted-2-aminopyrroles
  • the synthesis of 1,4-disubstituted-2-aminopyrroles may be accomplished in a manner analogous to Brodrick et al. ( J. Chem. Soc. Perkin Trans. I, 1975, 1910), and as illustrated in Scheme XII.
  • the potassium salt of formylnitrile XLV in water is treated with amine XLVI and acetic acid and the mixture heated at 50-90° C. for 5-30 min.
  • the aminonitrile XLVII is collected by filtration upon cooling and then is stirred at room temperature with a base such as ethanolic potassium ethoxide for 2-5 hr and the volatiles removed.
  • the residue is diluted with water and extracted with an organic solvent.
  • the combined extracts are dried with agents such as MgSO 4 and Na 2 SO 4 and the volatiles removed.
  • the product (XLVIII) may be carried forward without further
  • 2,4-Disubstituted 5-aminooxazoles may be prepared in a manner analogous to the procedure described by Poupaert et al. (Synthesis, 1972, 622) and illustrated in Scheme XIV.
  • Acid chloride LI is added to a cold mixture of 2-aminonitrile LII and a base such as triethylamine in a non-protic solvent, such as THF, benzene, toluene or ether.
  • the preferred temperature is 0° C.
  • the mixture is stirred for 12-24 hr and washed with water.
  • the volatiles are removed and the product LIII treated with ethylmercaptan and dry hydrogen chloride in dry methylene chloride for 5-30 min.
  • the solid 5-imino-1,3-oxazole hydrochloride (LIV) is collected by filtration, dissolved in dry pyridine and the solution saturated with hydrogen sulfide during 4 hr at 0° C.
  • the mixture is diluted with an organic solvent and washed with water and dried. Removal of the volatiles provides the 5-amino-1,3-oxazole product (LV) which may be carried forward without further purification or be purified by silica gel chromatography.
  • the compounds of the invention effectively block inflammatory cytokine production from cells.
  • the inhibition of cytokine production is an attractive means for preventing and treating a variety of disorders associated with excess cytokine production, e.g., diseases and pathological conditions involving inflammation.
  • the compounds of the invention are useful for the treatment of such conditions. These encompass chronic inflammatory diseases including, but not limited to, osteoarthritis, multiple sclerosis, Guillain-Barre syndrome, Crohn's disease, ulcerative colitis, psoriasis, graft versus host disease, systemic lupus erythematosus and insulin-dependent diabetes mellitus.
  • the compounds of the invention can also be used to treat other disorders associated with the activity of elevated levels of proinflammatory cytokines such as responses to various infectious agents and a number of diseases of autoimmunity such as rheumatoid arthritis, toxic shock syndrome, diabetes and inflammatory bowel diseases unrelated to those listed above are discussed in the Background of the Invention.
  • the compounds of the invention being inhibitors of cytokine production are expected to block inducible cyclooxygenase (COX-2) expression.
  • COX-2 expression has been shown to be increased by cytokines and it is believed to be the
  • the compounds of the invention may be useful in the treatment of diseases mediated predominantly by neutrophils such as stroke and myocardial infarction, alone or following thrombolytic therapy, thermal injury, adult respiratory distress syndrome (ARDS), multiple organ injury secondary to trauma, acute glomerulonephritis, dermatoses with acute inflammatory components, acute purulent meningitis or other central nervous system disorders, hemodialysis, leukopherisis, granulocyte transfusion associated syndromes, and necrotizing entrerocolitis.
  • diseases mediated predominantly by neutrophils such as stroke and myocardial infarction, alone or following thrombolytic therapy, thermal injury, adult respiratory distress syndrome (ARDS), multiple organ injury secondary to trauma, acute glomerulonephritis, dermatoses with acute inflammatory components, acute purulent meningitis or other central nervous system disorders, hemodialysis, leukopherisis, granulocyte transfusion associated syndromes, and necrotizing entrerocolitis.
  • ARDS adult respiratory distress syndrome
  • the compounds of the invention may be administered in any conventional dosage form in any conventional manner.
  • Routes of administration include, but are not limited to, intravenously, intramuscularly, subcutaneously, intrasynovially, by infusion, sublingually, transdermally, orally, topically or by inhalation.
  • the preferred modes of administration are oral and intravenous.
  • the compounds of this invention may be administered alone or in combination with adjuvants that enhance stability of the inhibitors, facilitate administration of pharmaceutic compositions containing them in certain embodiments, provide increased dissolution or dispersion, increase inhibitory activity, provide adjunct therapy, and the like, including other active ingredients.
  • combination therapies utilize lower dosages of the conventional therapeutics, thus avoiding possible toxicity and adverse side effects incurred when those agents are used as monotherapies.
  • Compounds of the invention may be physically combined with the conventional therapeutics or other adjuvants into a single pharmaceutical composition.
  • the compounds may then be administered together in a single dosage form.
  • the pharmaceutical compositions comprising such combinations of compounds contain at least about 5%, but more preferably at least about 20%, of a compound of formula (I) (w/w) or a combination thereof.
  • the optimum percentage (w/w) of a compound of formula(I) may vary and is within the purview of those skilled in the art.
  • the compounds may be administered separately (either serially or in parallel). Separate dosing allows for greater flexibility in the dosing regime.
  • dosage forms of the compounds of this invention include pharmaceutically acceptable carriers and adjuvants known to those of ordinary skill in the art.
  • carriers and adjuvants include, for example, ion exchangers, alumina, aluminum stearate, lecithin, serum proteins, buffer substances, water, salts or electrolytes and cellulose-based substances.
  • Preferred dosage forms include, tablet, capsule, caplet, liquid, solution, suspension, emulsion, lozenges, syrup, reconstitutable powder, granule, suppository and transdermal patch. Methods for preparing such dosage forms are known (see, for example, H. C. Ansel and N. G.
  • Dosage levels and requirements are well-recognized in the art and may be selected by those of ordinary skill in the art from available methods and techniques suitable for a particular patient. In some embodiments, dosage levels range from about 10-1000 mg/dose for a 70 kg patient. Although one dose per day may be sufficient, up to 5 doses per day may be given. For oral doses, up to 2000 mg/day may be required. As the skilled artisan will appreciate, lower or higher doses may be required depending on particular factors. For instance, specific dosage and treatment regimens will depend on factors such as the patient's general health profile, the severity and course of the patient's disorder or disposition thereto, and the judgment of the treating physician.
  • LXXII A mixture of LXXI (0.74 g) and HCl (5 mL, 4.0 M in dioxane) in 10 mL anhydrous dioxane was stirred overnight. Collection of the precipitate by vacuum filtration provided LXXII.
  • LXXVII Example 1 (0.23 g), saturated NaHCO 3 (15 mL), dichloromethane (15 mL), phosgene (2.1 mL, 1.93M in toluene) and LXXII (0.32 g) were reacted according to Method B (Scheme I and Example 1).
  • LXXIX A mixture of LXXIX (1.6 g) and sodium iodide (5.0 g) in 10 mL acetone was heated at reflux for 4 days, cooled to room temperature and diluted with dichloromethane. The organics were washed with water, dried (Na 2 SO 4 ) and the volatiles removed in vacuo to provide LXXX.
  • a slurry of diethyl malonate (42 mL) and sodium (4.71 g) was warmed slowly to 90° C. and stirred at 90° C. for 2 hours and 120° C. for 30 min. before being cooled to room temperature.
  • Toluene (200 mL) and 2-chloro-5-nitropyridine (25.0 g) were added and the mixture was heated at 110° C. for 1.5 hours and ambient temperature for 17 h. After removal of the volatiles in vacuo, 6 N HCl (200 mL) was added and the mixture heated to reflux for 4 h and cooled to room temperature.
  • Table 1 illustrates additional compounds of the invention, which were prepared by methods analogous to those described above.
  • TABLE 1 Ex. No. R 1 R 3 Q—L— m.p. ° C. 15 tert-butyl 2-Cl-pyridin-5-yl 2-(morpholin-4-yl)ethoxy 123-125 16 tert-butyl 4-methyl-phenyl 2-(imidazol-1-yl)ethoxy 201-202 17 tert-butyl 2-methoxy-pyridin- 2-(morpholin-4-yl)ethoxy 108-110 5-yl 18 tert-butyl pyridin-3-yl 2-(morpholin-4-yl)ethoxy 191-192 19 tert-butyl 4-Cl-phenyl 2-(morpholin-4-yl)ethoxy 116-118 20 tert-butyl 4-methyl-phenyl pyridin-3-ylmethylamino 137-140 21 tert-butyl 4-methyl
  • the inhibition of cytokine production can be observed by measuring inhibition of TNF ⁇ in lipopolysaccharide stimulated THP cells. All cells and reagents were diluted in RPMI 1640 with phenol red and L-glutamine, supplemented with additional L-glutamine (total: 4 mM), penicillin and streptomycin (50 units/ml each) and fetal bovine serum (FBS, 3%) (GIBCO, all conc. final). Assay was performed under sterile conditions; only test compound preparation was nonsterile. Initial stock solutions were made in DMSO followed by dilution into RPMI 1640 2-fold higher than the desired final assay concentration. Confluent THP.

Abstract

Disclosed are novel aromatic heterocyclic compounds of the formula(I) wherein Ar1, Ar2, L, Q and X are described herein. The compounds are useful in pharmaceutic compositions for treating diseases or pathological conditions involving inflammation such as chronic inflammatory diseases. Also disclosed are processes of making such compounds.
Figure US20020058678A1-20020516-C00001

Description

    RELATED APPLICATION DATA
  • This application is a divisional of U.S. application Ser. No. 09/484,638 filed Jan. 18, 2000.[0001]
  • TECHNICAL FIELD OF THE INVENTION
  • This invention relates to novel aromatic heterocyclic compounds of formula (I): [0002]
    Figure US20020058678A1-20020516-C00002
  • wherein Ar[0003] 1, Ar2, X, L and Q are defined below, which inhibit production of cytokines involved in inflammatory processes and are thus usefuil for treating diseases and pathological conditions involving inflammation such as chronic inflammatory disease. This invention also relates to processes for preparing these compounds and to pharmaceutical compositions comprising these compounds.
  • BACKGROUND OF THE INVENTION
  • Tumor necrosis factor (TNF) and interleukin-1 (IL-1) are important biological entities collectively referred to as proinflammatory cytokines. These, along with several other related molecules, mediate the inflammatory response associated with the immunological recognition of infectious agents. The inflammatory response plays an important role in limiting and controlling pathogenic infections. [0004]
  • Elevated levels of proinflammatory cytokines are also associated with a number of diseases of autoimmunity such as toxic shock syndrome, rheumatoid arthritis, osteoarthritis, diabetes and inflammatory bowel disease (Dinarello, C. A., et al., 1984, [0005] Rev. Infect. Disease 6:51). In these diseases, chronic elevation of inflammation exacerbates or causes much of the pathophysiology observed. For example, rheumatoid synovial tissue becomes invaded with inflammatory cells that result in destruction to cartilage and bone (Koch, A. E., et al., 1995, J. Invest. Med. 43: 28-38). An important and accepted therapeutic approach for potential drug intervention in these diseases is the reduction of proinflammatory cytokines such as TNF (also referred to in its secreted cell-free form termed TNFα) and IL-1β. A number of anti-cytokine therapies are currently in clinical trials. Efficacy has been demonstrated with a monoclonal antibody directed against TNFα in a number of autoimmune diseases (Heath, P., “CDP571: An Engineered Human IgG4 Anti-TNFα Antibody” IBC Meeting on Cytokine Antagonists, Philadelphia, Pa., Apr. 24-5, 1997). These include the treatment of rheumatoid arthritis, Crohn's disease and ulcerative colitis (Rankin, E. C. C., et al., 1997, British J. Rheum. 35: 334-342 and Stack, W. A., et al., 1997, Lancet 349: 521-524). The monoclonal antibody is thought to function by binding to both soluble TNFα and to membrane bound TNF.
  • A soluble TNFα receptor has been engineered that interacts with TNFα. The approach is similar to that described above for the monoclonal antibodies directed against TNFα; both agents bind to soluble TNFα, thus reducing its concentration. One version of this construct, called Enbrel (Immunex, Seattle, Wash.) recently demonstrated efficacy in a Phase III clinical trial for the treatment of rheumatoid arthritis (Brower et al., 1997, [0006] Nature Biotechnology 15: 1240). Another version of the TNFα receptor, Ro 45-2081 (Hofftnan-LaRoche Inc., Nutley, N.J.) has demonstrated efficacy in various animal models of allergic lung inflammation and acute lung injury. Ro 45-2081 is a recombinant chimeric molecule constructed from the soluble 55 kDa human TNF receptor fused to the hinge region of the heavy chain IgG1 gene and expressed in eukaryotic cells (Renzetti, et al., 1997, Inflamm. Res. 46: S143). IL-1 has been implicated as an immunological effector molecule in a large number of disease processes. IL-1 receptor antagonist (IL-1ra) had been examined in human clinical trials. Efficacy has been demonstrated for the treatment of rheumatoid arthritis (Antril, Amgen). In a phase III human clinical trial IL-1ra reduced the mortality rate in patients with septic shock syndrome (Dinarello, 1995, Nutrution 11, 492). Osteoarthritis is a slow progressive disease characterized by destruction of the articular cartilage. IL-1 is detected in synovial fluid and in the cartilage matrix of osteoarthritic joints. Antagonists of IL-1 have been shown to diminish the degradation of cartilage matrix components in a variety of experimental models of arthritis (Chevalier, 1997, Biomed Pharmacother. 51, 58). Nitric oxide (NO) is a mediator of cardiovascular homeostasis, neurotransmission and immune function; recently it has been shown to have important effects in the modulation of bone remodeling. Cytokines such as IL-1 and TNF are potent stimulators of NO production. NO is an important regulatory molecule in bone with effects on cells of the osteoblast and osteoclast lineage (Evans, et al., 1996, J Bone Miner Res. 11, 300). The promotion of beta-cell destruction leading to insulin dependent diabetes mellitis shows dependence on IL-1. Some of this damage may be mediated through other effectors such as prostaglandins and thromboxanes. IL-1 can effect this process by controlling the level of both cyclooxygenase II and inducible nitric oxide synthetase expression (McDaniel et al., 1996, Proc Soc Exp Biol Med. 211, 24).
  • Inhibitors of cytokine production are expected to block inducible cyclooxygenase (COX-2) expression. COX-2 expression has been shown to be increased by cytokines and it is believed to be the isoform of cyclooxygenase responsible for inflammation (M. K. O'Banion et al., [0007] Proc. Natl. Acad. Sci. U.S.A, 1992, 89, 4888.) Accordingly, inhibitors of cytokines such as IL-1 would be expected to exhibit efficacy against those disorders currently treated with COX inhibitors such as the familiar NSAIDs. These disorders include acute and chronic pain as well as symptoms of inflammation and cardiovascular disease.
  • Elevation of several cytokines have been demonstrated during active inflammatory bowel disease (IBD). A mucosal imbalance of intestinal IL-1 and IL-1ra is present in patients with IBD. Insufficient production of endogenous IL-1ra may contribute to the pathogenesis of IBD (Cominelli, et al., 1996, [0008] Aliment Pharmacol Ther. 10, 49). Alzheimer disease is characterized by the presence of beta-amyloid protein deposits, neurofibrillary tangles and cholinergic dysfunction throughout the hippocampal region. The structural and metabolic damage found in Alzheimer disease is possibly due to a sustained elevation of IL-1 (Holden, et al., 1995, Med Hypootheses 45, 559). A role for IL-1 in the pathogenesis of human immunodeficiency virus (HIV) has been identified. IL-1ra showed a clear relationship to acute inflammatory events as well as to the different disease stages in the pathophysiology of HIV infection (Kreuzer, et al., 1997, Clin Exp Immunol. 109, 54). IL-1 and TNF are both involved in periodontal disease. The destructive process associated with periodontal disease may be due to a disregulation of both IL-1 and TNF (Howells, 1995, Oral Dis. 1, 266).
  • Proinflammatory cytokines such as TNFα and IL-1β are also important mediators of septic shock and associated cardiopulmonary dysfunction, acute respiratory distress syndrome (ARDS) and multiple organ failure. TNFα has also been implicated in cachexia and muscle degradation, associated with HIV infection (Lahdiverta et al., 1988, [0009] Amer. J. Med., 85, 289). Obesity is associated with an increase incidence of infection, diabetes and cardiovascular disease. Abnormalities in TNFα expression have been noted for each of the above conditions (Loffreda, et al., 1998, FASEB J. 12, 57). It has been proposed that elevated levels of TNFα are involved in other eating related disorders such as anorexia and bulimia nervosa. Pathophysiological parallels are drawn between anorexia nervosa and cancer cachexia (Holden, et al., 1996, Med Hypotheses 47, 423). An inhibitor of TNFα production, HU-211, was shown to improve the outcome of closed brain injury in an experimental model (Shohami, et al., 1997, J Neuroimmunol. 72, 169). Atherosclerosis is known to have an inflammatory component and cytokines such as IL-1 and TNF have been suggested to promote the disease. In an animal model an IL-1 receptor antagonist was shown to inhibit fatty streak formation (Elhage et al., 1998, Circulation, 97, 242).
  • The abnormal expression of inducible nitric oxide synthetase (iNOS) has been associated with hypertension in the spontaneously hypertensive rat (Chou et al., 1998, [0010] Hypertension, 31, 643). IL-1 has a role in the expression of iNOS and therefore may also have a role in the pathogenesis of hypertension (Singh et al., 1996, Amer. J. Hypertension, 9, 867).
  • IL-1 has also been shown to induce uveitis in rats which could be inhibited with IL-1 blockers. (Xuan et al., 1998, [0011] J. Ocular Pharmacol. and Ther., 14, 31). Cytokines including IL-1, TNF and GM-CSF have been shown to stimulate proliferation of acute myelogenous leukemia blasts (Bruserud, 1996, Leukemia Res. 20, 65). IL-1 was shown to be essential for the development of both irritant and allergic contact dermatitis. Epicutaneous sensitization can be prevented by the administration of an anti- IL-1 monoclonal antibody before epicutaneous application of an allergen (Muller, et al., 1996, Am J Contact Dermat. 7, 177). Data obtained from IL-1 knock out mice indicates the critical involvement in fever for this cytokine (Kluger et al., 1998, Clin Exp Pharmacol Physiol. 25, 141). A variety of cytokines including TNF, IL-1, IL-6 and IL-8 initiate the acute-phase reaction which is stereotyped in fever, malaise, myalgia, headaches, cellular hypermetabolism and multiple endocrine and enzyme responses (Beisel, 1995, Am J Clin Nutr. 62, 813). The production of these inflammatory cytokines rapidly follows trauma or pathogenic organism invasion.
  • Other proinflammatory cytokines have been correlated with a variety of disease states. IL-8 correlates with influx of neutrophils into sites of inflammation or injury. Blocking antibodies against IL-8 have demonstrated a role for IL-8 in the neutrophil associated tissue injury in acute inflammation (Harada et al., 1996, [0012] Molecular Medicine Today 2, 482). Therefore, an inhibitor of IL-8 production may be useful in the treatment of diseases mediated predominantly by neutrophils such as stroke and myocardial infarction, alone or following thrombolytic therapy, thermal injury, adult respiratory distress syndrome (ARDS), multiple organ injury secondary to trauma, acute glomerulonephritis, dermatoses with acute inflammatory components, acute purulent meningitis or other central nervous system disorders, hemodialysis, leukopherisis, granulocyte transfusion associated syndromes, and necrotizing entrerocolitis. Rhinovirus triggers the production of various proinflammatory cytokines, predominantly IL-8, which results in symptomatic illnesses such as acute rhinitis (Winther et al., 1998, Am J Rhinol. 12, 17).
  • Other diseases that are effected by IL-8 include myocardial ischemia and reperfusion, inflammatory bowel disease and many others. [0013]
  • The proinflammatory cytokine IL-6 has been implicated with the acute phase response. IL-6 is a growth factor in a number in oncological diseases including multiple myeloma and related plasma cell dyscrasias (Treon, et al., 1998, [0014] Current Opinion in Hematology 5: 42). It has also been shown to be an important mediator of inflammation within the central nervous system. Elevated levels of IL-6 are found in several neurological disorders including AIDS dememtia complex, Alzheimer's disease, multiple sclerosis, systemic lupus erythematosus, CNS trauma and viral and bacterial meningitis (Gruol, et al., 1997, Molecular Neurobiology 15: 307). IL-6 also plays a significant role in osteoporosis. In murine models it has been shown to effect bone resorption and to induce osteoclast activity (Ershler et al., 1997, Development and Comparative Immunol. 21: 487). Marked cytokine differences, such as IL-6 levels, exist in vivo between osteoclasts of normal bone and bone from patients with Paget's disease (Mills, et al., 1997, Calcif Tissue Int. 61, 16). A number of cytokines have been shown to be involved in cancer cachexia. The severity of key parameters of cachexia can be reduced by treatment with anti IL-6 antibodies or with IL-6 receptor antagonists (Strassmann, et al., 1995, Cytokins Mol Ther. 1, 107). Several infectious diseases, such as influenza, indicate IL-6 and IFN alpha as key factors in both symptom formation and in host defense (Hayden, et al., 1998, J Clin Invest. 101, 643). Overexpression of IL-6 has been implicated in the pathology of a number of diseases including multiple myeloma, rheumatoid arthritis, Castleman's disease, psoriasis and post-menopausal osteoporosis (Simpson, et al., 1997, Protein Sci. 6, 929). Compounds that interfered with the production of cytokines including IL-6, and TNF were effective in blocking a passive cutaneous anaphylaxis in mice (Scholz et al., 1998, J. Med. Chem., 41, 1050).
  • GM-CSF is another proinflammatory cytokine with relevance to a number of therapeutic diseases. It influences not only proliferation and differentiation of stem cells but also regulates several other cells involved in acute and chronic inflammation. Treatment with GM-CSF has been attempted in a number of disease states including burn-wound healing, skin-graft resolution as well as cytostatic and radiotherapy induced mucositis (Masucci, 1996, [0015] Medical Oncology 13: 149). GM-CSF also appears to play a role in the replication of human immunodeficiency virus (HIV) in cells of macrophage lineage with relevance to AIDS therapy (Crowe et al., 1997, Journal of Leukocyte Biology 62, 41). Bronchial asthma is characterised by an inflammatory process in lungs. Involved cytokines include GM-CSF amongst others (Lee, 1998, J R Coll Physicians Lond 32, 56).
  • Interferon γ (IFN γ) has been implicated in a number of diseases. It has been associated with increased collagen deposition that is a central histopathological feature of graft-versus-host disease (Parkman, 1998, [0016] Curr Opin Hematol. 5, 22). Following kidney transplantation, a patient was diagnosed with acute myelogenous leukemia. Retrospective analysis of peripheral blood cytokines revealed elevated levels of GM-CSF and IFN γ. These elevated levels coincided with a rise in peripheral blood white cell count (Burke, et al., 1995, Leuk Lymphoma. 19, 173). The development of insulin-dependent diabetes (Type 1) can be correlated with the accumulation in pancreatic islet cells of T-cells producing IFN γ (Ablumunits, et al., 1998, J Autoimmun. 11, 73). IFN γ along with TNF, IL-2 and IL-6 lead to the activation of most peripheral T-cells prior to the development of lesions in the central nervous system for diseases such as multiple sclerosis (MS) and AIDS dementia complex (Martino et al., 1998, Ann Neurol. 43, 340). Atherosclerotic lesions result in arterial disease that can lead to cardiac and cerebral infarction.
  • Many activated immune cells are present in these lesions, mainly T-cells and macrophages. These cells produce large amounts of proinflammatory cytokines such as TNF, IL-1 and IFN γ. These cytokines are thought to be involved in promoting apoptosis or programmed cell death of the surrounding vascular smooth muscle cells resulting in the atherosclerotic lesions (Geng, 1997, [0017] Heart Vessels Suppl 12, 76). Allergic subjects produce mRNA specific for IFN γ following challenge with Vespula venom (Bonay, et al., 1997, Clin Exp Immunol. 109, 342). The expression of a number of cytokines, including IFN γ has been shown to increase following a delayed type hypersensitivity reaction thus indicating a role for IFN γ in atopic dermatitis (Szepietowski, et al., 1997, Br J Dermatol. 137, 195). Histopathologic and immunohistologic studies were performed in cases of fatal cerebral malaria. Evidence for elevated IFN γ amongst other cytokines was observed indicating a role in this disease (Udomsangpetch et al., 1997, Am J Trop Med Hyg. 57, 501). The importance of free radical species in the pathogenesis of various infectious diseases has been established. The nitric oxide synthesis pathway is activated in response to infection with certain viruses via the induction of proinflammatory cytokines such as IFN γ (Akaike, et al., 1998, Proc Soc Exp Biol Med. 217, 64). Patients, chronically infected with hepatitis B virus (HBV) can develop cirrhosis and hepatocellular carcinoma. Viral gene expression and replication in HBV transgenic mice can be suppressed by a post-transcriptional mechanism mediated by IFN γ, TNF and IL-2 (Chisari, et al., 1995, Springer Semin Immunopathol. 17, 261). IFN γ can selectively inhibit cytokine induced bone resorption. It appears to do this via the intermediacy of nitric oxide (NO) which is an important regulatory molecule in bone remodeling. NO may be involved as a mediator of bone disease for such diseases as: the rheumatoid arthritis, tumor associated osteolysis and postmenopausal osteoporosis (Evans, et al., 1996, J Bone Miner Res. 11, 300). Studies with gene deficient mice have demonstrated that the IL-12 dependent production of IFN γ is critical in the control of early parasitic growth. Although this process is independent of nitric oxide the control of chronic infection does appear to be NO dependent (Alexander et al., 1997, Philos Trans R Soc Lond B Biol Sci 352, 1355). NO is an important vasodilator and convincing evidence exists for its role in cardiovascular shock (Kilbourn, et al., 1997, Dis Mon. 43, 277). IFN γ is required for progression of chronic intestinal inflammation in such diseases as Crohn's disease and inflammatory bowel disease (IBD) presumably through the intermediacy of CD4 + lymphocytes probably of the TH1 phenotype (Sartor 1996, Aliment Pharmacol Ther. 10 Suppl 2, 43). An elevated level of serum IgE is associated with various atopic diseases such as bronchial asthma and atopic dermatitis. The level of IFN γ was negatively correlated with serum IgE suggesting a role for IFN γ in atopic patients (Teramoto et al., 1998, Clin Exp Allergy 28, 74).
  • Compounds which modulate release of one or more of the aforementioned inflammatory cytokines can be useful in treating diseases associated with release of these cytokines. For example, WO 98/52558 discloses heteroaryl urea compounds which are indicated to be useful in treating cytokine mediated diseases. [0018]
  • U.S. Pat. No. 5,162,360 discloses N-substituted aryl-N′-heterocyclic substituted urea compounds which are described as being useful for treating hypercholesterolemia and atheroclerosis. [0019]
  • The work cited above supports the principle that inhibition of cytokine production will be beneficial in the treatment of various disease states. Some protein therapeutics are in late development or have been approved for use in particular diseases. Protein therapeutics are costly to produce and have bioavailability and stability problems. Therefore a need exists for new small molecule inhibitors of cytokine production with optimized efficacy, pharmacokinetic and safety profiles. [0020]
  • BRIEF SUMMARYOF THE INVENTION
  • The work cited above supports the principle that inhibition of cytokine production will be beneficial in the treatment of various disease states. [0021]
  • It is therefore an object of the invention to provide novel compounds which inhibit the release of inflammatory cytokines such as interleukin-1 and tumor necrosis factor. [0022]
  • It is a further object of the invention to provide methods for treating diseases and pathological conditions involving inflammation such as chronic inflammatory disease. [0023]
  • It is yet a further object of the invention to provide processes of preparation of the above-mentioned novel compounds. [0024]
  • DETAILED DESCRIPTION OF THE INVENTION
  • The present invention is directed to compounds of the formula (I): [0025]
    Figure US20020058678A1-20020516-C00003
  • wherein [0026]
  • Ar[0027] 1 is a heterocyclic group selected from the group consisting of pyrrole, pyrrolidine, pyrazole, imidazole, oxazole, thiazole, furan and thiophene;
  • and wherein Ar[0028] 1 may be substituted by one or more R1, R2 or R3;
  • Ar[0029] 2 is:
  • phenyl, naphthyl, quinoline, isoquinoline, tetrahydronaphthyl, tetrahydroquinoline, tetrahydroisoquinoline, benzimidazole, benzofuran, indanyl, indenyl or indole each being optionally substituted with one to three R[0030] 2 groups;
  • L, a linking group, is a: [0031]
  • C[0032] 1-10 saturated or unsaturated branched or unbranched carbon chain;
  • wherein one or more methylene groups are optionally independently replaced by O, N or S; and [0033]
  • wherein said linking group is optionally substituted with 0-2 oxo groups and one or more C[0034] 1-4 branched or unbranched alkyl which may be substituted by one or more halogen atoms;
  • Q is selected from the group consisting of: [0035]
  • a) phenyl, naphthyl, pyridine, pyrimidine, pyridazine, imidazole, benzimidazole, furan, thiophene, pyran, naphthyridine, oxazo[4,5-b]pyridine and imidazo[4,5-b]pyridine, which are optionally substituted with one to three groups selected from the group consisting of halogen, C[0036] 1-6 alkyl, C1-6 alkoxy, hydroxy, mono- or di-(C1-3 alkyl)amino, C1-6 alkyl-S(O)m and phenylamino wherein the phenyl ring is optionally substituted with one to two groups consisting of halogen, C1-6 alkyl and C1-6 alkoxy;
  • b) tetrahydropyran, tetrahydrofuran, 1,3-dioxolanone, 1,3-dioxanone, 1,4-dioxane, morpholine, thiomorpholine, thiomorpholine sulfoxide, thiomorpholine sulfone, piperidine, piperidinone, tetrahydropyrimidone, cyclohexanone, cyclohexanol, pentamethylene sulfide, pentamethylene sulfoxide, pentamethylene sulfone, tetramethylene sulfide, tetramethylene sulfoxide and tetramethylene sulfone which are optionally substituted with one to three groups selected from the group consisting of C[0037] 1-6 alkyl, C1-6 alkoxy, hydroxy, mono- or di-(C1-3 alkyl)amino-C1-3 alkyl, phenylamino-C1-3 alkyl and C1-3 alkoxy-C1-3 alkyl;
  • c) C[0038] 1-6 alkoxy, secondary or tertiary amine wherein the amino nitrogen is covalently bonded to groups selected from the group consisting of C1-3 alkyl and C1-5 alkoxyalkyl and phenyl wherein the phenyl ring is optionally substituted with one to two groups consisting of halogen, C1-6 alkoxy, hydroxy or mono- or di-(C1-3 alkyl)amino, C1-6 alkyl-S(O)r, phenyl-S(O)t, wherein the phenyl ring is optionally substituted with one to two groups consisting of halogen, C1-6 alkoxy, hydroxy or mono- or di-(C1-3 alkyl)amino;
  • R[0039] 1 is selected from the group consisting of:
  • (a) C[0040] 3-10 branched or unbranched alkyl, which may optionally be partially or fully halogenated, and optionally substituted with one to three phenyl, naphthyl or heterocyclic groups selected from the group consisting of pyridinyl, pyrimidinyl, pyrazinyl, pyridazinyl, pyrrolyl, imidazolyl, pyrazolyl, thienyl, furyl, isoxazolyl and isothiazolyl; each such phenyl, naphthyl or heterocycle selected from the group hereinabove described, being substituted with 0 to 5 groups selected from the group consisting of halogen, C1-6 branched or unbranched alkyl which is optionally partially or fully halogenated, C3-8 cycloalkyl, C5-8 cycloalkenyl, hydroxy, cyano, C1-3 alkyloxy which is optionally partially or fully halogenated, NH2C(O) and di(C1-3)alkylaminocarbonyl;
  • (b) C[0041] 3-7 cycloalkyl selected from the group consisting of cyclopropyl, cyclobutyl, cyclopentanyl, cyclohexanyl, cycloheptanyl, bicyclopentanyl, bicyclohexanyl and bicycloheptanyl, which may optionally be partially or fully halogenated and which may optionally be substituted with one to three C1-3 alkyl groups, or an analog of such cycloalkyl group wherein one to three ring methylene groups are replaced by groups independently selected from O, S, CHOH, >C═O, >C═S and NH;
  • (c) C[0042] 3-10 branched alkenyl which may optionally be partially or fully halogenated, and which is optionally substituted with one to three C1-5 branched or unbranched alkyl, phenyl, naphthyl or heterocyclic groups, with each such heterocyclic group being independently selected from the group consisting of pyridinyl, pyrimidinyl, pyrazinyl, pyridazinyl, pyrrolyl, imidazolyl, pyrazolyl, thienyl, furyl, isoxazolyl and isothiazolyl, and each such phenyl, naphthyl or heterocyclic group being substituted with 0 to 5 groups selected from halogen, C1-6 branched or unbranched alkyl which is optionally partially or fully halogenated, cyclopropyl, cyclobutyl, cyclopentanyl, cyclohexanyl, cycloheptanyl, bicyclopentanyl, bicyclohexanyl and bicycloheptanyl, hydroxy, cyano, C1-3 alkyloxy which is optionally partially or fully halogenated, NH2C(O), mono- or di(C1-3)alkylaminocarbonyl;
  • (d) C[0043] 5-7 cycloalkenyl selected from the group consisting of cyclopentenyl, cyclohexenyl, cyclohexadienyl, cycloheptenyl, cycloheptadienyl, bicyclohexenyl and bicycloheptenyl, wherein such cycloalkenyl group may optionally be substituted with one to three C1-3 alkyl groups;
  • (e) cyano; and, [0044]
  • (f) methoxycarbonyl, ethoxycarbonyl and propoxycarbonyl; [0045]
  • R[0046] 2 is selected from the group consisting of:
  • a C[0047] 1-6 branched or unbranched alkyl which may optionally be partially or fully halogenated, acetyl, aroyl, C1-4 branched or unbranched alkoxy, which may optionally be partially or fully halogenated, halogen, methoxycarbonyl and phenylsulfonyl;
  • R[0048] 3 is selected from the group consisting of:
  • a) a phenyl, naphthyl or heterocyclic group selected from the group consisting of pyridinyl, pyrimidinyl, pyrazinyl, pyridazinyl, pyrrolyl, imidazolyl, pyrazolyl, thienyl, furyl, tetrahydrofuryl, isoxazolyl, isothiazolyl, quinolinyl, isoquinolinyl, indolyl, benzimidazolyl, benzofuranyl, benzoxazolyl, benzisoxazolyl, benzpyrazolyl, benzothiofuranyl, cinnolinyl, pterindinyl, phthalazinyl, naphthypyridinyl, quinoxalinyl, quinazolinyl, purinyl and indazolyl; wherein such phenyl, naphthyl or heterocyclic group is optionally substituted with one to five groups selected from the group consisting of a C[0049] 1-6 branched or unbranched alkyl, phenyl, naphthyl, heterocycle selected from the group hereinabove described, C1-6 branched or unbranched alkyl which is optionally partially or fully halogenated, cyclopropyl, cyclobutyl, cyclopentanyl, cyclohexanyl, cycloheptanyl, bicyclopentanyl, bicyclohexanyl, bicycloheptanyl, phenyl C1-5 alkyl, naphthyl C1-5 alkyl, halo, hydroxy, cyano, C1-3 alkyloxy which may optionally be partially or fully halogenated, phenyloxy, naphthyloxy, heteraryloxy wherein the heterocyclic moiety is selected from the group hereinabove described, nitro, amino, mono- or di-(C1-3)alkylamino, phenylamino, naphthylamino, heterocyclylamino wherein the heterocyclyl moiety is selected from the group hereinabove described, NH2C(O), a mono- or di-(C1-3)alkyl aminocarbonyl, C1-5 alkyl-C(O)—C1-4 alkyl, amino-C1-5alkyl, mono- or di-(C1-3)alkylamino-C1-5alkyl, amino-S(O)2, di-(C1-3)alkylamino-S(O)2, R4-C1-5 alkyl, R5 -C1-5 alkoxy, R6-C(O)—C1-5 alkyl and R7 -C1-5 alkyl(R8)N;
  • b) a fused aryl selected from the group consisting of benzocyclobutanyl, indanyl, indenyl, dihydronaphthyl, tetrahydronaphthyl, benzocycloheptanyl and benzocycloheptenyl, or a fused heterocyclyl selected from the group consisting of cyclopentenopyridine, cyclohexanopyridine, cyclopentanopyrimidine, cyclohexanopyrimidine, cyclopentanopyrazine, cyclohexanopyrazine, cyclopentanopyridazine, cyclohexanopyridazine, cyclopentanoquinoline, cyclohexanoquinoline, cyclopentanoisoquinoline, cyclohexanoisoquinoline, cyclopentanoindole, cyclohexanoindole, cyclopentanobenzimidazole, cyclohexanobenzimidazole, cyclopentanobenzoxazole, cyclohexanobenzoxazole, cyclopentanoimidazole, cyclohexanoimidazole, cyclopentanothiophene and cyclohexanothiophene; wherein the fused aryl or fused heterocyclyl ring is substituted with 0 to 3 groups independently selected from phenyl, naphthyl and heterocyclyl selected from the group consisting of pyridinyl, pyrimidinyl, pyrazinyl, pyridazinyl, pyrrolyl, imidazolyl, pyrazolyl, thienyl, furyl, isoxazolyl, and isothiazolyl, C[0050] 1-6 branched or unbranched alkyl which is optionally partially or fully halogenated, halo, cyano, C1-3 alkyloxy which is optionally partially or fully halogenated, phenyloxy, naphthyloxy, heterocyclyloxy wherein the heterocyclyl moiety is selected from the group hereinabove described, nitro, amino, mono- or di-(C1-3)alkylamino, phenylamino, naphthylamino, heterocyclylamino wherein the heterocyclyl moiety is selected from the group hereinabove described, NH2C(O), a mono- or di-(C1-3)alkyl aminocarbonyl, C1-4 alkyl-OC(O), C1-5 alkyl-C(O)—C1-4 branched or unbranched alkyl, an amino-C1-5 alkyl, mono- or di-(C1-3)alkylamino-C1-5 alkyl, R9-C1-5 alkyl, R10-C1-5 alkoxy, R11-C(O)—C1-5 alkyl, and R12-C15 alkyl(R13)N;
  • c) cycloalkyl selected from the group consisting of cyclopentanyl, cyclohexanyl, cycloheptanyl, bicyclopentanyl, bicyclohexanyl and bicycloheptanyl, which the cycloalkyl may optionally be partially or fully halogenated and which may optionally be substituted with one to three C[0051] 1-3 alkyl groups;
  • d) C[0052] 5-7 cycloalkenyl, selected from the group consisting of cyclopentenyl, cyclohexenyl, cyclohexadienyl, cycloheptenyl, cycloheptadienyl, bicyclohexenyl and bicycloheptenyl, wherein such cycloalkenyl group may optionally be substituted with one to three C1-3 alkyl groups; and
  • e) acetyl, aroyl, alkoxycarbonylalkyl or phenylsulfonyl; [0053]
  • f) C[0054] 1-6 branched or unbranched alkyl which may optionally be partially or fully halogenated; wherein
  • or R[0055] 1 and R2 taken together may optionally form a fused phenyl or pyridinyl ring,
  • each R[0056] 8, R13 is independently selected from the group consisting of:
  • hydrogen and C[0057] 1-4 branched or unbranched alkyl which may optionally be partially or fully halogenated;
  • each R[0058] 4, R5, R6, R7, R9, R10, R11, and R12 is independently selected from the group consisting of:
  • morpholine, piperidine, piperazine, imidazole and tetrazole; [0059]
  • m=0, 1, 2; [0060]
  • r=0, 1, 2; [0061]
  • t=0, 1, 2; [0062]
  • X=O or S and [0063]
  • physiologically acceptable acids or salts thereof. [0064]
  • A preferred subgeneric aspect of the invention comprises compounds of the formula(I) wherein Ar[0065] 2 is naphthyl, tetrahydronaphthyl, indanyl or indenyl.
  • A more preferred subgeneric aspect of the invention comprises compounds of the formula(I) wherein Ar[0066] 2 is naphthyl.
  • A yet more preferred subgeneric aspect of the invention comprises compounds of the formula (I), as described in the immediate previous paragraph, wherein: [0067]
  • Ar[0068] 1 is thiophene or pyrazole;
  • Ar[0069] 2 is 1-naphthyl;
  • L is C[0070] 1-6 saturated or unsaturated branched or unbranched carbon chain wherein
  • one or more methylene groups are optionally independently replaced by O, N or S; and [0071]
  • wherein said linking group is optionally substituted with 0-2 oxo groups and one or more C[0072] 1-4 branched or unbranched alkyl which may be substituted by one or more halogen atoms;
  • R[0073] 1 is selected from the group consisting of C1-4alkyl branched or unbranched, cyclopropyl and cyclohexyl which may optionally be partially or fully halogenated and which may optionally be substituted with one to three C1-3 alkyl groups;
  • R[0074] 3 is selected from the group consisting of C1-4alkyl branched or unbranched, cyclopropyl, phenyl, pyridinyl each being optionally substituted as described above, alkoxycarbonylalkyl; C1-6alkyl branched or unbranched; cyclopropyl or cyclopentyl optionally substituted as described above.
  • A yet further preferred subgeneric aspect of the invention comprises compounds of the formula (I), as described in the immediate previous paragraph, wherein Ar[0075] 1 is pyrazole.
  • A still yet further preferred subgeneric aspect of previous the invention comprises compounds of the formula (I), as described in the immediate paragraph, wherein L is C[0076] 1-5 saturated carbon chain wherein one or more methylene groups are optionally independently replaced by O, N or S; and
  • wherein said linking group is optionally substituted with 0-2 oxo groups and one or more C[0077] 1-4 branched or unbranched alkyl which may be substituted by one or more halogen atoms;
  • Particularly preferred embodiments of L are propoxy, ethoxy, methoxy, methyl, propyl, C[0078] 3-5 acetylene or methylamino each being optionally substituted are described herein.
  • A more particularly preferred embodiment of L is ethoxy optionally substituted. [0079]
  • The following compounds are representative of the compounds of formula(I): [0080]
  • 1-[5-tert-Butyl-2-p-tolyl-2H-pyrazol-3-yl]-3-[4-(2-morpholin-4-yl-ethoxy)naphthalen-1-yl]-urea; [0081]
  • 1-[5-tert-Butyl-2-p-tolyl-2H-pyrazol-3-yl]-3-[4-(2-(cis-2,6-dimethylmorpholin-4-yl)ethoxy)naphthalen-1-yl]-urea; [0082]
  • 1-[5-tert-Butyl-2-p-tolyl-2H-pyrazol-3-yl]-3-[4-(2-(trans-2,6-dimethylmorpholin-4-yl)ethoxy)naphthalen-1-yl]-urea; [0083]
  • 1-[5-tert-Butyl-2-p-tolyl-2H-pyrazol-3-yl]-3-[4-(2-(2-(methoxymethyl)morpholin-4-yl)ethoxy)naphthalen-1-yl]-urea; [0084]
  • 1-[5-tert-Butyl-2-p-tolyl-2H-pyrazol-3-yl]-3-[4-(2-(morpholin-4-yl)-2-oxoethoxy)naphthalen-1-yl]-urea; [0085]
  • 1-[5-tert-Butyl-2-p-tolyl-2H-pyrazol-3-yl]-3-[4-(2-(morpholin-4-yl)-2-methylethoxy)naphthalen-1-yl]-urea; [0086]
  • 1-[5-tert-Butyl-2-p-tolyl-2H-pyrazol-3-yl]-3-[4-(2-(morpholin-4-yl)-1-methylethoxy)naphthalen-1-yl]-urea; [0087]
  • 1-[5-tert-Butyl-2-p-tolyl-2H-pyrazol-3-yl]-3-[4-(2-thiomorpholin-4-yl-ethoxy)naphthalen-1-yl]-urea; [0088]
  • 1-[5-tert-Butyl-2-p-tolyl-2H-pyrazol-3-yl]-3-[4-(2-(1-oxothiomorpholin-4-yl)ethoxy)naphthalen-1-yl]-urea; [0089]
  • 1-[5-tert-Butyl-2-p-tolyl-2H-pyrazol-3-yl]-3-[4-(2-morpholin-4-yl-ethoxy)-3-methylnaphthalen-1-yl]-urea; [0090]
  • 1-[5-tert-Butyl-2-p-tolyl-2H-pyrazol-3-yl]-3-[4-(2-piperidin-4-yl-ethoxy)naphthalen-1-yl]-urea; [0091]
  • 1-[5-tert-Butyl-2-p-tolyl-2H-pyrazol-3-yl]-3-[4-(2-(1-acetylpiperidin-4-yl)ethoxy)naphthalen-1-yl]-urea; [0092]
  • 1-[5-tert-Butyl-2-p-tolyl-2H-pyrazol-3-yl]-3-[4-(2-thiazolidin-3-yl-ethoxy)naphthalen-1-yl]-urea; [0093]
  • 1-[5-tert-Butyl-2-p-tolyl-2H-pyrazol-3-yl]-3-[4-(2-(morpholin-4-yl-carbonyloxo)ethoxy)naphthalen-1-yl]-urea; [0094]
  • 1-[5-tert-Butyl-2-p-tolyl-2H-pyrazol-3-yl]-3-[4-(2-(tetrahydropyran-4-yl)ethoxy)naphthalen-1-yl]-urea; [0095]
  • 1-[5-tert-Butyl-2-p-tolyl-2H-pyrazol-3-yl]-3-[4-(2-(N-methyl-2-methoxyethylamino)ethoxy)naphthalen-1-yl]-urea; [0096]
  • 1-[5-tert-Butyl-2-p-tolyl-2H-pyrazol-3-yl]-3-[4-(2-(1-oxo-tetrahydrothiophen-3-yl)ethoxy)naphthalen-1-yl]-urea; [0097]
  • 1-[5-tert-Butyl-2-p-tolyl-2H-pyrazol-3-yl]-3-[4-(3-morpholin-4-yl-propyl)naphthalen-1-yl]-urea; [0098]
  • 1-[5-tert-Butyl-2-p-tolyl-2H-pyrazol-3-yl]-3-[4-(morpholin-4-yl-methyl)naphthalen-1-yl]-urea; [0099]
  • 1-[5-tert-Butyl-2-p-tolyl-2H-pyrazol-3-yl]-3-[4-(3-thiazolidin-3-yl-propyl)naphthalen-1-yl]-urea; [0100]
  • 1-[5-tert-Butyl-2-p-tolyl-2H-pyrazol-3-yl]-3-[4-(3-(tetrahydopyran-2-yl-oxy)propyl)naphthalen-1-yl]-urea; [0101]
  • 1-[5-tert-Butyl-2-p-tolyl-2H-pyrazol-3-yl]-3-[4-(2-pyridin-4-yl-ethyl)naphthalen-1-yl]-urea; [0102]
  • 1-[5-tert-Butyl-2-p-tolyl-2H-pyrazol-3-yl]-3-[4-(2-pyridin-4-yl-ethenyl)naphthalen-1-yl]-urea; [0103]
  • 1-[5-tert-Butyl-2-p-tolyl-2H-pyrazol-3-yl]-3-[4-(3-(morpholin-4-yl)propyn-1-yl)naphthalen-1-yl]-urea; [0104]
  • 1-[5-tert-Butyl-2-p-tolyl-2H-pyrazol-3-yl]-3-[4-(3-(tetrahydropyran-2-yl-oxy)propyn-1-yl)naphthalen-1-yl]-urea; [0105]
  • 1-[5-tert-Butyl-2-p-tolyl-2H-pyrazol-3-yl]-3-[4-(3-(methoxymethyloxy)propyn-1-yl)naphthalen-1-yl]-urea; [0106]
  • 1-[5-tert-Butyl-2-p-tolyl-2H-pyrazol-3-yl]-3-[4-(3-(morpholin-4-yl)-3-methylpropyn-1-yl)naphthalen-1-yl]-urea; [0107]
  • 1-[5-tert-Butyl-2-p-tolyl-2H-pyrazol-3-yl]-3-[4-(3-(morpholin-4-yl)-3,3-dimethylpropyn-1-yl)naphthalen-1-yl]-urea; [0108]
  • 1-[5-tert-Butyl-2-p-tolyl-2H-pyrazol-3-yl]-3-[4-(3-(tetrahydropyran-2-yl-oxy)butyn-1-yl)naphthalen-1-yl]-urea; [0109]
  • 1-[5-tert-Butyl-2-p-tolyl-2H-pyrazol-3-yl]-3-[4-(3-(furan-2-ylcarbonyloxy)propyn-1-yl)naphthalen-1-yl]-urea; [0110]
  • 1-[5-tert-Butyl-2-p-tolyl-2H-pyrazol-3-yl]-3-[4-(3-(piperdin-1-yl)propyn-1-yl)naphthalen-1-yl]-urea; [0111]
  • 1-[5-tert-Butyl-2-p-tolyl-2H-pyrazol-3-yl]-3-[4-(3-(2-methoxymethylmorpholin-4-yl)propyn-1-yl)naphthalen-1-yl]-urea; [0112]
  • 1-[5-tert-Butyl-2-p-tolyl-2H-pyrazol-3-yl]-3-[4-(pyridin-4-yl-methoxy)naphthalen-1-yl]-urea; [0113]
  • 1-[5-tert-Butyl-2-p-tolyl-2H-pyrazol-3-yl]-3-[4-(2-pyridin-4-yl-ethoxy)naphthalen-1-yl]-urea; [0114]
  • 1-[5-tert-Butyl-2-p-tolyl-2H-pyrazol-3-yl]-3-[4-(3-pyridin-4-yl-propoxy)naphthalen-1-yl]-urea; [0115]
  • 1-[5-tert-Butyl-2-p-tolyl-2H-pyrazol-3-yl]-3-[4-(2-imidazol-1-yl-ethoxy)naphthalen-1-yl]-urea; [0116]
  • 1-[5-tert-Butyl-2-p-tolyl-2H-pyrazol-3-yl]-3-[4-(2-benzimidazol-1-yl-ethoxy)naphthalen-1-yl]-urea; [0117]
  • 1-[5-tert-Butyl-2-p-tolyl-2H-pyrazol-3-yl]-3-[4-(2-(3,4-dimethoxyphenyl)-ethoxy)naphthalen-1-yl]-urea; [0118]
  • 1-[5-tert-Butyl-2-p-tolyl-2H-pyrazol-3-yl]-3-[4-(pyridin-4-yl-methylamino)naphthalen-1-yl]-urea; [0119]
  • 1-[5-tert-Butyl-2-p-tolyl-2H-pyrazol-3-yl]-3-[4-(pyridin-4-yl-carbonylamino)naphthalen-1-yl]-urea; [0120]
  • 1-[5-tert-Butyl-2-p-tolyl-2H-pyrazol-3-yl]-3-[4-(morpholin-4-yl-acetamido)naphthalen-1-yl]-urea; [0121]
  • 1-[5-tert-Butyl-2-p-tolyl-2H-pyrazol-3-yl]-3-[4-(pyridin-3-yl-methylamino)naphthalen-1-yl]-urea; [0122]
  • 1-[5-tert-Butyl-2-p-tolyl-2H-pyrazol-3-yl]-3-[4-(pyridin-3-yl-carbonylamino)naphthalen-1-yl]-urea; [0123]
  • 1-[5-iso-Propyl-2-phenyl-2H-pyrazol-3-yl]-3-[4-(2-morpholin-4-yl-ethoxy)naphthalen-1-yl]-urea; [0124]
  • 1-[5-(Tetrahydropyran-3-yl)-2-phenyl-2H-pyrazol-3-yl]-3-[4-(2-morpholin-4-yl-ethoxy)naphthalen-1-yl]-urea; [0125]
  • 1-[5-cyclohexyl-2-phenyl-2H-pyrazol-3-yl]-3-[4-(2-morpholin-4-yl-ethoxy)naphthalen-1-yl]-urea; [0126]
  • 1-[5-(2,2,2-trifluoroethyl)-2-phenyl-2H-pyrazol-3-yl]-3-[4-(2-morpholin-4-yl-ethoxy)naphthalen-1-yl]-urea; [0127]
  • 1-[5-(1-methylcycloprop-1-yl)-2-phenyl-2H-pyrazol-3-yl]-3-[4-(2-morpholin-4-yl-ethoxy)naphthalen-1-yl]-urea; [0128]
  • 1-[5-ethoxycarbonyl-2-phenyl-2H-pyrazol-3-yl]-3-[4-(2-morpholin-4-yl-ethoxy)naphthalen-1-yl]-urea; [0129]
  • 1-[5-(1-methylcyclohex-1-yl)-2-phenyl-2H-pyrazol-3-yl]-3-[4-(2-morpholin-4-yl-ethoxy)naphthalen-1-yl]-urea; [0130]
  • 1-[5-tert-butyl-2-methyl-2H-pyrazol-3-yl]-3-[4-(2-morpholin-4-yl-ethoxy)naphthalen-1-yl]-urea; [0131]
  • 1-[5-tert-butyl-2-benzyl-2H-pyrazol-3-yl]-3-[4-(2-morpholin-4-yl-ethoxy)naphthalen-1-yl]-urea; [0132]
  • 1-[5-tert-butyl-2-(4-chlorophenyl)-2H-pyrazol-3-yl]-3-[4-(2-morpholin-4-yl-ethoxy)naphthalen-1-yl]-urea; [0133]
  • 1-[5-tert-butyl-2-butyl-2H-pyrazol-3-yl]-3-[4-(2-morpholin-4-yl-ethoxy)naphthalen-1-yl]-urea; [0134]
  • 1-[5-tert-butyl-2-(ethoxycarbonylmethyl)-2H-pyrazol-3-yl]-3-[4-(2-morpholin-4-yl-ethoxy)naphthalen-1-yl]-urea; [0135]
  • 1-[5-tert-butyl-2-(4-methyl-3-carbamylphenyl)-2H-pyrazol-3-yl]-3-[4-(2-morpholin-4-yl-ethoxy)naphthalen-1-yl]-urea; [0136]
  • 1-[5-tert-butyl-2-(4-methyl-3-(2-ethoxyarbonylvinyl)phenyl)-2H-pyrazol-3-yl]-3-[4-(2-morpholin-4-yl-ethoxy)naphthalen-1-yl]-urea; [0137]
  • 1-[5-tert-butyl-2-(4-methyl-3-(morpholin-4-yl)methylphenyl)-2H-pyrazol-3-yl]-3-[4-(2-morpholin-4-yl-ethoxy)naphthalen-1-yl]-urea; [0138]
  • 1-[5-tert-butyl-2-(4-methyl-3-dimethylaminomethylphenyl)-2H-pyrazol-3-yl]-3-[4-(2-morpholin-4-yl-ethoxy)naphthalen-1-yl]-urea; [0139]
  • 1-[5-tert-butyl-2-(3-(2-morpholin-4-yl-ethyl)phenyl)-2H-pyrazol-3-yl]-3-[4-(2-morpholin-4-yl-ethoxy)naphthalen-1-yl]-urea; [0140]
  • 1-[5-tert-butyl-2-(3-(tetrahydropyran-4-ylamino)phenyl)-2H-pyrazol-3 -yl]-3-[4-(2-morpholin-4-yl-ethoxy)naphthalen-1-yl]-urea; [0141]
  • 1-[5-tert-butyl-2-(3-dimethylaminomethylphenyl)-2H-pyrazol-3-yl]-3-[4-(2-morpholin-4-yl-ethoxy)naphthalen-1-yl]-urea; [0142]
  • 1-[5-tert-butyl-2-(4-(tetrahydropyran-4-ylamino)phenyl)-2H-pyrazol-3-yl]-3-[4-(2-morpholin-4-yl-ethoxy)naphthalen- 1-yl]-urea; [0143]
  • 1-[5-tert-butyl-2-(4-(3-benzylureido)phenyl)-2H-pyrazol-3-yl]-3-[4-(2-morpholin-4-yl-ethoxy)naphthalen-1-yl]-urea; [0144]
  • 1-[5-tert-butyl-2-(2-chloropyridin-5-yl)-2H-pyrazol-3-yl]-3-[4-(2-morpholin-4-yl-ethoxy)naphthalen-1-yl]-urea; [0145]
  • 1-[5-tert-butyl-2-(2-methylpyridin-5-yl)-2H-pyrazol-3-yl]-3-[4-(2-morpholin-4-yl-ethoxy)naphthalen-1-yl]-urea; [0146]
  • 1-[5-tert-butyl-2-(2-methoxypyridin-5-yl)-2H-pyrazol-3-yl]-3-[4-(2-morpholin-4-yl-ethoxy)naphthalen-1-yl]-urea; [0147]
  • 1-[5-tert-butyl-2-(pyridin-3-yl)-2H-pyrazol-3-yl]-3-[4-(2-morpholin-4-yl-ethoxy)naphthalen-1-yl]-urea; [0148]
  • 1-[5-tert-butyl-2-(2-methylpyridin-5-yl)-2H-pyrazol-3-yl]-3-[4-(2-pyridin-4-yl-ethoxy)naphthalen-1-yl]-urea; [0149]
  • 1-[5-tert-butyl-2-(2-methylpyridin-5-yl)-2H-pyrazol-3-yl]-3-[4-(2-(trans-2,6-dimethylmorpholin-4-yl)ethoxy)naphthalen-1-yl]-urea; [0150]
  • 1-[5-tert-butyl-2-(2-methylpyridin-5-yl)-2H-pyrazol-3-yl]-3-[4-(3-morpholin-4-yl-propyn-1-yl)naphthalen-1-yl]-urea; [0151]
  • 1-[5-tert-Butyl-2-p-tolyl-2H-pyrazol-3 -yl]-3-[4-(2-(2-dimethylaminomethylmorpholin-4-yl)ethoxy)naphthalen-1-yl]-urea; [0152]
  • 1-[5-tert-butyl-2-iso-propyl-2H-pyrazol-3-yl]-3-[4-(2-morpholin-4-yl-ethoxy)naphthalen-1-yl]-urea; [0153]
  • 1-[5-tert-butyl-2-cyclopropyl-2H-pyrazol-3-yl]-3-[4-(2-morpholin-4-yl-ethoxy)naphthalen-1-yl]-urea; [0154]
  • 1-[5-tert-butyl-2-(thiophen-3-yl)-2H-pyrazol-3-yl]-3-[4-(2-morpholin-4-yl-ethoxy)naphthalen-1-yl]-urea; [0155]
  • 1-[5-tert-butyl-2-cyclopentyl-2H-pyrazol-3-yl]-3-[4-(2-morpholin-4-yl-ethoxy)naphthalen-1-yl]-urea; [0156]
  • 1-[5-tert-butyl-2-iso-propyl-2H-pyrazol-3-yl]-3-[4-(tetrahyropyran-4-yl-ethoxy)naphthalen-1-yl]-urea; [0157]
  • 1-[5-tert-butyl-2-cyclopropyl-2H-pyrazol-3-yl]-3-[4-(1-oxo-tetrahydrothiophen-3-yl-ethoxy)naphthalen-1-yl]-urea; [0158]
  • 1-[5-tert-butyl-2-(thiophen-3-yl)-2H-pyrazol-3-yl]-3-[4-(2-pyridinyl-4-yl-ethoxy)naphthalen-1-yl]-urea; [0159]
  • 1 -[5-tert-butyl-2-cyclopentyl-2H-pyrazol-3-yl]-3-[4-(pyridin-4-yl-methoxy)naphthalen-1-yl]-urea; [0160]
  • 1-[5-tert-Butyl-2-p-tolyl-2H-pyrazol-3-yl]-3-[4-(3-(pyridin-4-yl)propyn-1-yl)naphthalen-1-yl]-urea; [0161]
  • 1-[5-tert-Butyl-2-p-tolyl-2H-pyrazol-3-yl]-3-[4-(3-(2-methylaminopyridin-4-yl)propyn-1-yl)naphthalen-1-yl]-urea; [0162]
  • 1-[5-tert-Butyl-2-p-tolyl-2H-pyrazol-3-yl]-3-[4-(3-(1-oxo-tetrahydothiophen-3-yl)propyn-1-yl)naphthalen-1-yl]-urea; [0163]
  • 1-[5-tert-Butyl-2-p-tolyl-2H-pyrazol-3-yl]-3-[4-(3-(thiazolidin-3-yl)propyn- 1-yl)naphthalen-1-yl]-urea; [0164]
  • 1-[5-tert-Butyl-2-p-tolyl-2H-pyrazol-3-yl]-3-[4-(3-(tetrahydropyran-4-yl)propyn- 1-yl)naphthalen-1-yl]-urea; [0165]
  • 1-[5-tert-Butyl-2-p-tolyl-2H-pyrazol-3-yl]-3-[4-(2-methylaminopyrimidin-4-yl-methoxy)naphthalen-1-yl]-urea; [0166]
  • 1-[5-tert-Butyl-2-p-tolyl-2H-pyrazol-3-yl]-3-[4-(2-(2-methylaminopyrimidin-4-yl)ethoxy)naphthalen-1-yl]-urea; [0167]
  • 1-[5-tert-Butyl-2-p-tolyl-2H-pyrazol-3-yl]-3-[4-(2-(4-methoxybenzimidazol- 1-yl)ethoxy)naphthalen-1-yl]-urea; [0168]
  • 1-[5-tert-Butyl-2-p-tolyl-2H-pyrazol-3-yl]-3-[4-(2-(4-methylaminobenzimidazol-1-yl)ethoxy)naphthalen-1-yl]-urea; [0169]
  • 1-[5-tert-Butyl-2-p-tolyl-2H-pyrazol-3-yl]-3-[4-(2-(2-imidazo[4,5-b]pyridin-1-yl)ethoxy)naphthalen-1-yl]-urea; [0170]
  • 1-[5-tert-Butyl-2-p-tolyl-2H-pyrazol-3-yl]-3-[4-(2-[1,8]naphthyridin-4-yl)ethoxy)naphthalen-1-yl]-urea; [0171]
  • 1-[5-tert-Butyl-2-p-tolyl-2H-pyrazol-3-yl]-3-[4-(2-(3,4-dihydro-2H-pyrano[2,3-b]pyridin-5-yl)ethoxy)naphthalen-1-yl]-urea; [0172]
  • 1-[5-tert-Butyl-2-pyridin-3-yl-2H-pyrazol-3-yl]-3-[4-(2-methylaminopyrimidin-4-yl-methoxy)naphthalen-1-yl]-urea; [0173]
  • 1-[5-tert-Butyl-2-(2-methylpyridin-5-yl) -2H-pyrazol-3-yl]-3-[4-(2-(2-methylaminopyrimidin-4-yl)ethoxy)naphthalen-1-yl]-urea; [0174]
  • 1-[5-tert-Butyl-2-(2-methylpyridin-5-yl)-2H-pyrazol-3-yl]-3-[4-(2-(4-methoxybenzimidazol-1-yl)ethoxy)naphthalen-1-yl]-urea; [0175]
  • 1-[5-tert-Butyl-2-(2-methylpyridin-5-yl)-2H-pyrazol-3-yl]-3-[4-(2-(4-methylaminobenzimidazol-1-yl)ethoxy)naphthalen-1-yl]-urea; [0176]
  • 1-[5-tert-Butyl-2-(2-methylpyridin-5-yl)-2H-pyrazol-3-yl]-3-[4-(2-(2-imidazo[4,5-b]pyridin-1-yl)ethoxy)naphthalen-1-yl]-urea; [0177]
  • 1-[5-tert-Butyl-2-(2-methylpyridin-5-yl)-2H-pyrazol-3-yl]-3-[4-(2-[1,8]naphthyridin-4-yl)ethoxy)naphthalen-1-yl]-urea; [0178]
  • 1-[5-tert-Butyl-2-(2-methylpyridin-5-yl)-2H-pyrazol-3-yl]-3-[4-(2-(3,4-dihydro-2H-pyrano[2,3-b]pyridin-5-yl)ethoxy)naphthalen-1-yl]-urea; [0179]
  • 1-[5-tert-Butyl-2-cyclopropyl-2H-pyrazol-3-yl]-3-[4-(2-methylaminopyrimidin-4-yl-methoxy)naphthalen-1-yl]-urea; [0180]
  • 1-[5-tert-Butyl-2-cyclopropyl-2H-pyrazol-3-yl]-3-[4-(2-(2-methylaminopyrimidin-4-yl)ethoxy)naphthalen-1-yl]-urea; [0181]
  • 1-[5-tert-Butyl-2-cyclopropyl-2H-pyrazol-3-yl]-3-[4-(2-(4-methoxybenzinidazol-1-yl)ethoxy)naphthalen-1-yl]-urea; [0182]
  • 1-[5-tert-Butyl-2-cyclopropyl-2H-pyrazol-3-yl]-3-[4-(2-(4-methylaminobenzimidazol-1-yl)ethoxy)naphthalen-1-yl]-urea; [0183]
  • 1-[5-tert-Butyl-2-methyl-2H-pyrazol-3-yl]-3-[4-(2-(2-imidazo[4,5-b]pyridin-1-yl)ethoxy)naphthalen-1-yl]-urea; [0184]
  • 1-[5-tert-Butyl-2-methyl-2H-pyrazol-3-yl]-3-[4-(2-[1,8]naphthyridin-4-yl)ethoxy)naphthalen-1-yl]-urea; [0185]
  • 1-[5-tert-Butyl-2-methyl-2H-pyrazol-3-yl]-3-[4-(2-(3,4-dihydro-2H-pyrano[2,3-b]pyridin-5-yl)ethoxy)naphthalen-1-yl]-urea [0186]
  • and their physiologically acceptable acids or salts thereof. [0187]
  • Preferred compounds of the formula(I) are: [0188]
  • 1-[5-tert-Butyl-2-p-tolyl-2H-pyrazol-3-yl]-3-[4-(2-morpholin-4-yl-ethoxy)naphthalen-1-yl]-urea; [0189]
  • 1-[5-tert-Butyl-2-p-tolyl-2H-pyrazol-3-yl]-3-[4-(2-(cis-2,6-dimethylmorpholin-4-yl)ethoxy)naphthalen-1-yl]-urea; [0190]
  • 1-[5-tert-Butyl-2-p-tolyl-2H-pyrazol-3-yl]-3-[4-(2-(trans-2, 6-dimethylmorpholin-4-yl)ethoxy)naphthalen-1-yl]-urea; [0191]
  • 1-[5-tert-Butyl-2-p-tolyl-2H-pyrazol-3-yl]-3-[4-(2-(2-(methoxymethyl)morpholin-4-yl)ethoxy)naphthalen-1-yl]-urea; [0192]
  • 1-[5-tert-Butyl-2-p-tolyl-2H-pyrazol-3-yl]-3-[4-(2-(morpholin-4-yl)-2-oxoethoxy)naphthalen-1-yl]-urea; [0193]
  • 1-[5-tert-Butyl-2-p-tolyl-2H-pyrazol-3-yl]-3-[4-(2-(morpholin-4-yl)-2-methylethoxy)naphthalen-1-yl]-urea; [0194]
  • 1-[5-tert-Butyl-2-p-tolyl-2H-pyrazol-3-yl]-3-[4-(2-(morpholin-4-yl)-1-methylethoxy)naphthalen-1-yl]-urea; [0195]
  • 1-[5-tert-Butyl-2-p-tolyl-2H-pyrazol-3-yl]-3-[4-(2-thiomorpholin-4-yl-ethoxy)naphthalen-1-yl]-urea; [0196]
  • 1-[5-tert-Butyl-2-p-tolyl-2H-pyrazol-3-yl]-3-[4-(2-(1-oxothiomorpholin-4-yl)ethoxy)naphthalen-1-yl]-urea; [0197]
  • 1-[5-tert-Butyl-2-p-tolyl-2H-pyrazol-3-yl]-3-[4-(2-morpholin-4-yl-ethoxy)-3-methylnaphthalen-1-yl]-urea; [0198]
  • 1-[5-tert-Butyl-2-p-tolyl-2H-pyrazol-3-yl]-3-[4-(2-(morpholin-4-yl-carbonyloxo)ethoxy)naphthalen-1-yl]-urea; [0199]
  • 1-[5-tert-Butyl-2-p-tolyl-2H-pyrazol-3-yl]-3-[4-(2-(tetrahydropyran-4-yl)ethoxy)naphthalen-1-yl]-urea; [0200]
  • 1-[5-tert-Butyl-2-p-tolyl-2H-pyrazol-3-yl]-3-[4-(2-(1-oxo-tetrahydrothiophen-3-yl)ethoxy)naphthalen-1-yl]-urea; [0201]
  • 1-[5-tert-Butyl-2-p-tolyl-2H-pyrazol-3-yl]-3-[4-(3-morpholin-4-yl-propyl)naphthalen-1-yl]-urea; [0202]
  • 1-[5-tert-Butyl-2-p-tolyl-2H-pyrazol-3-yl]-3-[4-(morpholin-4-yl-methyl)naphthalen-1-yl]-urea; [0203]
  • 1-[5-tert-Butyl-2-p-tolyl-2H-pyrazol-3-yl]-3-[4-(2-pyridin-4-yl-ethyl)naphthalen-1-yl]-urea; [0204]
  • 1-[5-tert-Butyl-2-p-tolyl-2H-pyrazol-3-yl]-3-[4-(3-(morpholin-4-yl)propyn- 1-yl)naphthalen-1-yl]-urea; [0205]
  • 1-[5-tert-Butyl-2-p-tolyl-2H-pyrazol-3-yl]-3-[4-(3-(tetrahydropyran-2-yl-oxy)propyn-1-yl)naphthalen-1-yl]-urea; [0206]
  • 1-[5-tert-Butyl-2-p-tolyl-2H-pyrazol-3-yl]-3-[4-(3-(tetrahydropyran-2-yl-oxy)butyn-1-yl)naphthalen-1-yl]-urea; [0207]
  • 1-[5-tert-Butyl-2-p-tolyl-2H-pyrazol-3-yl]-3-[4-(3-(piperdin-1-yl)propyn-1-yl)naphthalen-1-yl]-urea; [0208]
  • 1-[5-tert-Butyl-2-p-tolyl-2H-pyrazol-3-yl]-3-[4-(3-(2-methoxymethylmorpholin-4-yl)propyn-1-yl)naphthalen-1-yl]-urea; [0209]
  • 1-[5-tert-Butyl-2-p-tolyl-2H-pyrazol-3-yl]-3-[4-(pyridin-4-yl-methoxy)naphthalen-1-yl]-urea; [0210]
  • 1-[5-tert-Butyl-2-p-tolyl-2H-pyrazol-3-yl]-3-[4-(2-pyridin-4-yl-ethoxy)naphthalen-1-yl]-urea; [0211]
  • 1-[5-tert-Butyl-2-p-tolyl-2H-pyrazol-3-yl]-3-[4-(3-pyridin-4-yl-propoxy)naphthalen-1-yl]-urea; [0212]
  • 1-[5-tert-Butyl-2-p-tolyl-2H-pyrazol-3-yl]-3-[4-(2-imidazol-1-yl-ethoxy)naphthalen-1-yl]-urea; [0213]
  • 1-[5-tert-Butyl-2-p-tolyl-2H-pyrazol-3-yl]-3-[4-(2-(3,4-dimethoxyphenyl)-ethoxy)naphthalen-1-yl]-urea; [0214]
  • 1-[5-tert-Butyl-2-p-tolyl-2H-pyrazol-3-yl]-3-[4-(pyridin-4-yl-methylamino)naphthalen-1-yl]-urea; [0215]
  • 1-[5-iso-Propyl-2-phenyl-2H-pyrazol-3-yl]-3-[4-(2-morpholin-4-yl-ethoxy)naphthalen-1-yl]-urea; [0216]
  • 1-[5-cyclohexyl-2-phenyl-2H-pyrazol-3-yl]-3-[4-(2-morpholin-4-yl-ethoxy)naphthalen-1-yl]-urea; [0217]
  • 1-[5-(2,2,2-trifluoroethyl)-2-phenyl-2H-pyrazol-3-yl]-3-[4-(2-morpholin-4-yl-ethoxy)naphthalen-1-yl]-urea; [0218]
  • 1-[5-(1-methylcycloprop-1-yl)-2-phenyl-2H-pyrazol-3-yl]-3-[4-(2-morpholin-4-yl-ethoxy)naphthalen-1-yl]-urea; [0219]
  • 1-[5-(1-methylcyclohex-1-yl)-2-phenyl-2H-pyrazol-3-yl]-3-[4-(2-morpholin-4-yl-ethoxy)naphthalen-1-yl]-urea; [0220]
  • 1-[5-tert-butyl-2-methyl-2H-pyrazol-3-yl] -3-[4-(2-morpholin-4-yl-ethoxy)naphthalen-1-yl]-urea; [0221]
  • 1-[5-tert-butyl-2-(4-chlorophenyl)-2H-pyrazol-3-yl]-3-[4-(2-morpholin-4-yl-ethoxy)naphthalen-1-yl]-urea; [0222]
  • 1-[5-tert-butyl-2-butyl-2H-pyrazol-3-yl]-3-[4-(2-morpholin-4-yl-ethoxy)naphthalen-1-yl]-urea; [0223]
  • 1-[5-tert-butyl-2-(4-methyl-3-carbamylphenyl)-2H-pyrazol-3-yl]-3-[4-(2-morpholin-4-yl-ethoxy)naphthalen-1-yl]-urea; [0224]
  • 1-[5-tert-butyl-2-(4-methyl-3-(morpholin-4-yl)methylphenyl)-2H-pyrazol-3-yl]-3-[4-(2-morpholin-4-yl-ethoxy)naphthalen-1-yl]-urea; [0225]
  • 1-[5-tert-butyl-2-(4-methyl-3-dimethylaminomethylphenyl)-2H-pyrazol-3-yl]-3-[4-(2-morpholin-4-yl-ethoxy)naphthalen-1-yl]-urea; [0226]
  • 1-[5-tert-butyl-2-(3-dimethylaminomethylphenyl)-2H-pyrazol-3-yl]-3-[4-(2-morpholin-4-yl-ethoxy)naphthalen-1-yl]-urea; [0227]
  • 1-[5-tert-butyl-2-(2-chloropyridin-5-yl)-2H-pyrazol-3-yl]-3-[4-(2-morpholin-4-yl-ethoxy)naphthalen-1-yl]-urea; [0228]
  • 1-[5-tert-butyl-2-(2-methylpyridin-5-yl)-2H-pyrazol-3-yl]-3-[4-(2-morpholin-4-yl-ethoxy)naphthalen-1-yl]-urea; [0229]
  • 1-[5-tert-butyl-2-(2-methoxypyridin-5-yl)-2H-pyrazol-3-yl]-3-[4-(2-morpholin-4-yl-ethoxy)naphthalen-1-yl]-urea; [0230]
  • 1-[5-tert-butyl-2-(pyridin-3-yl)-2H-pyrazol-3-yl]-3-[4-(2-morpholin-4-yl-ethoxy)naphthalen-1-yl]-urea; [0231]
  • 1- [5-tert-butyl-2-(2-methylpyridin-5-yl)-2H-pyrazol-3-yl]-3- [4-(2-pyridin-4-yl-ethoxy)naphthalen-1-yl]-urea; [0232]
  • 1-[5-tert-butyl-2-(2-methylpyridin-5-yl)-2H-pyrazol-3-yl]-3-[4-(2-(trans-2,6-dimethylmorpholin-4-yl)ethoxy)naphthalen-1-yl]-urea; [0233]
  • 1-[5-tert-butyl-2-(2-methylpyridin-5-yl)-2H-pyrazol-3-yl-3-[4-(3-morpholin-4-yl-propyn-1-yl)naphthalen-1-yl]-urea. [0234]
  • Particularly preferred compounds of the formula(I) are: [0235]
  • 1-[5-tert-Butyl-2-p-tolyl-2H-pyrazol-3-yl]-3-[4-(2-morpholin-4-yl-ethoxy)naphthalen-1-yl]-urea; [0236]
  • 1-[5-tert-Butyl-2-p-tolyl-2H-pyrazol-3-yl]-3-[4-(2-(1 -oxothiomorpholin-4-yl)ethoxy)naphthalen-1-yl]-urea; [0237]
  • 1-[5-tert-butyl-2-(2-methylpyridin-5-yl)-2H-pyrazol-3-yl]-3-[4-(2-pyridin-4-yl-ethoxy)naphthalen-1-yl]-urea; [0238]
  • 1-[5-tert-butyl-2-(2-methoxypyridin-5-yl)-2H-pyrazol-3-yl]-3-[4-(2-morpholin-4-yl-ethoxy)naphthalen-1-yl]-urea or [0239]
  • 1-[5-tert-butyl-2-methyl-2H-pyrazol-3-yl]-3-[4-(2-morpholin-4-yl-ethoxy)naphthalen-1-yl]-urea. [0240]
  • Any compounds of this invention containing one or more asymmetric carbon atoms may occur as racemates and racemic mixtures, single enantiomers, diastereomeric mixtures and individual diastereomers. All such isomeric forms of these compounds are expressly included in the present invention. Each stereogenic carbon may be in the R or S configuration, or a combination of configurations. [0241]
  • Some of the compounds of formula (I) can exist in more than one tautomeric form. The invention includes all such tautomers. [0242]
  • The term “aroyl” as used in the present specification shall be understood to mean “benzoyl” or “naphthoyl”. [0243]
  • The invention includes pharmaceutically acceptable derivatives of compounds of formula (I). A “pharmaceutically acceptable derivative” refers to any pharmaceutically acceptable salt or ester of a compound of this invention, or any other compound which, upon administration to a patient, is capable of providing (directly or indirectly) a compound of this invention, a pharmacologically active metabolite or pharmacologically active residue thereof. [0244]
  • The term “metabolite” shall be understood to mean any of the compounds of the formula (I) which are capable of being hydroxylated or oxidized, enzymatically or chemically, as will be appreciated by those skilled in the art. Nonlimiting examples of metabolites of the formula (I) are shown in the table below: [0245]
    Structure Name
    Figure US20020058678A1-20020516-C00004
    1-[5-(2-hydroxy-1,1-dimethyl-ethyl)-2-p- tolyl-2H-pyrazol-3-yl]-3-[4-(2-morpholin- 4-yl-ethoxy)-naphthalen-1-yl]-urea
    Figure US20020058678A1-20020516-C00005
    1-[5-tert-butyl-2-(3-hydroxy-4-methyl- phenyl)-2H-pyrazol-3-yl]-3-[4-(2- morpholin-4-yl-ethoxy)-naphthalen-1-yl]- urea
    Figure US20020058678A1-20020516-C00006
    1-[5-tert-butyl-2-(4-hydroxymethyl- phenyl)-2H-pyrazol-3-yl]-3-[4-(2- morpholin-4-yl-ethoxy)-naphthalen-1-yl]- urea
    Figure US20020058678A1-20020516-C00007
    1-[5-tert-butyl-2-p-tolyl-2H-pyrazol-3-yl]- 3-{4-[2-(3-oxo-morpholin-4-yl)-ethoxy]- naphthalen-1-yl}-urea
    Figure US20020058678A1-20020516-C00008
    1-[5-tert-butyl-2-p-tolyl-2H-pyrazol-3-yl]- 3-{4-[2-(4-hydroxy-morpholin-4-yl)- ethoxy]-naphthalen-1-yl}-urea
    Figure US20020058678A1-20020516-C00009
    1-[5-(2-hydroxy-1,1-dimethyl-ethyl)-2-(6- methyl-pyridin-3-yl)-2H-pyrazol-3-yl]-3- [4-(2-morpholin-4-yl-ethoxy)-naphthalen- 1-yl]-urea
    Figure US20020058678A1-20020516-C00010
    1-[5-tert-butyl)-2-(1-hydroxy-6-methyl- pyridin-3-yl)-2H-pyrazol-3-yl]-3-[4-(2- morpholin-4-yl-ethoxy)-naphthalen-1-yl]- urea
    Figure US20020058678A1-20020516-C00011
    1-[5-tert-butyl)-2-(6-methyl-pyridin-3-yl)- 2H-pyrazol-3-yl]-3-{4-[2-(4-hydroxy- morpholin-4-yl)-ethoxy]-naphthalen-1-yl}- urea
    Figure US20020058678A1-20020516-C00012
    1-[5-(2-hydroxy-1,1-dimethyl-ethyl)-2-(6- methyl-pyridin-3-yl)-2H-pyrazol-3-yl]-3- [4-(2-pyridin-4-yl-ethoxy)-naphthalen-1- yl]-urea
    Figure US20020058678A1-20020516-C00013
    1-[5-tert-butyl)-2-(6-methyl-pyridin-3-yl)- 2H-pyrazol-3-yl]-3-[4-(2-hydroxy-2- pyridin-4-yl-ethoxy)-naphthalen-1-yl]-urea
    Figure US20020058678A1-20020516-C00014
    1-[5-tert-butyl)-2-(6-methyl-pyridin-3-yl)- 2H-pyrazol-3-yl]-3-{4-[2-(1-hydroxy- pyridin-4-yl)-ethoxy]-naphthalen-1-yl}- urea
    Figure US20020058678A1-20020516-C00015
    1-[5-(2-hydroxy-1,1-dimethyl-ethyl)-2-p- tolyl-2H-pyrazol-3-yl]-3-{4-[2-(1-oxo- thiomorpholin-4-yl)-ethoxy]-naphthalen-1- yl}-urea
    Figure US20020058678A1-20020516-C00016
    1-[5-tert-butyl-2-(4-hydroxymethyl- phenyl)-2H-pyrazol-3-yl]-3-{4-[2-(1-oxo- thiomorpholin-4-yl)-ethoxy]-naphthalen-1- yl}-urea
    Figure US20020058678A1-20020516-C00017
    1-[5-tert-butyl-2-p-tolyl-2H-pyrazol-3-yl]- 3-{4-[2-(1,3 dioxo-thiomorpholin-4-yl)- ethoxy]-naphthalen-1-yl}-urea
    Figure US20020058678A1-20020516-C00018
    1-[5-(2-hydroxy-1,1-dimethyl-ethyl)-2- methyl-2H-pyrazol-3-yl]-3-[4-(2- morpholin-4-yl-ethoxy)-naphthalen-1-yl]urea
    Figure US20020058678A1-20020516-C00019
    1-[5-tert-butyl-2-methyl-2H-pyrazol-3-yl]- 3- {4-[2-(4-hydroxy-morpholin-4-yl)- ethoxy]-naphthalen-1-yl}-urea
  • Pharmaceutically acceptable salts of the compounds of this invention include those derived from pharmaceutically acceptable inorganic and organic acids and bases. Examples of suitable acids include hydrochloric, hydrobromic, sulfuric, nitric, perchloric, fumaric, maleic, phosphoric, glycolic, lactic, salicylic, succinic, toluene-p-sulfuric, tartaric, acetic, citric, methanesulfonic, formic, benzoic, malonic, naphthalene-2-sulfuric and benzenesulfonic acids. Other acids, such as oxalic acid, while not themselves pharmaceutically acceptable, may be employed in the preparation of salts useful as intermediates in obtaining the compounds of this invention and their pharmaceutically acceptable acid addition salts. Salts derived from appropriate bases include alkali metal (e.g., sodium), alkaline earth metal (e.g., magnesium), ammonium and N-(C[0246] 1-C4 alkyl)4 + salts.
  • In addition, the compounds of this invention include prodrugs of compounds of the formula (I). Prodrugs include those compounds that, upon simple chemical transformation, are modified to produce a compound of formula (I). Simple chemical transformations include hydrolysis, oxidation and reduction. Specifically, when a prodrug of this invention is administered to a patient, the prodrug may be transformed into a compound of formula (I), thereby imparting the desired pharmacological effect. [0247]
  • General Synthetic Methods
  • The compounds of the invention may be prepared by Method A, B, or C as illustrated in Scheme I, preferably method C. [0248]
    Figure US20020058678A1-20020516-C00020
  • In Method A, a mixture of an aminoheterocycle of formula II and an arylisocyanate of formula III is dissolved in a non-protic, anhydrous solvent such as THF, ether, toluene, dioxane or ethyl acetate. The preferred solvent is THF. The mixture is stirred at between 0-45° C., preferably at 25° C., for 2-24 hr, and the volatiles are removed. Purification of the residue by recrystallization from an appropriate solvent such as ethyl acetate/hexanes, ethyl acetate/methanol, THF/petroleum ether, ethanol/water, or by silica gel chromatography, using for example, hexanes and ethyl acetate as eluents, provides the product of formula I. [0249]
  • In Method B, an aminoheterocycle of formula II is dissolved in a halogenated solvent, such as methylene chloride, chloroform or dichloroethane. The preferred solvent is methylene chloride. The mixture is diluted with aqueous alkali, such as sodium bicarbonate or potassium carbonate, cooled in an ice bath and phosgene is added. [0250]
  • The mixture is vigorously stirred for 5-30 min, with 10 min being preferable. The organic layer is dried, with agents such as MgSO[0251] 4 or Na2SO4, and the volatiles removed to provide the corresponding isocyanate of formula II. The isocyanate and arylamine IV are mixed in a non-protic, anhydrous solvent such as THF, ether, toluene, dioxane, methylene chloride or ethyl acetate. The preferred solvent is THF. The mixture is stirred at between 0-45° C., preferably at 25° C., for 2-24 hr, and the volatiles are removed. Purification of the residue by recrystallization or by silica gel chromatography, as above, provides the product of formula I.
  • In Method C, an aminoheterocycle of formula II is dissolved in a halogenated solvent, such as methylene chloride, chloroform or dichloroethane. The preferred solvent is methylene chloride. A suitable base such as triethylamine may be added, followed by phenyl chloroformate. The mixture is stirred at between 0-85° C., preferably at reflux temperature, for 2-24 hr, and the volatiles are removed providing carbamate V. The carbamate and arylamine IV are mixed in a non-protic, anhydrous solvent such as THF, ether, toluene, dioxane, methylene chloride or ethyl acetate. The preferred solvent is THF. The mixture is stirred at between 0-110 C., preferably at reflux temperature, for 2-24 hr, and the volatiles are removed. Purification of the residue as above provides the product of formula I. [0252]
  • The method used to produce an aminoheterocycle of formula II will depend on the nature of the desired heterocycle. In general, intermediates of formula II can be made by methods known to those skilled in the art. Some general methods are illustrated in the schemes below. Compounds G—NCO or G—NH[0253] 2 in Scheme I may be commercially available, or may be prepared by methods known to those skilled in the art. If G is a precursor of Ar2—L—Q ,the desired final product of formula (I) may be constructed by methods known to those skilled in the art. Illustrative examples are contained in the Synthetic Examples section below.
  • Desired aminopyrazoles of formula XIII can be prepared as described in Scheme II. A hydrazine of formula VIII, bearing substituent R[0254] 3, may be prepared by Method D or E. In Method D, an aryl bromide of formula VI is dissolved in a non-protic, inert solvent, such as THF, 1,4-dioxane or diethyl ether, and cooled to low temperature under an inert atmosphere. The preferred temperature for the solution is −77° C. A strong base dissolved in a non-protic, inert solvent, such as hexanes, THF or ether, is added dropwise while maintaing a reaction temperature below 0° C. and preferrably below −60° C. The preferred bases are alkyl lithium reagents and the most preferred is sec-butyl lithium. After the addition of the base, the reaction mixture is stirred for a period of time between thirty and ninety minutes or until all the starting aryl bromide has been consumed. An excess of dialkyl azodicarboxylate is added while maintaining a reaction temperature below 0° C. and preferrably below −60° C. The preferred dialkyl azodicarboxylate is di-tert-butyl azodicarboxylate. The reaction is stirred at cold temperatures and warmed to room temperature after 0.5 hr to 2 hr. The reaction is quenched with the addition of water and the product extracted into a non-protic solvent, such as ethyl acetate, diethyl ether or chloroform. The organic layers are dried with agents such as MgSO4 or Na2SO4 and the volatiles removed. The residue is dissolved in protic solvents, such as methanol or iso-propanol, cooled, preferably to 0-5° C. and treated with acid. Preferred acids are hydrochloric, hydrobromic, sulfuric and trifluoroacetic. The most preferred is hydrochloric in gaseous form. After the addition of excess acid the mixture is heated at the reflux temperature of the solvent until all starting material has been consumed. After cooling the product aryl-hydrazine of formula VIII salt is filtered and dried.
    Figure US20020058678A1-20020516-C00021
  • In Method E, an aryl amine bearing R[0255] 3 of formula VII is dissolved in a concentrated aqueous acid such as hydrochloric, hydrobromic or sulfuric and cooled to ice bath temperatures. The most preferred acid is hydrochloric with concentrations between 3-8N with the most preferred concentration of 6N. A nitrosating reagent in water is added dropwise while maintaining a cold temperature. The preferred temperature is 0-5° C. The preferred reagent is sodium nitrite. The reaction is stirred between 10-90 min and a reducing agent is added while maintaing cold temperatures. The preferred temperature is 0-5° C. Reducing agents include zinc, iron, samarium iodide and tin(II) chloride. The most preferred agent is tin(II) chlroride dissolved in aqueous hydrochloride with a concentration of 3-8 N with a most preferred concentration of 6N. The reaction is stirred between 0.5-3 hr and quenched with alkali to a pH between 12-14. Alkali reagents include sodium hydroxide, potassium hydroxide, lithium hydroxide and calcium hydroxide. The most preferred alkali reagent is potassium hydroxide. The aqueous solution is extracted with a non-protic organic solvent, such as diethyl ether, chloroform, ethyl acetate and methylene chloride. The organic layers are dried with agents such as MgSO4 and Na2SO4 and the volatiles removed to provide the aryl-hydrazine (VIII) which can be carried forward without further purification.
  • A β-ketonitrile bearing R[0256] 1 (XII) may be prepared by Method F or G. In Method F, a metal hydride, such as sodium hydride, potassium hydride or lithium hydride, is suspended in an anhydrous, inert, non-protic solvent, such as diethyl ether, THF and dioxane, at temperatures between 35-85° C. The most preferred metal hydride is sodium hydride and the most preferred solvent is THF at a temperature of 75° C. An alkyl ester, preferably a methyl ester (IX), and acetonitrile is dissolved in an anhydrous, inert, non-protic solvent, such as diethyl ether, THF or dioxane and added dropwise to the metal hydride suspension. The preferred solvent is THF. The mixture is kept at elevated temperatures between 3-24 hours, cooled to room temperature and diluted with a non-protic solvent and aqueous acid. The organic layer is washed with water and brine, dried, with agents such as MgSO4 and Na2SO4, and the volatiles removed to provide the β-ketonitrile (XII) which could be used without further purification.
  • Alternatively, following Method G, a solution of a strong base, such as alkyl lithium reagents and metal amide reagents, such as n-butyl lithium, sec-butyl lithium, methyl lithium and lithium diisopropylamide, in an anhydrous, inert, non-protic solvent, such as diethyl ether, THF and dioxane, is cooled below 0° C. The preferred base is n-butyl lithium, the preferred solvent is THF and the preferred temperature is −77° C. A solution of cyanoacetic acid (X) in an anhydrous, inert, non-protic solvent, such as diethyl ether, THF and dioxane, and most preferrably THF, is added dropwise while maintaining a reaction temperature below 0° C. and preferrably at −77° C. The reaction is stirred between 10-45 min while warming to 0° C. The solution of the dianion of cyanoacetic is cooled to temperatures below −25° C. and preferrably at −77° C. An alkyl acid chloride (XI) dissolved in an anhydrous, inert, non-protic solvent, such as diethyl ether, THF and dioxane, and most preferrably THF, is added. The reaction mixture is warmed to 0° C. betweeen 10-30 min. and quenched with aqueous acid. The product is extracted with an organic solvent, such as chloroform, ethyl acetate, ether and methylene chloride. The combined organic extracts are dried, with agents such as MgSO[0257] 4 and Na2SO4, and the volatiles removed to provide the β-ketonitrile (XII) which could be used without further purification.
  • The desired aminopyrazole (XIII) may then be prepared by Method H or I. In Method H, aryl hydrazine VIII and β-ketonitrile XII are mixed in an organic solvent, such as toluene, ethanol, iso-propanol or t-butanol. The preferred solvent is ethanol. An acid, such as hydrochloric acid, p-toluene sulfonic acid or sulfuric acid, is added, The preferred acid is concentrated hydrochloric acid. The mixture is heated to temperatures between 50-100° C., preferrably at 80° C., for 10-24 hr and cooled to room temperature. The mixture is diluted with non-protic organic solvent, such as ethyl acetate, ether, chloroform and methylene chloride, and washed with aqueous alkali, such as sodium bicarbonate and potassium carbonate. The organic layer is dried, with agents such as MgSO[0258] 4 and Na2SO4, and the volatiles removed to provide a residue which is purified by recrystallization or silica gel chromatography using hexanes and ethyl acetate as eluents. The product-rich fractions are collected and the volatiles removed to provide the desired amonopyrazole (XIII).
  • Alternatively, using Method I, aryl hydrazine VIII and β-ketonitrile XII are mixed in an organic solvent, such as toluene, ethanol, iso-propanol or t-butanol. The preferred solvent is toluene. The mixture is heated at reflux temperatures for 3-24 hrs with azeotropic removal of water and worked up as described above providing the aminopyrazole XIII. [0259]
  • A general synthesis for desired aminothiophenes is illustrated in Scheme III, Method J. [0260]
    Figure US20020058678A1-20020516-C00022
  • A mixture of 1-aryl-5-alkyl-butane-1,4-dione (XIV) and a sulfating reagent, such as Lawesson's reagent or phosphorous (V) sulfide, and preferrably Lawesson's reagent, is dissolved in a non-protic, anhydrous solvent, such as toluene, THF and dioxane. The preferred solvent is toluene. The mixture is heated at elevated temperatures and preferably at a solvent-refluxing temperature for 1-10 hr. The volatiles are removed and the residue is purified by silica gel chromatography using hexanes and ethyl acetate as eluent. The product-rich fractions are collected and the volatiles removed to provide the substituted thiophene XV. [0261]
  • A mixture of substituted thiophene XV is dissolved in a solvent such as acetic anhydride or acetic acid. The preferred solvent is acetic anhydride. The mixture is cooled to 0-30° C. and preferrably to −10° C. A solution of concentrated nitric acid in a solvent such as acetic anhydride or acetic acid, with the preferred solvent being acetic anhydride is added while cooling 0-30° C. and preferrably to −10° C. The mixture is stirred between 10-120 min, poured onto ice and extracted with a non-protic solvent such as diethyl ether, chloroform, ethyl acetate or methylene chloride. The organic extracts are washed with aqueous alkali, dried with agents such as MgSO[0262] 4 and Na2SO4 and the volatiles removed. The residue is purified by silica gel chromatography using hexanes and ethyl acetate as eluents. The product-rich fractions are collected and the volatiles removed to provide the 2-aryl-5-alkyl-3-nitrothiophene. The 2-aryl-5-alkyl-3-nitrothiophene is reduced by metals, such as iron, tin and zinc or catalytic hydrogenation. The preferred reduction occurs with iron in acetic acid at temperatures between 50-110° C. and preferrably at 100° C. for 5-30 min. After cooling to room temperature the reaction is diluted with water, neutralized with alkali, such as sodium hydroxide, potassium hydroxide, potassium carbonate or sodium bicarbonate, and extracted with a non-protic solvent such as diethyl ether, ethyl acetate or methylene chloride. The organic extracts are dried with agents such as MgSO4 and NA2SO4 and the volatiles removed to provide the desired aminothiophene XVI.
  • Other desired aminoheterocycles can be prepared by methods known in the art and described in the literature. The examples that follow are illustrative and, as recognized by one skilled in the art, particular reagents or conditions could be modified as needed for individual compounds. Intermediates used in the schemes below are either commercially available or easily prepared from commercially available materials by those skilled in the art. [0263]
  • Scheme IV outlines a general scheme for desired aminofurans as described by Stevenson et al. (J. Am. Chem. Soc., 1937, 59, 2525). An ethyl aroylacetate (XVII) is dissolved in a non-protic solvent, such as ether or THF, and treated with a strong base, such as sodium, sodium ethoxide or sodium hydride, and the anion is reacted with a bromomethyl alkylketone (XVIII) at low temperatures, such as 0° C. After stirring the reaction until no starting material remains, it is poured onto cold water and extracted with a non-protic solvent. The combined extracts are dried with agents such as MgSO[0264] 4 or Na2SO4. The diketo-ester (XIX) may be carried forward without further purification or purified by distillation or silica gel chromatography. The diketo-ester in a protic solvent, such as ethanol, is heated in the presence of a mineral acid, such as sulfuric or hydrochloric, for 5-10 hr. and extracted with a non-protic solvent. The combined extracts are dried with agents such as MgSO4 or Na2SO4. The furan-ester (XX) may be carried forward without further purification or purified by distillation or silica gel chromatography. The furan-ester in a protic solvent, such as ethanol, is treated with hydrazine hydrate and the mixture heated for 2-5 days. The hydrazide is isloated as above and treated with hot formic acid and the resulting furan-amine (XXI) purified by distillation or silica gel chromatography.
    Figure US20020058678A1-20020516-C00023
  • The synthesis of substituted 4-aminooxazoles may be achieved analogous to a procedure described by Lakhan et al. ([0265] J. Het. Chem., 1988, 25, 1413) and illustrated in Scheme V. A mixture of aroyl cyanide (XXII), aldeyde (XXIII) and anhydrous ammonium acetate in acetic acid is heated at 100-110° C. for 3-6 hr, cooled to room temperature and quenched with water. Extraction by a non-protic solvent provides the product XXIV which can be carried forward without further purification or purified by recrystallization or silica gel chromatography.
    Figure US20020058678A1-20020516-C00024
  • The synthesis of substituted 3-aminopyrroles (XXVIII) may be achieved in a manner analogous to Aiello et al., [0266] J. Chem. Soc. Perkins Trans. I, 1981, 1. This is outlined in Scheme VI. A mixture of aryldioxoalkane (XXV) and amine (XXVI) in acetic acid is heated at 100-110° C. for 3-6 hr and worked up in the usual manner. The product (XXVII) in acetic acid is treated with a nitrating agent, such as nitric acid and potassium nitrate in concentrated sulfuric acid. The mixture is poured onto cold water and extracted with a non-protic solvent. The combined extracts are dried with agents such as MgSO4 and Na2SO4. Removal of the volatiles provides the nitro-pyrrole which which may be carried forward without further purification or purified by recrystallization or silica gel chromatography. The nitro-pyrrole is reduced to the amine with iron in acetic acid or by catalytic hydrogenation using palladium on activated carbon. The aminopyrrole (XXVIII) may be carried forward without further purification or purified by recrystallization or silica gel chromatography.
    Figure US20020058678A1-20020516-C00025
  • In an analogous fashion, a mixture of amine XXIX and 3-aryl-2,5-dioxoalkane (XXX) in acetic acid is heated between 80-110° C. for 2-24 hr. The reaction is diluted with water and extracted with an organic solvent. The combined extracts are dried with agents such as MgSO[0267] 4 or Na2SO4 and the volatiles removed. The resulting pyrrole is treated with a nitrating agent and subsequently reduced to XXXI as described above. The product may be carried forward without further purification or purified by recrystallization or silica gel chromatography. This process is illustrated in Scheme VII.
    Figure US20020058678A1-20020516-C00026
  • Substituted 5-aminothiazoles (XXXV) may be prepared in a manner analogous to Gerwald et al., [0268] J. Prakt. Chem. 1973, 315, 539. As illustrated in Scheme VIII, to a mixture of aminocyanide XXXII, aldehyde XXXIII and sulfur in an anhydrous solvent, such as ethanol and methanol, is added dropwise a base, such as triethylamine. The mixture is heated at 50° C. for 1-3 hr. The mixture is cooled and the excess sulfur removed. Acetic acid is added to neutralize the mixture and the solid collected. The imine XXXIV is treated with acid, such as hydrchloric and toluenesulfonic acid, in water and an organic solvent. After the starting material is consumed the reaction is worked up and the product XXXV may be carried forward without further purification or purified by recrystallization or silica gel chromatography.
    Figure US20020058678A1-20020516-C00027
  • A synthesis of substituted 2-aminothiophenes (XXXVII), analogous to a procedure described by Gewald et al. ([0269] J. Prakt. Chem., 1973, 315, 539) is illustrated in Scheme IX. A mixture of disubstituted thiophene-3-carboxylic acid (XXXVI) in a protic solvent, such as acetic acid, at a temperature of 0-50° C. is treated with a nitrating agent, such as nitric acid or potassium nitrate in concentrated sulfuric acid. After the starting material has been consumed the reaction is poured onto ice and the product extracted with a non-protic solvent. The combined extracts are dried with agents such as MgSO4 and Na2SO4 and the volatiles removed. The nitrothiophene is reduced to the amine with iron in acetic acid or by catalytic hydrogenation using palladium on activated carbon. The amino-thiophene may be carried forward without further purification or purified by recrystallization or silica gel chromatography.
    Figure US20020058678A1-20020516-C00028
  • 1,5-Disubstituted-3-aminopyrazoles (XL) may be prepared as shown in Scheme X, in a fashion analogous to the procedure described by Ege et al. (J. Het. Chem., 1982, 19, 1267). Potassium is added to anhydrous t-butanol and the mixture cooled to 5° C. Hydrazine XXXVIII is added, followed by cyanodibromoalkane XXXIX. The mixture is heated at refluxing temperatures for 3-10 hr. The mixture is cooled to room temperature and poured onto ice water. The product is extracted with an organic solvent. The combined extracts are dried with agents such as MgSO[0270] 4 or Na2SO4 and the volatiles removed. The product XL may be carried forward without further purification or purified by recrystallization or silica gel chromatography.
    Figure US20020058678A1-20020516-C00029
  • The synthesis of 2-amino-3,5-disubstituted thiophenes shown in Scheme XI, is done in a fashion analogous to Knoll et al., [0271] J. Prakt. Chem., 1985, 327, 463. A mixture of substituted N-(3-aminothioacryloyl)-formamidine (XLI) and substituted bromide (XLII) in a protic solvent, such as methanol or ethanol, is heated, preferably at a reflux temperature, for 5-30 min and cooled below room temperature. The product thiophene-imine is filtered and dried. The thiophene-imine XLIII is converted to the thiophene-amine (XLIV) by treatment with aqueous acid.
    Figure US20020058678A1-20020516-C00030
  • The synthesis of 1,4-disubstituted-2-aminopyrroles (XLVIII) may be accomplished in a manner analogous to Brodrick et al. ([0272] J. Chem. Soc. Perkin Trans. I, 1975, 1910), and as illustrated in Scheme XII. The potassium salt of formylnitrile XLV in water is treated with amine XLVI and acetic acid and the mixture heated at 50-90° C. for 5-30 min. The aminonitrile XLVII is collected by filtration upon cooling and then is stirred at room temperature with a base such as ethanolic potassium ethoxide for 2-5 hr and the volatiles removed. The residue is diluted with water and extracted with an organic solvent. The combined extracts are dried with agents such as MgSO4 and Na2SO4 and the volatiles removed. The product (XLVIII) may be carried forward without further purification or purified by recrystallization or silica gel chromatography.
    Figure US20020058678A1-20020516-C00031
  • The preparation of 1,2-disubstituted-4-aminoimidazoles (L) by reduction of the corresponding nitro compound (XLIX), for example with iron in acetic acid or catalytic hydrogenation may be accomplished as described by Al-Shaar et al. ([0273] J.
  • Chem. Soc. Perkin Trans. I, 1992, 2779) and illustrated in Scheme XIII. [0274]
    Figure US20020058678A1-20020516-C00032
  • 2,4-Disubstituted 5-aminooxazoles (LV) may be prepared in a manner analogous to the procedure described by Poupaert et al. (Synthesis, 1972, 622) and illustrated in Scheme XIV. Acid chloride LI is added to a cold mixture of 2-aminonitrile LII and a base such as triethylamine in a non-protic solvent, such as THF, benzene, toluene or ether. The preferred temperature is 0° C. The mixture is stirred for 12-24 hr and washed with water. The volatiles are removed and the product LIII treated with ethylmercaptan and dry hydrogen chloride in dry methylene chloride for 5-30 min. The solid 5-imino-1,3-oxazole hydrochloride (LIV) is collected by filtration, dissolved in dry pyridine and the solution saturated with hydrogen sulfide during 4 hr at 0° C. The mixture is diluted with an organic solvent and washed with water and dried. Removal of the volatiles provides the 5-amino-1,3-oxazole product (LV) which may be carried forward without further purification or be purified by silica gel chromatography. [0275]
    Figure US20020058678A1-20020516-C00033
  • The synthesis of 1,4-disubstituted-2-aminopyrazoles may be accomplished as illustrated in Scheme XV and described in Lancini et al., [0276] J. Het. Chem., 1966, 3, 152.
    Figure US20020058678A1-20020516-C00034
  • Methods of Therapeutic Use
  • The compounds of the invention effectively block inflammatory cytokine production from cells. The inhibition of cytokine production is an attractive means for preventing and treating a variety of disorders associated with excess cytokine production, e.g., diseases and pathological conditions involving inflammation. Thus, the compounds of the invention are useful for the treatment of such conditions. These encompass chronic inflammatory diseases including, but not limited to, osteoarthritis, multiple sclerosis, Guillain-Barre syndrome, Crohn's disease, ulcerative colitis, psoriasis, graft versus host disease, systemic lupus erythematosus and insulin-dependent diabetes mellitus. The compounds of the invention can also be used to treat other disorders associated with the activity of elevated levels of proinflammatory cytokines such as responses to various infectious agents and a number of diseases of autoimmunity such as rheumatoid arthritis, toxic shock syndrome, diabetes and inflammatory bowel diseases unrelated to those listed above are discussed in the Background of the Invention. [0277]
  • In addition, the compounds of the invention being inhibitors of cytokine production are expected to block inducible cyclooxygenase (COX-2) expression. COX-2 expression has been shown to be increased by cytokines and it is believed to be the [0278]
  • To a mixture of substituted aminoketone (LVI) and cyanamide in water and acetic acid was added aqueous sodium hydroxide until pH 4.5 is reached. The mixture is heated at 50-90° C. for 1-5 hr, cooled and basicified with ammonium hydroxide. The product LVII is collected by filtration and dried. [0279]
    Figure US20020058678A1-20020516-C00035
  • As in the cases described above, the synthesis of many other aminoheterocycles useful as intermediates may be accomplished by methods similar to those described in the literature or known to those skilled in the art. Several additional examples are illustrated in Scheme XVI. 2,5-Disubstituted-3-aminotriazoles (LVIII) have been described by Plenkiewicz et al. ([0280] Bull. Chem. Soc. Belg. 1987, 96, 675). 1,3-Disubstituted-4-aminopyrazoles (LIX) have been described by Guarneri et al. (Gazz. Chim. Ital. 1968, 98, 569). Damany et al. (Tetrahedron, 1976, 32, 2421) describe a 2-amino-3-substituted benzothiophene (LX). A 3-aminoindole (LXI) is described by Foresti et al.. (Gazz. Chim. Ital., 1975, 125, 151). Bristow et al. (J. Chem. Soc., 1954, 616) describe an imidazo[1,2-a]pyridin-2-yl amine (LXII).
  • Scheme XVI isoform of cyclooxygenase responsible for inflammation (M. K. O'Banion et al., [0281] Proc. Natl. Acad. Sci. U.S.A, 1992, 89, 4888.) Accordingly, the present novel compounds would be expected to exhibit efficacy against those disorders currently treated with COX inhibitors such as the familiar NSAIDs. These disorders include acute and chronic pain as well as symptoms of inflammation and cardiovascular disease.
  • As discussed in the Background of the Invention, IL-8 plays a role in the influx of neutrophils into sites of inflammation or injury. Therefore, in a yet further aspect of the invention, the compounds of the invention may be useful in the treatment of diseases mediated predominantly by neutrophils such as stroke and myocardial infarction, alone or following thrombolytic therapy, thermal injury, adult respiratory distress syndrome (ARDS), multiple organ injury secondary to trauma, acute glomerulonephritis, dermatoses with acute inflammatory components, acute purulent meningitis or other central nervous system disorders, hemodialysis, leukopherisis, granulocyte transfusion associated syndromes, and necrotizing entrerocolitis. [0282]
  • For therapeutic use, the compounds of the invention may be administered in any conventional dosage form in any conventional manner. Routes of administration include, but are not limited to, intravenously, intramuscularly, subcutaneously, intrasynovially, by infusion, sublingually, transdermally, orally, topically or by inhalation. The preferred modes of administration are oral and intravenous. [0283]
  • The compounds of this invention may be administered alone or in combination with adjuvants that enhance stability of the inhibitors, facilitate administration of pharmaceutic compositions containing them in certain embodiments, provide increased dissolution or dispersion, increase inhibitory activity, provide adjunct therapy, and the like, including other active ingredients. Advantageously, such combination therapies utilize lower dosages of the conventional therapeutics, thus avoiding possible toxicity and adverse side effects incurred when those agents are used as monotherapies. Compounds of the invention may be physically combined with the conventional therapeutics or other adjuvants into a single pharmaceutical composition. Advantageously, the compounds may then be administered together in a single dosage form. In some embodiments, the pharmaceutical compositions comprising such combinations of compounds contain at least about 5%, but more preferably at least about 20%, of a compound of formula (I) (w/w) or a combination thereof. The optimum percentage (w/w) of a compound of formula(I) may vary and is within the purview of those skilled in the art. Alternatively, the compounds may be administered separately (either serially or in parallel). Separate dosing allows for greater flexibility in the dosing regime. [0284]
  • As mentioned above, dosage forms of the compounds of this invention include pharmaceutically acceptable carriers and adjuvants known to those of ordinary skill in the art. These carriers and adjuvants include, for example, ion exchangers, alumina, aluminum stearate, lecithin, serum proteins, buffer substances, water, salts or electrolytes and cellulose-based substances. Preferred dosage forms include, tablet, capsule, caplet, liquid, solution, suspension, emulsion, lozenges, syrup, reconstitutable powder, granule, suppository and transdermal patch. Methods for preparing such dosage forms are known (see, for example, H. C. Ansel and N. G. Popovish, [0285] Pharmaceutical Dosage Forms and Drug Delivery Systems, 5th ed., Lea and Febiger (1990)). Dosage levels and requirements are well-recognized in the art and may be selected by those of ordinary skill in the art from available methods and techniques suitable for a particular patient. In some embodiments, dosage levels range from about 10-1000 mg/dose for a 70 kg patient. Although one dose per day may be sufficient, up to 5 doses per day may be given. For oral doses, up to 2000 mg/day may be required. As the skilled artisan will appreciate, lower or higher doses may be required depending on particular factors. For instance, specific dosage and treatment regimens will depend on factors such as the patient's general health profile, the severity and course of the patient's disorder or disposition thereto, and the judgment of the treating physician.
  • SYNTHETIC EXAMPLES Example 1 1-[5-tert-Butyl-2-p-tolyl-2H-pyrazol-3-yl]-3-[4-(2-morpholin-4-yl-ethoxy)naphthalen-1-yl]-urea:
  • [0286]
    Figure US20020058678A1-20020516-C00036
  • A mixture of 4-methylphenyl hydrazine hydrochloride (10.0 g) and 4,4-dimethyl-3-oxopentanenitrile (8.67 g) in 150 mL ethanol and 7 mL concentrated HCl was heated at reflux overnight, cooled to room temperature, basified to pH 12 with alkali and extracted with diethyl ether. The combined organic extracts were washed with brine and dried (MgSO[0287] 4). Removal of the volatiles in vacuo left a residue which was triturated with hot petroleum ether (100 mL) and provided 12.5 g of LXVII.
  • To a mixture of 4-amino-1-naphthol hydrochloride (LXIII) (172.1 g) in 750 mL anhydrous THF at −78° C. was added dropwise over 60 min n-butyl lithium (490 mL of a 1.60 M solution in hexanes). After the addition was complete the mixture was allowed to warm to room temperature and then cooled to −78° C. and di-tert-butyl dicarbonate ((BOC)[0288] 2O, 192 g) in 200 mL THF was added over 20 min. The mixture was slowly warmed to room temperature and stirred for 3 h and most of the volatiles removed in vacuo. The residue was diluted with ethyl acetate (1 L) and washed with water (2×200 mL) and brine (200 mL) and filtered through celite and dried (MgSO4). Removal of the volatiles in vacuo provided LXIV (226.1 g).
  • A mixture of LXIV (0.397 g), 4-chloromethylpyridine hydrochloride (0.237 g) and potassium carbonate (0.996 g, powdered) in 10 mL of acetonitrile was heated at 80° C. for 6 hr, cooled to room temperature and diluted with water and ethyl acetate. The organic layer was washed with water and brine and dried (MgSO[0289] 4). Removal of the volatiles in vacuo and purification of the residue with flash chromatography using ethyl acetate as the eluent provided 0.277 g LXV. A mixture of LXV (0.26 g) and HCl (0.6 mL of 4M HCl in dioxane) in 5 mL dioxane was stirred at room temperature for 18 hr. Removal of the volatiles in vacuo provided LXVI.
  • As outlined in Method B (Scheme I), a mixture of LXVII ( 0.076 g) and phosgene (0.68 mL of a 1.93 M solution in toluene) in 10 mL methylene chloride and 10 mL saturated sodium bicarbonate was stirred rapidly for 15 min at 0-5° C. and the organic layer dried (MgSO[0290] 4). Removal of the volatiles in vacuo left a residue which was added to a mixture of the dihydrochloride salt from above (0.104 g) and N,N-di-iso-propylethylamine (0.32 mL) in 5 mL anhydrous THF. The mixture was stirred overnight and diluted with ethyl acetate and water. The organic layer was washed with water and brine and dried (MgSO4). Removal of the volatiles in vacuo and purification of the residue with flash chromatography using ethyl acetate as the eluent and recrystallization of the solid with water and ethanol gave 1, m.p. 132-133° C.
  • Example 2 1-[5-tert-Butyl-2-p-tolyl-2H-pyrazol-3-yl]-3-[4-(3-(tetrahydropyran-2-yl-oxy)propyn-1-yl)naphthalen-1-yl]-urea:
  • [0291]
    Figure US20020058678A1-20020516-C00037
  • Tetrahydro-2-(2-propynyloxy)-2H-pyran (LXVIII) (2.50 mL; 17.8 mmol) in 100 mL anhydrous THF at −78° C. under inert atmosphere was treated with n-butyllithium (7.1 mL of a 2.5 M solution in hexanes), added via syringe. The reaction was warmed to −20° C. and after 1 h stirring, tributyltin chloride (4.8 mL, 17.8 mmol) was added. After stirring at −20° C. for 1 h the reaction mixture was quenched with dilute NaHCO[0292] 3 solution (75 mL) and extracted with ethyl ether (3×50 mL). The combined ethereal extracts were washed with brine and dried (MgSO4). After filtration all volatiles were removed in vacuo to produce LXIX as a yellow oil (4.7 g; 11.0 mmol or 62% yield).
  • A mixture LXVII (Example 1) (1.00 g; 3.76 mmol) and phosgene (5.6 mL of a 2 M solution in toluene) and 4-bromonaphthylamine were reacted according to Method B (Scheme I and Example 1). The product was purified by trituration with hot heptane to afford LXX, mp 193-194° C. (1.75 g, 3.67 mmol, 97% yield). [0293]
  • A mixture of LXX (970 mg, 2.03 mmol) and LXIX (1.31 g, 3.05 mmol) and BHT (50 mg) in 50 mL toluene at reflux under inert atmosphere was treated with tetrakis(triphenylphosphine)palladium(0) (350 mg, 0.305 mmol). The reaction mixture slowly changed color to black. After 40 min heating was stopped and, when the reaction mixture had cooled to ambient temperature, a 5 M aqueous solution of KF (˜75 mL) was added. The mixture was stirred vigorously for 6 h, then the product was extracted with ethyl acetate (3×50 mL). The combined organic extracts were washed with brine and dried (MgSO[0294] 4), filtered and all volatiles were removed in vacuo. Column chromatography, using 25% ethyl acetate in hexane eluant, followed by recrystallization from hot ethyl acetate/hexane afforded 780 mg of 2, mp 159-160° C., (1.45 mmol, 72% yield).
  • Example 3 1-[5-tert-butyl-2-p-tolyl-2H-pyrazol-3-yl]-3-[4-(3-pyridin-4-yl-propoxy)naphthalen-1-yl]-urea (3):
  • [0295]
    Figure US20020058678A1-20020516-C00038
  • To a mixture of LXIV (Example 1) (0.51 g), 4-pyridinyl-1-propanol (0.76 mL), and triphenylphosphine (1.5 g) in 10 mL anhydrous THF was added dropwise diethyl azodicarboxylate (DEADC, 0.90 mL). After stirring overnight, the volatiles were removed in vacuo. Purification of the residue by flash chromatography using 25% hexanes in ethyl acetate as the eluent and concentration of the product-rich fractions in vacuo provided ether LXXI. A mixture of LXXI (0.74 g) and HCl (5 mL, 4.0 M in dioxane) in 10 mL anhydrous dioxane was stirred overnight. Collection of the precipitate by vacuum filtration provided LXXII. LXXVII (Example 1) (0.23 g), saturated NaHCO[0296] 3 (15 mL), dichloromethane (15 mL), phosgene (2.1 mL, 1.93M in toluene) and LXXII (0.32 g) were reacted according to Method B (Scheme I and Example 1). Purification of the residue by flash chromatography using 25% hexanes in ethyl acetate as the eluent, concentration of the product-rich fractions in vacuo, followed by recrystallization from ethyl acetate/methanol provided urea 3, m.p. 205-207° C.
  • Example 4 1-[5-tert-butyl-2-p-tolyl-2H-pyrazol-3-yl]-3-[4-(2-(morpholin-4-yl)-2-oxoethoxy)naphthalen-1-yl]-urea (4):
  • [0297]
    Figure US20020058678A1-20020516-C00039
  • To a solution of morpholine (0.55 mL) in 5 mL of anhydrous ether at 0° C. was added chloroacetyl chloride. Collection of the precipitate by vacuum filtration provided amide LXXIII. A mixture of LXIV (Example 1) (0.44 g), LXXIII (0.30 g), and powered potassium carbonate (0.70 g) in 10 mL acetonitrile was heated to 80° C. for 3.5 hours, cooled to room temperature, and diluted with ethyl acetate and water. The organic layer was washed with water, saturated NaHCO[0298] 3, brine, dried (MgSO4) and the volatiles removed in vacuo. Purification of the residue by flash chromatography using 20% ethyl acetate in hexanes as the eluent and concentration of the product-rich fractions in vacuo provided ether LXXIV. A mixture of LXXIV (0.26 g) and HCl (0.7 mL, 4.0 M in dioxane) in 4 mL anhydrous dioxane was stirred overnight. Collection of the precipitate by vacuum filtration provided LXXV. LXVII (Example 1), (0.13 g), and LXXV were reacted according to Method B (Scheme I and Example 1). Trituration of the residue in hot methanol/water followed by collection of the solid by vacuum filtration provided urea 4, m.p. 240-241° C.
  • Example 5 1-[5-tert-butyl-2-p-tolyl-2H-pyrazol-3-yl]-3-[4-(2-(1-oxothiomorpholin-4-yl)ethoxy)naphthalen-1-yl]-urea (5):
  • [0299]
    Figure US20020058678A1-20020516-C00040
  • A suspension of LXIV (Example 1) (5.0 g), powdered potassium carbonate (13.3 g) and 1-bromo-2-chloroethane (5.5 g) in 100 mL acetonitrile was heated at 80° C. overnight, cooled to room temperature and partitioned between ethyl acetate and water. The organic layer was washed with brine, dried (MgSO[0300] 4) and the volatiles removed in vacuo. Purification of the residue by flash chromatography using 25% ethyl acetate in hexanes as the eluent and concentration of the product-rich fractions in vacuo provided ether LXXVI. A mixture of LXVI (2.0 g) and HCl (15 mL, 4.0 M in dioxane) in 10 mL anhydrous dioxane was stirred overnight. Ether was added and the precipitate collected by vacuum filtration to afford LXXVII. As outlined in Method C, (Scheme I) a solution of LXXVII (1.6 g), phenyl carbamate LXXVIII, prepared from LXVII, phenyl chloroformate (1.05 equiv), pyridine (3 equiv.) in THF, (2.3 g) and diisopropylethylamine (3.1 g) in 10 mL anhydrous DMSO was stirred for one hour and diluted with ethyl acetate and water. The organic layer was washed with water, 50% NaHCO3, brine, dried (MgSO4), and the volatiles removed in vacuo. Purification of the residue by flash chromatography using 33% ethyl acetate in hexanes as the eluent, concentration of the product-rich fractions in vacuo, followed by trituration with 33% ethyl acetate in hexanes provided LXXIX. A mixture of LXXIX (1.6 g) and sodium iodide (5.0 g) in 10 mL acetone was heated at reflux for 4 days, cooled to room temperature and diluted with dichloromethane. The organics were washed with water, dried (Na2SO4) and the volatiles removed in vacuo to provide LXXX.
  • To a solution of thiomorpholine (0.50 g) in 25 mL of dichloromethane was added di-tert-butyldicarbonate. The mixture was stirred for 18 h at room temperature and the volatiles removed in vacuo. Recrystallization of the residue from hexanes provided LXXXI. To a solution of LXXXI (0.40 g) in 8 mL ethanol at 0° C. was added sodium periodate. The mixture was stirred at 0° C. one hour, warmed to room temperature and stirred five days. The mixture was diluted with water and extracted with dichloromethane. The organic layer was washed with brine, dried (Na[0301] 2SO4) and the volatiles removed in vacuo. Trituration of the residue with hexanes provided sulfoxide LXXXIII. To a solution of LXXXIII (0.15 g) in 5 mL dichloromethane was added trifluoroacetic acid (TFA, 0.52 mL). The mixture was stirred 5 h and the volatiles removed in vacuo. The residue was dissolved in methanol and HCl (4.0 M in dioxane) was added. The volatiles were removed in vacuo to provide sulfoxide LXXXIII. A mixture of LXXXIII (0.05 g), LXXX (0.19 g), DIPEA (0.06 mL) in 2.5 mL DMF was stirred overnight. The mixture was partitioned between ethyl acetate and water and the aqueous layer extracted with ethyl acetate. The combined organic layers were washed with brine, dried (MgSO4) and the volatiles removed in vacuo. Purification of the residue by flash chromatography using 9% methanol in ethyl acetate as the eluent and concentration of the product-rich fractions in vacuo provided urea 5, m.p. 205-207° C.
  • Example 6 1-[5-tert-butyl-2-p-tolyl-2H-pyrazol-3-yl]-3-[4-(2-pyridin-4-yl-ethenyl)naphthalen-1-yl]-urea (6):
  • [0302]
    Figure US20020058678A1-20020516-C00041
  • A mixture of 4-bromoaminonaphthalene (5.0 g) and di-tert-butyldicarbonate (5.9 g) in 100 mL toluene was heated at 70° C. for 15 hours, cooled to room temperature and the volatiles removed in vacuo. The residue was dissolved in ethyl acetate, washed with 0.1M HCl and brine, dried (MgSO[0303] 4) and the volatiles removed in vacuo. Recrystallization of the residue from hot petroleum ether provided LXXXIV. 4-Vinylpyridine (0.86 mL) was added to a suspension of LXXXIV (2.0 g) in 5 mL of triethylamine, followed by palladium (II) acetate (0.014 g) and tri-ortho-tolylphosphine (0.038 g). The mixture was heated at 110° C. for four hours, cooled to room temperature, diluted with water and ethyl acetate. The organic layer was washed with brine, dried (MgSO4) and the volatiles removed in vacuo. Purification of the residue by flash chromatography using 50% ethyl acetate in hexanes as the eluent and concentration of the product-rich fractions in vacuo provided naphthalene LXXXV. A solution of LXXXV (0.34 g) in 10 mL TFA was stirred one hour and the volatiles removed in vacuo to provided LXXXVI. LXXXVI and LXVII (Example 1) were reacted according to Method B to provide 6, m.p. 203° C. (dec).
  • Example 7 1-[5-tert-butyl-2-p-tolyl-2H-pyrazol-3-yl]-3-[4-(2-(2-(methoxymethyl)morpholin-4-yl)ethoxy)naphthalen-1-yl]-urea (7):
  • [0304]
    Figure US20020058678A1-20020516-C00042
  • A mixture of LXXXVII (prepared by the method of Y. Jinbo et al; [0305] J. Med. Chem., 1994, 37, 2791) (0.044 g), LXXX (see Example 5) (0.15 g) and DIPEA (0.068 g) was stirred overnight, diluted with ether and water. The organic layer was washed with brine, dried (MgSO4) and the volatiles removed in vacuo. Purification of the residue by flash chromatography using a gradient of 1-4% methanol in ethyl acetate as the eluent and concentration of the product-rich fractions in vacuo provided 7, m.p. 85-90° C.
  • Example 8 1-[5-tert-butyl-2-p-tolyl-2H-pyrazol-3-yl]-3-[4-(2-morpholin-4-yl-ethoxy)naphthalen-1-yl]-urea (8):
  • [0306]
    Figure US20020058678A1-20020516-C00043
  • A mixture of LXIV (Example 1) (0.464 g), 4-(2-chloroethyl)morpholine hydrochloride (0.3435 g) and powdered potassium carbonate (0.93 g) was heated in acetonitrile (15 mL) at 80° C. for 3 hours, cooled to room temperature and diluted with ethyl acetate and water. The organic layer was washed with water, brine, dried (MgSO[0307] 4) and the volatile removed in vacuo. Purification of the residue by flash chromatography using 12% hexanes in ethyl acetate as the eluent and concentration in vacuo of the product-rich fractions afforded LXXXVIII. A solution of LXXXVIII (0.511 g) and HCl (1 mL of a 4M dioxane solution) in 5 mL dioxane was stirred at room temperature 20 hours. Removal of the volatiles in vacuo provided the product LXXXIX, which was reacted with LXVII (Example 1) according to Method B to provide 8, m.p. 142-143° C.
  • Example 9 1-[5-tert-butyl-2-(2-methylpyridin-5-yl)-2H-pyrazol-3-yl]-3-[4-(2-morpholin-4-yl-ethoxy)naphthalen-1-yl]-urea (9):
  • [0308]
    Figure US20020058678A1-20020516-C00044
  • A slurry of diethyl malonate (42 mL) and sodium (4.71 g) was warmed slowly to 90° C. and stirred at 90° C. for 2 hours and 120° C. for 30 min. before being cooled to room temperature. Toluene (200 mL) and 2-chloro-5-nitropyridine (25.0 g) were added and the mixture was heated at 110° C. for 1.5 hours and ambient temperature for 17 h. After removal of the volatiles in vacuo, 6 N HCl (200 mL) was added and the mixture heated to reflux for 4 h and cooled to room temperature. The solution was neutralized with solid sodium carbonate, extracted with ethyl acetate (6×100 mL), dried over solid magnesium sulfate, and concentrated to a dark solid. This material was purified by flash chromatography using 20% ethyl acetate in petroleum ether as the eluent. Concentration in vacuo of the product-rich fractions afforded 2-methyl-5-nitropyridine. A mixture of 2-methyl-5-nitropyridine (13.0 g) and 10% Pd on activated carbon (0.1 g) in 1,4-dioxane (150 mL) was hydrogenated at 50 psi for 24 hours and filtered over celite. Removal of the volatiles in vacuo provided 2-methyl-5-aminopyridine. A solution of this compound (9.90 g) was dissolved in 6 N HCl (100 mL), cooled to 0° C., and vigorously stirred throughout the procedure. Sodium nitrite (6.32 g) in water (50 mL) was added. After 30 min, tin (II) chloride dihydrate (52.0 g) in 6 N HCl (100 mL) was added and the reaction slurry was stirred at 0° C. for 3 hours. The pH was adjusted to pH 14 with 40% aqueous potassium hydroxide solution and extracted with ethyl acetate. The combined organic extracts were dried (MgSO[0309] 4) and removal of the volatiles in vacuo provided hydrazine XC. A solution of XC (8.0 g) and 4,4-dimethyl-3-oxopentanenitrile (10.0 g) in ethanol (200 mL) and 6 N HCl (6 mL) was refluxed for 17 hours and cooled to room temperature. Solid sodium hydrogen carbonate was added to neutralize the solution. The slurry was filtered and removal of the volatiles in vacuo provided a residue which was purified by column chromatography using ethyl acetate as the eluent. Concentration in vacuo of the product-rich fractions afforded XCI, which was reacted with LXXXIX (Example 8) according to Method B to provide 9, m.p. 121-123° C.
  • Example 10 1-[5-tert-butyl-2-(2-methylpyridin-5-yl)-2H-pyrazol-3-yl]-3-[4-(2-pyridin-4-yl-ethoxy)naphthalen-1-yl]-urea (10)
  • [0310]
    Figure US20020058678A1-20020516-C00045
  • To a solution of LXIV (Example 1) (0.962 g), 2-(pyridin-4-yl)ethanol (1.4 g) and triphenylphosphine (2.90 g) in THF (25 mL) was added dropwise DEADC (1.8 mL). The mixture was stirred overnight and the volatiles removed in vacuo. Purification of the residue with flash chromatography using ethyl acetate as the eluent and concentration in vacuo of the product-rich fractions provided XCII. To a solution of XCII (1.4 g) in dioxane (15 mL) was added HCl (10 mL of a 4M dioxane solution). The solution was stirred overnight and product XCIII was filtered and dried. This was reacted with XCI (Example 9) according to Method B to provide 10, m.p. 189-190° C. [0311]
  • Example 11 1-[5-(1-methylcyclohex-1-yl)-2-phenyl-2H-pyrazol-3-yl]-3-[4-(2-morpholin-4-yl-ethoxy)naphthalen-1-yl]-urea (11):
  • [0312]
    Figure US20020058678A1-20020516-C00046
  • To a solution of cyclohexane-1-methyl-1-carboxylic acid (1.31 g) in 5 mL methylene chloride was added oxalyl chloride solution (5.5 mL of a 2.0 M methylene chloride solution) and 1 drop of anhydrous DMF. The mixture was refluxed for 3 hours under inert atmosphere and cooled to room temperature. Cyanoacetic acid (1.57 g) in ethyl acetate was dried (MgSO[0313] 4) and the volatiles removed in vacuo. The residue and 2,2-bipyridine (˜10 mg) in anhydrous THF (70 mL) was cooled to −70° C. and treated with n-BuLi (2.5 M in Hexanes) slowly, while allowing the reaction mixture to reach 0° C. When the red color persists at 0° C. (ie. after 15.0 mL of n-BuLi solution), the solution was recooled to −70° C. and the acid chloride solution from above (9.21 mmol) was added via syringe in one portion. The mixture was warmed to room temperature, stirred 0.5 hours, poured onto 1 N aq. HCl (200 mL) and extracted with chloroform (3×100 mL). The combined organic layers were washed with saturated aqueous NaHCO3, brine and dried (MgSO4). Removal of volatiles in vacuo provided a residue which was purified by column chromatography using hexanes and ethyl acetate as the eluent. Concentration in vacuo of the product-rich fractions provided XCIV . A solution of XCIV (0.80 g) and phenylhydrazine (0.48 mL) in toluene (5 mL) was heated with azeotropic removal of water overnight and the volatiles removed in vacuo. Purification of the residue with flash chromatography using ethyl acetate and hexanes as the eluent and concentration in vacuo of the product-rich fractions provided XCV, which was reacted with LXXXIX (Example 8) according to Method B to provide 11, m.p. 147-149° C.
  • Example 12 1-[5-tert-butyl-2-p-tolyl-2H-pyrazol-3-yl]-3-[4-(pyridin-4-yl-methylamino)naphthalen-1-yl]-urea (12):
  • [0314]
    Figure US20020058678A1-20020516-C00047
  • Isonicotinic acid (1.13 g) and DCC (2.5 g) were mixed together in methylene chloride (80 mL) under an inert atmosphere and at room temperature. After 30 min 4-nitro-1-naphthylamine (1.70 g) was added to this suspension as well as a catalytic amount of DMAP (˜50 mg). After 2 days the suspension was filtered through Celite, the volatiles removed in vacuo and the residue purified by column chromatography to afford XCVI. A mixture of XCVI (0.299 g) in acetic acid (6 mL) was treated at room temperature with a solution of tin chloride (1.55 g) in 6 mL of concentrated HCl. After stirring for 1.5 hours, the mixture was poured slowly into 200 mL 15% aqueous NaOH solution and extracted with ethyl acetate (3×100 mL). Drying (MgSO[0315] 4), removal of volatiles in vacuo and purification of the residue by column chromatography using 5% methanol in ethyl acetate as the eluent afforded XCVII, which was reacted with LXVII (Example 1) according to Method B to provide XCVIII. To a suspension of XCVIII (0.101 g) in anhydrous THF (7 mL) at room temperature was added dropwise Red-Al (65% w/w solution in toluene; 0.27 mL) under an inert atmosphere. The mixture was then refluxed for 1 h (dark red color), cooled and methanol was added dropwise until no more evolution of H2 was detected. Removal of most of the solvent in vacuo provided a residue which was purified by column chromatography using hexanes, 50% ethyl ecetate in hexanes and finally ethyl acetate as the eluents. Concentration of the product-rich fractions in vacuo furnished solid 12, m.p. 174-177° C.
  • Example 13 1-[5-tert-butyl-2-(3-(2-morpholin-4-yl-ethyl)phenyl)-2H-pyrazol-3-yl]-3-[4-(2-morpholin-4-yl-ethoxy)naphthalen-1-yl]-urea (13):
  • [0316]
    Figure US20020058678A1-20020516-C00048
  • A mixture of 3-nitrophenylacetic acid (5.02 g), morpholine (4.83 mL) and EDC (10.62 g) in 80 mL DMF at room temperature was stirred for 6 hours and diluted with water and extracted with ethyl acetate. The combined organic extracts were washed with water and brine and dried (MgSO[0317] 4). Removal of the volatiles in vacuo provided XCIX. A mixture of XCIX (6.7 g) and 10% Pd on carbon (0.1 g) in ethyl acetate (100 mL) was hydrogenated at 45 psi for 15 hours and filtered over celite. Removal of the volatiles in vacuo furnished an amine (5.7 g) which was dissolved in 6 N HCl (40 mL), cooled to 0° C., and vigorously stirred. Sodium nitrite (2.11 g) in water (5 mL) was added in a dropwise fashion. After 30 min, tin (II) chloride dihydrate (52.0 g) in 6 N HCl (100 mL) was added via addition funnel and the reaction slurry was stirred at 0° C. for 3 hours. The pH was adjusted to 14 with 40% aqueous sodium hydroxide solution and the solution extracted with ethyl acetate. The organic layers were dried (MgSO4). Removal of the volatiles in vacuo provided C. A solution of C (2 g) and 4,4-dimethyl-3-oxopentanenitrile (1.1 g) in ethanol (80 mL) containing 6 N HCl (2 mL) was refluxed for 17 hours, cooled to room temperature and the pH was adjusted to 14 with 40% aqueous sodium hydroxide solution. The mixture was extracted with ethyl acetate and the combined organic extracts were dried (MgSO4). Removal of the volatile in vacuo provided CI. To a solution of CI (150 mg) in dry THF (10 mL) at 0° C. was added dropwise a solution of LAH in ether (2.13 mL of a 1M solution). The mixture was slowly warmed to 60° C., stirred for 5 hours, cooled to room temperature and stirred 16 hours. The reaction was quenched with the addition of 10% aqueous NaOH solution until a neutral pH was achieved The mixture was extracted with ethyl acetate and the combined organic extracts were dried (MgSO4). Removal of the volatile in vacuo provided a residue which was purified by column chromatography using ethyl acetate as the eluent. Concentration of the product-rich fractions in vacuo furnished CII, which was reacted with LXXXIX (Example 8) according to Method B to provide 13, as an oil.
  • Example 14 1-[5-tert-butyl-2-p-tolyl-2H-pyrazol-3-yl]-3-[4-(3-morpholin-4-yl-propyl)naphthalen-1-yl]-urea (14):
  • [0318]
    Figure US20020058678A1-20020516-C00049
  • A mixture of LXX (Example 2) (3.0 g), CIII (prepared by the procedure of J. W. Labadie et al; 1983, J. Org. Chem. 48, 4634) (3.0 g) and tetrakistriphenylphosphinepalladium (0.15 g) in 18 mL toluene was heated to 100° C. for 30 min. Another 0.050 g of catalyst was added. The mixture was heated three hours, cooled to room temperature, diluted with ether and washed with 5% NH[0319] 4OH, water, brine, dried (MgSO4) and the volatiles removed in vacuo. Purification of the residue by flash chromatography using 1% methanol in dichloromethane as the eluent and concentration of the product-rich fractions in vacuo provided CIV. To CIV (2.2 g), and hydrazine (4.9 g) in 50 mL ethanol and 10 mL THF at 0° C. was added dropwise a solution of sodium periodate (8.1 g) in 15 mL water. The mixture was warmed to room temperature, stirred six hours, heated to 40° C. for two hours and diluted with dichloromethane, washed with 1N sodium hydroxide, water, brine and dried (MgSO4). Removal of the volatiles in vacuo provided the saturated olefin. A mixture of this alkane (2.1 g) and tetrabutylammonium flouride (14.4 mL, 1M in THF) and acetic acid (1.1 g) was stirred overnight, diluted with ethyl acetate and washed with water, brine, and dried (MgSO4). Removal of the volatiles in vacuo, purification of the residue by flash chromatography using 33% hexanes in ethyl acetate as the eluent and concentration of the product-rich fractions in vacuo provided the alcohol. To a solution of this alcohol (0.60 g) in acetonitrile at 0° C. was added triphenylphosphine (0.52 g) then carbon tetrabromide (0.65 g). The mixture was stirred at room temperature for two days and the volatiles removed in vacuo. Purification of the residue by flash chromatography using 33% ethyl acetate in hexanes as the eluent and concentration of the product-rich fractions in vacuo provided CV. A mixture of CV (0.23 g), morpholine (0.039 g), KI (0.073 g) and DIPEA (0.1 mL) in DMF (3 mL) was stirred 6 hours at room temperature and diluted with ether and water. The organic layer was washed with brine and dried (MgSO4). Removal of the volatiles in vacuo provided a residue which was purified by flash chromatography using ethyl acetate as the eluent. Concentration in vacuo of the product-rich fractions provided 14 which was recrystallized from hexanes and ethyl acetate, m.p. 147-149° C.
  • Table 1 illustrates additional compounds of the invention, which were prepared by methods analogous to those described above. [0320]
    TABLE 1
    Figure US20020058678A1-20020516-C00050
    Figure US20020058678A1-20020516-C00051
    Figure US20020058678A1-20020516-C00052
    Ex.
    No. R1 R3 Q—L— m.p. ° C.
    15 tert-butyl 2-Cl-pyridin-5-yl 2-(morpholin-4-yl)ethoxy 123-125
    16 tert-butyl 4-methyl-phenyl 2-(imidazol-1-yl)ethoxy 201-202
    17 tert-butyl 2-methoxy-pyridin- 2-(morpholin-4-yl)ethoxy 108-110
    5-yl
    18 tert-butyl pyridin-3-yl 2-(morpholin-4-yl)ethoxy 191-192
    19 tert-butyl 4-Cl-phenyl 2-(morpholin-4-yl)ethoxy 116-118
    20 tert-butyl 4-methyl-phenyl pyridin-3-ylmethylamino 137-140
    21 tert-butyl 4-methyl-phenyl morpholin-4-yl-methyl 174
    22 tert-butyl 4-methyl-phenyl 2-(pyridin-4-yl)ethoxy 187-190
    23 tert-butyl 4-methyl-phenyl 3-(pyridin-3-yl)-n-propoxy 162-163
    24 tert-butyl 4-methyl-phenyl morpholine-4-carbonyloxyethoxy 176-177
    25 tert-butyl 4-methyl-phenyl 2-(morpholin-4-yl)ethoxy 176-177
    (Ar2 = 3-methylnapth-1-yl)
    26 tert-butyl 4-methyl-phenyl 2-(pyridin-4-yl)ethyl 117-120
    27 tert-butyl methyl 2-(morpholin-4-yl)ethoxy 201-202
    28 tert-butyl 4-methyl-phenyl 2-(thiomorpholin-4-yl)ethoxy 122-124
    29 tert-butyl 4-methyl-phenyl 2-(piperazin-1-yl)ethoxy 190
    30 tert-butyl 4-methyl-phenyl 2-(morpholin-4-yl)-n-propoxy 110-111
    31 tert-butyl 4-methyl-phenyl 2-(4-tetrahydropyran-4-yl)ethoxy 174-175
    32 tert-butyl 4-methyl-phenyl 3-(morpholin-4-yl)propyn-1-yl 120-121
    33 tert-butyl 4-methyl-phenyl 3-(piperidin-1-yl)propyn-1-yl 109-112
    34 tert-butyl 4-methyl-phenyl 4-[4-(tetrahydropyran-2-yloxy)but- 180-181
    1-ynyl]
    35 tert-butyl 4-methyl-phenyl 2-(3,4-dimethoxyphenyl)ethoxy 183-184
    36 tert-butyl 4-methyl-phenyl (pyridine-4-carbonyl)amino >250
    37 i-Pr phenyl 2-(morpholin-4-yl)ethyl 177-178
    38 CF3CH2 4-methyl-phenyl 2-(morpholin-4-yl)ethyl 176-178
    39 3- phenyl 2-(morpholin-4-yl)ethyl 155-156
    tetrahydro-
    pyranyl
    40 cyclohexyl phenyl 2-(morpholin-4-yl)ethyl 191-192
    41 tert-butyl n-butyl 2-(morpholin-4-yl)ethyl 81-83
    42 tert-butyl benzyl 2-(morpholin-4-yl)ethyl 180-181
    43 tert-butyl 4-methyl-3- 2-(morpholin-4-yl)ethyl oil
    morpholin-
    4-yl-methylphenyl
    44 tert-butyl 4-methyl-3- 2-(morpholin-4-yl)ethyl oil
    C(O)NH2
    phenyl
    45 tert-butyl 4-methyl-3- 2-(morpholin-4-yl)ethyl oil
    (dimethyl)NCH2
    phenyl
    46 tert-butyl 4-methyl-phenyl pyridin-4-yl-oxy
    47 1-methyl- 4-methyl-phenyl 2-(morpholin-4-yl)ethoxy 146-8 
    cycloprop-
    1-yl
    48 tert-butyl 4-methyl-phenyl 2-(morpholin-4-yl)ethoxy 99-100
    Ar2 = 5,6,7,8-
    tetrahydronaphthalene
    49 tert-butyl 4-methyl-phenyl 2-(trans-2,6-dimethy-morpholin- 137-139
    4-yl)ethoxy
    50 tert-butyl 4-methyl-phenyl 2-(cis-2,6-dimethyl-morpholin-4- 153-154
    yl)ethoxy
    51 tert-butyl 4-methyl-phenyl 2-(2-methoxymethylmorpholin-4- 85-90
    yl)ethoxy
    52 tert-butyl 4-methyl-phenyl 2-(1-oxo-thiomorpholin-4- 205-207
    yl)ethoxy
    53 tert-butyl 4-methyl-phenyl 2-(1-oxo-thiazolidin-3-yl)ethoxy 193-195
    54 tert-butyl 4-methyl-phenyl 5-methylamino-5-oxo-butyloxy 117-119
    55 tert-butyl 4-methyl-phenyl 5-amino-5-oxo-butyloxy foam
    56 tert-butyl 4-methyl-phenyl 5-(morpholin-4-yl)-5-oxo-butyloxy foam
    57 tert-butyl 2-methyl-pyridin-5- pyridin-4-yl-thio
    yl
  • Assessment of Biological Properties
  • Inhibition of TNF Production in THP Cells [0321]
  • The inhibition of cytokine production can be observed by measuring inhibition of TNFα in lipopolysaccharide stimulated THP cells. All cells and reagents were diluted in RPMI 1640 with phenol red and L-glutamine, supplemented with additional L-glutamine (total: 4 mM), penicillin and streptomycin (50 units/ml each) and fetal bovine serum (FBS, 3%) (GIBCO, all conc. final). Assay was performed under sterile conditions; only test compound preparation was nonsterile. Initial stock solutions were made in DMSO followed by dilution into RPMI 1640 2-fold higher than the desired final assay concentration. Confluent THP. 1 cells (2×10[0322] 6 cells/ml, final conc.; American Type Culture Company, Rockville, Md.) were added to 96 well polypropylene round bottomed culture plates (Costar 3790; sterile) containing 125 μl test compound (2 fold concentrated) or DMSO vehicle (controls, blanks). DMSO concentration did not exceed 0.2% final. Cell mixture was allowed to preincubate for 30 min, 37° C., 5% CO2 prior to stimulation with lipopolysaccharide (LPS; 1 μg/ml final; Siga L-2630, from E.coli serotype 0111.B4; stored as 1 mg/ml stock in endotoxin screened distilled H2O at −80° C.). Blanks (unstimulated) received H2O vehicle; final incubation volume was 250 μl. Overnight incubation (18-24 hr) proceeded as described above. Assay was terminated by centrifuging plates 5 min, room temperature, 1600 rpm (400×g); supernatants were transferred to clean 96 well plates and stored −80° C. until analyzed for human TNFα by a commercially available ELISA kit (Biosource #KHC3015, Camarillo, Calif.). Data was analyzed by non-linear regression (Hill equation) to generate a dose response curve using SAS Software System (SAS institute, Inc., Cary, N.C.). The calculated IC50 value is the concentration of the test compound that caused a 50% decrease in the maximal TNFα production.
  • Representative compounds from the synthetic examples above and Table 1 were evaluated and all had IC[0323] 50<10 μM in this assay.
  • Inhibition of other cytokines [0324]
  • By similar methods using peripheral blood monocytic cells, appropriate stimuli, and commercially available ELISA kits for a particular cytokine, inhibition of IL-1β, GM-CSF, IL-6 and IL-8 was demonstrated by representatives from the synthetic examples and Table 1. [0325]

Claims (15)

What is claimed is:
1. A compound of the formula (I) shown below, wherein said compound is a hydroxylated or oxidized metabolite compound of the formula (I);
Figure US20020058678A1-20020516-C00053
wherein
Ar1 is pyrazole optionally substituted by one or more R1, R2 or R3;
Ar2 is:
phenyl, naphthyl, quinoline, isoquinoline, tetrahydronaphthyl, tetrahydroquinoline, tetrahydroisoquinoline, benzimidazole, benzofuran, indanyl, indenyl or indole each being optionally substituted with one to three R2 groups;
L is a C1-10 saturated or unsaturated branched or unbranched carbon chain;
wherein one or more methylene groups are optionally independently replaced by O, N or S; and
wherein said linking group is optionally substituted with 0-2 oxo groups and one or more C1-4 branched or unbranched alkyl which may be substituted by one or more halogen atoms;
Q is selected from the group consisting of:
a) pyridine, pyrimidine, pyridazine, imidazole, benzimidazole, oxazo[4,5-b]pyridine and imidazo[4,5-b]pyridine, which are optionally substituted with one to three groups selected from the group consisting of halogen, C1-6 alkyl, C1-6 alkoxy, hydroxy, mono- or di-(C1-3 alkyl)amino, C1-6 alkyl-S(O)m and phenylamino wherein the phenyl ring is optionally substituted with one to two groups consisting of halogen, C1-6 alkyl and C1-6 alkoxy;
b) morpholine, thiomorpholine, thiomorpholine sulfoxide, thiomorpholine sulfone, piperidine, piperidinone or tetrahydropyrimidone which are optionally substituted with one to three groups selected from the group consisting of C1-6 alkyl, C1-6 alkoxy, hydroxy, mono- or di-(C1-3 alkyl)amino-C1-3 alkyl, phenylamino-C1-3 alkyl and C1-3 alkoxy-C1-3 alkyl;
R1 is selected from the group consisting of:
a) C3-10 branched or unbranched alkyl, which may optionally be partially or fully halogenated, and optionally substituted with one to three phenyl, naphthyl or heterocyclic groups selected from the group consisting of pyridinyl, pyrimidinyl, pyrazinyl, pyridazinyl, pyrrolyl, imidazolyl, pyrazolyl, thienyl, furyl, isoxazolyl and isothiazolyl; each such phenyl, naphthyl or heterocycle selected from the group hereinabove described, being substituted with 0 to 5 groups selected from the group consisting of halogen, C1-6 branched or unbranched alkyl which is optionally partially or fully halogenated, C3-8 cycloalkyl, C5-8 cycloalkenyl, hydroxy, cyano, C1-3 alkyloxy which is optionally partially or fully halogenated, NH2C(O) and di(C1-3)alkylaminocarbonyl;
b) C3-7 cycloalkyl selected from the group consisting of cyclopropyl, cyclobutyl, cyclopentanyl, cyclohexanyl, cycloheptanyl, bicyclopentanyl, bicyclohexanyl and bicycloheptanyl, which may optionally be partially or fully halogenated and which may optionally be substituted with one to three C1-3 alkyl groups, or an analog of such cycloalkyl group wherein one to three ring methylene groups are replaced by groups independently selected from O, S, CHOH, >C═O, >C═S and NH;
c) C3-10 branched alkenyl which may optionally be partially or fully halogenated, and which is optionally substituted with one to three C1-5 branched or unbranched alkyl, phenyl, naphthyl or heterocyclic groups, with each such heterocyclic group being independently selected from the group consisting of pyridinyl, pyrimidinyl, pyrazinyl, pyridazinyl, pyrrolyl, imidazolyl, pyrazolyl, thienyl, furyl, isoxazolyl and isothiazolyl, and each such phenyl, naphthyl or heterocyclic group being substituted with 0 to 5 groups selected from halogen, C1-6 branched or unbranched alkyl which is optionally partially or fully halogenated, cyclopropyl, cyclobutyl, cyclopentanyl, cyclohexanyl, cycloheptanyl, bicyclopentanyl, bicyclohexanyl and bicycloheptanyl, hydroxy, cyano, C1-3 alkyloxy which is optionally partially or fully halogenated, NH2C(O), mono- or di(C1-3)alkylaminocarbonyl;
d) C5-7 cycloalkenyl selected from the group consisting of cyclopentenyl, cyclohexenyl, cyclohexadienyl, cycloheptenyl, cycloheptadienyl, bicyclohexenyl and bicycloheptenyl, wherein such cycloalkenyl group may optionally be substituted with one to three C1-3 alkyl groups;
e) cyano; and,
f) methoxycarbonyl, ethoxycarbonyl and propoxycarbonyl;
R2 is selected from the group consisting of:
a C1-6 branched or unbranched alkyl which may optionally be partially or fully halogenated, acetyl, aroyl, C1-4 branched or unbranched alkoxy, which may optionally be partially or fully halogenated, halogen, methoxycarbonyl and phenylsulfonyl;
R3 is selected from the group consisting of:
a) a phenyl, naphthyl or heterocyclic group selected from the group consisting of pyridinyl, pyrimidinyl, pyrazinyl, pyridazinyl, pyrrolyl, imidazolyl, pyrazolyl, thienyl, furyl, tetrahydrofuryl, isoxazolyl, isothiazolyl, quinolinyl, isoquinolinyl, indolyl, benzimidazolyl, benzofuranyl, benzoxazolyl, benzisoxazolyl, benzpyrazolyl, benzothiofuranyl, cinnolinyl, pterindinyl, phthalazinyl, naphthypyridinyl, quinoxalinyl, quinazolinyl, purinyl and indazolyl; wherein such phenyl, naphthyl or heterocyclic group is optionally substituted with one to five groups selected from the group consisting of a C1-6 branched or unbranched alkyl, phenyl, naphthyl, heterocycle selected from the group hereinabove described, C1-6 branched or unbranched alkyl which is optionally partially or fully halogenated, cyclopropyl, cyclobutyl, cyclopentanyl, cyclohexanyl, cycloheptanyl, bicyclopentanyl, bicyclohexanyl, bicycloheptanyl, phenyl C1-5 alkyl, naphthyl C1-5 alkyl, halo, hydroxy, cyano, C1-3 alkyloxy which may optionally be partially or fully halogenated, phenyloxy, naphthyloxy, heteraryloxy wherein the heterocyclic moiety is selected from the group hereinabove described, nitro, amino, mono- or di-(C1-3)alkylamino, phenylamino, naphthylamino, heterocyclylamino wherein the heterocyclyl moiety is selected from the group hereinabove described, NH2C(O), a mono- or di-(C1-3)alkyl aminocarbonyl, C1-5 alkyl-C(O)—C1-4 alkyl, amino-C1-5 alkyl, mono- or di-(C1-3)alkylamino-C1-5 alkyl, amino-S(O)2, di-(C1-3)alkylamino-S(O)2, R4-C1-5 alkyl, R5 -C1-5 alkoxy, R6-C(O)—C1-5 alkyl and R7-C1-5 alkyl(R8)N;
b) a fused aryl selected from the group consisting of benzocyclobutanyl, indanyl, indenyl, dihydronaphthyl, tetrahydronaphthyl, benzocycloheptanyl and benzocycloheptenyl, or a fused heterocyclyl selected from the group consisting of cyclopentenopyridine, cyclohexanopyridine, cyclopentanopyrimidine, cyclohexanopyrimidine, cyclopentanopyrazine, cyclohexanopyrazine, cyclopentanopyridazine, cyclohexanopyridazine, cyclopentanoquinoline, cyclohexanoquinoline, cyclopentanoisoquinoline, cyclohexanoisoquinoline, cyclopentanoindole, cyclohexanoindole, cyclopentanobenzimidazole, cyclohexanobenzimidazole, cyclopentanobenzoxazole, cyclohexanobenzoxazole, cyclopentanoirnidazole, cyclohexanoimidazole, cyclopentanothiophene and cyclohexanothiophene; wherein the fused aryl or fused heterocyclyl ring is substituted with 0 to 3 groups independently selected from phenyl, naphthyl and heterocyclyl selected from the group consisting of pyridinyl, pyrimidinyl, pyrazinyl, pyridazinyl, pyrrolyl, imidazolyl, pyrazolyl, thienyl, furyl, isoxazolyl, and isothiazolyl, C1-6 branched or unbranched alkyl which is optionally partially or fully halogenated, halo, cyano, C1-3 alkyloxy which is optionally partially or fully halogenated, phenyloxy, naphthyloxy, heterocyclyloxy wherein the heterocyclyl moiety is selected from the group hereinabove described, nitro, amino, mono- or di-(C1-3)alkylamino, phenylamino, naphthylamino, heterocyclylamino wherein the heterocyclyl moiety is selected from the group hereinabove described, NH2C(O), a mono- or di-(C1-3)alkyl aminocarbonyl, C1-4 alkyl-OC(O), C1-5 alkyl-C(O)—C1-4 branched or unbranched alkyl, an amino-C1-5 alkyl, mono- or di-(C1-3)alkylamino-C1-5 alkyl, R9-C1-5 alkyl, R10-C1-5 alkoxy, R11-C(O)—C1-5 alkyl and R12-C1-5alkyl(R13)N;
c) cycloalkyl selected from the group consisting of cyclopentanyl, cyclohexanyl, cycloheptanyl, bicyclopentanyl, bicyclohexanyl and bicycloheptanyl, which the cycloalkyl may optionally be partially or fully halogenated and which may optionally be substituted with one to three C1-3 alkyl groups;
d) C5-7 cycloalkenyl, selected from the group consisting of cyclopentenyl, cyclohexenyl, cyclohexadienyl, cycloheptenyl, cycloheptadienyl, bicyclohexenyl and bicycloheptenyl, wherein such cycloalkenyl group may optionally be substituted with one to three C1-3 alkyl groups; and
e) acetyl, aroyl, alkoxycarbonylalkyl or phenylsulfonyl;
f) C1-6 branched or unbranched alkyl which may optionally be partially or fully halogenated; wherein
or R1 and R2 taken together may optionally form a fused phenyl or pyridinyl ring,
each R8, R13 is independently selected from the group consisting of:
hydrogen and C1-4 branched or unbranched alkyl which may optionally be partially or fully halogenated;
each R4, R5, R6, R7, R9, R10, R11, and R12 is independently selected from the group consisting of:
morpholine, piperidine, piperazine, imidazole and tetrazole;
m=0, 1, 2;
X=O or S and
physiologically acceptable acids or salts thereof.
2. The metabolite compound according to claim 1 where in the formula (I) Ar2 is naphthyl, tetrahydronaphthyl, indanyl or indenyl.
3. The metabolite compound according to claim 2 where in the formula (I) Ar2 is naphthyl.
4. The metabolite compound according to claim 3 where in the formula (I):
Ar2 is 1-naphthyl;
L is C1-6 saturated or unsaturated branched or unbranched carbon chain wherein
one or more methylene groups are optionally independently replaced by O, N or S; and
wherein said linking group is optionally substituted with 0-2 oxo groups and one or more C1-4 branched or unbranched alkyl which may be substituted by one or more halogen atoms;
R1 is selected from the group consisting of C3-10alkyl branched or unbranched, cyclopropyl and cyclohexyl which may optionally be partially or fully halogenated and which may optionally be substituted with one to three C1-3 alkyl groups;
R3 is selected from the group consisting of C1-4alkyl branched or unbranched, cyclopropyl, phenyl, pyridinyl each being optionally substituted as described in claim 1, alkoxycarbonylalkyl, C1-6alkyl branched or unbranched, cyclopropyl and cyclopentyl optionally substituted as described in claim 1.
5. The metabolite compound according to claim 4 where in the formula (I) L is C1-5 saturated carbon chain wherein one or more methylene groups are optionally independently replaced by O, N or S;
wherein said linking group is optionally substituted with 0-2 oxo groups and one or more C1-4 branched or unbranched alkyl which may be substituted by one or more halogen atoms; and X═O.
6. The metabolite compound according to claim 5 where in the formula (I) L is propoxy, ethoxy or methoxy each being optionally substituted with 0-2 oxo groups and one or more C1-4 branched or unbranched alkyl which may be substituted by one or more halogen atoms.
7. The metabolite compound according to claim 6 where in the formula (I) L is ethoxy optionally substituted with 0-2 oxo groups and one or more C1-4 branched or unbranched alkyl which may be substituted by one or more halogen atoms.
8. The metabolite compound according to claim 5 where in the formula (I) L is methyl or propyl each being optionally substituted with 0-2 oxo groups and one or more C1-4 branched or unbranched alkyl which may be substituted by one or more halogen atoms.
9. The metabolite compound according to claim 5 where in the formula (I) L is C3-5 acetylene optionally substituted with 0-2 oxo groups and one or more C1-4 branched or unbranched alkyl which may be substituted by one or more halogen atoms.
10. The metabolite compound according to claim 5 where in the formula (I) L is methylamino optionally substituted with 0-2 oxo groups and one or more C1-4 branched or unbranched alkyl which may be substituted by one or more halogen atoms.
11. A compound selected from the group consisting of:
1-[5-(2-hydroxy-1,1-dimethyl-ethyl)-2-p-tolyl-2H-pyrazol-3-yl]-3-[4-(2-morpholin-4-yl-ethoxy)-naphthalen-1-yl]-urea;
1-[5-tert-butyl-2-(3 -hydroxy-4-methyl-phenyl)-2H-pyrazol-3-yl]-3-[4-(2-morpholin-4-yl-ethoxy)-naphthalen-1-yl]-urea;
1-[5-tert-butyl-2-(4-hydroxymethyl-phenyl)-2H-pyrazol-3 -yl]-3-[4-(2-morpholin-4-yl-ethoxy)-naphthalen-1-yl]-urea;
1-[5-tert-butyl-2-p-tolyl-2H-pyrazol-3-yl]-3-{4-[2-(3-oxo-morpholin-4-yl)-ethoxy]-naphthalen-1-yl}-urea;
1-[5-tert-butyl-2-p-tolyl-2H-pyrazol-3-yl]-3-{4-[2-(4-hydroxy-morpholin-4-yl)-ethoxy]-naphthalen-1-yl}-urea;
1-[5-(2-hydroxy-1,1-dimethyl-ethyl)-2-(6-methyl-pyridin-3-yl)-2H-pyrazol-3-yl]-3-[4-(2-morpholin-4-yl-ethoxy)-naphthalen-1-yl]-urea;
1-[5-tert-butyl)-2-(1-hydroxy-6-methyl-pyridin-3-yl)-2H-pyrazol-3-yl]-3-[4-(2-morpholin-4-yl-ethoxy)-naphthalen-1-yl]-urea;
1-[5-tert-butyl)-2-(6-methyl-pyridin-3-yl)-2H-pyrazol-3-yl]- 3-{4-[2-(4-hydroxy-morpholin-4-yl)-ethoxy]-naphthalen-1-yl}-urea;
1-[5-(2-hydroxy-1,1-dimethyl-ethyl)-2-(6-methyl-pyridin-3-yl)-2H-pyrazol-3-yl]-3-[4-(2-pyridin-4-yl-ethoxy)-naphthalen-1-yl]-urea;
1-[5-tert-butyl)-2-(6-methyl-pyridin-3-yl)-2H-pyrazol-3-yl]-3-[4-(2-hydroxy-2-pyridin-4-yl-ethoxy)-naphthalen-1-yl]-urea;
1-[5-tert-butyl)-2-(6-methyl-pyridin-3-yl)-2H-pyrazol-3-yl]-3-{4-[2-(1-hydroxy-pyridin-4-yl)-ethoxy]-naphthalen-1-yl}-urea;
1-[5-(2-hydroxy-1,1-dimethyl-ethyl)-2-p-tolyl-2H-pyrazol-3-yl]- 3-{4-[2-(1 -oxo-thiomorpholin-4-yl)-ethoxy]-naphthalen-1-yl}-urea;
1-[5-tert-butyl-2-(4-hydroxymethyl-phenyl)-2H-pyrazol-3-yl]- 3-{4-[2-(1-oxo-thiomorpholin-4-yl)-ethoxy]-naphthalen-1-yl}-urea;
1-[5-tert-butyl-2-p-tolyl-2H-pyrazol-3-yl]- 3-{4-[2-(1,3 dioxo-thiomorpholin-4-yl)-ethoxy]-naphthalen-1-yl}-urea;
1-[5-(2-hydroxy-1,1-dimethyl-ethyl)-2-methyl-2H-pyrazol-3-yl]-3-[4-(2-morpholin-4-yl-ethoxy)-naphthalen-1-yl]-urea and
1-[5-tert-butyl-2-methyl-2H-pyrazol-3-yl]-3-{4-[2-(4-hydroxy-morpholin-4-yl)-ethoxy]-naphthalen-1-yl}-urea.
12. A method of treating a cytokine mediated disease or condition which comprises administering to a patient in need of such treatment a therapeutically effective amount of a metabolite compound according to claim 1.
13. The method according to claim 12 wherein the cytokine mediated disease or condition is selected from the groups consisting of rheumatoid arthritis, multiple sclerosis, Guillain-Barre syndrome, Crohn's disease, ulcerative colitis, psoriasis, graft versus host disease, systemic lupus erythematosus and insulin-dependent diabetes mellitus, toxic shock syndrome, osteoarthritis, diabetes, inflammatory bowel diseases, acute and chronic pain, contact dermatitis, atherosclerosis, glomerulonephritis, reperfusion injury, bone resorption diseases, asthma, stroke, myocardial infarction, thermal injury, adult respiratory distress syndrome (ARDS), multiple organ injury secondary to trauma, dermatoses with acute inflammatory components, acute purulent meningitis, necrotizing entrerocolitis and syndromes associated with hemodialysis, septic shock, leukopherisis and granulocyte transfusion
14. The method according to claim 13 wherein the cytokine mediated disease or condition is selected from the groups consisting of as rheumatoid arthritis, Crohn's disease and psoriasis.
15. A pharmnaceutical composition comprising a metabolite compound according to claim 1 or the pharmaceutically acceptable derivatives thereof.
US09/879,776 1999-01-19 2001-06-12 Aromatic heterocyclic compounds as antiinflammatory agents Abandoned US20020058678A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US09/879,776 US20020058678A1 (en) 1999-01-19 2001-06-12 Aromatic heterocyclic compounds as antiinflammatory agents
US10/165,372 US6525046B1 (en) 2000-01-18 2002-06-07 Aromatic heterocyclic compounds as antiinflammatory agents

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US11640099P 1999-01-19 1999-01-19
US09/484,638 US6319921B1 (en) 1999-01-19 2000-01-18 Aromatic heterocyclic compound as antiinflammatory agents
US09/879,776 US20020058678A1 (en) 1999-01-19 2001-06-12 Aromatic heterocyclic compounds as antiinflammatory agents

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US09/484,638 Division US6319921B1 (en) 1999-01-19 2000-01-18 Aromatic heterocyclic compound as antiinflammatory agents

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US10/165,372 Continuation-In-Part US6525046B1 (en) 2000-01-18 2002-06-07 Aromatic heterocyclic compounds as antiinflammatory agents

Publications (1)

Publication Number Publication Date
US20020058678A1 true US20020058678A1 (en) 2002-05-16

Family

ID=22366966

Family Applications (6)

Application Number Title Priority Date Filing Date
US09/484,638 Expired - Lifetime US6319921B1 (en) 1999-01-19 2000-01-18 Aromatic heterocyclic compound as antiinflammatory agents
US09/871,559 Expired - Lifetime US6333325B1 (en) 1999-01-19 2001-05-31 Method of treating cytokine mediated diseases or conditions
US09/879,776 Abandoned US20020058678A1 (en) 1999-01-19 2001-06-12 Aromatic heterocyclic compounds as antiinflammatory agents
US09/891,820 Expired - Lifetime US6506748B2 (en) 1999-01-19 2001-06-26 Aromatic heterocyclic compounds as antiinflammatory agents
US09/891,579 Expired - Lifetime US6329415B1 (en) 1999-01-19 2001-06-26 Aromatic heterocyclic compounds as antiinflammatory agents
US09/920,899 Expired - Lifetime US6372773B1 (en) 1999-01-19 2001-08-02 Aromatic heterocyclic compounds as antiinflammatory agents

Family Applications Before (2)

Application Number Title Priority Date Filing Date
US09/484,638 Expired - Lifetime US6319921B1 (en) 1999-01-19 2000-01-18 Aromatic heterocyclic compound as antiinflammatory agents
US09/871,559 Expired - Lifetime US6333325B1 (en) 1999-01-19 2001-05-31 Method of treating cytokine mediated diseases or conditions

Family Applications After (3)

Application Number Title Priority Date Filing Date
US09/891,820 Expired - Lifetime US6506748B2 (en) 1999-01-19 2001-06-26 Aromatic heterocyclic compounds as antiinflammatory agents
US09/891,579 Expired - Lifetime US6329415B1 (en) 1999-01-19 2001-06-26 Aromatic heterocyclic compounds as antiinflammatory agents
US09/920,899 Expired - Lifetime US6372773B1 (en) 1999-01-19 2001-08-02 Aromatic heterocyclic compounds as antiinflammatory agents

Country Status (33)

Country Link
US (6) US6319921B1 (en)
EP (1) EP1147104B1 (en)
JP (1) JP3793694B2 (en)
KR (1) KR100669839B1 (en)
CN (1) CN1333767A (en)
AR (1) AR034089A1 (en)
AT (1) ATE549328T1 (en)
AU (1) AU770581B2 (en)
BG (1) BG64971B1 (en)
BR (1) BR9916930A (en)
CA (1) CA2352524C (en)
CO (1) CO5150218A1 (en)
CZ (1) CZ20012635A3 (en)
EE (1) EE04527B1 (en)
HK (1) HK1041484A1 (en)
HR (1) HRP20010516A2 (en)
HU (1) HUP0201406A3 (en)
IL (2) IL143394A0 (en)
MX (1) MXPA01005628A (en)
MY (1) MY128063A (en)
NO (1) NO20013559D0 (en)
NZ (1) NZ513525A (en)
PE (1) PE20001399A1 (en)
PL (1) PL351764A1 (en)
RU (1) RU2220142C2 (en)
SA (1) SA00210018B1 (en)
SK (1) SK10192001A3 (en)
TR (1) TR200102072T2 (en)
TW (1) TW546297B (en)
UA (1) UA73492C2 (en)
WO (1) WO2000043384A1 (en)
YU (1) YU39101A (en)
ZA (1) ZA200104656B (en)

Cited By (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040180906A1 (en) * 2002-12-31 2004-09-16 Flynn Daniel L Anti-inflammatory medicaments
US20080261965A1 (en) * 2007-04-20 2008-10-23 Deciphera Pharmaceuticals, Llc Kinase inhibitors useful for the treatment of myleoprolific diseases and other proliferative diseases
US20090312349A1 (en) * 2004-12-23 2009-12-17 Deciphera Pharmaceuticals, Llc Anti-inflammatory medicaments
US20100120806A1 (en) * 2008-10-29 2010-05-13 Flynn Daniel L Cyclopropane amides and analogs exhibiting anti-cancer and anti-proliferative activities
US7790756B2 (en) 2006-10-11 2010-09-07 Deciphera Pharmaceuticals, Llc Kinase inhibitors useful for the treatment of myleoproliferative diseases and other proliferative diseases
US20110189167A1 (en) * 2007-04-20 2011-08-04 Flynn Daniel L Methods and Compositions for the Treatment of Myeloproliferative Diseases and other Proliferative Diseases
US8163756B2 (en) 2004-12-23 2012-04-24 Deciphera Pharmaceuticals, Llc Enzyme modulators and treatments
US8188113B2 (en) 2006-09-14 2012-05-29 Deciphera Pharmaceuticals, Inc. Dihydropyridopyrimidinyl, dihydronaphthyidinyl and related compounds useful as kinase inhibitors for the treatment of proliferative diseases
US8940756B2 (en) 2012-06-07 2015-01-27 Deciphera Pharmaceuticals, Llc Dihydronaphthyridines and related compounds useful as kinase inhibitors for the treatment of proliferative diseases
US10927079B2 (en) 2017-04-27 2021-02-23 Mochida Pharmaceutical Co., Ltd. Intermediate compound of novel tetrahydronaphthyl urea derivative
US10966966B2 (en) 2019-08-12 2021-04-06 Deciphera Pharmaceuticals, Llc Methods of treating gastrointestinal stromal tumors
US11185535B2 (en) 2019-12-30 2021-11-30 Deciphera Pharmaceuticals, Llc Amorphous kinase inhibitor formulations and methods of use thereof
US11266635B2 (en) 2019-08-12 2022-03-08 Deciphera Pharmaceuticals, Llc Methods of treating gastrointestinal stromal tumors
US11395818B2 (en) 2019-12-30 2022-07-26 Deciphera Pharmaceuticals, Llc Compositions of 1-(4-bromo-5-(1-ethyl-7-(methylamino)-2-oxo-1,2-dihydro-1,6-naphthyridin-3-yl)-2-fluorophenyl)-3-phenylurea
US11779572B1 (en) 2022-09-02 2023-10-10 Deciphera Pharmaceuticals, Llc Methods of treating gastrointestinal stromal tumors
US11969414B2 (en) 2022-07-20 2024-04-30 Deciphera Pharmaceuticals, Llc Methods of treating gastrointestinal stromal tumors

Families Citing this family (182)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7301021B2 (en) 1997-07-02 2007-11-27 Smithkline Beecham Corporation Substituted imidazole compounds
US7329670B1 (en) * 1997-12-22 2008-02-12 Bayer Pharmaceuticals Corporation Inhibition of RAF kinase using aryl and heteroaryl substituted heterocyclic ureas
US7517880B2 (en) * 1997-12-22 2009-04-14 Bayer Pharmaceuticals Corporation Inhibition of p38 kinase using symmetrical and unsymmetrical diphenyl ureas
EP1112070B1 (en) 1998-08-20 2004-05-12 Smithkline Beecham Corporation Novel substituted triazole compounds
US6525069B1 (en) 1998-12-18 2003-02-25 Bristol-Myers Squibb Pharma Co. N-ureidoalkyl-piperidines as modulators of chemokine receptor activity
RU2319693C9 (en) * 1999-01-13 2008-08-20 Байер Копэрейшн Derivatives of urea (variants), pharmaceutical composition (variants) and method for treatment of diseases associated with cancer cells growth (variants)
WO2000041698A1 (en) 1999-01-13 2000-07-20 Bayer Corporation φ-CARBOXY ARYL SUBSTITUTED DIPHENYL UREAS AS p38 KINASE INHIBITORS
US8124630B2 (en) 1999-01-13 2012-02-28 Bayer Healthcare Llc ω-carboxyaryl substituted diphenyl ureas as raf kinase inhibitors
WO2000042012A1 (en) * 1999-01-13 2000-07-20 Bayer Corporation φ-CARBOXYARYL SUBSTITUTED DIPHENYL UREAS AS RAF KINASE INHIBITORS
JP2002537397A (en) 1999-02-22 2002-11-05 ベーリンガー インゲルハイム ファーマシューティカルズ インコーポレイテッド Polycyclic heterocyclic derivatives as anti-inflammatory agents
DE60023853T2 (en) 1999-03-12 2006-05-24 Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield AROMATIC HETEROCYCLIC COMPOUNDS OF ANTI-INFLAMMATORY ACTIVE SUBSTANCES
IL144897A0 (en) 1999-03-12 2002-06-30 Boehringer Ingelheim Pharma Compounds useful as anti-inflammatory agents
JP4162406B2 (en) 1999-07-09 2008-10-08 ベーリンガー インゲルハイム ファーマシューティカルズ インコーポレイテッド Novel synthesis method of heteroaryl-substituted urea compounds
US6759410B1 (en) * 1999-11-23 2004-07-06 Smithline Beecham Corporation 3,4-dihydro-(1H)-quinazolin-2-ones and their use as CSBP/p38 kinase inhibitors
US6525046B1 (en) 2000-01-18 2003-02-25 Boehringer Ingelheim Pharmaceuticals, Inc. Aromatic heterocyclic compounds as antiinflammatory agents
US6608052B2 (en) 2000-02-16 2003-08-19 Boehringer Ingelheim Pharmaceuticals, Inc. Compounds useful as anti-inflammatory agents
WO2001098270A2 (en) * 2000-06-21 2001-12-27 Bristol-Myers Squibb Pharma Company N-ureidoalkyl-piperidines as modulators of chemokine receptor activity
US6565880B2 (en) 2000-07-24 2003-05-20 Boehringer Ingelheim Pharmaceuticals, Inc. Oral dosage formulations of 1-(5-tert-butyl-2-p-tolyl-2H-pyrazol-3-yl)-3-[4-(2-morpholin-4-yl-ethoxy)-naphthalen-1-yl]-urea
ATE355275T1 (en) 2000-10-20 2006-03-15 Eisai R&D Man Co Ltd NITROGEN CONTAINING AROMATIC RING COMPOUNDS FOR THE TREATMENT OF TUMOR DISEASES
JP2004518739A (en) * 2001-02-15 2004-06-24 ベーリンガー インゲルハイム ファーマシューティカルズ インコーポレイテッド Method for synthesizing heteroaryl-substituted urea compounds useful as anti-inflammatory agents
JP2004530690A (en) * 2001-05-16 2004-10-07 ベーリンガー インゲルハイム ファーマシューティカルズ インコーポレイテッド Diaryl urea derivatives useful as anti-inflammatory drugs
MXPA03010724A (en) 2001-05-25 2004-03-02 Boehringer Ingelheim Pharma Carbamate and oxamide compounds as inhibitors of cytokine production.
ATE338035T1 (en) 2001-06-05 2006-09-15 Boehringer Ingelheim Pharma 1,4-DISUBSTITUTED BENZOCONDENSED CYCLOALKYL UREA COMPOUNDS
EP1709965A3 (en) 2001-07-11 2006-12-27 Boehringer Ingelheim Pharmaceuticals, Inc. Methods of treating cytokine mediate diseases
ES2284887T3 (en) * 2001-07-11 2007-11-16 Boehringer Ingelheim Pharmaceuticals Inc. METHODS TO TREAT DISEASES TRANSMITTED BY CITOCINES.
US6808721B2 (en) * 2001-07-11 2004-10-26 Boehringer Ingelheim Pharmaceuticals, Inc. Methods for coating pharmaceutical core tablets
CA2454913A1 (en) * 2001-08-20 2003-02-27 Boehringer Ingelheim Pharmaceuticals, Inc. Parenteral formulations of 1-(5-tert-butyl-2-p-tolyl-2h-pryrazol-3-yl)-3-¬4-(2-morpholin-4-yl-ethoxy)-naphthalen-1-yl|-urea and a cyclodextrin
CN1315781C (en) * 2001-08-31 2007-05-16 拜尔化学品股份公司 Preparation of 4-amino-1-naphthol ethers
JP2005503400A (en) * 2001-09-13 2005-02-03 ベーリンガー インゲルハイム ファーマシューティカルズ インコーポレイテッド Method for treating cytokine-mediated diseases
JP2005506350A (en) 2001-10-18 2005-03-03 ベーリンガー インゲルハイム ファーマシューティカルズ インコーポレイテッド 1,4-Disubstituted benzo-condensed urea compounds as cytokine inhibitors
US20030118575A1 (en) * 2001-12-11 2003-06-26 Boehringer Ingelheim Pharmaceuticals, Inc. Method for administering BIRB 796 BS
US20040023961A1 (en) * 2002-02-11 2004-02-05 Bayer Corporation Aryl ureas with raf kinase and angiogenisis inhibiting activity
PT1478358E (en) 2002-02-11 2013-09-11 Bayer Healthcare Llc Sorafenib tosylate for the treatment of diseases characterized by abnormal angiogenesis
CN1318404C (en) 2002-02-11 2007-05-30 拜耳制药公司 Aryl ureas as kinase inhibitors
AU2003209119A1 (en) 2002-02-11 2003-09-04 Bayer Pharmaceuticals Corporation Pyridine, quinoline, and isoquinoline n-oxides as kinase inhibitors
EP1480973B1 (en) * 2002-02-25 2008-02-13 Boehringer Ingelheim Pharmaceuticals Inc. 1,4-disubstituted benzofused cycloalkyl urea compounds useful in treating cytokine mediated diseases
US20030225089A1 (en) * 2002-04-10 2003-12-04 Boehringer Ingelheim Pharma Gmbh & Co. Kg Pharmaceutical compositions based on anticholinergics and p38 kinase inhibitors
US20030236287A1 (en) * 2002-05-03 2003-12-25 Piotrowski David W. Positive allosteric modulators of the nicotinic acetylcholine receptor
US20040044020A1 (en) * 2002-07-09 2004-03-04 Boehringer Ingelheim Pharma Gmbh & Co. Kg Pharmaceutical compositions based on novel anticholinergics and p38 kinase inhibitors
DE60310730T2 (en) 2002-07-09 2007-05-16 Boehringer Ingelheim Pharma Gmbh & Co. Kg PHARMACEUTICAL COMPOSITIONS OF ANTICHOLINERGICA AND P38 KINASE INHIBITORS FOR THE TREATMENT OF RESPIRATORY DISEASES
JP2006504667A (en) * 2002-08-08 2006-02-09 ベーリンガー インゲルハイム ファーマシューティカルズ インコーポレイテッド Fluorinated phenyl-naphthalenyl-urea compounds as inhibitors of cytokines involved in inflammatory processes
US7232828B2 (en) * 2002-08-10 2007-06-19 Bethesda Pharmaceuticals, Inc. PPAR Ligands that do not cause fluid retention, edema or congestive heart failure
US20040110755A1 (en) * 2002-08-13 2004-06-10 Boehringer Ingelheim Pharmaceuticals, Inc. Combination therapy with p38 MAP kinase inhibitors and their pharmaceutical compositions
US20040033222A1 (en) * 2002-08-14 2004-02-19 Boehringer Ingelheim Pharmaceuticals, Inc. Anticoagulant and fibrinolytic therapy uning p38 MAP kinase inhibitors
EP1567507B1 (en) * 2002-11-27 2006-06-14 Boehringer Ingelheim Pharmaceuticals Inc. 1,2,3- triazole amide derivatives as cytokine inhibitors
US20040138216A1 (en) * 2002-12-23 2004-07-15 Boehringer Ingelheim Pharma Gmbh & Co. Kg Process for the preparation of an essentially pure polymorph of an n-pyrazolyl-n'-naphthyl-urea
US7279576B2 (en) 2002-12-31 2007-10-09 Deciphera Pharmaceuticals, Llc Anti-cancer medicaments
US20080045706A1 (en) * 2002-12-31 2008-02-21 Flynn Daniel L Anti-inflammatory medicaments
US7202257B2 (en) 2003-12-24 2007-04-10 Deciphera Pharmaceuticals, Llc Anti-inflammatory medicaments
NZ750285A (en) * 2003-02-21 2020-08-28 ResMed Pty Ltd Nasal assembly
US7557129B2 (en) 2003-02-28 2009-07-07 Bayer Healthcare Llc Cyanopyridine derivatives useful in the treatment of cancer and other disorders
JP4594925B2 (en) * 2003-02-28 2010-12-08 ニッポネックス インコーポレイテッド 2-Oxo-1,3,5-perhydrotriazapine derivatives useful for the treatment of hyperproliferative, angiogenic and inflammatory diseases
US7135575B2 (en) 2003-03-03 2006-11-14 Array Biopharma, Inc. P38 inhibitors and methods of use thereof
GB0320244D0 (en) 2003-05-06 2003-10-01 Aventis Pharma Inc Pyrazoles as inhibitors of tumour necrosis factor
EP2390262A1 (en) 2003-05-16 2011-11-30 Intermune, Inc. Synthetic chemokine receptor ligands and methods of use thereof
JPWO2004101529A1 (en) * 2003-05-19 2006-07-13 小野薬品工業株式会社 Nitrogen-containing heterocyclic compound and pharmaceutical use thereof
PL1626714T3 (en) 2003-05-20 2007-12-31 Bayer Healthcare Llc Diaryl ureas for diseases mediated by pdgfr
US7405210B2 (en) 2003-05-21 2008-07-29 Osi Pharmaceuticals, Inc. Pyrrolopyridine-2-carboxylic acid amide inhibitors of glycogen phosphorylase
WO2004113352A1 (en) * 2003-06-19 2004-12-29 Amedis Pharmaceuticals Ltd. Silylated heterocyclylurea derivatives as cytokine-inhibitors
TWI372050B (en) 2003-07-03 2012-09-11 Astex Therapeutics Ltd (morpholin-4-ylmethyl-1h-benzimidazol-2-yl)-1h-pyrazoles
CN1856469B (en) 2003-07-23 2013-03-06 拜耳医药保健有限责任公司 Fluoro substituted omega-carboxyaryl diphenyl urea for the treatment and prevention of diseases and conditions
CN1838958A (en) * 2003-08-22 2006-09-27 贝林格尔·英格海姆药物公司 Methods of treating COPD and pulmonary hypertension
JP4895811B2 (en) 2003-09-11 2012-03-14 ケミア,インコーポレイテッド Cytokine inhibitor
WO2005044788A1 (en) 2003-11-11 2005-05-19 Eisai Co., Ltd. Urea derivative and process for producing the same
US7767670B2 (en) 2003-11-13 2010-08-03 Ambit Biosciences Corporation Substituted 3-carboxamido isoxazoles as kinase modulators
WO2005063715A1 (en) * 2003-12-18 2005-07-14 Boehringer Ingelheim Pharmaceuticals, Inc. Polymorph of birb 796, a p38map kinase inhibitor
US20070191336A1 (en) * 2003-12-24 2007-08-16 Flynn Daniel L Anti-inflammatory medicaments
ES2353309T3 (en) 2004-03-08 2011-03-01 Prosidion Ltd. HYDRAZIDS OF THE PIRROLOPIRIDIN-2-CARBOXYL ACID AS INHIBITORS OF GLUCOGEN PHOSPHORILASE.
CN101010315A (en) * 2004-04-30 2007-08-01 拜耳制药公司 Substituted pyrazolyl urea derivatives useful in the treatment of cancer
EP1609789A1 (en) * 2004-06-23 2005-12-28 Eli Lilly And Company Ureido-pyrazole derivatives and their use as kinase inhibitors
ES2308523T3 (en) 2004-06-23 2008-12-01 Eli Lilly And Company QUINASE INHIBITORS.
US20060035893A1 (en) 2004-08-07 2006-02-16 Boehringer Ingelheim International Gmbh Pharmaceutical compositions for treatment of respiratory and gastrointestinal disorders
US7597884B2 (en) 2004-08-09 2009-10-06 Alios Biopharma, Inc. Hyperglycosylated polypeptide variants and methods of use
AU2005273612B2 (en) 2004-08-12 2010-10-14 Pfizer Inc. Triazolopyridinylsulfanyl derivatives as p38 map kinase inhibitors
AU2005283422C1 (en) 2004-09-17 2017-02-02 Eisai R & D Management Co., Ltd. Medicinal composition
DE102004055633A1 (en) * 2004-11-12 2006-05-18 Schering Ag New 5-substituted quinoline and isoquinoline derivatives are glucocorticoid receptor binders useful for treating inflammatory diseases e.g. alveolitis, Sjogren's syndrome, atopic dermatitis, acute lymphatic leukemia, rhinitis and emesis
US20080009519A1 (en) * 2004-11-17 2008-01-10 Lawrence Steinman Method of modulating t cell functioning
BRPI0518432A2 (en) * 2004-11-17 2008-11-25 Angiogen Pharmaceuticals Pty L Method of Modulating Cell B Operation
US20090099176A1 (en) 2004-12-02 2009-04-16 Thomas Martin Krulle Pyrrolopyridine-2-carboxylic acid amides
EP1824843A2 (en) 2004-12-07 2007-08-29 Locus Pharmaceuticals, Inc. Inhibitors of protein kinases
WO2006062982A2 (en) 2004-12-07 2006-06-15 Locus Pharmaceuticals, Inc. Urea inhibitors of map kinases
PE20060777A1 (en) 2004-12-24 2006-10-06 Boehringer Ingelheim Int INDOLINONE DERIVATIVES FOR THE TREATMENT OR PREVENTION OF FIBROTIC DISEASES
CA2593993C (en) 2004-12-30 2014-07-29 Astex Therapeutics Limited Pyrazole compounds that modulate the activity of cdk, gsk and aurora kinases
JP2008540526A (en) * 2005-05-12 2008-11-20 ベーリンガー インゲルハイム インターナショナル ゲゼルシャフト ミット ベシュレンクテル ハフツング Bisamination of aryl halides
EP1925676A4 (en) 2005-08-02 2010-11-10 Eisai R&D Man Co Ltd Method for assay on the effect of vascularization inhibitor
WO2007044490A2 (en) * 2005-10-06 2007-04-19 Boehringer Ingelheim International Gmbh Process for making heteroaryl amine intermediate compounds
TW200800194A (en) 2005-10-28 2008-01-01 Lilly Co Eli Kinase inhibitors
GB2431927B (en) 2005-11-04 2010-03-17 Amira Pharmaceuticals Inc 5-Lipoxygenase-activating protein (FLAP) inhibitors
US7977359B2 (en) 2005-11-04 2011-07-12 Amira Pharmaceuticals, Inc. 5-lipdxygenase-activating protein (FLAP) inhibitors
US8399666B2 (en) 2005-11-04 2013-03-19 Panmira Pharmaceuticals, Llc 5-lipoxygenase-activating protein (FLAP) inhibitors
US20110195110A1 (en) * 2005-12-01 2011-08-11 Roger Smith Urea compounds useful in the treatment of cancer
EP1968579A1 (en) 2005-12-30 2008-09-17 Astex Therapeutics Limited Pharmaceutical compounds
EP1973408A2 (en) * 2006-01-04 2008-10-01 Locus Pharmaceuticals, Inc. Inhibitors of protein kinases
DK1981851T3 (en) 2006-01-31 2012-03-19 Array Biopharma Inc KINASE INHIBITORS AND PROCEDURES FOR USE THEREOF
CA2640665A1 (en) * 2006-02-10 2007-08-16 Pfizer Products Inc. Pyridinone pyrazole urea and pyrimidinone pyrazole urea derivatives
JP5273038B2 (en) 2006-05-08 2013-08-28 アリアド・ファーマシューティカルズ・インコーポレイテッド Monocyclic heteroaryl compound
WO2007133560A2 (en) 2006-05-08 2007-11-22 Ariad Pharmaceuticals, Inc. Acetylenic heteroaryl compounds
CN101443009A (en) 2006-05-18 2009-05-27 卫材R&D管理有限公司 Antitumor agent for thyroid cancer
EP2049119A2 (en) 2006-06-29 2009-04-22 Astex Therapeutics Limited Pharmaceutical combinations of 1-cyclopropyl-3-[3-(5-morphoolin-4-ylmethyl-1h-benzoimidazol-2-yl)-1h-1-pyrazol-4-yl]-urea
EP2065372B1 (en) 2006-08-28 2012-11-28 Eisai R&D Management Co., Ltd. Antitumor agent for undifferentiated gastric cancer
US7737149B2 (en) * 2006-12-21 2010-06-15 Astrazeneca Ab N-[5-[2-(3,5-dimethoxyphenyl)ethyl]-2H-pyrazol-3-yl]-4-(3,5-dimethylpiperazin-1-yl)benzamide and salts thereof
WO2008093855A1 (en) 2007-01-29 2008-08-07 Eisai R & D Management Co., Ltd. Composition for treatment of undifferentiated-type of gastric cancer
CN101636397B (en) * 2007-04-13 2012-06-13 中国人民解放军军事医学科学院毒物药物研究所 Urea compounds, preparation methods and pharmaceutical uses thereof
EP1992344A1 (en) 2007-05-18 2008-11-19 Institut Curie P38 alpha as a therapeutic target in pathologies linked to FGFR3 mutation
US7868001B2 (en) * 2007-11-02 2011-01-11 Hutchison Medipharma Enterprises Limited Cytokine inhibitors
CA2704000C (en) 2007-11-09 2016-12-13 Eisai R&D Management Co., Ltd. Combination of anti-angiogenic substance and anti-tumor platinum complex
EP2111861A1 (en) 2008-04-21 2009-10-28 Ranbaxy Laboratories Limited Compositions of phosphodiesterase type IV inhibitors
CN102137858B (en) 2008-05-23 2014-07-23 潘米拉制药有限责任公司 5-lipoxygenase-activating protein inhibitor
US8637575B2 (en) 2008-06-05 2014-01-28 Novotyr Therapeutics Ltd. Modulators of protein kinase signaling
US8293748B2 (en) 2008-10-02 2012-10-23 Respivert Ltd. p38 MAP kinase inhibitors
GB0818033D0 (en) 2008-10-02 2008-11-05 Respivert Ltd Novel compound
FR2937973B1 (en) * 2008-11-04 2010-11-05 Galderma Res & Dev MELANOCORTIN RECEPTOR MODULATORS, PROCESS FOR THEIR PREPARATION AND THEIR USE IN HUMAN MEDICINE AND COSMETICS
WO2010056311A1 (en) 2008-11-12 2010-05-20 Ariad Pharmaceuticals, Inc. Pyrazinopyrazines and derivatives as kinase inhibitors
BRPI0922962A2 (en) 2008-12-11 2019-09-24 Respivert Ltd p38 map kinase inhibitors
MX2011010105A (en) 2009-03-27 2012-01-12 Pathway Therapeutics Inc Pyrimidinyl and 1,3,5-triazinyl benzimidazole sulfonamides and their use in cancer therapy.
GB0905955D0 (en) * 2009-04-06 2009-05-20 Respivert Ltd Novel compounds
JP5573677B2 (en) 2009-09-30 2014-08-20 東レ株式会社 2,3-Dihydro-1H-inden-2-ylurea derivative and pharmaceutical use thereof
GB0921730D0 (en) 2009-12-11 2010-01-27 Respivert Ltd Method of treatment
GB0921731D0 (en) 2009-12-11 2010-01-27 Respivert Ltd Theraputic uses
GB201005589D0 (en) 2010-04-01 2010-05-19 Respivert Ltd Novel compounds
FR2993780B1 (en) * 2012-07-26 2015-02-13 Assist Publ Hopitaux De Paris METHOD OF TREATING PLATE SCLEROSIS
JP5787977B2 (en) 2010-04-08 2015-09-30 レスピバート・リミテツド P38 MAP kinase inhibitor
US9260410B2 (en) 2010-04-08 2016-02-16 Respivert Ltd. P38 MAP kinase inhibitors
WO2011150198A1 (en) 2010-05-27 2011-12-01 Ambit Biosciences Corporation Azolyl urea compounds and methods of use thereof
GB201009731D0 (en) * 2010-06-10 2010-07-21 Pulmagen Therapeutics Inflamma Kinase inhibitors
WO2011158044A2 (en) 2010-06-17 2011-12-22 Respivert Limited Respiratory formulations and compounds for use therein
ES2573515T3 (en) 2010-06-25 2016-06-08 Eisai R&D Management Co., Ltd. Anti-tumor agent that uses compounds with combined kinase inhibitory effect
US9090592B2 (en) * 2010-12-30 2015-07-28 AbbVie Deutschland GmbH & Co. KG Heterocyclic compounds and their use as glycogen synthase kinase-3 inhibitors
RU2580609C2 (en) 2011-04-18 2016-04-10 Эйсай Ар Энд Ди Менеджмент Ко., Лтд. Anticancer therapeutic agent
LT2712358T (en) 2011-05-13 2017-01-25 Array Biopharma, Inc. Pyrrolidinyl urea, pyrrolidinyl thiourea and pyrrolidinyl guanidine compounds as trka kinase inhibitors
ES2841809T3 (en) 2011-06-03 2021-07-09 Eisai R&D Man Co Ltd Biomarkers to predict and evaluate the degree of response of subjects with thyroid and kidney cancer to lenvatinib compounds
PT2763984T (en) 2011-10-03 2016-07-25 Respivert Ltd 1-pyrazolyl-3-(4-((2-anilinopyrimidin-4-yl)oxy)napththalen-1-yl)ureas as p38 map kinase inhibitors
EP2578582A1 (en) 2011-10-03 2013-04-10 Respivert Limited 1-Pyrazolyl-3-(4-((2-anilinopyrimidin-4-yl)oxy)napththalen-1-yl)ureas as p38 MAP kinase inhibitors
CA2853279C (en) 2011-10-24 2021-03-23 Endo Pharmaceuticals Inc. Cyclohexylamines
CN103987708B (en) * 2011-12-09 2016-06-22 奇斯药制品公司 The derivant of 4-hydroxyl-1,2,3,4-naphthane-1-base urea and the purposes in treatment of respiratory diseases thereof
EP2799537B1 (en) 2011-12-28 2021-09-22 Kyoto Prefectural Public University Corporation Normalization of culture of corneal endothelial cells
GB201214750D0 (en) 2012-08-17 2012-10-03 Respivert Ltd Compounds
EP2890460B1 (en) 2012-08-29 2017-02-22 Respivert Limited Kinase inhibitors
US9783556B2 (en) 2012-08-29 2017-10-10 Respivert Limited Kinase inhibitors
WO2014033447A2 (en) 2012-08-29 2014-03-06 Respivert Limited Kinase inhibitors
GB201215357D0 (en) 2012-08-29 2012-10-10 Respivert Ltd Compounds
WO2014078328A1 (en) 2012-11-13 2014-05-22 Array Biopharma Inc. N-bicyclic aryl,n'-pyrazolyl urea, thiourea, guanidine and cyanoguanidine compounds as trka kinase inhibitors
NZ708028A (en) 2012-11-13 2018-12-21 Array Biopharma Inc N-pyrrolidinyl, n’-pyrazolyl- urea, thiourea, guanidine and cyanoguanidine compounds as trka kinase inhibitors
DK2920166T3 (en) 2012-11-13 2017-02-06 Array Biopharma Inc SUITABLE BICYCLIC UREA, THIOUREA, GUANIDINE AND CYANOGUANIDE COMPOUNDS FOR PAIN TREATMENT
US9828360B2 (en) 2012-11-13 2017-11-28 Array Biopharma Inc. Pyrrolidinyl urea, thiourea, guanidine and cyanoguanidine compounds as TrkA kinase inhibitors
WO2014078325A1 (en) 2012-11-13 2014-05-22 Array Biopharma Inc. N-(monocyclic aryl),n'-pyrazolyl-urea, thiourea, guanidine and cyanoguanidine compounds as trka kinase inhibitors
US9969694B2 (en) 2012-11-13 2018-05-15 Array Biopharma Inc. N-(arylalkyl)-N′-pyrazolyl-urea, thiourea, guanidine and cyanoguanidine compounds as TrkA kinase inhibitors
US9809578B2 (en) 2012-11-13 2017-11-07 Array Biopharma Inc. Pyrazolyl urea, thiourea, guanidine and cyanoguanidine compounds as trkA kinase inhibitors
WO2014078322A1 (en) 2012-11-13 2014-05-22 Array Biopharma Inc. Thiazolyl and oxazolyl urea, thiourea, guanidine and cyanoguanidine compounds as trka kinase inhibitors
WO2014078372A1 (en) 2012-11-13 2014-05-22 Array Biopharma Inc. Pyrrolidinyl urea, thiourea, guanidine and cyanoguanidine compounds as trka kinase inhibitors
WO2014078408A1 (en) 2012-11-13 2014-05-22 Array Biopharma Inc. Bicyclic heteroaryl urea, thiourea, guanidine and cyanoguanidine compounds as trka kinase inhibitors
US9732063B2 (en) 2012-11-16 2017-08-15 Respivert Limited Kinase inhibitors
AU2013364953A1 (en) 2012-12-21 2015-04-30 Eisai R&D Management Co., Ltd. Amorphous form of quinoline derivative, and method for producing same
EP2970190A1 (en) 2013-03-14 2016-01-20 Respivert Limited Kinase inhibitors
BR112015024671A2 (en) 2013-04-02 2017-07-18 Respivert Ltd urea derivatives useful as kinase inhibitors
EP2981534B1 (en) 2013-04-02 2017-07-19 Topivert Pharma Limited Kinase inhibitors based upon n-alkyl pyrazoles
US10517861B2 (en) 2013-05-14 2019-12-31 Eisai R&D Management Co., Ltd. Biomarkers for predicting and assessing responsiveness of endometrial cancer subjects to lenvatinib compounds
BR112015029504A2 (en) * 2013-06-06 2017-07-25 Chiesi Farm Spa kinase inhibitors
EP3021944B1 (en) 2013-07-14 2018-12-19 Yissum Research Development Company of the Hebrew University of Jerusalem, Ltd. Igf-1r signaling pathway inhibitors useful in the treatment of neurodegenerative diseases
US10813920B2 (en) 2013-11-14 2020-10-27 The Doshisha Drug for treating corneal endothelium by promoting cell proliferation or inhibiting cell damage
JP6514703B2 (en) 2013-12-20 2019-05-15 トピバート ファーマ リミテッド Urea derivatives useful as kinase inhibitors
TWI751099B (en) * 2014-02-14 2022-01-01 英商瑞斯比維特有限公司 Pyrazolyl ureas ask kinase inhibitors, pharmaceutical compositions comprising the same and use thereof
ES2746530T3 (en) 2014-05-15 2020-03-06 Array Biopharma Inc 1 - ((3S, 4R) -4- (3-fluorophenyl) -1- (2-methoxyethyl) pyrrolidin-3-yl) -3- (4-methyl-3- (2-methylpyrimidin-5-yl) - 1-phenyl-1H-pyrazol-5-yl) urea as a TrkA kinase inhibitor
BR112016026556A8 (en) 2014-05-16 2021-07-06 Atriva Therapeutics Gmbh mek, p38 and/or nfkb inhibitors, their uses, composition, in vitro test system, their use, and method for detecting molecules effective in the prophylaxis and/or treatment of a co-infection
HRP20221047T1 (en) 2014-08-28 2022-11-11 Eisai R&D Management Co., Ltd. High-purity quinoline derivative and method for manufacturing same
WO2016030852A1 (en) 2014-08-29 2016-03-03 Torrent Pharmaceuticals Limited P38 map kinase inhibiting indanyl urea compounds
MA40774A (en) 2014-10-01 2017-08-08 Respivert Ltd DIARYLEUREA DERIVATIVES AS KINASE P38 INHIBITORS
CA2975673A1 (en) 2015-02-05 2016-08-11 Tyrnovo Ltd. Combinations of irs/stat3 dual modulators and anti-cancer agents for treating cancer
FI3263106T3 (en) 2015-02-25 2024-01-03 Eisai R&D Man Co Ltd Method for suppressing bitterness of quinoline derivative
CA2978226A1 (en) 2015-03-04 2016-09-09 Merck Sharpe & Dohme Corp. Combination of a pd-1 antagonist and a vegfr/fgfr/ret tyrosine kinase inhibitor for treating cancer
CA2988707C (en) 2015-06-16 2023-10-10 Eisai R&D Management Co., Ltd. Combination of cbp/catenin inhibitor and immune checkpoint inhibitor for treating cancer
WO2017151409A1 (en) 2016-02-29 2017-09-08 University Of Florida Research Foundation, Incorporated Chemotherapeutic methods
EP3440073B1 (en) 2016-04-06 2022-06-08 Oxular Acquisitions Limited Kinase inhibitors
EP3582781A4 (en) 2017-02-15 2020-12-09 The University of Melbourne A method of treatment
US10342786B2 (en) 2017-10-05 2019-07-09 Fulcrum Therapeutics, Inc. P38 kinase inhibitors reduce DUX4 and downstream gene expression for the treatment of FSHD
CN114732910A (en) 2017-10-05 2022-07-12 弗尔康医疗公司 P38 kinase inhibitor reduces DUX4 and downstream gene expression for treatment of FSHD
CN111548313A (en) * 2019-02-11 2020-08-18 绍兴从零医药科技有限公司 Guanidine compounds as medicine for preventing and treating chronic pain
ES2789898B2 (en) 2019-04-25 2021-10-14 Fund Profesor Novoa Santos COMPOSITION FOR THE TREATMENT OF SIDE EFFECTS OF RADIOTHERAPY AND / OR CHEMOTHERAPY
TW202112368A (en) 2019-06-13 2021-04-01 荷蘭商法西歐知識產權股份有限公司 Inhibitor combinations for treatment of diseases related to dux4 expression
CN112961120B (en) * 2021-02-06 2022-02-08 河南省锐达医药科技有限公司 Naphthyl urea compound, preparation method and application thereof
WO2023280911A1 (en) 2021-07-06 2023-01-12 Westfälische Wilhelms-Universität Münster P38-inhibitors for the treatment of coronavirus infections and/or covid-19 cytokine storm
CN114702439B (en) * 2021-12-13 2023-11-10 河南省锐达医药科技有限公司 Naphthyl urea-piperazine compound and preparation method and application thereof

Family Cites Families (48)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE293352C (en)
US4105766A (en) 1977-08-19 1978-08-08 Sterling Drug Inc. 4,5-Dihydro-5-oxopyrazolo[1,5-a]quinazoline-3-carboxylic acid derivatives
HU185294B (en) 1980-12-29 1984-12-28 Chinoin Gyogyszer Es Vegyeszet Process for producing substituted urea derivatives
JPS61228444A (en) 1985-04-02 1986-10-11 Fuji Photo Film Co Ltd Silver halide color photographic sensitive material
EP0272866A1 (en) 1986-12-23 1988-06-29 Merck & Co. Inc. 1,4-Benzodiazepines with 5-membered heterocyclic rings
GB8908869D0 (en) 1989-04-19 1989-06-07 Shell Int Research A process for the preparation of aromatic ureas
WO1991004027A1 (en) 1989-09-15 1991-04-04 Pfizer Inc. New n-aryl and n-heteroarylamide and urea derivatives as inhibitors of acyl coenzyme a: cholesterol acyl transferase (acat)
DD293352A5 (en) 1990-04-06 1991-08-29 Adw,Zi Fuer Organische Chemie,De PROCESS FOR PREPARING 2,2-DISUBSTITUTED 1-ACYL AND / OR 9-ACYL-2,3-DIHYDRO-IMIDAZO / 1,2-A / BENZIMIDAZOLE-3-ONEN
US5162360A (en) * 1991-06-24 1992-11-10 Warner-Lambert Company 2-heteroatom containing urea and thiourea ACAT inhibitors
IL104369A0 (en) 1992-01-13 1993-05-13 Smithkline Beecham Corp Novel compounds and compositions
EP0642498A1 (en) 1992-05-28 1995-03-15 Pfizer Inc. NEW $i(N)-ARYL AND $i(N)-HETEROARYLUREA DERIVATIVES AS INHIBITORS OF ACYL COENZYME A:CHOLESTEROL ACYL TRANSFERASE (ACAT)
US5342942A (en) 1992-06-09 1994-08-30 Warner-Lambert Company Pyrazoloquinazolone derivatives as neurotrophic agents
GB9302275D0 (en) 1993-02-05 1993-03-24 Smithkline Beecham Plc Novel compounds
CA2159344A1 (en) * 1993-03-30 1994-10-13 Minoru Moriwaki Cell adhesion inhibitor and thienotriazolodiazepine compound
PT724588E (en) 1993-09-17 2000-05-31 Smithkline Beecham Corp PROTEIN FROM LIGACAO TO DRUGS
US5783664A (en) 1993-09-17 1998-07-21 Smithkline Beecham Corporation Cytokine suppressive anit-inflammatory drug binding proteins
US5869043A (en) 1993-09-17 1999-02-09 Smithkline Beecham Corporation Drug binding protein
JPH11503110A (en) 1995-02-17 1999-03-23 スミスクライン・ビーチャム・コーポレイション IL-8 receptor antagonist
US5739143A (en) 1995-06-07 1998-04-14 Smithkline Beecham Corporation Imidazole compounds and compositions
IL118544A (en) 1995-06-07 2001-08-08 Smithkline Beecham Corp Imidazole derivatives, process for their preparation and pharmaceutical compositions comprising them
JPH11514651A (en) 1995-10-31 1999-12-14 メルク エンド カンパニー インコーポレーテッド Substituted pyridylpyrroles, compositions containing said compounds and methods of use
US6074862A (en) 1995-12-20 2000-06-13 Signal Pharmaceuticals Inc. Mitogen-activated protein kinase kinase MEK6 and variants thereof
US6096748A (en) 1996-03-13 2000-08-01 Smithkline Beecham Corporation Pyrimidine compounds useful in treating cytokine mediated diseases
JP2000507545A (en) 1996-03-25 2000-06-20 スミスクライン・ビーチャム・コーポレイション Novel treatment for CNS injury
WO1997035856A1 (en) 1996-03-25 1997-10-02 Smithkline Beecham Corporation Novel treatment for cns injuries
US5948885A (en) 1996-05-20 1999-09-07 Signal Pharmaceuticals, Inc. Mitogen-activated protein kinase p38-2 and methods of use therefor
JP2002515744A (en) 1996-05-20 2002-05-28 シグナル ファーマシュウティカルズ,インコーポレイテッド Mitogen-activated protein kinase p38-2 and methods of use
EP0906307B1 (en) 1996-06-10 2005-04-27 Merck & Co., Inc. Substituted imidazoles having cytokine inhibitory activity
JP4373497B2 (en) 1996-06-19 2009-11-25 ローン−プーラン・ロレ・リミテツド Substituted azabicyclo compounds and their use as inhibitors of TNF and cyclic AMP phosphodiesterase production
EP0956018A4 (en) 1996-08-21 2000-01-12 Smithkline Beecham Corp Imidazole compounds, compositions and use
AU727733B2 (en) 1996-10-09 2000-12-21 Medical Research Council Map kinases: polypeptides, polynucleotides and uses thereof
AU5135998A (en) 1996-12-03 1998-06-29 Banyu Pharmaceutical Co., Ltd. Novel urea derivatives
US6147080A (en) 1996-12-18 2000-11-14 Vertex Pharmaceuticals Incorporated Inhibitors of p38
US6376214B1 (en) 1997-02-18 2002-04-23 Smithkline Beecham Corporation DNA encoding a novel homolog of CSBP/p38 MAP kinase
ES2151467T3 (en) * 1997-05-23 2005-03-01 Bayer Corporation ARILURES FOR THE TREATMENT OF INFLAMMATORY OR IMMUNOMODULATING DISEASES.
EP0986382B1 (en) 1997-05-23 2008-06-25 Bayer Pharmaceuticals Corporation Raf kinase inhibitors
US6093742A (en) 1997-06-27 2000-07-25 Vertex Pharmaceuticals, Inc. Inhibitors of p38
US5851812A (en) 1997-07-01 1998-12-22 Tularik Inc. IKK-β proteins, nucleic acids and methods
JP2001521934A (en) * 1997-11-03 2001-11-13 ベーリンガー インゲルハイム ファーマシューティカルズ インコーポレイテッド Aromatic heterocyclic compounds as anti-inflammatory drugs
CA2223075A1 (en) 1997-12-02 1999-06-02 Smithkline Beecham Corporation Drug binding protein
EP1042305B1 (en) 1997-12-22 2005-06-08 Bayer Pharmaceuticals Corp. INHIBITION OF p38 KINASE USING SYMMETRICAL AND UNSYMMETRICAL DIPHENYL UREAS
DE69836563T2 (en) * 1997-12-22 2007-05-16 Bayer Pharmaceuticals Corp., West Haven INHIBITION OF p38 KINASE ACTIVITY THROUGH THE USE OF ARYL AND HETEROARYL SUBSTITUTED UREAS
ATE328873T1 (en) * 1997-12-22 2006-06-15 Bayer Pharmaceuticals Corp INHIBITION OF RAF KINASE USING ARYL- AND HETEROARYL-SUBSTITUTED HETEROCYCLIC UREAS
ATE529109T1 (en) * 1997-12-22 2011-11-15 Bayer Healthcare Llc INHIBITION OF P38 KINASE ACTIVITY BY SUBSTITUTED HETEROCYCLIC UREAS
TR200002618T2 (en) * 1997-12-22 2001-04-20 Bayer Corporation Inhibition of raf kinase using substituted heterocyclic ureas
AU2713799A (en) 1998-03-12 1999-09-27 Novo Nordisk A/S Modulators of protein tyrosine phosphatases (ptpases)
JP2002537397A (en) 1999-02-22 2002-11-05 ベーリンガー インゲルハイム ファーマシューティカルズ インコーポレイテッド Polycyclic heterocyclic derivatives as anti-inflammatory agents
DE60023853T2 (en) * 1999-03-12 2006-05-24 Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield AROMATIC HETEROCYCLIC COMPOUNDS OF ANTI-INFLAMMATORY ACTIVE SUBSTANCES

Cited By (41)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7144911B2 (en) * 2002-12-31 2006-12-05 Deciphera Pharmaceuticals Llc Anti-inflammatory medicaments
US20040180906A1 (en) * 2002-12-31 2004-09-16 Flynn Daniel L Anti-inflammatory medicaments
US7737283B2 (en) 2002-12-31 2010-06-15 Deciphera Pharmaceuticals, Llc Anti-inflammatory medicaments
US20090312349A1 (en) * 2004-12-23 2009-12-17 Deciphera Pharmaceuticals, Llc Anti-inflammatory medicaments
US8163756B2 (en) 2004-12-23 2012-04-24 Deciphera Pharmaceuticals, Llc Enzyme modulators and treatments
US8188113B2 (en) 2006-09-14 2012-05-29 Deciphera Pharmaceuticals, Inc. Dihydropyridopyrimidinyl, dihydronaphthyidinyl and related compounds useful as kinase inhibitors for the treatment of proliferative diseases
US7790756B2 (en) 2006-10-11 2010-09-07 Deciphera Pharmaceuticals, Llc Kinase inhibitors useful for the treatment of myleoproliferative diseases and other proliferative diseases
US20080261965A1 (en) * 2007-04-20 2008-10-23 Deciphera Pharmaceuticals, Llc Kinase inhibitors useful for the treatment of myleoprolific diseases and other proliferative diseases
US20080269267A1 (en) * 2007-04-20 2008-10-30 Deciphera Pharmaceuticals, Llc Kinase inhibitors useful for the treatment of myleoprolific diseases and other proliferative diseases
US20110189167A1 (en) * 2007-04-20 2011-08-04 Flynn Daniel L Methods and Compositions for the Treatment of Myeloproliferative Diseases and other Proliferative Diseases
US8143293B2 (en) 2007-04-20 2012-03-27 Deciphera Pharmaceuticals, Llc Kinase inhibitors useful for the treatment of myleoprolific diseases and other proliferative diseases
US8278331B2 (en) 2008-10-29 2012-10-02 Deciphera Pharmaceuticals, Llc N-acyl ureas exhibiting anti-cancer and anti-proliferative activities
US20100120806A1 (en) * 2008-10-29 2010-05-13 Flynn Daniel L Cyclopropane amides and analogs exhibiting anti-cancer and anti-proliferative activities
USRE48731E1 (en) 2012-06-07 2021-09-14 Deciphera Pharmaceuticals, Llc Dihydronaphthyridines and related compounds useful as kinase inhibitors for the treatment of proliferative diseases
US8940756B2 (en) 2012-06-07 2015-01-27 Deciphera Pharmaceuticals, Llc Dihydronaphthyridines and related compounds useful as kinase inhibitors for the treatment of proliferative diseases
US10927079B2 (en) 2017-04-27 2021-02-23 Mochida Pharmaceutical Co., Ltd. Intermediate compound of novel tetrahydronaphthyl urea derivative
US11266635B2 (en) 2019-08-12 2022-03-08 Deciphera Pharmaceuticals, Llc Methods of treating gastrointestinal stromal tumors
US11813251B2 (en) 2019-08-12 2023-11-14 Deciphera Pharmaceuticals, Llc Methods of treating gastrointestinal stromal tumors
US11576904B2 (en) 2019-08-12 2023-02-14 Deciphera Pharmaceuticals, Llc Methods of treating gastrointestinal stromal tumors
US11344536B1 (en) 2019-08-12 2022-05-31 Deciphera Pharmaceuticals, Llc Methods of treating gastrointestinal stromal tumors
US10966966B2 (en) 2019-08-12 2021-04-06 Deciphera Pharmaceuticals, Llc Methods of treating gastrointestinal stromal tumors
US11426390B2 (en) 2019-08-12 2022-08-30 Deciphera Pharmaceuticals, Llc Methods of treating gastrointestinal stromal tumors
US11433056B1 (en) 2019-08-12 2022-09-06 Deciphera Pharmaceuticals, Llc Methods of treating gastrointestinal stromal tumors
US11529336B2 (en) 2019-08-12 2022-12-20 Deciphera Pharmaceuticals, Llc Methods of treating gastrointestinal stromal tumors
US11534432B2 (en) 2019-08-12 2022-12-27 Deciphera Pharmaceuticals, Llc Methods of treating gastrointestinal stromal tumors
US11395818B2 (en) 2019-12-30 2022-07-26 Deciphera Pharmaceuticals, Llc Compositions of 1-(4-bromo-5-(1-ethyl-7-(methylamino)-2-oxo-1,2-dihydro-1,6-naphthyridin-3-yl)-2-fluorophenyl)-3-phenylurea
US11844788B1 (en) 2019-12-30 2023-12-19 Deciphera Pharmaceuticals, Llc Compositions of 1-(4-bromo-5-(1-ethyl-7-(methylamino)-2-oxo-1,2-dihydro-1,6-naphthyridin-3-yl)-2-fluoropheyl)-3-phenylurea
US11612591B2 (en) 2019-12-30 2023-03-28 Deciphera Pharmaceuticals, Llc Compositions of 1-(4-bromo-5-(1-ethyl-7-(methylamino)-2-oxo-1,2-dihydro-1,6-naphthyridin-3-yl)-2-fluorophenyl)-3-phenylurea
US11918564B1 (en) 2019-12-30 2024-03-05 Deciphera Pharmaceuticals, Llc Compositions of 1-(4-bromo-5-(1-ethyl-7-(methylamino)-2-oxo-1,2-dihydro-1,6-naphthyridin-3-yl)-2-fluoropheyl)-3-phenylurea
US11793795B2 (en) 2019-12-30 2023-10-24 Deciphera Pharmaceuticals, Llc Compositions of 1-(4-bromo-5-(1-ethyl-7-(methylamino)-2-oxo-1,2-dihydro-1,6-naphthyridin-3-yl)-2-fluorophenyl)-3-phenylurea
US11801237B2 (en) 2019-12-30 2023-10-31 Deciphera Pharmaceuticals, Llc Amorphous kinase inhibitor formulations and methods of use thereof
US11185535B2 (en) 2019-12-30 2021-11-30 Deciphera Pharmaceuticals, Llc Amorphous kinase inhibitor formulations and methods of use thereof
US11576903B2 (en) 2019-12-30 2023-02-14 Deciphera Pharmaceuticals, Llc Amorphous kinase inhibitor formulations and methods of use thereof
US11850240B1 (en) 2019-12-30 2023-12-26 Deciphera Pharmaceuticals, Llc Compositions of 1-(4-bromo-5-(1-ethyl-7-(methylamino)-2-oxo-1,2-dihydro-1,6-naphthyridin-3-yl)-2-fluoropheyl)-3-phenylurea
US11850241B1 (en) 2019-12-30 2023-12-26 Deciphera Pharmaceuticals, Llc Compositions of 1-(4-bromo-5-(1-ethyl-7-(methylamino)-2-oxo-1,2-dihydro-1,6-naphthyridin-3-yl)-2-fluoropheyl)-3-phenylurea
US11896585B2 (en) 2019-12-30 2024-02-13 Deciphera Pharmaceuticals, Llc Compositions of 1-(4-bromo-5-(1-ethyl-7-(methylamino)-2-oxo-1,2-dihydro-1,6-naphthyridin-3-yl)-2-fluorophenyl)-3-phenylurea
US11903933B2 (en) 2019-12-30 2024-02-20 Deciphera Pharmaceuticals, Llc Compositions of 1-(4-bromo-5-(1-ethyl-7-(methylamino)-2-oxo-1,2-dihydro-1,6-naphthyridin-3-yl)-2-fluoropheyl)-3-phenylurea
US11911370B1 (en) 2019-12-30 2024-02-27 Deciphera Pharmaceuticals, Llc Compositions of 1-(4-bromo-5-(1-ethyl-7-(methylamino)-2-oxo-1,2-dihydro-1,6-naphthyridin-3-yl)-2-fluoropheyl)-3-phenylurea
US11969414B2 (en) 2022-07-20 2024-04-30 Deciphera Pharmaceuticals, Llc Methods of treating gastrointestinal stromal tumors
US11779572B1 (en) 2022-09-02 2023-10-10 Deciphera Pharmaceuticals, Llc Methods of treating gastrointestinal stromal tumors
US11969415B1 (en) 2023-11-22 2024-04-30 Deciphera Pharmaceuticals, Llc (methylamino)-2-oxo-1,2-dihydro-1,6-naphthyridin-3-yl)-2-fluoropheyl)-3-phenylurea

Also Published As

Publication number Publication date
UA73492C2 (en) 2005-08-15
US6329415B1 (en) 2001-12-11
NZ513525A (en) 2004-05-28
KR100669839B1 (en) 2007-01-18
SA00210018B1 (en) 2006-08-12
US6319921B1 (en) 2001-11-20
BR9916930A (en) 2001-10-30
ATE549328T1 (en) 2012-03-15
HRP20010516A2 (en) 2002-08-31
HUP0201406A2 (en) 2002-08-28
BG105653A (en) 2002-01-31
HK1041484A1 (en) 2002-07-12
IL143394A (en) 2011-08-31
PE20001399A1 (en) 2000-12-20
US6333325B1 (en) 2001-12-25
PL351764A1 (en) 2003-06-16
BG64971B1 (en) 2006-11-30
US20020065285A1 (en) 2002-05-30
JP2003535023A (en) 2003-11-25
EE200100376A (en) 2002-10-15
NO20013559L (en) 2001-07-18
HUP0201406A3 (en) 2003-11-28
NO20013559D0 (en) 2001-07-18
TR200102072T2 (en) 2004-12-21
CA2352524A1 (en) 2000-07-27
CN1333767A (en) 2002-01-30
WO2000043384A1 (en) 2000-07-27
MY128063A (en) 2007-01-31
YU39101A (en) 2005-06-10
CZ20012635A3 (en) 2001-11-14
CO5150218A1 (en) 2002-04-29
IL143394A0 (en) 2002-04-21
JP3793694B2 (en) 2006-07-05
EP1147104B1 (en) 2012-03-14
US6506748B2 (en) 2003-01-14
RU2220142C2 (en) 2003-12-27
US6372773B1 (en) 2002-04-16
EP1147104A1 (en) 2001-10-24
TW546297B (en) 2003-08-11
KR20010094748A (en) 2001-11-01
SK10192001A3 (en) 2001-12-03
MXPA01005628A (en) 2002-04-24
AU1752200A (en) 2000-08-07
ZA200104656B (en) 2003-02-10
EE04527B1 (en) 2005-08-15
CA2352524C (en) 2010-06-01
AR034089A1 (en) 2004-02-04
AU770581B2 (en) 2004-02-26

Similar Documents

Publication Publication Date Title
US6329415B1 (en) Aromatic heterocyclic compounds as antiinflammatory agents
US6492529B1 (en) Bis pyrazole-1H-pyrazole intermediates and their synthesis
US6525046B1 (en) Aromatic heterocyclic compounds as antiinflammatory agents
US6916814B2 (en) Methods of treating cytokine mediated diseases
US6297381B1 (en) Aromatic heterocyclic compounds as anti-inflammatory agents
US6432995B2 (en) Aromatic heterocyclic compounds and their use as anti-inflammatory agents
US6492393B1 (en) Compounds useful as anti-inflammatory agents
US6242453B1 (en) Polycyclo heterocyclic derivatives as antiinflammatory agents
EP1709965A2 (en) Methods of treating cytokine mediate diseases
EP1473292A1 (en) Aromatic heterocyclic compounds as anti-inflammatory agents

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

Free format text: EXPRESSLY ABANDONED -- DURING EXAMINATION