WO2001038314A1 - 3,4-dihydro-(1h)quinazolin-2-one compounds as csbp/p38 kinase inhibitors - Google Patents

3,4-dihydro-(1h)quinazolin-2-one compounds as csbp/p38 kinase inhibitors Download PDF

Info

Publication number
WO2001038314A1
WO2001038314A1 PCT/US2000/031894 US0031894W WO0138314A1 WO 2001038314 A1 WO2001038314 A1 WO 2001038314A1 US 0031894 W US0031894 W US 0031894W WO 0138314 A1 WO0138314 A1 WO 0138314A1
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
optionally substituted
aryl
heteroaryl
compound
Prior art date
Application number
PCT/US2000/031894
Other languages
French (fr)
Inventor
Jerry L. Adams
Michael J. Bower
Jeffrey Charles Boehm
Don E. Griswold
David C. Underwood
Original Assignee
Smithkline Beecham Corporation
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Smithkline Beecham Corporation filed Critical Smithkline Beecham Corporation
Priority to JP2001540077A priority Critical patent/JP2003514900A/en
Priority to DE60020595T priority patent/DE60020595T2/en
Priority to AU17832/01A priority patent/AU1783201A/en
Priority to US10/129,888 priority patent/US7053098B1/en
Priority to AT00980587T priority patent/ATE296809T1/en
Priority to EP00980587A priority patent/EP1233951B1/en
Publication of WO2001038314A1 publication Critical patent/WO2001038314A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/70Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings condensed with carbocyclic rings or ring systems
    • C07D239/72Quinazolines; Hydrogenated quinazolines
    • C07D239/78Quinazolines; Hydrogenated quinazolines with hetero atoms directly attached in position 2
    • C07D239/80Oxygen atoms
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • 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
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/02Drugs for disorders of the urinary system of urine or of the urinary tract, e.g. urine acidifiers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/02Drugs for dermatological disorders for treating wounds, ulcers, burns, scars, keloids, or the like
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/06Antigout agents, e.g. antihyperuricemic or uricosuric agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • A61P19/10Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease for osteoporosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • 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
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/16Antivirals for RNA viruses for influenza or rhinoviruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P33/00Antiparasitic agents
    • A61P33/02Antiprotozoals, e.g. for leishmaniasis, trichomoniasis, toxoplasmosis
    • A61P33/06Antimalarials
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/02Antithrombotic agents; Anticoagulants; Platelet aggregation inhibitors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • Intracellular signal transduction is the means by which cells respond to extracellular stimuli Regardless of the nature of the cell surface receptor (e g protein tyrosine kinase or seven-transmembrane G-protein coupled), protein kinases and phosphatases along with phopholipases are the essential machinery by which the signal is further transmitted within the cell [Marshall, J C Cell , 80, 179-278 (1995)] Protein kinases can be categorized into five classes with the two major classes being, tyrosine kinases and serine / threonine kinases depending upon whether the enzyme phosphorylates its substrate(s) on specific tyros ⁇ ne(s) or serine / threonine(s) residues [Hunter, T . Methods in Enzymology (Protein Kinase Classification) p 3, Hunter, T , Sefton, B M , eds vol 200, Academic Press, San Diego, 1991 ]
  • pharmaceutically acceptable salts of compounds of Formula (I) may also be formed with a pharmaceutically acceptable cation, for instance, if a substituent group comprises a carboxy moiety.
  • Suitable pharmaceutically acceptable cations are well known to those skilled in the art and include alkaline, alkaline earth, ammonium and quaternary ammonium cations.
  • the term "halo" or “halogens” is used herein to mean the halogens, chloro, fluoro, bromo and iodo
  • CSAID for the treatment of CSBP mediated disease states, can include, but not be limited to neurodegenerative diseases, such as Alzheimer's disease (as noted above), Parkinson's disease and multiple sclerosis, etc
  • the compounds of Formula (I) may also be used topically in the treatment or prophylaxis of topical disease states mediated by or exacerbated by excessive cytokine production, such as by IL-1 or TNF respectively, such as inflamed joints, eczema, psoriasis and other inflammatory skin conditions such as sunburn; inflammatory eye conditions including conjunctivitis; pyresis, pain and other conditions associated with inflammation.
  • Periodontal disease has also been implemented in cytokine production, both topically and systemically.
  • use of compounds of Formula (I) to control the inflammation associated with cytokine production in such peroral diseases such as gingivitis and periodontitis is another aspect of the present invention.
  • novel compounds of Formula (I) may also be used in association with the veterinary treatment of mammals, other than humans, in need of inhibition of CSBP/p38 or cytokine inhibition or production
  • CSBP/p38 mediated diseases for treatment, therapeutically or prophylactically, in animals include disease states such as those noted herein in the Methods of Treatment section, but in particular viral infections
  • viruses include, but are not limited to, lentivirus infections such as, equine infectious anaemia virus, caprine arthritis virus, visna virus, or aedi virus or retrovirus infections, such as but not limited to feline immunodeficiency virus (FIV), bovine immunodeficiency virus, or canine immunodeficiency virus or other retroviral infections
  • mice are treated with a p38 kinase inhibitor, a compound of Formula (I) at approximatetly a 30 mg/kg, p o b i d Reduction in lung KC (a murine homolog of IL-8) levels are assesed 1 day after exposure (prior to neutrophilia), and attenuated airway neutrophilia and lung IL-6 levels are assessed following 3 days of cigarette exposure

Landscapes

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

Abstract

Novel substituted quinazoline compounds and compositions for use in therapy as CSBP/p38 kinase inhibitors.

Description

3,4-DIHYDRO-(lH)QUINAZOLIN-2-ONE COMPOUNDS AS CSBP/P38 KINASE INHIBITORS
FIELD OF THE INVENTION
This invention relates to a novel group of 6-thio aryl -3,4-dihydro-( lH) quinazolin-2-one compounds, processes for the preparation thereof, the use thereof in treating CSBP/p 8 kinase mediated diseases and pharmaceutical compositions for use in such therapy
BACKGROUND OF THE INVENTION
Intracellular signal transduction is the means by which cells respond to extracellular stimuli Regardless of the nature of the cell surface receptor (e g protein tyrosine kinase or seven-transmembrane G-protein coupled), protein kinases and phosphatases along with phopholipases are the essential machinery by which the signal is further transmitted within the cell [Marshall, J C Cell , 80, 179-278 (1995)] Protein kinases can be categorized into five classes with the two major classes being, tyrosine kinases and serine / threonine kinases depending upon whether the enzyme phosphorylates its substrate(s) on specific tyrosιne(s) or serine / threonine(s) residues [Hunter, T . Methods in Enzymology (Protein Kinase Classification) p 3, Hunter, T , Sefton, B M , eds vol 200, Academic Press, San Diego, 1991 ]
For most biological responses, multiple intracellular kinases are involved and an individual kinase can be involved in more than one signaling event These kinases are often cytosolic and can translocate to the nucleus or the ribosomes where they can affect transcriptional and translational events, respectively The involvement of kinases in transcriptional control is presently much better understood than their effect on translation as illustrated by the studies on growth factor induced signal transduction involving MAP/ERK kinase [Marshall, C J CeJL, 80, 179 (1995), Herskowitz, I Cell - 80, 187 (1995), Hunter, T Cell , 80, 225 (1 95),Seger, R , and Krebs, E G FASEB J , 726-735 (1995)]
While many signaling pathways are part of cell homeostasis, numerous cytokines (e g , IL-1 and TNF) and certain other mediators of inflammation (e g , COX-2, and iNOS) are produced only as a response to stress signals such as bacterial lipopolysaccharide (LPS) The first indications suggesting that the signal transduction pathway leading to LPS-induced cytokine biosynthesis involved protein kinases came from studies of Weinstein [Weinstein, el al , J Immunol 151, 3829(1993)] but the specific protein kinases involved were not identified. Working from a similar perspective, Han [Han, et al, Science 265. 808(1994)] identified murine p38 as a kinase which is tyrosine phosphorylated in response to LPS. Definitive proof of the involvement of the p38 kinase in LPS-stimulated signal transduction pathway leading to the initiation of proinflammatory cytokine biosynthesis was provided by the independent discovery of p38 kinase by Lee [Lee; et al, Nature. 372, 739(1994)] as the molecular target for a novel class of anti-inflammatory agents. The discovery of p38 (termed by Lee as CSBP 1 and 2) provided a mechanism of action of a class of anti-inflammatory compounds for which SK&F 86002 was the prototypic example. These compounds inhibited IL-1 and TNF synthesis in human monocytes at concentrations in the low uM range [Lee, et al, Int. J. Immunopharmac. 10(7), 835(1988)] and exhibited activity in animal models which are refractory to cyclooxygenase inhibitors [Lee; et al, Annals N. Y. Acad. Sci., 696, 149(1993)].
It is now firmly established that CSBP/p38 is a one of several kinases involved in a stress-response signal transduction pathway which is parallel to and largely independent of the analogous mitogen-activated protein kinase (MAP) kinase cascade (Figure 1). Stress signals, including LPS, pro-inflammatory cytokines, oxidants, UV light and osmotic stress, activate kinases upstream from CSBP/p38 which in turn phosphorylate CSBP/p38 at threonine 180 and tyrosine 182 resulting in CSBP/p38 activation. MAPKAP kinase-2 and MAPKAP kinase-3 have been identified as downstream substrates of CSBP/p38 which in turn phosphorylate heat shock protein Hsp 27 (Figure 1). It is not yet known whether MAPKAP-2, MAPKAP-3, Mnkl or Mnk2 are involved in cytokine biosynthesis or alternatively that inhibitors of CSBP/p38 kinase might regulate cytokine biosynthesis by blocking a yet unidentified substrate downstream from CSBP/p38 [Cohen, P. Trends Cell Biol.. 353-361( 1997)].
What is known, however, is that in addition to inhibiting IL-1 and TNF,
CSBP/p38 kinase inhibitors (SK&F 86002 and SB 203580) also decrease the synthesis of a wide variety of pro-inflammatory proteins including, IL-6, IL-8, GM-CSF and
COX-2. Inhibitors of CSBP/p38 kinase have also been shown to suppress the TNF- induced expression of VCAM-1 on endothelial cells, the TNF-induced phosphorylation and activation of cytosolic PLA2 and the IL- 1 -stimulated synthesis of collagenase and stromelysin. These and additional data demonstrate that CSBP/p38 is involved not only cytokine synthesis, but also in cytokine signaling [CSBP P38 kinase reviewed in Cohen, P. Trends Cell Biol.. 353-361(1997)]. Interleukin-1 (IL-1) and Tumor Necrosis Factor (TNF) are biological substances produced by a variety of cells, such as monocytes or macrophages. IL-1 has been demonstrated to mediate a variety of biological activities thought to be important in immunoregulation and other physiological conditions such as inflammation [See, e.g., Dinarello et al., Rev. Infect. Disease, 6, 51 (1984)]. The myriad of known biological activities of IL-1 include the activation of T helper cells, induction of fever, stimulation of prostaglandin or collagenase production, neutrophil chemotaxis, induction of acute phase proteins and the suppression of plasma iron levels.
There are many disease states in which excessive or unregulated IL-1 production is implicated in exacerbating and/or causing the disease. These include rheumatoid arthritis, osteoarthritis, endotoxemia and/or toxic shock syndrome, other acute or chronic inflammatory disease states such as the inflammatory reaction induced by endotoxin or inflammatory bowel disease; tuberculosis, atherosclerosis, muscle degeneration, cachexia, psoriatic arthritis, Reiter's syndrome, rheumatoid arthritis, gout, traumatic arthritis, rubella arthritis, and acute synovitis. Recent evidence also links IL-1 activity to diabetes and pancreatic β cells [review of the biological activities which have been attributed to IL-1 Dinarello, J. Clinical Immunology. 5 (5), 287-297 (1985)].
Excessive or unregulated TNF production has been implicated in mediating or exacerbating a number of diseases including rheumatoid arthritis, rheumatoid spondylitis, osteoarthritis, gouty arthritis and other arthritic conditions; sepsis, septic shock, endotoxic shock, gram negative sepsis, toxic shock syndrome, adult respiratory distress syndrome, cerebral malaria, chronic pulmonary inflammatory disease, silicosis, pulmonary sarcoisosis, bone resorption diseases, reperfusion injury, graft vs. host reaction, allograft rejections, fever and myalgias due to infection, such as influenza, cachexia secondary to infection or malignancy, cachexia, secondary to acquired immune deficiency syndrome (AIDS), AIDS, ARC (AIDS related complex), keloid formation, scar tissue formation, Crohn's disease, ulcerative colitis, or pyresis.
Interleukin-8 (IL-8) is a chemotactic factor produced by several cell types including mononuclear cells, fibroblasts, endothelial cells, and keratinocytes. Its production from endothelial cells is induced by IL-1, TNF, or lipopolysachharide (LPS). IL-8 stimulates a number of functions in vitro. It has been shown to have chemoattractant properties for neutrophils, T-lymphocytes, and basophils. In addition it induces histamine release from basophils from both normal and atopic individuals as well as lysozomal enzyme release and respiratory burst from neutrophils. IL-8 has also been shown to increase the surface expression of Mac- 1 (CD1 lb/CD 18) on neutrophils without de novo protein synthesis, this may contribute to increased adhesion of the neutrophils to vascular endothelial cells. Many diseases are characterized by massive neutrophil infiltration. Conditions associated with an increased in IL-8 production (which is responsible for chemotaxis of neutrophil into the inflammatory site) would benefit by compounds, which are suppressive of IL-8 production
IL-1 and TNF affect a wide variety of cells and tissues and these cytokines as well as other leukocyte derived cytokines are important and critical inflammatory mediators of a wide variety of disease states and conditions The inhibition of these cytokines is of benefit in controlling, reducing and alleviating many of these disease states
Inhibition of signal transduction via CSBP/p38, which in addition to IL- 1 , TNF and IL-8 described above is also required for the synthesis and/or action of several additional pro-inflammatory proteins (i e , IL-6, GM-CSF, COX-2, collagenase and stromelysin), is expected to be a highly effective mechanism for regulating the excessive and destructive activation of the immune system This expectation is supported by the potent and diverse anti-inflammatory activities described for CSBP/p38 kinase inhibitors
[Badger, et al . J Pharm Exp Thera. 279 (3) 1453-1461 (1996), Griswold, et al,
Pharmacol Comm 7, 323-229 ( 1996)] There remains a need for treatment, in this field, for compounds that are cytokine suppressive anti-inflammatory drugs, i e compounds that are capable of inhibiting the CSBP/p38/RK kinase
BRIEF DESCRIPTION OF THE DRAWINGS Figure 1 demonstrates the p38 kinase cascade
Figure 2 demonstrates a Citric Acid Induced Cough Model
Figure 3 demonstrates an Antigen- or LTD4 - Induced Hypertussive Model in the
Guinea Pig Figure 4 demonstrates Effects of Dextromethorphan or Codeine On Citric Acid- Induced Cough in Guinea Pigs
SUMMARY OF THE INVENTION
This invention relates to the novel compounds of Formula (I), and pharmaceutical compositions comprising a compound of Formula (I), and a pharmaceutically acceptable diluent or carrier
This invention relates to a method of treating a CSBP/RK/p38 kinase mediated disease in a mammal in need thereof, which comprises administering to said mammal an effective amount of a compound of Formula (I)
This invention also relates to a method of inhibiting cytokines and the treatment of a cytokine mediated disease, in a mammal in need thereof, which comprises administering to said mammal an effective amount of a compound of Formula (I).
This invention more specifically relates to a method of inhibiting the production of EL-1 in a mammal in need thereof which comprises administering to said mammal an effective amount of a compound of Formula (I).
This invention more specifically relates to a method of inhibiting the production of IL-6 in a mammal in need thereof which comprises administering to said mammal an effective amount of a compound of Formula (I).
This invention more specifically relates to a method of inhibiting the production of IL-8 in a mammal in need thereof which comprises administering to said mammal an effective amount of a compound of Formula (I).
This invention more specifically relates to a method of inhibiting the production of TNF in a mammal in need thereof which comprises administering to said mammal an effective amount of a compound of Formula (I). Accordingly, the present invention provides a compound of Formula (I):
- ?S ' HN
O ..J, f\T
2 (0 wherein
Rl is a phenyl, naphth-1-yl, naphth-2-yl, heterocyclic or heteroaryl ring, which ring is optionally substituted independently by one or more substituents selected from halogen, C ]-4 alkyl, halo-substituted-C i-4 alkyl, cyano, nitro,
(CRi 0R20)vNR4Rl4, (CRι0R2θ)vC(Z)NR4Rl4, (CRι0R2θ)vC(Z)OR8, (CRioR20)vCOR3, (CRi0R2θ)vC(O)H, SR5, S(O)R5, S(O) R5, (CRioR20)vOR8, ZC(Z)Rι 1, NRι0C(Z)Rι 1, or NRioS(O)2R7. R2 is a Cι _ιo alkyl, aryl, arylCi _io alkyl, heteroaryl, heteroarylC 1. ] Q alkyl, heterocyclic, or heterocyclylCι _ιo alkyl moiety, which moieties are optionally substituted one or more times independently with Ci-io alkyl, halo- substituted Cι _ιo alkyl, C2-10 alkenyl, C2-10 alkynyl, C3.7 cycloalkyl, C3.7cycloalkylC1.jo alkyl, C5.7 cycloalkenyl, C5.7 cycloalkenyl Cι_ιo alkyl, halogen, (CRι0R2θ)nO 6, (CRι0R2θ)nSH, (CR10R2θ)nS(O)mR7, (CR10R2θ)n HS(O)2R7, (CRιoR2o)nNR4Rl4, (CR10R2θ)nCN,
(CR10R20)n S(O)2NR4R14, (CR10R2o)nC(Z)R6, (CR10R2θ)nOC(Z)R6, (CR10R2θ)nC(Z)OR6, (CR1oR2θ)nC(Z)NR4R14) (CR10R2θ)n Rl θC(Z)R6, (CRι 0R2θ)nNRlθC(=NRι0) NR4R-I4, (CR10R20)nOC(Z)NR4R14, (CR10R2θ)nNRlθC(Z) NR4R14, or (CRι0R2θ)nNRl θC(Z)OR7,
Z is oxygen or sulfur, n is 0, or an integer having a value of 1 to 10, m is 0, or the integer 1 or 2, v is 0, or an integer having a value of 1 or 2, t is an integer having a value of 1 to 3,
R3 is C 1-4 alkyl, halo-substituted C i -4 alkyl, C2-4 alkenyl, C2-4 alkynyl, C3-.7 cycloalkyl, C5-7 cycloalkenyl, aryl, arylC ι .4 alkyl, heteroaryl, heteroarylC 1.4 alkyl, heterocyclyl, heterocyclylC 1.4 alkyl, (CRloR2θ)vOR7, (CRιoR2θ)vS(O)mR7, (CRιoR2θ)vNHS(O) R7, or (CRifjR2θ)vNR4Rl4. and wherein the aryl, arylalkyl, heteroaryl, or heteroaryl alkyl may be optionally substituted, R4 and R14 is each independently selected from hydrogen or optionally substituted Ci-4 alkyl, optionally substituted aryl or optionally substituted aryl-Ci -4 alkyl, or together with the nitrogen which they are attached form a heterocyclic ring of 5 to 7 members which ring optionally contains an additional heteroatom selected from oxygen, sulfur or NR9, R5 is hydrogen, C 1.4 alkyl, C2-4 alkenyl, C2-4 alkynyl or NR4R14, excluding the moieties SR5 being SNR4R14, S(O)2Rs being SO H and S(O)R5 being
SOH,
R6 is hydrogen, C i -i o alkyl, C3-7 cycloalkyl, heterocyclyl, heterocyclyl C i- i fjalkyl, aryl, arylC i - io alkyl, heteroaryl or heteroarylC 1 - 10 alkyl, wherein these moieties may be optionally substituted,
R7 is C i-6alkyl, aryl, arylC 1 -6alkyl, heterocyclic, heterocyclylC 1 -6 alkyl, heteroaryl, or heteroarylC ι _6alkyl, and wherein each of these moieties may be optionally substituted, 8 is hydrogen, C i-4 alkyl, halo-substituted C i-4 alkyl, C2-4 alkenyl, C2-4 alkynyl, C3-7 cycloalkyl, C5-.7 cycloalkenyl, aryl, arylC i -4 alkyl, heteroaryl, heteroarylC ι_4 alkyl, heterocyclyl, heterocyclylC 1.4 alkyl, (CRι oR2θ)tOR7, (CRi oR20)tS(O)mR7, (CRι 0R2θ)tNHS(O)2R7, or (CRι0R2θ)tNR4 l4, and wherein the aryl, arylalkyl, heteroaryl, or heteroaryl alkyl may be optionally substituted, RQ is hydrogen, C(Z)R6 or optionally substituted C ] _ o alkyl, optionally substituted aryl or optionally substituted aryl-C i -4 alkyl, RlO and R20 are each independently selected from hydrogen or C 1.4 alkyl;
Rl 1 is C 1-4 alkyl, halo-substituted d .4 alkyl, C -4 alkenyl, C2-4 alkynyl, C3-7 cycloalkyl, C5-7 cycloalkenyl, aryl, arylCi-4 alkyl, heteroaryl, heteroarylC 1-4 alkyl, heterocyclyl, heterocyclylC 1.4 alkyl, (CRιoR2θ)tOR7, (CRioR20)tS(O)mR7, (CRι0R20)tNHS(O)2R7, or (CRιoR2θ)vNR4Rl4. and wherein the aryl, arylalkyl, heteroaryl, heteroaryl alkyl may be optionally substituted; or a pharmaceutically acceptable salt thereof.
DETAILED DESCRIPTION OF THE INVENTION
The present invention is directed to novel compounds of Formula (I), or pharmaceutically acceptable salts thereof.
Suitably, for compounds of Formula (I) Ri is a phenyl, naphth-1-yl, naphth-2-yl, heterocyclic or a heteroaryl ring. The Ri moiety may be optionally substituted independently by one or more substituents, preferably from one to three substituents each independently selected from halogen, C 1.4 alkyl, halo-substituted-C 1.4 alkyl, cyano, nitro, (CRioR20)vNR4Rl4, (CRioR2θ)vC(Z)NR4Rl4, (CRioR20)vC(Z)ORg, (CRι0R2θ)vCOR3, (CRι0R2θ)vC(O)H, SR5, S(O)R5, S(O)2R5, (CRιoR2θ)vORg, ZC(Z)Rι 1, NRιoC(Z)Rι 1, or NRι0S(O)2R7. Preferably, the Ri moiety is an aryl ring. More preferably, the aryl ring is an optionally substituted phenyl ring.
Preferably the Rj moiety is substituted by one or more halogens, such as fluorine or chlorine; an alkyl, hydroxy, alkoxy, amino, or halosubstituted alkyl, such as CF3. More preferably substituted by one or more halogens. Suitably, for compounds of Formula (I), R2 is an C ι_ιo lkyl aryl, arylC i.jo alkyl, heteroaryl, heteroarylC 1 _ j 0 alkyl, heterocyclic, or heterocyclylC \ _ \ Q alkyl moiety. These moieties may be optionally substituted one or more times, preferably 1 to 3 times, independently with Ci-io alkyl, halo-substituted C J . I Q alkyl, C2-10 alkenyl, C2-10 alkynyl, C3.7 cycloalkyl, C3.7cycloalkylC1.10 alkyl, C5.7 cycloalkenyl, C5.7 cycloalkenyl Cj_ιo lkyl halogen, (CRιoR2θ)nOR-6> (CR10R20)nSH, (CRι0R20)nS(O)mR7, (CRι0R2θ)n HS(O)2R7, (CRι0R20)nN 4Rl4, (CRι0R2θ)nCN, (CRι0R θ)n S(O)2NR4Rι4, (CR, 0R2θ)nC(Z)R6, (CRι0R2θ)nOC(Z)R6, (CRι0R2θ)nC(Z)OR6, (CR10R2θ)nC(Z)NR4Ri45 (CRιoR2θ)nNRlθC(Z)R6, (CRιoR2θ)n RioC(=NR10) NI^R^, (CRιoR2θ)nOC(Z)NR4Ri4, (CRι0R2θ)n RιoC(Z) NR4R14, or (CRι0R2θ)pNRi()C(Z)OR7. Preferably, R2 is an optionally substituted aryl moiety, more preferably an optionally substituted phenyl
Preferably the R moiety is substituted by one or more halogens, such as fluorine or chlorine, an alkyl, hydroxy, alkoxy, amino, or halosubstituted alkyl, such as CF3. More preferably, by one or more halogens
Suitably, n is 0, or an integer having a value of 1 to 10
Suitably, m is 0, or the integer 1 or 2
Suitably, v is 0, or an integer having a value of 1 or 2
Suitably, t is an integer having a value of 1 to 3 Suitably, Z is oxygen or sulfur, preferably oxygen
Suitably, R3 is a C i -4 alkyl, halo-substituted C i -4 alkyl, C2-4 alkenyl, C2-4 alkynyl, C3-7 cycloalkyl, C5-7 cycloalkenyl, aryl, arylC i -4 alkyl, heteroaryl, heteroarylC 1 -4 alkyl, heterocyclyl, heterocyclylC 1.4 alkyl, (CRiθ 2θ)v 7, (CRl 0R20)vS(O)mR7, (CRι 0R2θ) NHS(O)2R7, or (CR] θ 2θ)vNR4Rl4, and wherein the aryl, arylalkyl, heteroaryl, heteroaryl alkyl may be optionally substituted
Suitably, R4 and R14 is each independently selected from hydrogen or optionally substituted C 1.4 alkyl, optionally substituted aryl or optionally substituted aryl-C 1.4 alkyl, or together with the nitrogen which they are attached form a heterocyclic ring of 5 to 7 members which ring optionally contains an additional heteroatom selected from oxygen, sulfur or NR9
Suitably, R5 is hydrogen, C i -4 alkyl, C2-4 alkenyl, C2-4 alkynyl or NR4R14, excluding the moieties SR5 being S ^R^ S(O)2 s being SO2H and S(O)R5 being SOH
Suitably, R6 is hydrogen, C i -io alkyl, C3-7 cycloalkyl, heterocyclyl, heterocyclyl C 1 _ 1 oalkyl, aryl, arylC 1.10 alkyl, heteroaryl or heteroarylC 1 - 10 alkyl, wherein these moieties may be optionally substituted
Suitably, R7 is C i -6alkyl, aryl, arylC 1 -6alkyl, heterocyclic, heterocyclylC 1 -6 alkyl, heteroaryl, or heteroarylC i -6alkyl, and wherein each of these moieties may be optionally substituted Suitably, R8 is hydrogen, C 1-4 alkyl, halo-substituted C i-4 alkyl, C2-4 alkenyl, C -4 alkynyl, C3-7 cycloalkyl, C5-7 cycloalkenyl, aryl, arylC i-4 alkyl, heteroaryl, heteroarylC 1.4 alkyl, heterocyclyl, heterocyclylC 1.4 alkyl, (CR]0R20)tOR7, (CRl0R20)tS(O)mR7, (CRi0R2θ)tNHS(O)2R7, or (CRι oR2θ)t R4Ri4, and wherein the aryl, arylalkyl, heteroaryl, heteroaryl alkyl may be optionally substituted Suitably, R9 is hydrogen, C(Z)R6 or optionally substituted Ci-io alkyl, optionally substituted aryl or optionally substituted aryl-Cι_4 alkyl.
Suitably, Rio and R20 are each independently selected from hydrogen or C 1-4 alkyl. Suitably, Ri 1 is C i-4 alkyl, halo-substituted C i-4 alkyl, C2-4 alkenyl, C2-4 alkynyl, C3-7 cycloalkyl, C5-7 cycloalkenyl, aryl, arylC i-4 alkyl, heteroaryl, heteroarylC 1 -4 alkyl, heterocyclyl, heterocyclylC 1.4 alkyl, (CRιoR2θ)tOR7, (CRl 0R20)tS(O)mR7, (CRi 0R2θ)tNHS(O)2R7, or (CRι0 2θ)vNR4Rl4; and wherein the aryl, arylalkyl, heteroaryl, heteroaryl alkyl may be optionally substituted. As used herein, "optionally substituted" unless specifically defined shall mean such groups as halogen, such as fluorine, chlorine, bromine or iodine; hydroxy; hydroxy substituted Ci-ioalkyl; C i-io alkoxy, such as methoxy or ethoxy; halosubstituted C i-io alkoxy; S(O)m alkyl, such as methyl thio, methylsulfinyl or methyl sulfonyl; NR7R17, such as amino or mono or -disubstituted C j _4 alkyl or wherein the R7R17 can cyclize together with the nitrogen to which they are attached to form a 5 to 7 membered ring which optionally contains an additional heteroatom selected from O/N/S; C i- io alkyl, C3_7cycloalkyl, or C3_7cycloalkyl C i-io alkyl group, such as methyl, ethyl, propyl, isopropyl, t-butyl, etc. or cyclopropyl methyl; halosubstituted C i -io alkyl, such CF2CF2H, or CF3; an optionally substituted aryl, such as phenyl, or an optionally substituted arylalkyl, such as benzyl or phenethyl, wherein these aryl containing moieties may also be substituted one to two times by halogen; hydroxy; hydroxy substituted alkyl; C i-io alkoxy; S(O)malkyl; amino, mono & di-substituted C 1.4 alkyl amino, such as in the NR7R17 group; C ] _4 alkyl, or CF3. Suitable pharmaceutically acceptable salts are well known to those skilled in the art and include basic salts of inorganic and organic acids, such as hydrochloric acid, hydrobromic acid, sulphuric acid, phosphoric acid, methane sulphonic acid, ethane sulphonic acid, acetic acid, malic acid, tartaric acid, citric acid, lactic acid, oxalic acid, succinic acid, fumaric acid, maleic acid, benzoic acid, salicylic acid, phenylacetic acid and mandelic acid.
In addition, pharmaceutically acceptable salts of compounds of Formula (I) may also be formed with a pharmaceutically acceptable cation, for instance, if a substituent group comprises a carboxy moiety. Suitable pharmaceutically acceptable cations are well known to those skilled in the art and include alkaline, alkaline earth, ammonium and quaternary ammonium cations. The term "halo" or "halogens" is used herein to mean the halogens, chloro, fluoro, bromo and iodo
The term "Ci.joalkyl" or "alkyl" or "alkyl _ιo" 's use(^ herein to mean both straight and branched chain radicals of 1 to 10 carbon atoms, unless the chain length is otherwise limited, including, but not limited to, methyl, ethyl, «-propyl, .so-propyl, «-butyl, .sec-butyl, /-rø-butyl, tert-butyl, «-pentyl and the like
The term "cycloalkyl" is used herein to mean cyclic radicals, preferably of 3 to 8 carbons, including but not limited to cyclopropyl, cyclopentyl, cyclohexyl, and the like The term "cycloalkenyl" is used herein to mean cyclic radicals, preferably of 5 to 8 carbons, which have at least one bond including but not limited to cyclopentenyl, cyclohexenyl, and the like
The term "alkenyl" is used herein at all occurrences to mean straight or branched chain radical of 2-10 carbon atoms, unless the chain length is limited thereto, including, but not limited to ethenyl, 1-propenyl, 2-propenyl, 2-methyl-l- propenyl, 1 -butenyl, 2-butenyl and the like
The term "aryl" is used herein to mean phenyl and naphthyl The term "heteroaryl" (on its own or in any combination, such as "heteroaryloxy", or "heteroaryl alkyl") is used herein to mean a 5-10 membered aromatic ring system in which one or more rings contain one or more heteroatoms selected from the group consisting of N, O or S, such as, but not limited, to pyrrole, pyrazole, furan, thiophene, quinoline, isoquinoline, quinazolinyl, pyridine, pyrimidine, oxazole, thiazole, thiadiazole, tetrazole, indole, triazole, imidazole, benzimidazole, benzofuran, or benzothiophene The term "heterocyclic" (on its own or in any combination, such as
"heterocyclylalkyl") is used herein to mean a saturated or partially unsaturated 4-10 membered ring system in which one or more rings contain one or more heteroatoms selected from the group consisting of N, O, or S, such as, but not limited to, pyrrolidine, piperidine, piperazine, morpholine, tetrahydropyran, imidazolidine, or pyrazolidine
The term "aralkyl" or "heteroarylalkyl" or "heterocyclicalkyl" is used herein to mean C i-4 alkyl as defined above attached to an aryl, heteroaryl or heterocyclic moiety as also defined herein unless otherwise indicate
The term "sulfinyl" is used herein to mean the oxide S (O) of the corresponding sulfide, the term "thio" refers to the sulfide, and the term "sulfonyl" refers to the fully oxidized S (O)2 moiety The term "aroyl" is used herein to mean C(O)Ar, wherein Ar is as phenyl, naphthyl, or aryl alkyl derivatn e such as defined above, such group include but are not limited to benzyl and phenethyl
The term "alkanoyl" is used herein to mean C(O)Ci-io alkyl wherein the alkyl is as defined above
It is recognized that the compounds of the present invention may exist as stereoisomers, regioisomers, or diastereiomers These compounds may contain one or more asymmetric carbon atoms and may exist in racemic and optically active forms All of these compounds are included within the scope of the present invention
Exemplified compounds of Formula (I), or pharmaceutically acceptable salts thereof, include
1 -Phenyl-2-oxo-3 ,4-dihydro-6-phenylthioquinazoline, l-(2,6-Dichlorophenyl)-2-oxo-3,4-dihydro-6-(4-fluorophenyl)thioquinazoline, l-(2,6-Dichlorophenyl)-6-(2,4-difluorophenylsulfanyl)-3,4-dihydro-l-H-quinazolin-2- one
The compounds of Formula (I) may be obtained by applying synthetic procedures, described herein The synthesis provided for is applicable to producing compounds of Formula (I) having a variety of different Ri, and R2, groups which are reacted, employing optional substituents which are suitably protected, to achieve compatibility with the reactions outlined herein Subsequent deprotection, in those cases, then affords compounds of the nature generally disclosed
Once the nucleus has been established, further compounds of Formula (I) may be prepared by applying standard techniques for functional group interconversion, well known in the art For instance C(O)NR4R 14 from CO2CH3 by heating with or without catalytic metal cyanide, e g NaCN, and HNR4R14 in CH3OH, OC(O)R3 from OH with e g , ClC(O)R3 in pyridine, NRιo-C(S)NR4Rl4 from NHR10 with an alkylisothiocyante or thiocyanic acid, NRιoC(O)OR7 from NHR10 with the alkyl chloroformate, NRιoC(O)NR4Ri4 from NHR10 by treatment with an isocyanate, e g HN=C=O or RιoN=C=O, NRιo-C(O)R7 from NHRio by treatment with Cl- C(O)R7 in pyridine, C(=NRιo)NR4R]4 from C(NR4Ri4)SR3 with H3NR3+OAc" by heating in alcohol, C(NR4Ri4)SR3 from C(S)NR4Ri4 with R6-I in an inert solvent, e g acetone, C(S)NR4Ri4 (where R4 or R14 is not hydrogen) from C(S)NH2 with HNR4Ri4-C(=NCN)-NR4Ri4 from C(=NR4Ri4)-SR3 with NH2CN by heating in anhydrous alcohol, alternatively from C(=NH)-NR4Ri4 by treatment with BrCN and NaOEt in EtOH,
Figure imgf000012_0001
from NHR10 by treatment with (R8S)2C=NCN, NR10SO2R3 from NHRj o by treatment with CISO2R3 by heating in pyridine, NRιoC(S)R6 from NRιoC(O)R6 by treatment with Lawesson's reagent [2,4-6/s(4-methoxyphenyl)-l ,3,2,4-dithiadiphosphetane-2,4-disulfide], NR10SO2CF3 from NHRό with triflic anhydride and base wherein R3, R6, R7, RlO, R4 and R14 are as defined in Formula (I) herein
Precursors of the groups R\ , and R can be other Ri , and R groups which can be interconverted by applying standard techniques for functional group interconversion For example wherein a moiety is a halo substituted C J. J Q alkyl can be converted to the corresponding C J . I Q alkylN3 derivative by reacting with a suitable azide salt, and thereafter if desired can be reduced to the corresponding
C 1. j QalkylNH2 compound, which in turn can be reacted with R7S(0)2X wherein X is halo (e g , chloro) to yield the corresponding C ] _ιoalkylNHS(0)2R7 compound
Alternatively wherein the moiety is a halo-substituted C j.i o-alkyl it can be reacted with an amine R4R] 4NH to yield the corresponding C ι _ ιo-alkylNR4Ri4 compound, or can be reacted with an alkali metal salt of R7SH to yield the corresponding C ι _ joalkylSR7 compound
Suitable protecting groups for use with hydroxyl groups and nitrogen groups are well known in the art and described in many references, for instance, Protecting Groups in Organic Synthesis, Greene T W, Wiley-Interscience, New York, 1981 Suitable examples of hydroxyl protecting groups include silyl ethers, such as t- butyldimethyl or t-butyldiphenyl, and alkyl ethers, such as methyl connected by an alkyl chain of variable link, (CRι oR2θ)n
Pharmaceutically acid addition salts of compounds of Formula (I) may be obtained in known manner, for example by treatment thereof with an appropriate amount of acid in the presence of a suitable solvent
Figure imgf000014_0001
Scheme I
The above noted scheme depicts the synthesis compounds of Formula 1 wherein Ri and R are aryl substituents, however, as described below this methodology can also afford the compounds of formula I wherein either or both Ri and R2 may be alkyl. Thus, in the above noted scheme, 2-chloro-4-nitrobenzonitrile can be reacted at 120° with aniline, a variety of substituted anilines, or alkylmines in N-methylpyrollidine in the presence of excess tertiary amine for about 18 hour to afford the 2-anilino-4- nitrobenzonitriles in good yield. Alternatively, less reactive anilines can be converted to the sodium salts with NaH in DMSO and then reacted with 2-chloro-4-nitrobenzonitrile to form the 2-anilino-4-nitrobenzonitriles in high yield in a rapid reaction. The more reactive 2-fluoro-4-nitrobenzonitrile can be substituted for the 2-chloro-4- nitrobenzonitrile in the case of displacements with very weakly nucleophillic anilines.
Reduction of the nitro by any of the many well know procedures, such as hydrogenation at 1 atmosphere with sulfided 5% Pt on carbon, affords high yields of aniline 2. The primary arylamine may be diazotized using one of the many well known standard methods such as with fluoroboric acid and sodium nitrate. The diazonium ion is then displaced with sodium arylthiolate or alkylthiolate to afford the sulfide 3. The dihydro quinazolinone (4) was prepared by lithium aluminium hydride (LAH) reduction of the nitrile and cyclization of the di-amine with carbonyldiimidazole, phosgene, a phosgene equivalent (triphosgene) or other appropriately activated derivatives of carbonic acid, (i.e. chloroformate deriviatives) using either basic or acidic catalysis [pyridine as a base, tolene sulfonic acid] and heating [ such as room temperature, solvent refluxing conditions] in a variety of organic solvents as required to effect ring closure. Suitable organic solvents are halogenated solvents such as methylene chloride, chloroform; diethylether, THF, pyridine, toluene, or benzene. In cases where functionality is incompatible with LAH reduction, alternative, selective reduction methods for the nitrile include, but are not limited to, samarium di-iodide in H3PO or sodium borohydride-cobaltous chloride.
Another aspect of the present invention is the novel process of making a compound of formula (I) by reacting a compound of formula (II)
Figure imgf000015_0001
wherein R\ and R2 are as defined herein for Formula (I) with a suitable reducing agent and a cyclization agent to yield a compound of Formula (I).
METHODS OF TREATMENT
The compounds of Formula (I) or a pharmaceutically acceptable salt thereof can be used in the manufacture of a medicament for the prophylactic or therapeutic treatment of any disease state in a human, or other mammal, which is exacerbated or caused by excessive or unregulated cytokine production by such mammal's cell, such as but not limited to monocytes and/or macrophages.
Compounds of Formula (I) are capable of inhibiting proinflammatory cytokines, such as IL-1, IL-6, IL-8, and TNF and are therefore of use in therapy. IL-1 , IL-6, IL-8 and TNF affect a wide variety of cells and tissues and these cytokines, as well as other leukocyte-derived cytokines, are important and critical inflammatory mediators of a wide variety of disease states and conditions. The inhibition of these pro-inflammatory cytokines is of benefit in controlling, reducing and alleviating many of these disease states.
Accordingly, the present invention provides a method of treating a cytokine- mediated disease which comprises administering an effective cytokine-interfering amount of a compound of Formula (I) or a pharmaceutically acceptable salt thereof. Compounds of Formula (I) are capable of inhibiting inducible proinflammatory proteins, such as COX-2, also referred to by many other names such as prostaglandin endoperoxide synthase-2 (PGHS-2) and are therefore of use in therapy These proinflammatory lipid mediators of the cyclooxygenase (CO) pathway are produced by the inducible COX-2 enzyme Regulation, therefore of COX-2 which is responsible for the these products derived from arachidonic acid, such as prostaglandins affect a wide variety of cells and tissues are important and critical inflammatory mediators of a wide variety of disease states and conditions Expression of COX- 1 is not effected by compounds of Formula (I). This selective inhibition of COX-2 may alleviate or spare ulcerogenic liability associated with inhibition of COX- 1 thereby inhibiting prostoglandins essential for cytoprotective effects Thus inhibition of these pro-inflammatory mediators is of benefit in controlling, reducing and alleviating many of these disease states Most notably these inflammatory mediators, in particular prostaglandins, have been implicated in pain, such as in the sensitization of pain receptors, or edema This aspect of pain management therefore includes treatment of neuromuscular pain, headache, cancer pain, and arthritis pain Compounds of Formula (I) or a pharmaceutically acceptable salt thereof, are of use in the prophylaxis or therapy in a human, or other mammal, by inhibition of the synthesis of the COX-2 enzyme
Accordingly, the present invention provides a method of inhibiting the synthesis of COX-2 which comprises administering an effective amount of a compound of Formula (1) or a pharmaceutically acceptable salt thereof The present invention also provides for a method of prophylaxis treatment in a human, or other mammal, by inhibition of the synthesis of the COX-2 enzyme
In particular, compounds of Formula (I) or a pharmaceutically acceptable salt thereof are of use in the prophylaxis or therapy of any disease state in a human, or other mammal, which is exacerbated by or caused by excessive or unregulated IL-1, IL-6, IL-8 or TNF production by such mammal's cell, such as, but not limited to, monocytes and/or macrophages
Accordingly, in another aspect, this invention relates to a method of inhibiting the production of IL-1 in a mammal in need thereof which comprises administering to said mammal an effective amount of a compound of Formula (I) or a pharmaceutically acceptable salt thereof
There are many disease states in which excessive or unregulated IL-1 production is implicated in exacerbating and/or causing the disease These include rheumatoid arthritis, osteoarthritis, meningitis, ischemic and hemorrhagic stroke, neurotrauma/closed head injury, stroke, endotoxemia and/or toxic shock syndrome, other acute or chronic inflammatory disease states such as the inflammatory reaction induced by endotoxin or inflammatory bowel disease, tuberculosis, atherosclerosis, muscle degeneration, multiple sclerosis, cachexia, bone resorption, psoπatic arthritis, Reiter's syndrome, gout, traumatic arthritis, rubella arthritis and acute synovitis Recent evidence also links IL-1 activity to diabetes, pancreatic β cell diseases and Alzheimer's disease
Use of a CSAID for the treatment of CSBP mediated disease states, can include, but not be limited to neurodegenerative diseases, such as Alzheimer's disease (as noted above), Parkinson's disease and multiple sclerosis, etc
In a further aspect, this invention relates to a method of inhibiting the production of TNF in a mammal in need thereof which comprises administering to said mammal an effective amount of a compound of Formula (I) or a pharmaceutically acceptable salt thereof
Excessive or unregulated TNF production has been implicated in mediating or exacerbating a number of diseases including rheumatoid arthritis, rheumatoid spondylitis, osteoarthritis, gouty arthritis and other arthritic conditions sepsis, septic shock, endotoxic shock, gram negative sepsis, toxic shock syndrome, adult respiratory distress syndrome, chronic pulmonary inflammatory disease and chronic obstructuve pulmonary disease, silicosis, pulmonary sarcoisosis, bone resorption diseases, such as osteoporosis, cardiac, brain and renal reperfusion injury, graft vs host reaction, allograft rejections, fever and myalgias due to infection, such as influenza, brain infections including encephalitis (including HIV-induced forms), cerebral malaria, meningitis, ischemic and hemorrhagic stroke, cachexia secondary to infection or malignancy cachexia secondary to acquired immune deficiency syndrome (AIDS), AIDS, ARC (AIDS related complex), keloid formation, scar tissue formation, inflammatory bowel disease, Crohn's disease, ulcerative colitis and pyresis Compounds of Formula (I) are also useful in the treatment of inflammation associated with viral infections, where such viruses are sensitive to upregulation by TNF or will elicit TNF production m vivo The viruses contemplated for treatment herein are those that produce TNF as a result of infection, or those which are sensitive to inhibition, such as by decreased replication, directly or indirectly, by the TNF inhibiting-compounds of Formula (1) Such viruses include, but are not limited to HIV-1, HIV-2 and HIV-3, Cytomegalovirus (CMV), Influenza, adenovirus and the Herpes group of viruses, such as but not limited to, Herpes Zoster and Herpes Simplex Accordingly, in a further aspect, this invention relates to a method of treating a mammal afflicted with a human immunodeficiency virus (HIV) which comprises administering to such mammal an effective TNF inhibiting amount of a compound of Formula (I) or a pharmaceutically acceptable salt thereof
It is also recognized that both IL-6 and IL-8 are produced during rhinovirus (HRV) infections and contribute to the pathogenesis of common cold and exacerbation of asthma associated with HRV infection (Turner et al (1998), Clin Infec Dis , Vol 26, p 840, Teren et al (1997), Am J Respir Crit Care Med vol 155, p l362, Grunberg et al (1997), Am J Respir Crit Care Med 156 609 and Zhu et al, J Clin Invest (1996), 97 421) It has also been demonstrated in vitro that infection of pulmonary epithelial cells with HRV results in production of IL-6 and IL-8 (Subauste et al , J Clin Invest 1995, 96 549 ) Epithelial cells represent the primary site of infection of HRV
Therefore, another another aspect of the present invention relates to the use of a CSBP/p38 kinase inhibitor for the treatment, including prophylaxis, of the common cold, or respiratory viral infection caused by human rhinovirus infection (HRV), other enteroviruses, coronavirus, influenza virus, parainfluenza virus, respiratory syncytial virus, or adenovirus infection in a human in need thereof which method comprises administering to said human an effective amount of a CBSP/p38 inhibitor
Another aspect of the present invention relates to a method of treating, including prophylaxis of influenza induced pneumonia in a human in need thereof which method comprises administering to said human an effective amount of a CBSP/p38 inhibitor
The present invention also relates to the use of the CSBP/p38 kinase inhibitor for the treatment, including prophylaxis, of inflammation associated with a viral infection of a human rhinovirus (HRV), other enteroviruses, coronavirus, influenza virus, parainfluenza virus, respiratory syncytial virus, or adenovirus
In particular, the present invention is directed to the treatment of a viral infection in a human, which is caused by the human rhinovirus (HRV), other enterovirus, coronavirus, influenza virus, parainfluenza virus, respiratory syncytial virus, or an adenovirus In particular the invention is directed to respiratory viral infections that exacerbate asthma (induced by such infections), chronic bronchitis, chronic obstructive pulmonary disease, otitis media, and sinusitis While inhibiting IL-8 or other cytokines may be beneficial in treating a rhinovirus may be known, the use of an inhibitor of the p38 kinase for treating HRV or other respiratory viral infections causing the common cold is believed novel. It should be noted that the respiratory viral infection treated herein may also be associated with a secondary bacterial infection, such as otitis media, sinusitis, or pneumonia
For use herein treatment may include prophylaxis for use in a treatment group susceptible to such infections It may also include reducing the symptoms of, ameliorating the symptoms of, reducing the severity of, reducing the incidence of, or any other change in the condition of the patient, which improves the therapeutic outcome
It should be noted that the treatment herein is not directed to the elimination or treatment of the viral organism itself but is directed to treatment of the respiratory viral infection that exacerbates other diseases or symptoms of disease, such as asthma (induced bv such infections), chronic bronchitis, chronic obstructive pulmonary disease, otitis media and sinusitis It is a method of of treatment including prophylaxis to reduce inflammation associated with a rhinovirus infection and not necessarily a direct effect on virus itself
The CSA1D inhibitors of Formula (I) are useful in the treatment of symptoms associated with HRV, including exacerbations of underlying conditions such as asthma, COPD, sinusitis and otitis media amongst others
A preferred virus for treatment herein is the human rhinovirus infection (HRV) or respiratory syncytial virus (RSV)
For use in the common cold or respiratory viral infections, or in influenza, the CSBP/p38 inhibitors of Formula (I) may also be administered with a second therapeutic agent The second therapeutic agent may be an antiviral agent such as πbaviπn amantidine, πmantidine, Pleconaπl,AG 7088 or BTA-188, it may also be an antiviral agent such as an influenza neuraminidase inhibitor, such as zamamvar (Relenza), oseltamivir (Tamiflu) or RWJ-270201 , it may be an antihistamine, such as Benadryl®, chlorpheneramine and salts thereof, brompheneramme or salts thereof, and the generally accepted non-sedating antihistamines, such as loratadine (Claπtin®), descarboethoxyloratadine (DCL), fexofenadine (Allegra®), and cetiπzine hydrochloride (Zyrtec®) etc , a decongestant, such as phenylpropanolamine and salts thereof, pseudoephedreine or salts thereof, steroids, such as dexamethasone, prednisone, or prenisolone, etc , various antibiotics, such as the quinolones, cephalospoπns, β-lactamase inhibitors, etc , anti-inflammatory agents, such as an NSAID, a COX-1 or COX-2 inhibitor, ASA, or mdomethacin, etc It is recognized that the above noted agents may be administerd as immediate release, or as extended release dosage forms, either together with a suitable CSAID compound, or seperately. The compositions may be administered sequentially, in combination with, or contemporaneously with a CSAID agent. The administration route of the second agent may also differ from that of the CSAID agent, and hence the dosing schedule may vary accordingly. Compounds of Formula (I) may also be used in association with the veterinary treatment of mammals, other than in humans, in need of inhibition of TNF production. TNF mediated diseases for treatment, therapeutically or prophylactically, in animals include disease states such as those noted above, but in particular viral infections. Examples of such viruses include, but are not limited to, lentivirus infections such as, equine infectious anaemia virus, caprine arthritis virus, visna virus, or maedi virus or retrovirus infections, such as but not limited to feline immunodeficiency virus (FIV), bovine immunodeficiency virus, or canine immunodeficiency virus or other retroviral infections.
The compounds of Formula (I) may also be used topically in the treatment or prophylaxis of topical disease states mediated by or exacerbated by excessive cytokine production, such as by IL-1 or TNF respectively, such as inflamed joints, eczema, psoriasis and other inflammatory skin conditions such as sunburn; inflammatory eye conditions including conjunctivitis; pyresis, pain and other conditions associated with inflammation. Periodontal disease has also been implemented in cytokine production, both topically and systemically. Hence use of compounds of Formula (I) to control the inflammation associated with cytokine production in such peroral diseases such as gingivitis and periodontitis is another aspect of the present invention.
Compounds of Formula (I) have also been shown to inhibit the production of IL-8 (Interleukin-8, NAP). Accordingly, in a further aspect, this invention relates to a method of inhibiting the production of IL-8 in a mammal in need thereof which comprises administering to said mammal an effective amount of a compound of Formula (I) or a pharmaceutically acceptable salt thereof.
There are many disease states in which excessive or unregulated IL-8 production is implicated in exacerbating and/or causing the disease. These diseases are characterized by massive neutrophil infiltration such as, psoriasis, inflammatory bowel disease, asthma, cardiac, brain and renal reperfusion injury, adult respiratory distress syndrome, thrombosis and glomerulonephritis. All of these diseases are associated with increased IL-8 production which is responsible for the chemotaxis of neutrophils into the inflammatory site. In contrast to other inflammatory cytokines (IL-1, TNF, and IL-6), IL-8 has the unique property of promoting neutrophil chemotaxis and activation Therefore, the inhibition of IL-8 production would lead to a direct reduction in the neutrophil infiltration
The compounds of Formula (I) are administered in an amount sufficient to inhibit cytokine, in particular IL-1 , IL-6, IL-8 or TNF, production such that it is regulated down to normal levels, or in some case to subnormal levels, so as to ameliorate or prevent the disease state Abnormal levels of IL-1 , IL-6, IL-8 or TNF, for instance in the context of the present invention, constitute (i) levels of free (not cell bound) IL-1, IL-6, IL-8 or TNF greater than or equal to 1 picogram per ml, (ii) any cell associated IL-1 , IL-6, IL-8 or TNF, or (iii) the presence of IL-1, IL-6, IL-8 or TNF mRNA above basal levels in cells or tissues in which IL-1, IL-6, IL-8 or TNF, respectively, is produced
The discovery that the compounds of Formula (I) are inhibitors of cytokines, specifically IL-1 , IL-6, IL-8 and TNF is based upon the effects of the compounds of Formulas (I) on the production of the IL- 1 , IL-8 and TNF in in vitro assays which are described herein
As used herein, the term "inhibiting the production of IL-1 (IL-6, IL-8 or TNF)" refers to a) a decrease of excessive /// vivo levels of the cytokine (IL-1 , IL-6, IL-8 or TNF) in a human to normal or sub-normal levels by inhibition of the /// release of the cytokine by all cells, including but not limited to monocytes or macrophages, b) a down regulation, at the genomic level, of excessive in vivo levels of the cytokine (IL- 1 , IL-6, IL-8 or TNF) in a human to normal or sub-normal levels, c) a down regulation, by inhibition of the direct synthesis of the cytokine (IL- 1 , IL-6, IL-8 or TNF) as a postranslational event, or d) a down regulation, at the translational level, of excessive in vivo levels of the cytokine (IL- 1 , IL-6, IL-8 or TNF) in a human to normal or sub-normal levels
As used herein, the term "TNF mediated disease or disease state" refers to any and all disease states in which TNF plays a role, either by production of TNF itself, or by TNF causing another monokine to be released, such as but not limited to IL-1, IL- 6 or IL-8 A disease state in which, for instance, IL-1 is a major component, and whose production or action, is exacerbated or secreted in response to TNF, would therefore be considered a disease stated mediated by TNF
As used herein, the term "cytokine" refers to any secreted polypeptide that affects the functions of cells and is a molecule which modulates interactions between cells in the immune, inflammatory or hematopoietic response A cytokine includes, but is not limited to, monokines and lymphokines, regardless of which cells produce them. For instance, a monokine is generally referred to as being produced and secreted by a mononuclear cell, such as a macrophage and/or monocyte. Many other cells however also produce monokines, such as natural killer cells, fibroblasts, basophils, neutrophils, endothelial cells, brain astrocytes, bone marrow stromal cells, epideral keratinocytes and B-lymphocytes. Lymphokines are generally referred to as being produced by lymphocyte cells. Examples of cytokines include, but are not limited to, Interleukin-1 (IL-1), lnterleukin-6 (IL-6), Interleukin-8 (IL-8), Tumor Necrosis Factor-alpha (TNF-α) and Tumor Necrosis Factor beta (TNF-β).
As used herein, the term "cytokine interfering" or "cytokine suppressive amount" refers to an effective amount of a compound of Formula (I) which will cause a decrease in the in vivo levels of the cytokine to normal or sub-normal levels, when given to a patient for the prophylaxis or treatment of a disease state which is exacerbated by, or caused by, excessive or unregulated cytokine production.
As used herein, the cytokine referred to in the phrase "inhibition of a cytokine, for use in the treatment of a HIV-infected human" is a cytokine which is implicated in (a) the initiation and/or maintenance of T cell activation and/or activated T cell- mediated HIV gene expression and/or replication and/or (b) any cytokine-mediated disease associated problem such as cachexia or muscle degeneration.
As TNF-β (also known as lymphotoxin) has close structural homology with TNF-α (also known as cachectin) and since each induces similar biologic responses and binds to the same cellular receptor, both TNF-α and TNF-β are inhibited by the compounds of the present invention and thus are herein referred to collectively as "TNF" unless specifically delineated otherwise.
A new member of the MAP kinase family, alternatively termed CSBP, p38, or RK, has been identified independently by several laboratories. Activation of this novel protein kinase via dual phosphorylation has been observed in different cell systems upon stimulation by a wide spectrum of stimuli, such as physicochemical stress and treatment with lipopolysaccharide or proinflammatory cytokines such as interleukin-1 and tumor necrosis factor. The cytokine biosynthesis inhibitors, of the present invention, compounds of Formula (I) have been determined to be potent and selective inhibitors of CSBP/p38/RK kinase activity. These inhibitors are of aid in determining the signaling pathways involvement in inflammatory responses. In particular, for the first time a definitive signal transduction pathway can be prescribed to the action of lipopolysaccharide in cytokine production in macrophages. In addition to those diseases already noted, treatment of stroke, neurotrauma, cardiac and renal reperfusion injury, congestive heart failure, chronic renal failure, angiogenesis & related processes, such as cancer, thrombosis, glomerulonephritis, diabetes and pancreatic b cells, multiple sclerosis, muscle degeneration , eczema, psoriasis, sunburn, and conjunctivitis are also included.
The CSBP inhibitors were subsequently tested in a number of animal models for anti-inflammatory activity Model systems were chosen that were relatively insensitive to cyclooxygenase inhibitors in order to reveal the unique activities of cytokine suppressive agents The inhibitors exhibited significant activity in many such in vivo studies Most notable are its effectiveness in the collagen-induced arthritis model and inhibition of TNF production in the endotoxic shock model. In the latter study, the reduction in plasma level of TNF correlated with survival and protection from endotoxic shock related mortality Also of great importance is the compound effectiveness in inhibiting bone resorption in a rat fetal long bone organ culture system Griswold et al., ( 1988) Arthritis Rheum 31 : 1406-1412, Badger, et al , ( 1989) Circ. Shock 27, 51 -61 , Votta et al., (1994)/// vitro Bone 15, 533-538, Lee et al , (1993) B Ann. N. Y. Acad. Sci 696, 149-170
Chronic diseases which have an inappropriate angiogenic component are various ocular neovasularizations, such as diabetic retinopathy and macular degeneration Other chronic diseases which have an excessive or increased proliferation of vasculature are tumor growth and metastasis, atherosclerosis, and certain arthritic conditions Therefore CSBP kinase inhibitors will be of utility in the blocking of the angiogenic component of these disease states
The term "excessive or increased proliferation of vasculature inappropriate angiogenesis" as used herein includes, but is not limited to, diseases which are characterized by hemangiomas and ocular diseases The term "inappropriate angiogenesis" as used herein includes, but is not limited to, diseases which are characterized by vesicle proliferation with accompanying tissue proliferation, such as occurs in cancer, metastasis, arthritis and atherosclerosis
Another aspect of the present invention is directed to the treatment of inhaled smoke induced airway inflammation, lung chemokine production and cytokine production The invention may be directed to treatment of the airway induced inflammation which is secondary to other respiratory disorders such as viral infections that exacerbate asthma (induced by such infections), chronic bronchitis, chronic obstructive pulmonary disease, otitis media, and sinusitis A respiratory viral infection treated in conjunction with the smoke related airway inflammation may also be associated with a secondary bacterial infection, such as otitis media, sinusitis, or pneumonia.
It is noted that the inflammation may be due to cytokines and chemokine release from neutrophile activation and other leukocytes, as well as vascular and airway endothelial cell activiation.
For use herein treatment may include prophylaxis for use in a treatment group who may be susceptible to such airway inflammation. It may also include reducing the symptoms of, ameliorating the symptoms of, reducing the severity of, reducing the incidence of, or any other change in the condition of the patient, which improves the therapeutic outcome.
Suitable patient populations for whom this may be prophylatically beneficial could be firemen who routinely inhale smoke in the course of their duties; use in the military, and by civilians in wartime exposure.
As noted, smoke of natural causes, such as plant extracts, natural plants products, synthetic material, chemically treated natural materials, or natural products such as oil and gas or other fossil fuels, may be treated within the scope of this invention. Suitably, the treatment including prophylaxis is related to cigarette smoke or synthetic/composites, such as occur in fires associated with buring buildings or homes. Another aspect of the present invention relates to the use of a CSBP/p38 kinase inhibitor for the treatment, including prophylaxis, of the hypertussive activity associated withwith resulting airway inflammation and/or cough in a mammal in need thereof.
The present invention also relates to use of a CSBP/p38 kinase inhibitor for the treatment, including prophylaxis, of the inflammation enhanced cought related disorders in a mammal in need thereof.
The present invention is also directed to the use of a compound of Formula (I) in eosinophilic bronchitis, and in cough variant asthma.
The compounds of Formula (I) may also be used in the treatment, including prophylaxis, of eosinophilic inflammation in the airways and cough. Treatment, including prophylaxis is appropriate for eosinophilic bronchitis (as this differs from asthma) and for the treatment, including prophylaxis of cough variant asthma. These disorders may be directed to treatment of the airway induced inflammation which is secondary to other respiratory disorders such as viral infections that exacerbate asthma (induced by such infections), chronic bronchitis, chronic obstructive pulmonary disease, otitis media, and sinusitis. A respiratory viral infection treated in conjunction with the smoke related airway inflammation may also be associated with a secondary bacterial infection, such as otitis media, sinusitis, or pneumonia The hypertussive or inflammation enhanced cough related disorders may either be a direct result of or an association with eosinophilia activity It may also be a result of, or associated with the blocking production of certain cytokines which may mediate these phenomena
For use herein treatment may include prophylaxis for use in a treatment group who may be susceptible to such airway inflammation, and/or cough It may also include reducing the symptoms of, ameliorating the symptoms of, reducing the severity of, reducing the incidence of, or any other change in the condition of the patient, which improves the therapeutic outcome
Clinically, eosinophilic bronchitis presents as chronic cough and sputum eosinophilia, but without the abnormalities of airway function seen in asthma In contrast to cough in patients without sputum eosinophilia, the cough responds to anti-inflammatory therapy, such as inhaled corticosteroids (Niimi et al , Eosinophilic inflammation in cough variant asthma, European Respiratory Journal 1 1 (5) 1064-9, (1998))
Patients with cough-variant asthma may also have the following criteria. (1) have not been previously diagnosed as having asthma, (2) complain of a cough of at least a 3 -week duration, (3) do not complain of wheezing, shortness of breath, or chest tightness, (4) have normal results of physical examinations, (5) have normal or nearly normal results of spirometry, (6) have evidence of bronchial hyperresponsiveness during bronchoprovocation challenge testing, and (7) have a favorable response to asthma medications (Irwin et al , Interpretation of positive results of a methacholine inhalation challenge and 1 week of inhaled bronchodilator use in diagnosing and treating cough-variant asthma (Archives of Internal Medicine 157(17) 1981-1987, (1997))
Unlike conventional anti-tussive agents, such as codeine or dextromethorphan, a p38 kinase inhibitor appears to have no direct antitussive activity, but reduces the airway eosinophilia and normalizes the hypertussive state Therefore, use of a p38 inhibitor will reduce the added coughs, or hypertussive state, back to a normal level which can be suitably treated with conventional agents and/or therapies as appropriate Use of the p38 inhibitors will allow for the maintenance of patients who are subject to increased cough responsiveness, especially unproductive cough, due to other underlying disorders or treatments This increased cough responsiveness may be modulated, or decreased by use of this innovative anti-inflammatory therapy Accordingly, the present invention provides a method of treating a CSBP kinase mediated disease in a mammal in need thereof, preferably a human, which comprises administering to said mammal, an effective amount of a compound of Formula (I) or a pharmaceutically acceptable salt thereof In order to use a compound of Formula (I) or a pharmaceutically acceptable salt thereof in therapy, it will normally be formulated into a pharmaceutical composition in accordance with standard pharmaceutical practice This invention, therefore, also relates to a pharmaceutical composition comprising an effective, non- toxic amount of a compound of Formula (I) and a pharmaceutically acceptable carrier or diluent
Compounds of Formula (I), pharmaceutically acceptable salts thereof and pharmaceutical compositions incorporating such may conveniently be administered by any of the routes conventionally used for drug administration, for instance, orally, topically, parenterally or by inhalation The compounds of Formula (I) may be administered in conventional dosage forms prepared by combining a compound of Formula (I) with standard pharmaceutical carriers according to conventional procedures The compounds of Formula (I) may also be administered in conventional dosages in combination with a known, second therapeutically active compound These procedures may involve mixing, granulating and compressing or dissolving the ingredients as appropriate to the desired preparation It will be appreciated that the form and character of the pharmaceutically acceptable character or diluent is dictated by the amount of active ingredient with which it is to be combined, the route of administration and other well-known variables The carrier(s) must be "acceptable" in the sense of being compatible with the other ingredients of the formulation and not deleterious to the recipient thereof
The pharmaceutical carrier employed may be, for example, either a solid or liquid Exemplary of solid carriers are lactose, terra alba, sucrose, talc, gelatin, agar, pectin, acacia, magnesium stearate, stearic acid and the like Exemplary of liquid carriers are syrup, peanut oil, olive oil, water and the like Similarly, the carrier or diluent may include time delay material well known to the art, such as glyceryl mono- stearate or glyceryl distearate alone or with a wax
A wide variety of pharmaceutical forms can be employed Thus, if a solid carrier is used, the preparation can be tableted, placed in a hard gelatin capsule in powder or pellet form or in the form of a troche or lozenge The amount of solid carrier will vary widely but preferably will be from about 25mg to about Ig When a liquid carrier is used, the preparation will be in the form of a syrup, emulsion, soft gelatin capsule, sterile injectable liquid such as an ampule or nonaqueous liquid suspension
Compounds of Formula (I) may be administered topically, that is by non- systemic administration This includes the application of a compound of Formula (I) externally to the epidermis or the buccal cavity and the instillation of such a compound into the ear, eye and nose, such that the compound does not significantly enter the blood stream In contrast, systemic administration refers to oral, intravenous, intraperitoneal and intramuscular administration
Formulations suitable for topical administration include liquid or semi-liquid preparations suitable for penetration through the skin to the site of inflammation such as liniments, lotions, creams, ointments or pastes, and drops suitable for administration to the eye, ear or nose The active ingredient may comprise, for topical administration, from 0 001% to 10% w/w, for instance from 1% to 2% by weight of the formulation It may however comprise as much as 10% w/w but preferably will comprise less than 5% w/w, more preferably from 0 1% to 1% w/w of the formulation
Lotions according to the present invention include those suitable for application to the skin or eye An eye lotion may comprise a sterile aqueous solution optionally containing a bactericide and may be prepared by methods similar to those for the preparation of drops Lotions or liniments for application to the skin may also include an agent to hasten drying and to cool the skin, such as an alcohol or acetone, and/or a moisturizer such as glycerol or an oil such as castor oil or arachis oil
Creams, ointments or pastes according to the present invention are semi-solid formulations of the active ingredient for external application They may be made by mixing the active ingredient in finely-divided or powdered form, alone or in solution or suspension in an aqueous or non-aqueous fluid, with the aid of suitable machinery, with a greasy or non-greasy base The base may comprise hydrocarbons such as hard, soft or liquid paraffin, glycerol, beeswax, a metallic soap, a mucilage, an oil of natural origin such as almond, corn, arachis, castor or olive oil, wool fat or its derivatives or a fatty acid such as steric or oleic acid together with an alcohol such as propylene glycol or a macrogel The formulation may incorporate any suitable surface active agent such as an anionic, cationic or non-ionic surfactant such as a sorbitan ester or a polyoxyethylene derivative thereof Suspending agents such as natural gums, cellulose derivatives or inorganic materials such as silicaceous silicas, and other ingredients such as lanolin, may also be included Drops according to the present invention may comprise sterile aqueous or oily solutions or suspensions and may be prepared by dissolving the active ingredient in a suitable aqueous solution of a bactericidal and/or fungi cidal agent and/or any other suitable preservative, and preferably including a surface active agent The resulting solution may then be clarified by filtration, transferred to a suitable container which is then sealed and sterilized by autoclaving or maintaining at 98-100°C for half an hour Alternatively, the solution may be sterilized by filtration and transferred to the container by an aseptic technique Examples of bactericidal and fungicidal agents suitable for inclusion in the drops are phenylmercuric nitrate or acetate (0 002%), benzalkonium chloride (0 01%) and chlorhexidine acetate (0 01%) Suitable solvents for the preparation of an oily solution include glycerol, diluted alcohol and propylene glycol
Compounds of Formula (I) may be administered parenterally, that is by intravenous, intramuscular, subcutaneous intranasal, intrarectal, intravaginal or intraperitoneal administration The subcutaneous and intramuscular forms of parenteral administration are generally preferred Appropriate dosage forms for such administration may be prepared by conventional techniques Compounds of Formula (I) may also be administered by inhalation, that is by intranasal and oral inhalation administration Appropriate dosage forms for such administration, such as an aerosol formulation or a metered dose inhaler, may be prepared by conventional techniques For all methods of use disclosed herein for the compounds of Formula (I), the daily oral dosage regimen will preferably be from about 0 1 to about 80 mg/kg of total body weight, preferably from about 0 2 to 30 mg/kg, more preferably from about 0 5 mg to 15mg The daily parenteral dosage regimen about 0 1 to about 80 mg/kg of total body weight, preferably from about 0 2 to about 30 mg/kg, and more preferably from about 0 5 mg to 15mg/kg The daily topical dosage regimen will preferably be from 0 1 mg to 150 mg, administered one to four, preferably two or three times daily The daily inhalation dosage regimen will preferably be from about 0 01 mg/kg to about 1 mg/kg per day It will also be recognized by one of skill in the art that the optimal quantity and spacing of individual dosages of a compound of Formula (I) or a pharmaceutically acceptable salt thereof will be determined by the nature and extent of the condition being treated, the form, route and site of administration, and the particular patient being treated, and that such optimums can be determined by conventional techniques It will also be appreciated by one of skill in the art that the optimal course of treatment, i e., the number of doses of a compound of Formula (I) or a pharmaceutically ac;eptaDle salt thereof given per day for a defined number of days, can be ascertained by those skilled in the art using conventional course of treatment determination tests
The novel compounds of Formula (I) may also be used in association with the veterinary treatment of mammals, other than humans, in need of inhibition of CSBP/p38 or cytokine inhibition or production In particular, CSBP/p38 mediated diseases for treatment, therapeutically or prophylactically, in animals include disease states such as those noted herein in the Methods of Treatment section, but in particular viral infections Examples of such viruses include, but are not limited to, lentivirus infections such as, equine infectious anaemia virus, caprine arthritis virus, visna virus, or aedi virus or retrovirus infections, such as but not limited to feline immunodeficiency virus (FIV), bovine immunodeficiency virus, or canine immunodeficiency virus or other retroviral infections
The invention will now be described by reference to the following biological examples which are merely illustrative and are not to be construed as a limitation of the scope of the present invention
BIOLOGICAL EXAMPLES
The cytokine-inhibiting effects of compounds of the present invention may be determined by the following in vitro assays Assays for Interleukin - 1 (IL-1), lnterleukin -8 (IL-8 ), and Tumour Necrosis
Factor (TNF) are well known in the art, and may be found in a number of publications, and patents Representative suitable assays for use herein are described in Adams et al , US 5,593,992, whose disclosure is incorporated by reference in its entirety
Interleukin - 1 (IL-1)
Human peripheral blood monocytes are isolated and purified from either fresh blood preparations from volunteer donors, or from blood bank buffy coats, according to the procedure of Colotta et al, J Immunol, 132, 936 (1984) These monocytes (1x10^) are plated in 24-well plates at a concentration of 1-2 million/ml per well The cells are allowed to adhere for 2 hours, after which time non-adherent cells are removed by gentle washing Test compounds are then added to the cells for lh before the addition of lipopolysaccharide (50 ng/ml), and the cultures are incubated at 37°C for an additional 24h At the end of this period, culture supernatants are removed and clarified of cells and all debris Culture supernatants are then immediately assayed for IL-1 biological activity, either by the method of Simon et al , J Immunol Methods, 84, 85, (1985) (based on ability of IL-1 to stimulate a Interleukin 2 producing cell line (EL-4) to secrete IL-2, in concert with A23187 ionophore) or the method of Lee et al , J ImmunoTherapy, 6 (1), 1-12 (1990) (ELISA assay)
In vivo TNF assay: (1) Griswold et al , Drugs Under Exp and Clinical Res .XIX (6). 243-248
(1993); or
(2) Boehm, et al , Journal Of Medicinal Chemistry 39, 3929-3937 (1996) whose disclosures are incorporated by reference herein in their entirety
LPS-induced TNFα Production in Mice and Rats
In order to evaluate in vivo inhibition of LPS-induced TNFα production in rodents, both mice and rats are injected with LPS Mouse Method
Male Balb/c mice from Charles River Laboratories are pretreated (30 minutes) with compound or vehicle After the 30 min pretreat time, the mice are given LPS (lipopolysaccharide from Esherichia coli Serotype 055-85, Sigma
Chemical Co , St Louis, MO) 25 ug/mouse in 25 ul phosphate buffered saline (pH 7 0) intraperitoneally Two hours later the mice are killed by CO2 inhalation and blood samples are collected by exsanguination into heparinized blood collection tubes and stored on ice. The blood samples are centrifuged and the plasma collected and stored at -20°C until assayed for TNFα by ELISA
Rat Method
Male Lewis rats from Charles River Laboratories are pretreated at various times with compound or vehicle After a determined pretreat time, the rats are given
LPS (lipopolysaccharide from Esherichia coli Serotype 055-85, Sigma Chemical Co , St Louis, MO) 3 0 mg/kg intraperitoneally The rats are killed by CO2 inhalation and heparinized whole blood is collected from each rat by cardiac puncture 90 minutes after the LPS injection The blood samples are centrifuged and the plasma collected for analysis by ELISA for TNFα levels
ELISA Method
TNFα levels were measured using a sandwich ELISA, as described in Olivera et al., Circ. Shock, 37, 301-306, (1992), whose disclosure is incorporated by reference in its entirety herein, using a hamster monoclonal antimurine TNFα (Genzyme, Boston, MA) as the capture antibody and a polyclonal rabbit antimurine TNFa (Genzyme) as the second antibody For detection, a peroxidase-conjugated goat antirabbit antibody (Pierce, Rockford, EL) was added, followed by a substrate for peroxidase (1 mg/ml orthophenylenediamine with 1% urea peroxide) TNFα levels in the plasma samples from each animal were calculated from a standard curve generated with recombinant murine TNFα (Genzyme)
LPS-Stimuiated Cytokine Production in Human Whole Blood
Assay Test compound concentrations were prepared at 10 X concentrations and
LPS prepared at 1 ug/ml (final cone of 50 ng/ml LPS) and added in 50 uL volumes to 1 5 mL eppendorf tubes Heparinized human whole blood was obtained from healthy volunteers and was dispensed into eppendorf tubes containing compounds and LPS in 0 4 mL volumes and the tubes incubated at 37 C Following a 4 hour incubation, the tubes were centrifuged at 5000 rpm for 5 minutes in a TOMY microfuge, plasma was withdrawn and frozen at -80 C
Cytokine measurement: IL-I and/or TNF were quantified using a standardized ELISA technology An in-house ELISA kit was used to detect human IL-1 and TNF Concentrations of IL- 1 or TNF were determined from standard curves of the appropriate cytokine and IC50 values for test compound (concentration that inhibited 50%) of LPS-stimulated cytokine production) were calculated by linear regression analysis
CSBP/p38 Kinase Assay:
This assay measures the CSBP/p38-catalyzed transfer of - 2p from [a-32p]ATP to threonine residue in an epidermal growth factor receptor (EGFR)-derived peptide (T669) with the following sequence KRELVEPL7TJSGEAPNQALLR (residues 661 - 681 ) (See Gallagher et al , "Regulation of Stress Induced Cytokine Production by Pyridinyl Imidazoles Inhibition of CSBP Kinase", BioOrganic & Medicinal Chemistry, 1997, 5, 49-64)
Reactions were carried in round bottom 96 well plate (from Corning) in a 30 ml volume Reactions contained (in final concentration). 25 mM Hepes, pH 7 5, 8 mM MgCl2, 0 17 mM ATP (the Km[ATP] of p38 (see Lee et al , Nature 300, n72 pg. 639-746 (Dec 1994)), 2 5 uCi of [g-32P]ATP, 0 2 mM sodium orthovanadate, 1 mM DTT, 0 1% BSA, 10% glycerol, 0 67 mM T669 peptide, and 2-4 nM of yeast- expressed, activated and purified p38 Reactions were initiated by the addition of [gamma-32P]Mg/ATP, and incubated for 25 min at 37 °C Inhibitors (dissolved in DMSO) were incubated with the reaction mixture on ice for 30 minutes prior to adding the 32P-ATP Final DMSO concentration was 0 16% Reactions were terminated by adding 10 ul of 0.3 M phosphoric acid, and phosphorylated peptide was isolated from the reactions by capturing it on p81 phosphocellulose filters.
Filters were washed with 75 mM phosphoric acids, and incorporated 32P was quantified using beta scintillation counter. Under these conditions, the specific activity of p38 was 400-450 pmol/pmol enzyme, and the activity was linear for up to
2 hr of incubation. The kinase activity values were obtained after subtracting values generated in the absence of substrate which were 10-15% of total values.
Representative final compounds of Formula (I), Examples 1 to 3 demonstrated positive inhibitory activity of an IC50 of < 50uM in this assay or a similar assay.
Prostaglandin endoperoxide synthase-2 (PGHS-2) assay:
This assay describes a method for determining the inhibitory effects of compounds of Formula (I) on human PGHS-2 protein expression in LPS stimulated human monocytes. A suitable assay for PGHS-2 protein expression may be found in a number of publications, including US Patent 5,593,992 whose disclosure is incorporated herein by reference.
TNF-a in Traumatic Brain Injury Assay
This assay provides for examination of the expression of tumor necrosis factor mRNA in specific brain regions which follow experimentally induced lateral fluid- percussion traumatic brain injury (TBI) in rats. Since TNF- a is able to induce nerve growth factor (NGF) and stimulate the release of other cytokines from activated astrocytes, this post-traumatic alteration in gene expression of TNF- a plays an important role in both the acute and regenerative response to CNS trauma. A suitable assay may be found in WO 97/35856 whose disclosure is incorporated herein by reference.
CNS Injury model for IL-b mRNA
This assay characterizes the regional expression of interleukin- lβ (IL-lβ) mRNA in specific brain regions following experimental lateral fluid-percussion traumatic brain injury (TBI) in rats. Results from these assays indicate that following TBI, the temporal expression of IL-lβ mRNA is regionally stimulated in specific brain regions. These regional changes in cytokines, such as IL-lβ play a role in the post- traumatic pathologic or regenerative sequelae of brain injury. A suitable assay may be found in WO 97/35856 whose disclosure is incorporated herein by reference. Angiogenesis Assay:
Described in WO 97/32583, whose disclosure is incorporated herein by reference, is an assay for determination of inflammatory angiogenesis which may be used to show that cytokine inhibition will stop the tissue destruction of excessive or inappropriate proliferation of blood vessels
Rhinovirus Methods
Cell lines, rhinovirus serotype 39, and influenza virus A/PR/8/34 are purchased from American Type Culture Collection (ATCC) BEAS-2B cells were cultured according to instructions provided by ATCC using BEGM (bronchial epithelial growth media) purchased from Clonetics Corp HELA cell cultures, used for detection and titration of virus, are maintained in Eagle's minimum essential media containing 10% fetal calf serum, 2mM 1-glutamine, and 10 mM HEPES buffer (MEM) A modification of the method reported by Subauste et al , Supra, for in vitro infection of human bronchial epithelial cells with rhinovirus is used in these studies BEAS-2B cells (2xlONwell) are cultured in collagen-coated wells for 24 hours prior to infection with rhinovirus Rhinovirus serotype 39 is added to cell cultures for one hour incubation at 34°C after which inoculum is replaced with fresh media and cultures are incubated for an additional 72 hours at 34°C Supernatants are collected at 72 hours post-infection and are assayed for cytokine protein concentration by ELISA using commercially available kits (R&D Systems) Virus yield is also determined from culture supernatants using a microtitration assay in HELA cell cultures (Subauste et al., supra 1995) In cultures treated with p38 kinase inhibitors, drug is added 30 minutes prior to infection Stocks of compounds are prepared in DMSO (10 mM drug) and stored at -20°C
For detection of p38 kinase, cultures are incubated in basal media without growth factors and additives to reduce endogenous levels of activated p38 kinase Cells are harvested at various timepoints after addition of rhinovirus Detection of tyrosine phosphorylated p38 kinase by immunoblot is analyzed by a commercially available kit and is performed according to the manufacturer's instructions (PhosphoPlus p38 MAPK Antibody Kit New England BioLabs Inc )
In some experiments, BEAS-2B cells may be infected with influenza virus (strain A/PR/8/34) in place of rhinovirus Culture supernatant is harvested 48 and 72 hour post-infection and tested by ELISA for cytokine as described above Cells and Vims: Influenza A/PR/8/34 sub type H1N1 (VR-95 American Type Culture Collection, Rockville, MD) is grown in the allantoic cavity of 10 day old chicken eggs Following incubation at 37°C, and refrigeration for 2 1/2 hours at 4°C, allantoic fluid is harvested, pooled, and centrifuged (1,000 rcf, 15 min, 4°C) to remove cells Supernatent is aliquoted and stored at -70°C The titer of the stock culture of virus is 1 O x 1010 Tissue Culture Infective Dose/ml (TCID50) Inoculation procedure: Four-six week old female Balb/cAnNcrlBr mice are obtained from Charles River, Raleigh, NC Animals are infected intranasally Mice are anesthetized by intraperitioneal injection of Ketamine (40mg/kg, Fort Dodge Labs, Fort Dodge, la) and Xylazine (5 mg/kg, Miles, Shawnee Mission, Ks) and then inoculated with 100 TCID50 of PR8 diluted in PBS in 20 ul Animals are observed daily for signs of infection
Virus titration: At various times post infection, animals are sacrificed and lungs are aseptically harvested Tissues are homogenized, in vials containing 1 micron glass beads (Biospec Products, Bartlesville, OK) and 1 ml of Eagles minimal essential medium Cell debris is cleared by centrifugation at 1,000 rcf for 15 minutes at 4°C, and supernatants are serially diluted on Madin-Darby canine kidney (MDCK) cells After 5 days of incubation at 37°C (5% CO2), 50 μl of 0 5% chick red blood cells are added per well, and agglutination is read after 1 hour at room temperature The virus titer is expressed as 50% tissue culture infective dose (TCID50) calculated by logistic regression
ELISA: Cytokine levels are measured by quantitative ELISA using commercially available kits Ear samples are homogenized using a tissue minser in PBS Cell debris is cleared by centrifugation at 14,000 rpm for 5 minutes The cytokine concentrations and thresholds are determined as described by the manufacturer, IL-6, IFN-γ, and KC (R&D Systems, Minneapolis, MN)
Myeloperoxidase Assay: Myeloperoxidase (MPO) activity is determined kinetically as described by Bradley et al (1982) Briefly, rabbit cornea are homogenized in Hexadecyl Trimethyl-Ammonium Bromide (HTAB) (Sigma Chemical Co St Louis, Mo) which is dissolved in 0 5 m Potassium phosphate buffer (J T Baker Scientific, Phillipsburg, NJ) Following homogenization, the samples are subjected to freeze- thaw-sonication (Cole-Parmer 8853, Cole-Parmer, Vernon Hills, II) 3 times Suspensions are then cleared by centrifugation at 12,500 x g for 15 minutes at 4°C MPO enzymatic activity is determined by colormetric change in absorbance during a reaction of O-Dianisidine dihydrochloride (ODI) 0 175 mg/ml (Sigma Chemical Co St Louis, Mo) with 0002% Hydrogen peroxide (Sigma Chemical Co St Louis, Mo) Measurements are performed by using a Beckman Du 640 Spectrophotometer (Fullerton, Ca ) fitted with a temperature control device 50 ul of material to be assayed is added to 950 ul of ODI and change in absorbance is measured at a wave length of 460 nm for 2 minutes at 25°C Whole Body Plethysomography: Influenza virus infected mice are placed into a whole body plethysomograph box with an internal volume of approximately 350-ml A bias airflow of one l/min is applied to the box and flow changes were measured and recorded with a Buxco XA data acquisition and respiratory analysis system (Buxco Electronics, Sharon, CT) Animals are allowed to acclimate to the plethysmograph box for 2 min before airflow data is recorded Airway measurements are calculated as Penh (enhanced pause) Penh has previously been shown as an index of airway obstruction and coπelates with increased intrapleural pressure The algorithm for Penh calculation is as follows Penh = [(expiratory time / relaxation time)-l] x (peak expiratory flow / peak inspiratory flow) where relaxation time is the amount of time required for 70% of the tidal volume to be expired
Determination of arterial oxygen saturation. A Nonin veterinary hand held pulse oximeter 8500V with lingual sensor (Nonin Medical, Inc., Plymouth MN) is used to determine daily arterial oxygen saturation %SpO2 as described (Sidwell et al. 1992 Antimicrobial Agents and Chemotherapy 36 473-476)
Results:
Inhibition of cytokine production by specific inhibitors ofp38MAP kinase Consistent with published reports, IL-6, IL-8, and GM-CSF are detected 72 hours post-infection of BEAS-2B cells with rhinovirus-39 (multiplicity of infection, MOI 1 0) (figure 1) Production of IL-6, IL-8, and GM-CSF is not mediated through IL-1 or TNF produced in response to rhinovirus infection since addition of neutralizing antibodies to IL-1 and TNF to the infected cultures did not reduce the amount of IL-
6, IL-8 or GM-CSF produced (not shown) Productive infection of cells is confirmed by titering infectious supernatants from BEAS-2B cells on HELA monolayers There is low but consistent replication of virus during the 72 hour culture period resulting in 1 22 + 0 3 log ιo TCID50 increase over the initial input inoculum (n=6 experiments)
To investigate the role of p38 kinase signal transduction in rhinovirus-induced cytokine production by epithelial cells, specific p38 kinase inhibitors of Formula (I) may be tested for their ability to inhibit cytokine production in the rhinovirus-infected BEAS-2B cell cultures Further data on this method may be found in PCT application USOO/25386, filed September 15, 2000 whose disclosure is incorporated herein by reference in its entirety
Activation ofp38 kinase by rhinovirus infection
The presence of tyrosine phosphorylated p38 kinase is measured by immunoblot at various times after the addition of virus to BEAS-2B cultures Rhinovirus infection of BEAS-2B cells results in an increase in phosphorylated p38 kinase that was both dose and time-dependent Increases in phosphorylated p38 kinase is evident by 15 minutes post exposure to rhinovirus-39 (MOI 10), appeared to peak by 30 minutes after addition of virus and remained elevated 60 minutes post-infection (figure 3) In addition, rhinovirus-induced tyrosine phosphorylation of p38 kinase was dose- dependent When cells are cultured in the absence of virus, there was no increase in the amount of tyrosine phosphorylation of p38 kinase at any of the timepoints tested Overall levels of p38 kinase protein were comparable between all the groups indicating that virus infection caused phosphorylation of p38 kinase without de novo synthesis of protein
Effects on in vitro influenza virus infection Exposure of BEAS-2B cells with influenza virus (A/PR/8/34, MOI 1 0) also results in elaboration of IL-8 and IL-6 as measured 48 - 72 hours post-infection, although the secreted protein levels are lower than that obtained with rhinovirus infection
Cigarette Smoke Exposure Model
A murine model of cigarette smoke inhalation was developed to explore a relationship to leukocyte trafficking and lung chemokine and cytokine production Balb/c mice are exposed to smoke generated from commercial unfiltered cigarettes for a specified period of time and samples are obtained at varying times during the post-exposure This model is demonstrated in greater detail as shown below, in contrast to other smoke extract models known in the art
A model of cigarette smoke exposure in the mouse is established in which mice are placed 6 at a time into a small animal plexiglass dosing chamber attached to a peristaltic pump whose intake is connected to a holder for a commercial unfiltered cigarette (Lucky Strike^M) Along with fresh air, smoke is delivered into the chamber until the cigarette is consumed (approximately 5 minutes) Varying numbers of cigarettes (2-4 per day, 2-3 hr apart) are utilized for 1 -3 consecutive days Animals are euthanized by pentobarbital overdose approximately 18 hours after the final exposure Bronchoalveolar lavage with phosphate-buffered saline is performed for inflammatory cell enumeration, and BAL aliquots and lungs are frozen for cytokine analysis Smoke exposure results in time- and cigarette number-related increases in airway neutrophils, and lung chemokine (KC) and cytokine (IL-6) content
To evaluate the role of a p38 MAP kinase inhibitor in this inflammatory response, mice are treated with a p38 kinase inhibitor, a compound of Formula (I) at approximatetly a 30 mg/kg, p o b i d Reduction in lung KC (a murine homolog of IL-8) levels are assesed 1 day after exposure (prior to neutrophilia), and attenuated airway neutrophilia and lung IL-6 levels are assessed following 3 days of cigarette exposure
Hypertussive Cough Models Described below is an example of how to determine the usefulness of p38 inhibitors in the treatment of hypertussive disorders or inflammation enhanced cough The directed antitussive activity of the compound in question if first assessed, by a 10 to 30 minute pretreatment period by intraperitoneal injection or a 1 hour pretreatment period for oral administration The animals (guinea pigs) are then subjected to an inhaled citric acid-induced cough challenge The Citric Acid Induced Cough Model is shown in Figure 2
The effects of the compound are then assessed on the hypertussive response that occurs 72 hours post aerosol exposure to antigen or LTD4 exposure Treatment of the animals occurs with the drug prior and/or after antigen or LTD4 challenge, but not on the day of citric acid challenge The antigen or LTD4 induced hypertussive model is shown in Figure 3
The effects of known antitussive agents, dextromethorphan and codeine on Citric Acid Induced Cough in Guinea Pigs is shown in Figure 4
Inhalation of citric acid (CA, 0 4% for 1 minute) induces 1 1 to 15 coughs during the exposure and 12 -minute monitoring period in conscious guinea pigs
Exposure of sensitized animals to inhaled ovalbumin resulted in a hypertussive state (50-80% increae in CA-induced cough incidence) for several days, which positively correlated with airway esoinophilia determined by bronchoalveolar lavage
Similarly, inhalation of LTD4 (10 ug/ml for 1 minute) increases cough incidence and airway esoinophils 72 hours after exposure SYNTHETIC EXAMPLES
The invention will now be described by reference to the following examples which are merely illustrative and are not to be construed as a limitation of the scope of the present invention. All temperatures are given in degrees centigrade, all solvents are highest available purity and all reactions run under anhydrous conditions in an argon atmosphere unless otherwise indicated.
In the Examples, all temperatures are in degrees Centigrade (°C). Mass spectra were performed upon a VG Zab mass spectrometer using fast atom bombardment or on a micromass platform electrospray ionization mass spectrometer in the positive ion mode using 95:5 CH3CN/CH3OH with 1% formic acid as the caπier solvent, unless otherwise indicated. 1H-NMR (hereinafter "NMR") spectra were recorded at 250 MHz using a Bruker AM 250 or Am 400 spectrometer. Multiplicities indicated are: s=singlet, d=doublet, t=triplet, q=quartet, m=multiplet and br indicates a broad signal. Sat. indicates a saturated solution, eq indicates the proportion of a molar equivalent of reagent relative to the principal reactant.
Flash chromatography is run over Merck Silica gel 60 (230 - 400 mesh).
Example 1 1 -Phenyl-2-oxo-3 _4-dihydro-6-phenylthioquinazoline a) 5-nitro-2-phenylaminobenzonitrile
2-Chloro-5-nitrobenzonitrile (Aldrich) (3.64grams (hereinafter "g"), 20 millimoles (hereinafter "mmol"), aniline (2 milliliter (hereinafter "mL"), 22 mmol), N- methylpyrollidine ( 10 mL) and diisopropylethylamine (4 mL) were combined and heated to 120° under Ar for 18 hours (hereinafter "h"), cooled, poured into EtOAc (150 mL), washed with H2O (3X 50 mL) and saturated aqueous (hereinafter "satd. aq.) NaCl (50 mL), dried (Na2SO4), concentrated, and the residue was triturated with hexane, filtered and the solid was washed with hexane and dried in vacuo to afford 4.23 g (88%) of the title compound as a light green solid. >H NMR (400) MHz, δ: 8.45 (finely split doublet, 1), 8.19 (dd, 1), 7.48 (m,3), 7.28 (d,2), 7.06 (d, l), 6.96 (br s, l). b) 5-Amino-2-phenylaminobenzonitrile
The product of the preceding example (3.21g, 13.4 mmol), MeOH (200 mL) and 5% platinum, sulfided, (200 mg) was stirred under a balloon of H2 for 6 h, filtered and concentrated to afford 2.80 g (100%).of the title compound as a tan solid. ES+ MS m/z = 210 (MH+). c) 2-Phenylamino-5-phenylsulfanylbenzonitrile
The product of the preceding example (0 523 g, 2 5 mmol) was combined with 48% HBF4 (2 mL) and H2O (2 mL), the mixture was cooled to 4° and then NaNO2 (0 173 g, 2 5 mmol) in H O (2 5 mL) was added and the mixture turned dark Stirred on an ice bath for 20 min, filtered, the solid was dried in vacuo and then placed under an Ar atmosphere, dissolved in DMSO (5 mL) which had previously been deoxygenated with a stream of Ar to afford Solution A containing the 3-cyano-4-phenylamino- benzendiazonium fluoroborate
In a second flask Ar was bubbled through DMSO (10 mL) for 10 min and then thiophenol (1 1 mL, 10 mmol) was added with continued bubbling of Ar into the solution After 10 min the Ar stream was removed and the soln was kept under an Ar atmosphere and 60% NaH (0 36 g, 9 mmol) was added in one portion and the mixture was stirred 15 min to form the thiolate Solution A was added in one portion to the thiolate and the mixture was stirred 16 h, diluted with EtOAc, and the organic phase was washed with H2O, satd aq NaCl, dried (Na2SO ), concentrated, prebsorbed onto ca 10 g silica and chromatographed over an additional 100 g silica with 0-10%> EtOAc in hexane to afford 173 mg of the title compound ES+ MS m/z = 303 (MH+) d) l-Aminomethyl-2-phenylamino-5-phenylsulfanylbenzene
The product of the preceding example (0 138 g, 0 46 mmol) and 1M lithium aluminium hydride in Et2O ( 1 mL, 1 mmol) were combined and heated to Et2O reflux for 30 mm, cooled to 23°, poured into ice cold H2O, and extracted with EtOAc The EtOAc was washed with H2O, satd aq NaCl, dried (Na2SO ) and concentrated to afford the target molecule as a thick light green oil ES+ MS m/z = 307 (MH+) e) 1 -Phenyl-2-oxo-3 ,4-dihvdro-6-phenylfhioquinazoline
All of the product of the preceding example, carbonyldiimidazole (90 mg, 0 552 mmol) and THF were dissolved together and stirred for 3 days, concentrated and chromatographed on reverse phase hplc to afford 35 mg (23% from the product of example 1 c). ES+ MS m/z = 333 (MH+)
Example 2 l-(2.6-Dichlorophenyl)-2-oxo-3,4-dihydro-6-(4-fluorophenyl)thioquinazoline a) 2-(2,6-dichlorophenylamino)-5-nitrobenzonitrile 2,6-Dichloroaniline (2 84 g, 17 5 mmol) was dissolved in DMSO (5 mL) and 60% NaH (0 60 g, 15 mmol) was added The mixture was stirred 30 min and then 2- chloro-5-nitrobenzonitrile (0 91 g, 5 0 mmol) in DMSO (5 mL) was added The reaction darkened and warmed to 50° After cooling to 23° the reaction was diluted with EtOAc (200 mL) and washed with H2O (3X), satd. aq NaCl, and dried (Na SO4), and concentrated to afford 1 45 g (94%) of the title compound as an ivory colored powder ]H NMR (400) MHz, δ 8 49 (finely split doublet, 1), 8 20 (dd, 1), 7 51 (d,2), 7 34 (d, 2), 6 41 (d, l) b) 5-amino-2-(2.6-dichlorophenylamino)-benzonitrile
The product of the preceding example (1 0 g, 3 25 mmol), CH3OH (100 mL) and 5% Pt, sulfided, 5% wt on carbon (100 mg), were combined and the mixture was stirred under a balloon of H2 for 3 h, and then the mixture was diluted with CH2C12 (50 mL) and filtered through a pad of celite The filter pad was washed with 1 1 CH2C12 and CH OH (50 mL) and the combined filtrates were concentrated and the residue was triturated with hexane, filtered and dried in vacuo to afford 0 783 g (86%) of the title compound as a light yellow powder ] H NMR (400) MHz, δ 7 41 (d, 2), 7 17 (t, 1), 6 97 (finely split doublet, 1) 6 85 (m, 1), 6 38 (d, 1) c) 2-(2,6-Dichlorophenylamino)-5-(4-flυorophenylsulfanyl)benzonitrile
The product of the preceding example (0 556 g, 2 0 mmol) was combined with 48% HBF4 (75 mL) and H2O (75 mL), the mixture was cooled to 4° and then NaNO2 (0 173 g, 2 5 mmol) in H2O (2 0 mL) was added and the mixture turned dark Stirred on an ice bath for 20 min, filtered, the solid was dried /// vacuo for 16 h to afford 0 577g of a dry powder which was placed under an Ar atmosphere, dissolved in DMSO (10 mL) which had previously been deoxygenated with a stream of Ar to afford Solution A containing 3-cyano-4-(2,6-dichlorophenylamino)-benzene diazonium fluoroborate In a second flask Ar was bubbled through DMSO (10 mL) for 10 min and then 4-fluorothiophenol (1 1 mL, 10 mmol) was added with continued bubbling of Ar into the solution After 10 min the Ar stream was removed and the soln was kept under an Ar atmosphere and 60% NaH (0 36 g, 9 mmol) was added in one portion and the mixture was stirred 15 min to form the thiolate Solution A was added in one portion to the thiolate and the mixture was stirred 16 h, diluted with EtOAc, and the organic phase was washed with H2O, satd aq NaCl, dried (Na2SO4), concentrated, prebsorbed onto ca 10 g silica and chromatographed over an additional 100 g silica with 0-10%) EtOAc in hexane to afford 156 mg of the title compound. ES+ MS m/z = 389 (MH+) d) l-(2,6-dichlorophenylamino)-2-aminomethyl-4-(4-fluorophenylsulfanyl) benzene The product of the preceding example (0 140 g, 0 36 mmol) and 1M lithium aluminium hydride in Et2O ( 1 mL, 1 mmol) were combined and heated to Et2O reflux for 30 min, cooled to 23°, poured into ice cold H2O, and extracted with EtOAc The EtOAc was washed with H2O, satd aq NaCl, dried (Na2SO4) and concentrated to afford 1 17 mg of the target molecule as an oil ES+ MS m/z = 393 (MH+) e) l-l -(2.6-Dichlorophenyl)-6-(4-fluorophenylsulfanyl)-3,4-dihydro-lH- quinazolin-2-one
The product of the preceding example (1 17 mg, 0 30 mmol) was dissolved in toluene (4 mL) and a 20% solution of phosgene in toluene (2 mL) Pyridine (2 mL) was added and the reaction warmed and a precitpitate formed The mixture was diluted with more toluene ( 10 mL) and was stirred 5 min The reaction was diluted with EtOAc (75 mL) and the organic phase was washed with 10% aq NaOH, H2O, and satd aq NaCl, dried (Na2SO4), concentrated and filtered through a 5 g Varien silica gel column with 1% CH<OH in CH2C12 to afford 64 mg of a brown foam Further purification was achieved by reverse phase chromatography to afford the title compound as a white powder ES+ MS m/z = 419 (MH+)
Example 3 1 -(2,6-Dichlorophenyl)-6-(2,4-difluorophenylsulfanyl)-3,4-dihydro- 1 -H-quinazolin-2- one a) 5-(2,4-di-Fluorophenylsulfanyl)-2-(2,6-dichlorophenylamino)-benzonitrile 3- cyano-4-(2,6-dichlorophenylamino)-benzene diazonium fluoroborate, prepared as described in example 2c, was reacted with the sodium thiolate of 2,4-di- fluorothiophenol prepared as described in example 2c, except that 2,4- difluorothiophenol was the mercaptan in the present example, to afford the title compound as a brown solid ES+ MS m/z = 407 (MH+) b) l -(2,6-di-Chlorophenylamino)-2-aminomethyl-4-(2.4-di-fluorophenylsulfanyl) benzene
The product of the preceding example (0 204 g, 0 52 mmol) was dissolved in THF (4 mL) and 1 M Lithium aluminum hydride in Et2O was added dropwise and then the resulting solution was heated to reflux of the solvent mixture for 5 min The reaction was cooled, carefully diluted with EtOAc, and the organic phase was washed with 10% aq NaOH, H2O satd aq NaCl and dried (Na2SO4), and concentrated to afford 0 212 g of a gummy solid The crude product was purified by elution from a silica column with CH2C12 ES+ MS m/z = 41 1 (MH+) c) l-(2.6-Dichlorophenyl)-6-(2.4-difluorophenylsulfanyl)-3,4-dihydro-l-H- quinazolin-2-one The product of the preceding example was cyclized with phosgene by the method of example 2(e) to afford the title compound as a white powder after reverse phase chromatography. ES+ LC MS m/z = 437 (MH+) (>97%).
All publications, including but not limited to patents and patent applications, cited in this specification are herein incorporated by reference as if each individual publication were specifically and individually indicated to be incoφorated by reference herein as though fully set forth.
The above description fully discloses the invention including preferred embodiments thereof. Modifications and improvements of the embodiments specifically disclosed herein are within the scope of the following claims. Without further elaboration, it is believed that one skilled in the are can, using the preceding description, utilize the present invention to its fullest extent. Therefore, the Examples herein are to be construed as merely illustrative and not a limitation of the scope of the present invention in any way. The embodiments of the invention in which an exclusive property or privilege is claimed are defined as follows.

Claims

What is Claimed is:
1. A compound of the formula :
Figure imgf000043_0001
wherein Rl is a phenyl, naphth-1-yl, naphth-2-yl, heterocyclic or heteroaryl ring, which ring is optionally substituted, independently, by one or more substituents selected from halogen, C i-4 alkyl, halo-substituted-Ci-4 alkyl, cyano, nitro,
(CRi oR20)vNR4Rl4, (CRι0R2θ)vC(Z)NR4Rl4, (CRioR2θ)vC(Z)OR8, (CRi 0R20)vCOR3, (CRi 0R2θ)yC(O)H, SR5, S(O)R5, S(O)2R5, (CRi oR20)vOR8, ZC(Z)Rn, NRιoC(Z)Rn , or N ιoS(O)2R7;
R2 is a C ]_ ιo alkyl, aryl, arylC ι_ιo alkyl, heteroaryl, heteroarylC i . i Q alkyl, heterocyclic, or heterocyclylC I _ J O alkyl moiety, which moieties are optionally substituted one or more times independently with C l . l o alkyl, halosubstituted C ι_ιo alkyl, C2.10 alkenyl, C2-10 alkynyl, C3_7 cycloalkyl, C3.7cycloalkylC 1.10 alkyl, C5_7 cycloalkenyl, C5.7 cycloalkenyl C \ . \ Q alkyl, halogen, (CRι0R2θ)nOR6, (CR10R2θ)nSH, (CRι0R2θ)nS(O)mR7, (CR10R2θ)n HS(O)2R7, (CR10R2θ)nNR4Rl4. (CRι0R2θ)nCN, (CRι0R20)n S(O)2NR4R14, (CRι0R2θ)nC(Z)R6. (CR10R2θ)nOC(Z)R6, (CRιoR2θ)nC(Z)OR6, (CR10R2θ)nC(Z)NR4R14> (CR1 0R2θ)n RlθC(Z)R6, (CR1 0R2θ)n lθC(=NRlθ) R4R14,
(CRι0R20)nOC(Z)NR4R14, (CR,oR2θ)n TMθC(Z) NR4RM, or (CR1 0R2θ)nNRl θC(Z)OR7;
Z is oxygen or sulfur; n is 0, or an integer having a value of 1 to 10; m is 0, or the integer 1 or 2; v is 0, or an integer having a value of 1 or 2; t is an integer having a value of 1 to 3;
R3 is a C i_4 alkyl, halo-substituted C i -4 alkyl, C2-4 alkenyl, C -4 alkynyl, C3-7 cycloalkyl, C5-7 cycloalkenyl, aryl, arylC]-4 alkyl, heteroaryl, heteroarylC 1 -4 alkyl, heterocyclyl, heterocyclylC 1.4 alkyl, (CRιoR2θ)vOR7,
(CRioR20)vS(O)mR7, (CRι0R2θ)vNHS(O)2R7, or (CR10R20 NR4R14; and wherein the aryl, arylalkyl, heteroaryl, heteroaryl alkyl may be optionally substituted; R4 and R14 is each independently selected from hydrogen or optionally substituted
Cl-4 alkyl, optionally substituted aryl or optionally substituted aryl-Ci-4 alkyl, or together with the nitrogen which they are attached form a heterocyclic ring of 5 to 7 members which ring optionally contains an additional heteroatom selected from oxygen, sulfur or NR9; R5 is hydrogen, C i-4 alkyl, C2-4 alkenyl, C2-4 alkynyl or NR4R14, excluding the moieties SR5 being SNR4R14, S(O)2Rs being SO H and S(O)R5 being SOH;
R6 is hydrogen, Ci-io alkyl, C3-7 cycloalkyl, heterocyclyl, heterocyclyl Cj-ioalkyl, aryl, arylCl-io alkyl, heteroaryl or heteroarylCi-io alkyl, wherein these moieties may be optionally substituted; R7 is a Ci-6alkyl, aryl, arylC 1 -6alkyl, heterocyclic, heterocyclylC 1 -6 alkyl, heteroaryl, or heteroarylC 1 -όalkyl moiety; and wherein each of these moieties may be optionally substituted; R8 is hydrogen, Cl-4 alkyl, halo-substituted Ci-4 alkyl, C2-4 alkenyl, C2-4 alkynyl,
C3-7 cycloalkyl, C5-7 cycloalkenyl, aryl, arylCi-4 alkyl, heteroaryl, heteroarylC 1 -4 alkyl, heterocyclyl, heterocyclylC 1 -4 alkyl, (CR] θR2θ)tOR7, (CRioR20)tS(O)mR7, (CRioR20)tNHS(O)2R7, or (CRι0R2θ)tNR4Rl4. and wherein the aryl, arylalkyl, heteroaryl, heteroaryl alkyl may be optionally substituted; R9 is hydrogen, C(Z)R6 or optionally substituted Cj-io alkyl, optionally substituted aryl or optionally substituted aryl-C i-4 alkyl; io and R20 are each independently selected from hydrogen or Cι_4 alkyl;
Rl 1 is a Ci-4 alkyl, halo-substituted C i-4 alkyl, C2-4 alkenyl, C2-4 alkynyl, C3-7 cycloalkyl, C5-7 cycloalkenyl, aryl, arylCi-4 alkyl, heteroaryl, heteroarylC 1.4 alkyl, heterocyclyl, heterocyclylC 1.4 alkyl, (CRιoR2θ)tOR7,
(CRi0R20)tS(O)mR7, (CRi0R20)tNHS(O)2R7, or (CRι0R2θ)vNR4Rl4; and wherein the aryl, arylalkyl, heteroaryl, heteroaryl alkyl may be optionally substituted; or a pharmaceutically acceptable salt thereof.
2. The compound according to Claim 1 wherein Ri is an optionally substituted phenyl, naphth-1-yl, or aphth-2-yl. 3 The compound according to Claim 3 wherein the phenyl or napth-yl is substituted one or more times by halogen, alkyl, hydroxy, alkoxy, amino, or halosubstituted alkyl.
4 The compound according to Claim 3 wherein the phenyl is optionally substituted one or more times by halogen
5 The compound according to Claim 1 wherein R2 is an optionally substituted aryl or arylalkyl
6 The compound according to Claim 5 wherein the aryl is an optionally substituted phenyl
7 The compound according to Claim 6 wherein the phenyl is substituted one or more times by halogen, alkyl, hydroxy, alkoxy, amino, or halosubstituted alkyl
8 The compound according to Claim 7 wherein the phenyl is optionally substituted one or more times by halogen
9 The compound according to Claim 1 which is
I -Phenyl-2-oxo-3 ,4-dihydro-6-phenylthioquinazoline, l-(2,6-Dichlorophenyl)-2-oxo-3,4-dihydro-6-(4-fluorophenyl)thioquinazoline; l-(2,6-Dichlorophenyl)-6-(2,4-difluorophenylsulfanyl)-3,4-dihydro-l -H-quinazolin-2- one, or a pharmaceutically acceptable salt thereof
10 A pharmaceutical composition comprising an effective amount of a compound according to any one of Claims 1 to 9 and a pharmaceutically acceptable carrier or diluent
I I A method of treating a CSBP/RK7p38 kinase mediated disease in a mammal in need thereof, which method comprises administering to said mammal an effective amount of a compound of Formula (I) according to any one of Claims 1 to 9
12 The method according to Claim 9 wherein the CSBP/RK/p38 kinase mediated disease is psoriatic arthritis, Reiter's syndrome, gout, traumatic arthritis, rubella arthritis and acute synovitis, rheumatoid arthritis, rheumatoid spondylitis, osteoarthritis, gouty arthritis and other arthritic condition, sepsis, septic shock, endotoxic shock, gram negative sepsis, toxic shock syndrome, cerebral malaria, meningitis, ischemic and hemorrhagic stroke, neurotrauma/open or closed head injury, asthma, adult respiratory distress syndrome, chronic pulmonary inflammatory disease, chronic obstructive pulmonary disease, silicosis, pulmonary sarcososis, bone resorption disease, osteoporosis, restenosis, cardiac, brain and renal reperfusion injury, thrombosis, glomerularnephritis, chronic renal failure, diabetes, diabetic retinopathy, macular degeneration, graft vs host reaction, allograft rejection, inflammatory bowel disease, Crohn's disease, ulcerative colitis, neurodegenrative disease, muscle degeneration, diabetic retinopathy, macular degeneration, tumor growth and metastasis, angiogenic disease, rhinovirus infection, eczema, contact dermatitis, psoriasis, sunburn, and conjunctivitis
13 A method of treating the common cold or respiratory viral infection caused by human rhinovirus (HRV), other enteroviruses, coronavirus, influenza virus, parainfluenza virus, respiratory syncytial virus, or adenovirus in a human in need thereof which method comprises administering to said human an effective amount of a compound of Formula (I), according to Claim 1
14 The method according to Claim 13 wherein the respiratory viral infection exacerbates asthma, chronic bronchitis, chronic obstructive pulmonary disease, otitis media, or sinusitis
15 A method of treatment, including prophylaxis, of smoke induced airway inflammation in a human in need thereof, which comprises administering to said human an effective amount of a compound according to Claim 1
16 The method according to Claim 15 wherein the smoke induced airway inflammation is caused by inhalation of cigarette smoke, inhalation of smoke produced from a burning plant materiafor inhalation of buring smoke from fossil fuels
17 The method according to Claim 15 or 16 wherein the smoke induced airway inflammation exacerbates a pre-existing asthmatic condition, a pre-existing chronic bronchitis, or pre-existing chronic obstructive pulmonary disease in said human
18 A method of treatment, including prophylaxis, for inflammation enhanced cough in a mammal in need thereof, which comprises administering to said mammal an effective amount of a compound according to Claim 1
19 The method according to Claim 18 wherein the inflammation enhanced cough is cough variant asthma, or eosinophilic bronchitis
20 The method according to any one of Claims 1 1 to 19 wherein the compound of Formula (I) is administered with a second therapeutic agent
21 The method according to Claim 20 wherein the second therapeutic agent is an anti-tussive, an antihistamine, a steroid, a PDE4 agent, an antibiotic, an anti- inflammatory agent selected from an NSAID, a COX-1 or COX-2 inhibitor, ASA, or indomethacin, an antiviral agent ribavirin, amantidine, rimantidine, Pleconaril, AG 7088, or BTA-188, a decongestant, an influenza neuraminidase inhibitor selected from zamanivar (Relenza), oseltamivir (Tamiflu) or RWJ-270201.
22 The method according to any one of Claims 1 1 to 21 wherein the compound of Formula (I) agent is administered orally, topically (intranasal) or via inhalation (aerosol), or both topically and via inhalation
23 The method according to Claim 22 wherein a second therapeutic agent may be administered by a different route than the compound of Formula (I)
24 A process for making a compound according to Claim 1 which process comprises reacting a compound of the formula
Figure imgf000047_0001
wherein R\ and R are as defined herein for Formula (I) with a suitable reducing agent and cyclization agent sufficent to provide a ring closure to yield a compound of Formula (I)
25. The process according to Claim 24 wherein the reducing agent is lithium aluminium hydride (LAH).
26. The process according to Claim 24 wherein the cyclization agent is carbonyldiimidazole, phosgene, triphosgene, or other appropriately activated derivatives of carbonic acid.
PCT/US2000/031894 1999-11-23 2000-11-21 3,4-dihydro-(1h)quinazolin-2-one compounds as csbp/p38 kinase inhibitors WO2001038314A1 (en)

Priority Applications (6)

Application Number Priority Date Filing Date Title
JP2001540077A JP2003514900A (en) 1999-11-23 2000-11-21 3,4-Dihydro- (1H) -quinazolin-2-one compounds as CSBP / p38 kinase inhibitors
DE60020595T DE60020595T2 (en) 1999-11-23 2000-11-21 3,4-DIHYDRO- (1H) CHINAZOLIN-2-ON COMPOUNDS AS CSBP / P38 KINASE INHIBITORS
AU17832/01A AU1783201A (en) 1999-11-23 2000-11-21 3,4-dihydro-(1h)quinazolin-2-one compounds as csbp/p38 kinase inhibitors
US10/129,888 US7053098B1 (en) 1999-11-23 2000-11-21 3,4-Dihydro-(1H) quinazolin-2-one compounds as CSBP/P38 kinase inhibitors
AT00980587T ATE296809T1 (en) 1999-11-23 2000-11-21 3,4-DIHYDRO-(1H)CHINAZOLINE-2-ONE COMPOUNDS AS CSBP/P38 KINASE INHIBITORS
EP00980587A EP1233951B1 (en) 1999-11-23 2000-11-21 3,4-dihydro-(1h)quinazolin-2-one compounds as csbp/p38 kinase inhibitors

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US16697599P 1999-11-23 1999-11-23
US60/166,975 1999-11-23

Publications (1)

Publication Number Publication Date
WO2001038314A1 true WO2001038314A1 (en) 2001-05-31

Family

ID=22605428

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2000/031894 WO2001038314A1 (en) 1999-11-23 2000-11-21 3,4-dihydro-(1h)quinazolin-2-one compounds as csbp/p38 kinase inhibitors

Country Status (8)

Country Link
US (1) US7053098B1 (en)
EP (1) EP1233951B1 (en)
JP (1) JP2003514900A (en)
AT (1) ATE296809T1 (en)
AU (1) AU1783201A (en)
DE (1) DE60020595T2 (en)
ES (1) ES2241675T3 (en)
WO (1) WO2001038314A1 (en)

Cited By (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002050045A1 (en) * 2000-12-18 2002-06-27 Biota Scientific Management Pty Ltd Antiviral agents
EP1248624A1 (en) * 1999-11-23 2002-10-16 SmithKline Beecham Corporation 3,4-DIHYDRO-(1H)-QUINAZOLIN-2-ONES AND THEIR USE AS CSBP/p38 KINASE INHIBITORS
EP1707205A2 (en) 2002-07-09 2006-10-04 Boehringer Ingelheim Pharma GmbH & Co. KG Pharmaceutical compositions of anticholinergics and p38 kinase inhibitors in the treatment of respiratory diseases
WO2006137421A1 (en) * 2005-06-21 2006-12-28 Kyowa Hakko Kogyo Co., Ltd. Agent for topical administration
WO2008011032A1 (en) 2006-07-17 2008-01-24 Amgen Inc. Quinazoline and pyridopyrimidine derivatives as p38 kinase inhibitors
WO2008142031A1 (en) 2007-05-18 2008-11-27 Institut Curie P38alpha as a therapeutic target in bladder carcinoma
US7531545B2 (en) 2005-10-25 2009-05-12 Janssen Pharmaceutica, N.V. 2-amino-3,4-dihydro-pyrido[3,4-d]pyrimidine derivatives useful as inhibitors of β-secretase (BACE)
EP2116245A2 (en) 2004-08-07 2009-11-11 Boehringer Ingelheim International GmbH EGFR kinase inhibitor combinations for treating respiratory and gastrointestinal disorders
US7776882B2 (en) 2006-02-06 2010-08-17 Baxter Ellen W 2-amino-3,4-dihydro-quinoline derivatives useful as inhibitors of β-secretase (BACE)
US7868022B2 (en) 2006-02-06 2011-01-11 Janssen Pharmaceutica Nv 2-amino-quinoline derivatives useful as inhibitors of β-secretase (BACE)
US7932261B2 (en) 2006-02-06 2011-04-26 Janssen Pharmaceutica Nv Macrocycle derivatives useful as inhibitors of β-secretase (BACE)
EP2384751A1 (en) 2004-12-24 2011-11-09 Boehringer Ingelheim International Gmbh Medicaments for the treatment or prevention of fibrotic diseases
US8076358B2 (en) 2008-01-28 2011-12-13 Janssen Pharmaceutica Nv 6-substituted-thio-2-amino-quinoline derivatives useful as inhibitors of β-secretase (BACE)
US8383637B2 (en) 2004-08-06 2013-02-26 Jansssen Pharmaceutica N.V. 2-amino-quinazoline derivatives useful as inhibitors of β-secretase (BACE)
US8426429B2 (en) 2004-08-06 2013-04-23 Jansssen Pharmaceutica N.V. 2-amino-quinazoline derivatives useful as inhibitors of β-secretase (BACE)
US8436006B2 (en) 2004-08-06 2013-05-07 Jansssen Pharmaceutica N.V. 2-amino-quinazoline derivatives useful as inhibitors of β-secretase (BACE)
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
US11291659B2 (en) 2017-10-05 2022-04-05 Fulcrum Therapeutics, Inc. P38 kinase inhibitors reduce DUX4 and downstream gene expression for the treatment of FSHD
US11452713B2 (en) 2016-02-29 2022-09-27 University Of Florida Research Foundation, Incorporated Chemotherapeutic methods for treating low-proliferative disseminated tumor cells

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8916704B1 (en) 2013-10-02 2014-12-23 King Fahd University Of Petroleum And Minerals Dihydroquinone derivatives of piperidine and piperazine

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1999064400A1 (en) * 1998-06-12 1999-12-16 Vertex Pharmaceuticals Incorporated INHIBITORS OF p38

Family Cites Families (83)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2833779A (en) 1956-10-29 1958-05-06 American Cyanamid Co Substituted pyrazoles
US3707475A (en) 1970-11-16 1972-12-26 Pfizer Antiinflammatory imidazoles
US3940486A (en) 1971-05-10 1976-02-24 Ciba-Geigy Corporation Imidazole derivatives in the treatment of pain
US3929807A (en) 1971-05-10 1975-12-30 Ciba Geigy Corp 2-Substituted-4(5)-(aryl)-5(4)-(2,3 or -4-pyridyl)-imidazoles
US3703475A (en) 1971-06-21 1972-11-21 Continental Oil Co Preparation of oil soluble sulfonic acids useful as dispersant additives
US4058614A (en) 1973-12-04 1977-11-15 Merck & Co., Inc. Substituted imidazole compounds and therapeutic compositions therewith
US4199592A (en) 1978-08-29 1980-04-22 E. I. Du Pont De Nemours And Company Antiinflammatory 4,5-diaryl-2-nitroimidazoles
GR75287B (en) 1980-07-25 1984-07-13 Ciba Geigy Ag
WO1983002613A1 (en) 1981-07-20 1983-08-04 Sallmann, Alfred Trisubstituted oxazo compounds
US4503065A (en) 1982-08-03 1985-03-05 E. I. Du Pont De Nemours And Company Antiinflammatory 4,5-diaryl 1-2-halo imidazoles
JPS60226882A (en) 1984-04-24 1985-11-12 Nippon Zoki Pharmaceut Co Ltd Novel pyrimidopyrimidine derivative
US4565875A (en) 1984-06-27 1986-01-21 Fmc Corporation Imidazole plant growth regulators
US4686231A (en) 1985-12-12 1987-08-11 Smithkline Beckman Corporation Inhibition of 5-lipoxygenase products
IL83467A0 (en) 1986-08-15 1988-01-31 Fujisawa Pharmaceutical Co Imidazole derivatives,processes for their preparation and pharmaceutical compositions containing the same
JPH05508153A (en) 1990-06-12 1993-11-18 スミスクライン・ビーチャム・コーポレイション Inhibition of diseases mediated by the 5-lipoxygenase and cyclooxygenase pathways
FR2665898B1 (en) 1990-08-20 1994-03-11 Sanofi DERIVATIVES OF AMIDO-3 PYRAZOLE, PROCESS FOR THEIR PREPARATION AND PHARMACEUTICAL COMPOSITIONS CONTAINING THEM.
IE913473A1 (en) 1990-10-15 1992-04-22 Fujisawa Pharmaceutical Co Quinazoline derivatives and their preparation
JPH06504779A (en) 1990-12-13 1994-06-02 スミスクライン・ビーチャム・コーポレイション New CSAIDS
EP0563286A4 (en) 1990-12-13 1995-01-11 Smithkline Beecham Corp Novel csaids
JPH06502178A (en) 1990-12-31 1994-03-10 藤沢薬品工業株式会社 imidazotriazine derivatives
JP3375659B2 (en) 1991-03-28 2003-02-10 テキサス インスツルメンツ インコーポレイテツド Method of forming electrostatic discharge protection circuit
IL102764A0 (en) 1991-08-16 1993-01-31 Merck & Co Inc Quinazoline derivatives,and pharmaceutical compositions containing them
US5716972A (en) 1993-01-13 1998-02-10 Smithkline Beecham Corporation Pyridyl substituted imidazoles
US6008235A (en) 1992-01-13 1999-12-28 Smithkline Beecham Corporation Pyridyl substituted imidazoles
EP0624159B1 (en) 1992-01-13 1998-11-25 Smithkline Beecham Corporation Pyridyl substituted imidazoles
US5656644A (en) 1994-07-20 1997-08-12 Smithkline Beecham Corporation Pyridyl imidazoles
US5916891A (en) 1992-01-13 1999-06-29 Smithkline Beecham Corporation Pyrimidinyl imidazoles
MX9300141A (en) 1992-01-13 1994-07-29 Smithkline Beecham Corp NOVEL IMIDAZOLE COMPOUNDS, PROCEDURE FOR THE PREPARATION AND PHARMACEUTICAL COMPOSITIONS CONTAINING IT.
GB9303993D0 (en) 1993-02-26 1993-04-14 Fujisawa Pharmaceutical Co New heterocyclic derivatives
US5670527A (en) 1993-07-16 1997-09-23 Smithkline Beecham Corporation Pyridyl imidazole compounds and compositions
US5593992A (en) 1993-07-16 1997-01-14 Smithkline Beecham Corporation Compounds
IL110296A (en) 1993-07-16 1999-12-31 Smithkline Beecham Corp Imidazole compounds process for their preparation and pharmaceutical compositions containing them
US5593991A (en) 1993-07-16 1997-01-14 Adams; Jerry L. Imidazole compounds, use and process of making
WO1995003297A1 (en) 1993-07-21 1995-02-02 Smithkline Beecham Corporation Imidazoles for treating cytokine mediated disease
ATE186551T1 (en) 1993-09-17 1999-11-15 Smithkline Beecham Corp DRUG BINDING PROTEIN
US5869043A (en) 1993-09-17 1999-02-09 Smithkline Beecham Corporation Drug binding protein
US5783664A (en) 1993-09-17 1998-07-21 Smithkline Beecham Corporation Cytokine suppressive anit-inflammatory drug binding proteins
PL179417B1 (en) 1993-10-01 2000-09-29 Novartis Ag Pharmacologically active derivatives of pyridine and methods of obtaining them
ATE325113T1 (en) 1993-10-01 2006-06-15 Novartis Pharma Gmbh PHARMACOLOGICALLY ACTIVE PYRIMIDINE DERIVATIVES AND METHOD FOR THE PRODUCTION THEREOF
US5543520A (en) 1993-10-01 1996-08-06 Ciba-Geigy Corporation Pyrimidine derivatives
JPH08503971A (en) 1993-10-01 1996-04-30 チバ−ガイギー アクチェンゲゼルシャフト Pyrimidineamine derivatives and methods for their preparation
AU1046895A (en) 1993-11-05 1995-05-23 Merck & Co., Inc. New quinazolines as inhibitors of hiv reverse transcriptase
WO1995013067A1 (en) 1993-11-08 1995-05-18 Smithkline Beecham Corporation Oxazoles for treating cytokine mediated diseases
US5559137A (en) 1994-05-16 1996-09-24 Smithkline Beecham Corp. Compounds
US5545669A (en) 1994-06-02 1996-08-13 Adams; Jerry L. Anti-inflammatory compounds
PL321292A1 (en) 1995-01-12 1997-11-24 Smithkline Beecham Corp Novel xchemical compounds
IL118544A (en) 1995-06-07 2001-08-08 Smithkline Beecham Corp Imidazole derivatives, process for their preparation and pharmaceutical compositions comprising them
US5739143A (en) 1995-06-07 1998-04-14 Smithkline Beecham Corporation Imidazole compounds and compositions
WO1997005878A1 (en) 1995-08-10 1997-02-20 Merck & Co., Inc. 2,5-substituted aryl pyrroles, compositions containing such compounds and methods of use
EP0863757B1 (en) 1995-08-10 2003-03-26 Merck & Co., Inc. 2-substituted aryl pyrroles, compositions containing such compounds and methods of use
KR19990064117A (en) 1995-10-06 1999-07-26 폴락 돈나 엘. Substituted imidazole with anticancer activity and cytokine inhibitory activity
AU7482396A (en) 1995-10-31 1997-05-22 Merck & Co., Inc. Substituted aryl pyrroles, compositions containing such compounds and methods of use
AU7529096A (en) 1995-10-31 1997-05-22 Merck & Co., Inc. Substituted aryl pyrroles, compositions containing such compounds and methods of use
EP0859771A4 (en) 1995-10-31 2000-03-15 Merck & Co Inc Substituted pyridyl pyrroles, compositions containing such compounds and methods of use
ZA9610687B (en) 1995-12-22 1997-09-29 Smithkline Beecham Corp Novel synthesis.
ZA97175B (en) 1996-01-11 1997-11-04 Smithkline Beecham Corp Novel substituted imidazole compounds.
WO1997025047A1 (en) 1996-01-11 1997-07-17 Smithkline Beecham Corporation Novel cycloalkyl substituded imidazoles
AP9700912A0 (en) 1996-01-11 1997-01-31 Smithkline Beecham Corp Novel cycloalkyl substituted imidazoles
HUP9902460A3 (en) 1996-01-11 2000-03-28 Smithkline Beecham Corp Novel substituted imidazole compounds, their use, method for their preparation and pharmaceutical compositions containing them
US6372741B1 (en) 1996-03-08 2002-04-16 Smithkline Beecham Corporation Use of CSAID™ compounds as inhibitors of angiogenesis
JP2000506532A (en) * 1996-03-13 2000-05-30 スミスクライン・ビーチャム・コーポレイション Novel pyrimidine compounds useful in treating cytokine-mediated diseases
US6096739A (en) 1996-03-25 2000-08-01 Smithkline Beecham Corporation Treatment for CNS injuries
EP0889887A4 (en) 1996-03-25 2003-06-11 Smithkline Beecham Corp Novel treatment for cns injuries
WO1997036587A1 (en) 1996-04-03 1997-10-09 Merck & Co., Inc. A method of treating cancer
US6875769B2 (en) 1996-05-23 2005-04-05 Pfizer Inc. Substituted6,6-hetero-bicyclicderivatives
WO1997047618A1 (en) 1996-06-10 1997-12-18 Merck & Co., Inc. Substituted imidazoles having cytokine inhibitory activity
SI0912518T1 (en) 1996-07-18 2003-12-31 Merck Frosst Canada & Co. Substituted pyridines as selective cyclooxygenase-2 inhibitors
WO1998006715A1 (en) 1996-08-09 1998-02-19 Smithkline Beecham Corporation Novel piperazine containing compounds
WO1998007425A1 (en) 1996-08-21 1998-02-26 Smithkline Beecham Corporation Imidazole compounds, compositions and use
WO1998016230A1 (en) 1996-10-17 1998-04-23 Smithkline Beecham Corporation Methods for reversibly inhibiting myelopoiesis in mammalian tissue
ES2215242T3 (en) 1996-11-19 2004-10-01 Amgen Inc. ANTI-INFLAMMATORY AGENTS OF CONDENSED PIRROL REPLACED WITH ARILO AND HETEROARILO.
AU730295B2 (en) 1996-11-20 2001-03-01 Merck & Co., Inc. Triaryl substituted imidazoles as glucagon antagonists
WO1998024780A2 (en) 1996-12-05 1998-06-11 Amgen Inc. Substituted pyrimidinone and pyridinone compounds and their use
ZA9711092B (en) 1996-12-11 1999-07-22 Smithkline Beecham Corp Novel compounds.
US6147080A (en) 1996-12-18 2000-11-14 Vertex Pharmaceuticals Incorporated Inhibitors of p38
WO1998028292A1 (en) * 1996-12-23 1998-07-02 Smithkline Beecham Corporation Novel piperidine containing compounds
US5929076A (en) 1997-01-10 1999-07-27 Smithkline Beecham Corporation Cycloalkyl substituted imidazoles
CA2306077A1 (en) 1997-10-08 1999-04-15 Smithkline Beecham Corporation Novel cycloalkenyl substituted compounds
AU1924699A (en) 1997-12-19 1999-07-12 Smithkline Beecham Corporation Compounds of heteroaryl substituted imidazole, their pharmaceutical compositionsand uses
AU769989B2 (en) 1998-10-23 2004-02-12 F. Hoffmann-La Roche Ag Bicyclic nitrogen heterocycles
EP1233950B1 (en) * 1999-11-23 2005-10-05 Smithkline Beecham Corporation 3,4-DIHYDRO-(1H)QUINAZOLIN-2-ONE COMPOUNDS AS CSBP/P39 kINASE INHIBITORS
WO2001038312A1 (en) * 1999-11-23 2001-05-31 Smithkline Beecham Corporation 3,4-DIHYDRO-(1H)QUINAZOLIN-2-ONE COMPOUNDS AS CSBP/p38 KINASE INHIBITORS
EP1248624A4 (en) * 1999-11-23 2003-01-22 Smithkline Beecham Corp 3,4-DIHYDRO-(1H)-QUINAZOLIN-2-ONES AND THEIR USE AS CSBP/p38 KINASE INHIBITORS

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1999064400A1 (en) * 1998-06-12 1999-12-16 Vertex Pharmaceuticals Incorporated INHIBITORS OF p38

Cited By (31)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1248624A4 (en) * 1999-11-23 2003-01-22 Smithkline Beecham Corp 3,4-DIHYDRO-(1H)-QUINAZOLIN-2-ONES AND THEIR USE AS CSBP/p38 KINASE INHIBITORS
EP1248624A1 (en) * 1999-11-23 2002-10-16 SmithKline Beecham Corporation 3,4-DIHYDRO-(1H)-QUINAZOLIN-2-ONES AND THEIR USE AS CSBP/p38 KINASE INHIBITORS
US7829705B2 (en) 2000-12-18 2010-11-09 Biota Scientific Management Pty. Ltd. Antiviral agents
US7951955B2 (en) 2000-12-18 2011-05-31 Biota Scientific Management Pty Ltd Antiviral agents
US8217171B2 (en) 2000-12-18 2012-07-10 Biota Scientific Management Pty. Ltd. Antiviral agents
US7166604B2 (en) 2000-12-18 2007-01-23 Biota Scientific Management Pty Ltd Antiviral agents
WO2002050045A1 (en) * 2000-12-18 2002-06-27 Biota Scientific Management Pty Ltd Antiviral agents
US8624025B2 (en) 2000-12-18 2014-01-07 Biota Scientific Management Pty Ltd Antiviral agents
US7579465B2 (en) 2000-12-18 2009-08-25 Biota Scientific Managment Pty. Ltd. Antiviral agents
EP1707205A2 (en) 2002-07-09 2006-10-04 Boehringer Ingelheim Pharma GmbH & Co. KG Pharmaceutical compositions of anticholinergics and p38 kinase inhibitors in the treatment of respiratory diseases
US8436006B2 (en) 2004-08-06 2013-05-07 Jansssen Pharmaceutica N.V. 2-amino-quinazoline derivatives useful as inhibitors of β-secretase (BACE)
US8426429B2 (en) 2004-08-06 2013-04-23 Jansssen Pharmaceutica N.V. 2-amino-quinazoline derivatives useful as inhibitors of β-secretase (BACE)
US8383637B2 (en) 2004-08-06 2013-02-26 Jansssen Pharmaceutica N.V. 2-amino-quinazoline derivatives useful as inhibitors of β-secretase (BACE)
EP2116245A2 (en) 2004-08-07 2009-11-11 Boehringer Ingelheim International GmbH EGFR kinase inhibitor combinations for treating respiratory and gastrointestinal disorders
EP2384751A1 (en) 2004-12-24 2011-11-09 Boehringer Ingelheim International Gmbh Medicaments for the treatment or prevention of fibrotic diseases
EP2878297A1 (en) 2004-12-24 2015-06-03 Boehringer Ingelheim International GmbH Medicaments for the treatment or prevention of fibrotic diseases
WO2006137421A1 (en) * 2005-06-21 2006-12-28 Kyowa Hakko Kogyo Co., Ltd. Agent for topical administration
US7531545B2 (en) 2005-10-25 2009-05-12 Janssen Pharmaceutica, N.V. 2-amino-3,4-dihydro-pyrido[3,4-d]pyrimidine derivatives useful as inhibitors of β-secretase (BACE)
US7932261B2 (en) 2006-02-06 2011-04-26 Janssen Pharmaceutica Nv Macrocycle derivatives useful as inhibitors of β-secretase (BACE)
US7868022B2 (en) 2006-02-06 2011-01-11 Janssen Pharmaceutica Nv 2-amino-quinoline derivatives useful as inhibitors of β-secretase (BACE)
US7776882B2 (en) 2006-02-06 2010-08-17 Baxter Ellen W 2-amino-3,4-dihydro-quinoline derivatives useful as inhibitors of β-secretase (BACE)
AU2007275696B2 (en) * 2006-07-17 2011-11-10 Amgen Inc. Quinazoline and pyridopyrimidine derivatives as p38 kinase inhibitors
US7935709B2 (en) 2006-07-17 2011-05-03 Amgen Inc. 2-quinazolinone compounds and methods of use
WO2008011032A1 (en) 2006-07-17 2008-01-24 Amgen Inc. Quinazoline and pyridopyrimidine derivatives as p38 kinase inhibitors
WO2008142031A1 (en) 2007-05-18 2008-11-27 Institut Curie P38alpha as a therapeutic target in bladder carcinoma
US8076358B2 (en) 2008-01-28 2011-12-13 Janssen Pharmaceutica Nv 6-substituted-thio-2-amino-quinoline derivatives useful as inhibitors of β-secretase (BACE)
US11452713B2 (en) 2016-02-29 2022-09-27 University Of Florida Research Foundation, Incorporated Chemotherapeutic methods for treating low-proliferative disseminated tumor cells
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
US10537560B2 (en) 2017-10-05 2020-01-21 Fulcrum Therapeutics. Inc. P38 kinase inhibitors reduce DUX4 and downstream gene expression for the treatment of FSHD
US11291659B2 (en) 2017-10-05 2022-04-05 Fulcrum Therapeutics, Inc. P38 kinase inhibitors reduce DUX4 and downstream gene expression for the treatment of FSHD
US11479770B2 (en) 2017-10-05 2022-10-25 Fulcrum Therapeutics, Inc. Use of p38 inhibitors to reduce expression of DUX4

Also Published As

Publication number Publication date
ES2241675T3 (en) 2005-11-01
AU1783201A (en) 2001-06-04
US7053098B1 (en) 2006-05-30
ATE296809T1 (en) 2005-06-15
JP2003514900A (en) 2003-04-22
EP1233951A1 (en) 2002-08-28
DE60020595D1 (en) 2005-07-07
EP1233951A4 (en) 2003-05-21
EP1233951B1 (en) 2005-06-01
DE60020595T2 (en) 2006-03-16

Similar Documents

Publication Publication Date Title
US7629462B2 (en) Tetrasubstituted pyrimidine compounds as chemical intermediates
US7053098B1 (en) 3,4-Dihydro-(1H) quinazolin-2-one compounds as CSBP/P38 kinase inhibitors
US6774127B2 (en) Pyrazole and pyrazoline substituted compounds
US6362193B1 (en) Cycloalkenyl substituted compounds
EP1265900B1 (en) 1,5-disubstituted-3,4-dihydro-1h-pyrimido 4,5-d|pyrimidin-2-one compounds and their use in treating csbp/p38 kinase mediated diseases
US6982270B1 (en) 3,4-dihydro-(1H)quinazolin-2-one compounds as CSBP/p38 kinase inhibitors
AU2001239992A1 (en) 1,5-disubstituted-3,4-dihydro-1H-pyrimido[4,5-D]pyrimidin-2- one compounds and their use in treating CSBP/P38 kinase mediated diseases
US6861417B2 (en) Pyridin-4-YL or pyrimidin-4-YL substituted pyrazines
US6469018B1 (en) Compounds
EP1248624A1 (en) 3,4-DIHYDRO-(1H)-QUINAZOLIN-2-ONES AND THEIR USE AS CSBP/p38 KINASE INHIBITORS
US7629350B2 (en) Compounds
EP1235814B1 (en) 3,4-DIHYDRO-(1H)QUINAZOLIN-2-ONE COMPOUNDS AS CSBP/p38 KINASE INHIBITORS
US6759410B1 (en) 3,4-dihydro-(1H)-quinazolin-2-ones and their use as CSBP/p38 kinase inhibitors
EP1842851A2 (en) Novel compounds
NZ520906A (en) 2(alkyl)-, 5(4-pyrimidinyl)- substituted imidazole derivatives and pharmaceuticals thereof

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AE AL AU BA BB BG BR BZ CA CN CZ DZ EE GE GH GM HR HU ID IL IN IS JP KP KR LC LK LR LT LV MA MG MK MN MX MZ NO NZ PL RO SG SI SK SL TR TT TZ UA US UZ VN YU ZA

AL Designated countries for regional patents

Kind code of ref document: A1

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

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

Ref document number: 10129888

Country of ref document: US

ENP Entry into the national phase

Ref country code: JP

Ref document number: 2001 540077

Kind code of ref document: A

Format of ref document f/p: F

WWE Wipo information: entry into national phase

Ref document number: 2000980587

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 2000980587

Country of ref document: EP

WWG Wipo information: grant in national office

Ref document number: 2000980587

Country of ref document: EP