US20070020704A1 - Diaryl ureas with kinase inhibiting activity - Google Patents

Diaryl ureas with kinase inhibiting activity Download PDF

Info

Publication number
US20070020704A1
US20070020704A1 US10/571,100 US57110006A US2007020704A1 US 20070020704 A1 US20070020704 A1 US 20070020704A1 US 57110006 A US57110006 A US 57110006A US 2007020704 A1 US2007020704 A1 US 2007020704A1
Authority
US
United States
Prior art keywords
phenyl
compound
group
independently selected
canceled
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US10/571,100
Other languages
English (en)
Inventor
Scott Wilhelm
Jacques Dumas
Gaetan Ladouceur
Mark Lynch
William Scott
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Bayer Healthcare LLC
Original Assignee
Bayer Pharmaceuticals Corp
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=33545309&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20070020704(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Bayer Pharmaceuticals Corp filed Critical Bayer Pharmaceuticals Corp
Priority to US10/571,100 priority Critical patent/US20070020704A1/en
Assigned to BAYER PHARMACEUTICALS CORPORATION reassignment BAYER PHARMACEUTICALS CORPORATION ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: SCOTT, WILLIAM J., WHILHELM, SCOTT, LADOCUEUR, GAETAN, DUMAS, JAXQUES, LYNCH, MARK
Publication of US20070020704A1 publication Critical patent/US20070020704A1/en
Assigned to BAYER HEALTHCARE LLC reassignment BAYER HEALTHCARE LLC ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: BAYER PHARMACEUTICALS CORPORATION
Assigned to BAYER HEALTHCARE LLC reassignment BAYER HEALTHCARE LLC ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: BAYER PHARMACEUTICALS CORPORATION
Abandoned legal-status Critical Current

Links

Images

Classifications

    • 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
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/16Amides, e.g. hydroxamic acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/16Amides, e.g. hydroxamic acids
    • A61K31/165Amides, e.g. hydroxamic acids having aromatic rings, e.g. colchicine, atenolol, progabide
    • 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
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4412Non condensed pyridines; Hydrogenated derivatives thereof having oxo groups directly attached to the heterocyclic ring
    • 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
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4418Non condensed pyridines; Hydrogenated derivatives thereof having a carbocyclic group directly attached to the heterocyclic ring, e.g. cyproheptadine
    • 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
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/443Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a five-membered ring with oxygen as a ring hetero atom
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/66Phosphorus compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L31/00Materials for other surgical articles, e.g. stents, stent-grafts, shunts, surgical drapes, guide wires, materials for adhesion prevention, occluding devices, surgical gloves, tissue fixation devices
    • A61L31/14Materials characterised by their function or physical properties, e.g. injectable or lubricating compositions, shape-memory materials, surface modified materials
    • A61L31/16Biologically active materials, e.g. therapeutic substances
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/02Stomatological preparations, e.g. drugs for caries, aphtae, periodontitis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/08Drugs for disorders of the urinary system of the prostate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/10Drugs for disorders of the urinary system of the bladder
    • 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
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • A61P19/10Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease for osteoporosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • 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
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • A61P27/06Antiglaucoma agents or miotics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • 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
    • 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
    • A61P31/04Antibacterial agents
    • A61P31/06Antibacterial agents for tuberculosis
    • 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
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/20Antivirals for DNA viruses
    • A61P31/22Antivirals for DNA viruses for herpes viruses
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P33/00Antiparasitic agents
    • A61P33/02Antiprotozoals, e.g. for leishmaniasis, trichomoniasis, toxoplasmosis
    • A61P33/06Antimalarials
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P39/00General protective or antinoxious agents
    • A61P39/02Antidotes
    • 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
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/14Drugs for disorders of the endocrine system of the thyroid hormones, e.g. T3, T4
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/02Antithrombotic agents; Anticoagulants; Platelet aggregation inhibitors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/06Antianaemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/04Inotropic agents, i.e. stimulants of cardiac contraction; Drugs for heart failure
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/08Vasodilators for multiple indications
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/12Antihypertensives
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/502Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics for testing non-proliferative effects
    • G01N33/5041Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics for testing non-proliferative effects involving analysis of members of signalling pathways
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57407Specifically defined cancers
    • G01N33/5743Specifically defined cancers of skin, e.g. melanoma
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57407Specifically defined cancers
    • G01N33/57438Specifically defined cancers of liver, pancreas or kidney
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L2300/00Biologically active materials used in bandages, wound dressings, absorbent pads or medical devices
    • A61L2300/20Biologically active materials used in bandages, wound dressings, absorbent pads or medical devices containing or releasing organic materials
    • A61L2300/204Biologically active materials used in bandages, wound dressings, absorbent pads or medical devices containing or releasing organic materials with nitrogen-containing functional groups, e.g. aminoxides, nitriles, guanidines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L2300/00Biologically active materials used in bandages, wound dressings, absorbent pads or medical devices
    • A61L2300/40Biologically active materials used in bandages, wound dressings, absorbent pads or medical devices characterised by a specific therapeutic activity or mode of action
    • A61L2300/416Anti-neoplastic or anti-proliferative or anti-restenosis or anti-angiogenic agents, e.g. paclitaxel, sirolimus

Definitions

  • Raf kinase a downstream effector of ras, is a key mediator of these signals from cell surface receptors to the cell nucleus (Lowy, D. R. and Willumsen, B. M. Ann. Rev. Biochem. 1993, 62, 851; Bos, J. L. Cancer Res. 1989, 49, 4682).
  • tumor cells require a functional stroma, a support structure consisting of fibroblast, smooth muscle cells, endothelial cells, extracellular matrix proteins, and soluble factors (Folkman, J., Semin Oncol, 2002. 29(6 Suppl 16), 15-8).
  • Tumors induce the formation of stromal tissues through the secretion of soluble growth factor such as PDGF and transforming growth factor-beta (TGF-beta), which in turn stimulate the secretion of complimentary factors by host cells such as fibroblast growth factor (FGF), epidermal growth factor (EGF), and vascular endothelial growth factor (VEGF).
  • FGF fibroblast growth factor
  • EGF epidermal growth factor
  • VEGF vascular endothelial growth factor
  • PDGF vascular endothelial growth factor
  • FGF a chemo-attractant and mitogen for fibroblasts and endothelial cells
  • VEGF a potent regulator of vascularization
  • PDGF vascular endothelial growth factor
  • a or B chains which share 60% homology in their primary amino acid sequence.
  • the chains are disulfide cross linked to form the 30 kDa mature protein composed of either AA, BB or AB homo- or heterodimmers.
  • PDGF is found at high levels in platelets, and is expressed by endothelial cells and vascular smooth muscle cells. In addition, the production of PDGF is up regulated under low oxygen conditions such as those found in poorly vascularized tumor tissue (Kourembanas, S., et al., Kidney Int, 1997, 51(2), 438-43). PDGF binds with high affinity to the PDGF receptor, a 1106 amino acid 124 kDa transmembrane tyrosine kinase receptor (Heldin, C. H.; A. Ostman, and L.
  • PDGFR is found as homo- or heterodimer chains which have 30% homology overall in their amino acid sequence and 64% homology between their kinase domains (Heldin, C. H., et al. Embo J, 1988, 7(5), 1387-93).
  • PDGFR is a member of a family of tyrosine kinase receptor with split kinase-domains that includes VEGFR2 (KDR), c-Kit, and FLT3 which have all been found to have a role in promoting tumor angiogenesis, growth and survival.
  • the PDGF receptor is expressed primarily on fibroblast, smooth muscle cells, and pericytes and to a lesser extent on neurons, kidney mesangial, Leydig, and Schwann cells of the central nervous system. Upon binding to the receptor, PDGF induces receptor dimerization and undergoes auto- and trans-phosphorylation of tyrosine residues which increase the receptors' kinase activity and promotes the recruitment of downstream effectors through the activation of SH2 protein binding domains.
  • a number of signaling molecules form complexes with activated PDGFR including PI-3-kinase, phospholipase C-gamma, src and GAP (GTPase activating protein for p21-ras) (Soskic, V., et al. Biochemistry, 1999, 38(6), 1757-64).
  • PI-3-kinase phospholipase C-gamma
  • src GAP
  • PDGF blood vessel homeostasis
  • PDGF is found at high concentrations in platelets and is a potent chemoattractant for fibroblast, smooth muscle cells, neutrophils and macrophages. In addition to its role in wound healing PDGF helps maintain vascular homeostasis.
  • PDGF vascular endothelial growth factor
  • angiogenesis PDGF controls interstitial fluid pressure, regulating the permeability of vessels through its regulation of the interaction between connective tissue cells and the extracellular matrix. Inhibiting PDGFR activity can lower interstitial pressure and facilitate the influx of cytotoxics into tumors improving the anti-tumor efficacy of these agents (Pietras, K., et al. Cancer Res, 2002. 62(19), 5476-84; Pietras; K., et al. Cancer Res, 2001. 61(7), 2929-34).
  • PDGF can promote tumor growth through either the paracrine or autocrine stimulation of PDGFR receptors on stromal cells or tumor cells directly, or through the amplification of the receptor or activation of the receptor by recombination.
  • Over expressed PDGF can transform human melanoma cells and keratinocytes (Forsberg, K., et al. Proc Natl Acad Sci USA., 1993. 90(2), 393-7; Skobe, M. and N. E. Fusenig, Proc Natl Acad Sci USA, 1998. 95(3), 1050-5), two cell types that do not express PDGF receptors, presumably by the direct effect of PDGF on stroma formation and induction of angiogenesis.
  • CMML chronic myelomonocytic leukemia
  • PDGFR inhibitors will interfere with tumor stromal development and inhibit tumor growth and metastasis without undue side effects. Additional factors such as VEGF and FGF, secreted by stromal cells in response to tumor secreted PDGF, play key roles in stromal formation, angiogenesis and tumor progression.
  • VEGF vascular endothelial growth factor
  • VPF vascular permeability factor
  • VEGF expression is induced by hypoxia (Shweiki et al. Nature 1992, 359, 843), as well as by a variety of cytokines and growth factors, such as interleukin-1, interleukin-6, epidermal growth factor and transforming growth factor.
  • cytokines and growth factors such as interleukin-1, interleukin-6, epidermal growth factor and transforming growth factor.
  • VEGF and the VEGF family members have been reported to bind to one or more of three transmembrane receptor tyrosine kinases (Mustonen et al. J.
  • VEGF receptor-1 also known as flt-1 (fms-like tyrosine kinase-1)
  • VEGFR-2 also known as kinase insert domain containing receptor (KDR); the murine analogue of KDR is known as fetal liver kinase-1 (flk-1)), and VEGFR-3 (also known as flt-4).
  • KDR and flt-1 have been shown to have different signal transduction properties (Waltenberger et al. J. Biol. Chem. 1994, 269, 26988); Park et al. Oncogene 1995, 10, 135).
  • KDR undergoes strong ligand-dependant tyrosine phosphorylation in intact cells, whereas flt-1 displays a weak response.
  • binding to KDR is a critical requirement for induction of the full spectrum of VEGF-mediated biological responses.
  • VEGF plays a central role in vasculogenesis, and induces angiogenesis and permeabilization of blood vessels.
  • Deregulated VEGF expression contributes to the development of a number of diseases that are characterized by abnormal angiogenesis and/or hyperpermeability processes. Regulation of the VEGF-mediated signal transduction cascade will therefore provide a useful mode for control of abnormal angiogenesis and/or hyperpermeability processes.
  • Angiogenesis is regarded as an absolute prerequisite for growth of tumors beyond about 1-2 mm. Oxygen and nutrients may be supplied to cells in tumor smaller than this limit through diffusion. However, every tumor is dependent on angiogenesis for continued growth after it has reached a certain size. Tumorigenic cells within hypoxic regions of tumors respond by stimulation of VEGF production, which triggers activation of quiescent endothelial cells to stimulate new blood vessel formation. (Shweiki et al. Proc. Nat'l. Acad. Sci., 1995, 92, 768). In addition, VEGF production in tumor regions where there is no angiogenesis may proceed through the ras signal transduction pathway (Grugel et al. J. Biol.
  • VEGF mRNA is strongly upregulated in a wide variety of human tumors, including lung (Mattern et al. Br. J. Cancer 1996, 73, 931), thyroid (Viglietto et al. Oncogene 1995, 11, 1569), breast (Brown et al. Human Pathol. 1995, 26, 86), gastrointestional tract (Brown et al. Cancer Res. 1993, 53, 4727; Suzuki et al. Cancer Res. 1996, 56, 3004), kidney and bladder (Brown et al. Am. J. Pathol.
  • VEGF intraocular angiogenesis
  • ischemic retinal-vein occlusion a cascade of events
  • retinopathy of prematurity a number of retinopathies, including diabetic retinopathy, ischemic retinal-vein occlusion, and retinopathy of prematurity (Aiello et al. New Engl. J. Med. 1994, 331, 1480; Peer et al. Lab. Invest. 1995, 72, 638), and age-related macular degeneration (AMD; see, Lopez et al. Invest Opththalmol. Vis. Sci. 1996, 37, 855).
  • AMD age-related macular degeneration
  • rheumatoid arthritis In rheumatoid arthritis (RA), the in-growth of vascular pannus may be mediated by production of angiogenic factors. Levels of immunoreactive VEGF are high in the synovial fluid of RA patients, while VEGF levels were low in the synovial fluid of patients with other forms of arthritis of with degenerative joint disease (Koch et al. J. Immunol 1994, 152, 4149).
  • the angiogenesis inhibitor AGM-170 has been shown to prevent neovascularization of the joint in the rat collagen arthritis model (Peacock et al. J. Exper. Med. 1992, 175, 1135).
  • VEGF expression has also been shown in psoriatic skin, as well as bullous disorders associated with subepidermal blister formation, such as bullous pemphigoid, erythema multiforme, and dermatitis herpetiformis (Brown et al. J. Invest. Dermatol. 1995, 104, 744).
  • VEGF, VEGF-C, VEGF-D vascular endothelial growth factors
  • VEGFR2, VEGFR3 vascular endothelial growth factors
  • VEGF, VEGF-C and VEGF-D are expressed in most tumors, primarily during periods of tumor growth and, often at substantially increased levels.
  • VEGF expression is stimulated by hypoxia, cytokines, oncogenes such as ras, or by inactivation of tumor suppressor genes (McMahon, G. Oncologist 2000, 5(Suppl. 1), 3-10; McDonald, N. Q.; Hendrickson, W. A. Cell 1993, 73, 421-424)
  • VEGFR3 (also called flt-4) is predominantly expressed on lymphatic endothelium in normal adult tissues. VEGFR3 function is needed for new lymphatic vessel formation, but not for maintenance of the pre-existing lymphatics. VEGFR3 is also upregulated on blood vessel endothelium in tumors. Recently VEGF-C and VEGF-D, ligands for VEGFR3, have been identified as regulators of lymphangiogenesis in mammals. Lymphangiogenesis induced by tumor-associated lymphangiogenic factors could promote the growth of new vessels into the tumor, providing tumor cells access to systemic circulation.
  • VEGF-C vascular endothelial growth factor
  • VEGF-D vascular endothelial growth factor
  • VEGFR3 vascular endothelial growth factor-D
  • clinicopathological factors that relate directly to the ability of primary tumors to spread (e.g., lymph node involvement, lymphatic invasion, secondary metastases, and disease-free survival).
  • these studies demonstrate a statistical correlation between the expression of lymphangiogenic factors and the ability of a primary solid tumor to metastasize, (Skobe, M. et al. Nature Med. 2001, 7(2), 192-198; Stacker, S. A. et al. Nature Med. 2001, 7(2), 186-191; Makinen, T.
  • hypoxia appears to be an important stimulus for VEGF production in malignant cells.
  • Activation of p38 MAP kinase is required for VEGF induction by tumor cells in response to hypoxia (Blaschke, F. et al. Biochem. Biophys. Res. Commun. 2002, 296, 890-896; Shemirani, B. et al. Oral Oncology 2002, 38, 251-257).
  • p38 MAP kinase promotes malignant cell invasion, and migration of different tumor types through regulation of collagenase activity and urokinase plasminogen activator expression (Laferriere, J. et al. J. Biol.
  • Diarylureas are a class of serine-threonine kinase inhibitors as well as tyrosine kinase inhibitors well known in the art.
  • the following publications illustrate their utility as active ingredient in pharmaceutical compositions for the treatment of cancer, angiogenesis disorders, and inflammatory disorders:
  • FIG. 1 Qualitative assessment consisted of assessing the staining intensity, identifying the positively staining cells and the intracellular compartments involved in staining, and evaluating the overall slide quality. Separate evaluations were performed on the tumor cells and the tumor cell stroma. Intensity of staining was graded based upon the following scale: Negative, ⁇ (equivocal), 1+ (weak), 2+ (moderate), 3+ (strong), 4+ (intense). The intensity of staining was determined by evaluation of the entire specimen.
  • FIG. 2 Immunohistochemical Staining for phospho-ERK of Human Melanoma Biopsies pre- and post-treament.
  • the present invention provides methods for treating, ameliorating, preventing, modulating, etc., conditions and diseases in humans and other mammals which are associated with signal transduction pathways comprising, but not limited to, raf, VEGFR, PDGFR, p38, and/or FLT-3.
  • Preferred methods of the present invention provide for the modulation of diseases and conditions associated with raf, VEGFR2, VEGFR3, and/or PDGFR-beta.
  • the methods can comprise, e.g., administering an aryl urea compound as described below, pharmaceutically-acceptable salts thereof, derivatives thereof, etc.
  • the present invention also provides compositions and methods for identifying conditions and diseases which can be modulated with compounds of the present invention. These methods facilitate the selection of subjects who can be efficiently treated with compounds of the present invention. Additionally, the invention provides methods for monitoring subjects who have been administered a compound of the present invention. This includes, e.g., determining the efficacy of compounds of the present invention in the treatment of various diseases and conditions, and for determining treatment regimens.
  • aryl urea compounds employed in the methods of this invention comprise compounds of Formula I, pharmaceutically acceptable salts thereof, esters thereof, stereoisomers thereof (both isolated and in mixtures), prodrugs thereof, and any active derivatives thereof, which are collectively referred to herein as the “compounds, of the invention” and the like.
  • Formula I is as follows: B—NH—C(O)—NH-L-M-L 1 -(Q) 1-3 (I)
  • phenyl optionally substituted with 1-3 substituents independently selected from the group consisting of R 1 , OR 1 , NR 1 R 2 , S(O) q R 1 , SO 2 NR1R 2 , NR 1 SO 2 R 2 , C(O)R 1 , C(O)OR 1 , C(O)NR 1 R 2 , NR 1 C(O)R 2 , NR 1 C(O)OR 2 , halogen, cyano; and nitro;
  • a 5 or 6 membered monocyclic heteroaryl group having 1-3 heteroatoms independently selected from the group consisting of O, N and S, optionally substituted with 1-3 substituents independently selected from the group consisting of R 1 , OR 1 , NR 1 R 2 , S(O) q R 1 , SO 2 NR 1 R 2 , NR 1 SO 2 R 2 , C(O)R 1 , C(O)OR 1 , C(O)NR 1 R 2 ; NR 1 C(O)R 2 , NR 1 C(O)OR 2 , halogen, cyano, oxo, and nitro; or
  • the bicyclic heteroaryl group is optionally substituted with 1-3 substituents independently selected from the group consisting of R 1 , OR 1 , NR 1 R 2 , S(O) q R 1 , SO 2 NR 1 R 2 , NR 1 SO 2 R 2 , C(O)R 1 , C(O)OR 1 , C(O)NR 1 R 2 , NR 1 C(O)R 2 , NR 1 C(O)OR 2 , halogen, cyano, oxo, and nitro.
  • phenyl optionally substituted with 1-3 substituents independently selected from the group consisting of C 1 -C 5 linear or branched alkyl, C 1 -C 5 linear or branched haloalkyl, C 1 -C 3 alkoxy, hydroxy, amino, C 1 -C 3 alkylamino, C 1 -C 6 dialkylamino, halogen, cyano, and nitro;
  • naphthyl optionally substituted with 1-3 substituents independently selected from the group consisting of C 1 -C 5 linear or branched alkyl, C 1 -C 5 linear or branched haloalkyl, C 1 -C 3 alkoxy, hydroxy, amino, C 1 -C 3 alkylamino, C 1 -C 6 dialkylamino, halogen, cyano, and nitro;
  • a 5 or 6 membered monocyclic heteroaryl group having 1-3 heteroatoms independently selected from the group consisting of O, N and S, optionally substituted with 1-3 substituents independently selected from the group consisting of C 1 -C 5 linear or branched alkyl, C 1 -C 5 linear or branched haloalkyl, C 1 -C 3 alkoxy, hydroxy, amino, C 1 -C 3 alkylamino, C 1 -C 6 dialkylamino, halogen, cyano, and nitro; or
  • an 8 to 10 membered bicyclic heteroaryl group having 1-6 heteroatoms independently selected from the group consisting of O, N and S, optionally substituted with 1-3 substituents independently selected from the group consisting of C 1 -C 5 linear or branched alkyl, C 1 -C 5 linear or branched haloalkyl, C 1 -C 3 alkoxy, hydroxy, amino, C 1 -C 3 alkylamino, C 1 -C 6 dialkylamino, halogen, cyano, and nitro.
  • phenyl optionally substituted with 1-2 additional substituents other than Q, independently selected from the group consisting of R 1 , OR 1 , NR 1 R 2 , S(O) q R 1 , SO 2 NR 1 R 2 , NR 1 SO 2 R 2 , NR 1 C(O)R 2 , NR 1 C(O)OR 2 , halogen, cyano and nitro;
  • a 5 and 6 membered monocyclic heteroaryl group having 1-3 heteroatoms independently selected from the group consisting of O, N and S, optionally substituted with 1-2 additional substituents other than Q, independently selected from the group consisting of R 1 , OR 1 , NR 1 R 2 , S(O) q R 1 , SO 2 NR 1 R 2 , NR 1 SO 2 R 2 , NR 1 C(O)R 2 , NR 1 C(O)OR 2 , halogen, cyano and nitro and also oxides (e.g. ⁇ O, —O ⁇ or —OH);
  • an 8 to 10 membered bicyclic heteroaryl group having 1-6 heteroatoms independently selected from the group consisting of O, N and S, optionally substituted with 1-2 additional substituents other than Q, independently selected from the group consisting of R 1 , OR 1 , NR 1 R 2 , S(O) q R 1 , SO 2 NR 1 R 2 , NR 1 SO 2 R 2 , NR 1 C(O)R 2 , NR 1 C(O)OR 2 , halogen, cyano and nitro and also oxides (e.g. ⁇ O, —O ⁇ or —OH).
  • a saturated and partially saturated 5 and 6 membered monocyclic heterocyclic moiety having 1-3 heteroatoms independently selected from the group consisting of O, N and S, optionally substituted with 1-2 additional substituents other than Q, independently selected from the group consisting of R 1 , OR 1 , NR 1 R 2 , S(O) q R 1 , SO 2 NR 1 R 2 , NR 1 SO 2 R 2 N, R 1 C(O)R 2 , NR 1 C(O)OR 2 , halogen, cyano and nitro, and also oxides (e.g. ⁇ O, —O ⁇ or —OH); or
  • a saturated and partially saturated 8 to 10 membered bicyclic heterocyclic moiety having 1-6 heteroatoms independently selected from the group consisting of O, N and S, optionally substituted with 1-2 additional substituents other than Q, independently selected from the group consisting of R 1 , OR 1 , NR 1 R 2 , S(O) q R 1 , SO 2 NR 1 R 2 , NR 1 SO 2 R 2 , NR 1 C(O)R 2 , NR 1 C(O)OR 2 , halogen, cyano and nitro, and also oxides (e.g. ⁇ O, —O ⁇ or —OH);
  • each Q is independently C(O)R 4 , C(O)OR 4 and C(O)NR 4 R 5 ;
  • each R 1 -R 5 is independently selected from the group consisting of:
  • X is a 5 or 6 membered monocyclic heterocyclic ring, containing 1-4 atoms selected from oxygen, nitrogen and sulfur, which is saturated, partially saturated, or aromatic, or a 8-10 membered bicyclic heteroaryl having 1-4 heteroatoms selected from the group consisting of O, N and S; and wherein said alkyl moiety is optionally substituted with halogen up to per-halo,
  • each R 1 -R 5 other than per-halo substituted C 1 -C 5 linear or branched alkyl, is optionally substituted with 1-3 substituents independently selected from the group consisting of C 1 -C 5 linear or branched alkyl, up to perhalo substitiuted C 1 -C 5 linear or branched alkyl, C 1 -C 3 alkoxy, hydroxy, carboxy, amino, C 1 -C 3 alkylamino, C 1 -C 6 dialkylamino, halogen, cyano, and nitro;
  • variable p is an integer selected from 0, 1, or 2 and the variable q is an integer selected from 0, 1, 2, 3, or 4.
  • suitable hetaryl groups include, but are not limited to, 5-10 membered ring systems containing monocyclic and bicyclic rings, at least one of which is aromatic, in which one or more, e.g., 1-4 carbon atoms in one or more of the rings can be replaced by oxygen, nitrogen or sulfur atoms.
  • each ring can have from 3-7 atoms.
  • “Monocyclic heteroaryl” means an aromatic monocyclic ring having 5 to 6 ring atoms, at least one of which is a hetero atom selected from N, O and S, the remaining atoms being carbon. When more than one hetero atom is present in the moiety, they are selected independently from the other(s) so that they may be the same or different.
  • Monocyclic heteroaryl moieties include, but are not limited to pyrrole, furan, thiophene, imidazole, pyrazole, thiazole, oxazole, isoxazole, isothiazole, triazole, tetrazole, thiadiazole, oxadiazole, pyridine, pyrimidine, pyridazine, pyrazine, and triazine.
  • Bicyclic heteroaryl means fused bicyclic moieties where one of the rings is chosen from the monocyclic heteroaryl rings described above and the second ring is either benzene or another monocyclic heteroaryl ring described above. When both rings in the bicyclic moiety are heteroaryl rings, they may be the same or different, as long as they are chemically accessible by means known in the art.
  • Bicyclic heteroaryl rings include synthetically accessible 5-5, 5-6, or 6-6 fused bicyclic aromatic structures including, for example but not by way of limitation, benzoxazole (fused phenyl and oxazole), quinoline (fused phenyl and pyridine), imidazopyrimidine (fused imidazole and pyrimidine), and the like.
  • the phrase “5 or 6 membered heterocyclic ring, containing at least one atom selected from oxygen, nitrogen and sulfur, which is saturated, partially saturated, or aromatic” includes, by no way of limitation, tetrahydropyrane, tetrahydrofurane, 1,3-dioxolane, 1,4-dioxane, morpholine, thiomorpholine, piperazine, piperidine, piperidinone, tetrahydropyrimidone, pentamethylene sulfide, tetramethylene sulfide, dihydropyrane, dihydrofuran, dihydrothiophene, pyrrole, furan, thiophene, imidazole, pyrazole, thiazole, oxazole, isoxazole, isothiazole, triazole, pyridine, pyrimidine, pyridazine, pyrazine, triazine, and the like.
  • C 1 -C 3 alkyl-phenyl includes, by no way of limitation, 3-phenyl-propyl, 2-phenyl-1-methyl-ethyl. Substituted examples include 2-[2-chlorophenyl]ethyl, 3,4-dimethylphenyl-methyl, and the like.
  • Suitable substituted and unsubstituted heteroaryl groups for the compounds of this invention include, but are not limited to the following monocyclic heteroaryl groups:
  • benzofuryl benzothienyl, indolyl, benzimidazolyl, benzopyrazolyl, benzoxazolyl, benzisoxazolyl, benzothiazolyl, benzisothiazolyl, benz-1,3-oxadiazolyl, quinolinyl, isoquinolinyl, quinazolinyl, tetrahydroquinolinyl, tetrahydroisoquinolinyl, dihydrobenzofuryl, pyrazolo[3,4-b]pyrimidinyl, purinyl, benzodiazine, pterindinyl, pyrrolo[2,3-b]pyridinyl, pyrazolo[3,4-b]pyridinyl, oxazo[4,5-b]pyridinyl, imidazo[4,5-b]pyridinyl, cyclopentenopyridine, cyclohexanopyridine, cyclopent
  • Suitable aryl groups which do not contain heteroatoms include, for example, phenyl and 1- and 2-naphthyl, tetrahydronaphthyl, indanyl, indenyl, benzocyclobutanyl, benzocycloheptanyl and benzocycloheptenyl.
  • Suitable linear alkyl groups and alkyl portions of groups include methyl, ethyl, propyl, butyl, pentyl, etc.
  • Suitable branched alkyl groups include all branched isomers such as isopropyl, isobutyl, sec-butyl, tert-butyl, etc.
  • alkoxy means a straight or branched chain alkoxy group having saturated carbon atoms which may be linear or branched with single or multiple branching, and includes such groups as methoxy, ethoxy, n-propoxy, isopropoxy, and the like. It also includes halogenated groups such as 2,2-dichloroethoxy, trifluoromethoxy, and the like.
  • C 1 -C 3 alkylamino means methylamino, ethylamino, propylamino or isopropylamino.
  • Examples of C 1 -C 6 dialkylamino group include but are not limited to diethylamino, ethyl-isopropylamino, means methylamino, methyl-isobutylamino, dihexylamino.
  • Suitable halogens include F, Cl, Br, and/or I, from one to per-substitution (i.e. all H atoms on a group replaced by a halogen atom) being possible where an alkyl group is substituted by halogen, mixed substitution of halogen atom types also being possible on a given moiety.
  • Preferred halogens are Cl, Br and F.
  • perhalo substituted linear and branched alkyl includes alkyl groups having one alkyl hydrogen replaced with halogen, alkyl groups wherein all hydrogens are replaced with halogen, alkyl groups wherein more than one but less than all hydrogens are replaced by halogen and alkyl groups having alkyl hydrogens replaced by halogen and other substituents. Examples include chloromethyl, dichloromethyl, trichloromethyl, fluoromethyl, difluoromethyl, trifluoromethyl, and the like.
  • cycloalkyl refers to cyclic structures having 3-8 members in the ring such as cyclopropyl, cyclobutyl and cyclopentyl and cyclic structures having 3-8 members with alkyl substituents such that, for example, “C 3 cycloalkyl” includes methyl substituted cyclopropyl groups.
  • saturated carbocyclic moieties defines only the cyclic structure, i.e. cyclopentyl, cyclohexyl, etc. Any alkyl substitution on these cyclic structures is specifically identified.
  • Saturated monocyclic and bicyclic carbocyclic moieties include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, and decahydronaphthalene.
  • Partially saturated monocyclic and bicyclic carbocyclic moieties include cyclopentenyl, cyclohexenyl, cyclohexadienyl and tetrahydronaphthalene.
  • Saturated monocyclic and bicyclic heterocyclic moieties include tetrahydropyranyl, tetrahydrofuranyl, 1,3-dioxolane, 1,4-dioxanyl, morpholinyl, thiomorpholinyl, piperazinyl, piperidinyl, piperidinonyl, tetrahydropyrimidonyl, pentamethylene sulfide and tetramethylene sulfide.
  • Partially saturated monocyclic and bicyclic heterocyclic moieties include dihydropyranyl, dihydrofuranyl, dihydrothienyl, dihydropiperidinyl, and dihydropyrimidonyl.
  • any moiety When any moiety is “substituted”, it can have up to the highest number of indicated substituents, and each substituent can be located at any available position on the moiety and can be attached through any available atom on the substituent. “Any available position” means any position on the moiety that is chemically accessible through means known in the art or taught herein and that does not create an unduly unstable molecule. When there are two or more substituents on any moiety, each substituent is defined independently of any other substituent and can, accordingly, be the same or different.
  • L′ is pyridine
  • hydroxy as a pyridine substituent includes 2-, 3-, and 4-hydroxypyridine, but also includes those structures referred to in the art as 1-oxo-pyridine and 1-hydroxy-pyridine.
  • substituted structures of B and L′ are preferably each, independently, selected from the group consisting of
  • Each of the above substituents is optionally partially or fully halogenated, such as difluoromethyl sulfonyl.
  • An embodiment of this invention includes the administration of compounds of this invention wherein in formula I, L, B and L′ follow one of the following of combinations:
  • B phenyl
  • B phenyl
  • L pyridinyl and L′ is phenyl, pyridinyl, quinolinyl, isoquinolinyl or not present,
  • B phenyl, L naphthyl and L′ is phenyl, pyridinyl, quinolinyl, isoquinolinyl or not present,
  • B pyridinyl
  • L phenyl and L′ is phenyl, pyridinyl, quinolinyl, isoquinolinyl or not present,
  • B pyridinyl
  • L pyridinyl and L′ is phenyl, pyridinyl, quinolinyl, isoquinolinyl or not present,
  • B pyridinyl
  • B isoquinolinyl
  • L phenyl and L′ is phenyl, pyridinyl, quinolinyl, isoquinolinyl or not present,
  • B isoquinolinyl
  • L pyridinyl and L′ is phenyl, pyridinyl, quinolinyl, isoquinolinyl or not present,
  • B isoquinolinyl
  • L naphthyl and L′ is phenyl, pyridinyl, quinolinyl, isoquinolinyl or not present,
  • B quinolinyl
  • L phenyl and L′ is phenyl, pyridinyl, quinolinyl, isoquinolinyl or not present,
  • B quinolinyl
  • L pyridinyl and L′ is phenyl, pyridinyl, quinolinyl, isoquinolinyl or not present,
  • B quinolinyl
  • the structure M of formula I is preferably —O—, a single bond, —S—, —NH—, —N(CH 3 )—, —NHCH 2 —, —NC 2 H 4 —, —CH 2 —, —C(O)—, —CH(OH)—, —NHC(O)N(CH 3 )CH 2 —, —N(CH 3 )C(O)N(CH 3 )CH 2 —, —CH 2 C(O)N(CH 3 )—, —C(O)N(CH 3 )CH 2 —, —NHC(O)—, —N(CH 3 )C(O)—, —C(O)N(CH 3 )—, —C(O)NH—, —CH 2 O—, —CH 2 S—, —CH 2 N(CH 3 )—, —OCH 2 —, —CHF—, —CF 2 —, —CCl 2 —,
  • Compounds of the invention of particular interest also include those of formula I wherein B is phenyl or pyridyl, optionally substituted with 1-6 substituents independently selected from the group consisting of R 1 and halogen, L′ and Q are as defined above, M is —O— and L is phenyl, optionally substituted with 1-4 halogen.
  • Compounds of the invention of particular interest also include those of formula I wherein B is phenyl, optionally substituted with 1-6 substituents independently selected from the group consisting of R 1 and halogen, L′ and Q are as defined above, M is —O— and L is phenyl, optionally substituted with 1-4 halogen.
  • Compounds of the invention of particular interest also include those of formula I wherein B is 4-chloro(2-trifluoromethyl)phenyl, optionally substituted by the group consisting of R 1 and halogen, L′ and Q are as defined above, M is —O— and L is phenyl, optionally substituted with 1-4 halogen.
  • a number of the compounds of Formula I possess asymmetric centers, depending on the location a nature of various substituents. and can therefore exist in racemic and optically active forms as well as in the form of racemic or non-racemic mixtures thereof, and in the form of diastereomers and diastereomeric mixtures.
  • Asymmetric carbon atoms may be present in the (R) or (S) configuration or (R,S) configuration.
  • asymmetry may also be present due to restricted rotation about a given bond, for example, the central bond adjoining two substituted aromatic rings of the specified compounds.
  • All of these compounds including cis isomers, trans isomers, diastereomic mixtures, racemates, non-racemic mixtures of enantiomers, substantially pure, and pure enantiomers, are considered to be within the scope of the compounds of this invention and are collectively referred to when reference is made to compounds of this invention. Therefore, the methods of the present invention encompass the use of any isolated racemic or optically, active form of compounds described in Formula I which possess c-raf, b-raf, p38, VEGFR, PDGFR, and/or FLT-3 activity.
  • optical isomers can be obtained by resolution of the racemic mixtures according to conventional processes, for example, by the formation of diastereoisomeric salts using an optically active acid or base.
  • appropriate acids are tartaric, diacetyltartaric, dibenzoyltartaric, ditoluoyltartaric and camphorsulfonic acid.
  • Mixtures of diastereoisomers can be separated into their individual diastereomers on the basis of their physical chemical differences by methods known to those skilled in the art, for example, by chromatography or fractional crystallization.
  • the optically active bases or acids are liberated from the separated diastereomeric salts.
  • Another process for separation of optical isomers involves the use of a chiral chromatography column (e.g., chiral HPLC columns) optimally chosen to maximize the separation of the enantiomers.
  • a chiral HPLC column e.g., chiral HPLC columns
  • Suitable chiral HPLC columns are manufactured by Diacel, e.g., Chiracel OD and Chiracel OJ.
  • the optically active compounds of Formula (I) can likewise be obtained by utilizing optically active starting materials.
  • the present invention encompasses any separated, isolated, pure or partially purified isomers or racemic mixtures of the compounds of formula I which possess Raf, VEGFR, PDGFR, p38, and/or FLT-3 activity, and/or an efficacy in modulating any of the diseases and/or conditions mentioned herein.
  • stereoisomer is understood to encompass diastereoisomers, enantiomers, geometric isomers, etc.
  • Preferred compounds are those with the absolute configuration of the compound of Formula I which produce the more desirable biological activity are also included within the scope of the present invention.
  • the purification of said isomers and the separation of said isomeric mixtures can be accomplished by standard techniques known in the art.
  • substantially pure enantiomers is intended to mean that no more than 5% w/w of the corresponding opposite enantiomer is present.
  • pharmaceutically-acceptable salts of these compounds are also within the scope of the invention.
  • pharmaceutically acceptable salt refers to a relatively non-toxic, inorganic, or organic acid addition salt of a compound of the present invention. For example, see S. M. Berge, et al. “Pharmaceutical Salts,” J. Pharm. Sci. 1977, 66, 1-19.
  • Suitable salts are especially the pharmaceutically acceptable salts of compounds of formula (I) or such as, for example, organic or inorganic acid addition salts of compounds of formula (I).
  • Suitable acid addition salts include acetate, adipate, alginate, ascorbate, aspartate, benzoate, benzenesulfonate, bisulfate, butyrate, citrate, camphorate, camphorsulfonate, cinnamate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, fumarate, glucoheptanoate, glycerophosphate, hemisulfate, heptanoate, hexanoate, hydrochloride, hydrobromide, hydroiodide, 2-hydroxyethanesulfonate, itaconate, lactate, maleate, mandelate, methanesulfonate, 2-naphthalenesulf
  • Suitable inorganic acids include but are not limited to halogen acids (such as hydrochloric acid and hydrobromic acid), sulfuric acid, or phosphoric acid.
  • Suitable organic acids include but are not limited to carboxylic, phosphonic, sulfonic, or sulfamic acids, with examples including acetic acid, propionic acid, octanoic acid, decanoic acid, trifluoroacetic acid, dodecanoic acid, glycolic acid, lactic acid, 2- or 3-hydroxybutyric acid, ⁇ -aminobutyric acid (GABA), gluconic acid, glucosemonocarboxylic acid, benzoic acid, salicylic acid, phenylacetic acid and mandelic acid, fumaric acid, succinic acid, adipic acid, pimelic acid, suberic acid, azeiaic acid, maleic acid, tartaric acid, citric, acid, glucaric acid, galactaric acid, amino acids (such as glutamic
  • pharmaceutically acceptable salts include acid salts of inorganic bases, such as salts containing alkaline cations (e.g., Li + Na + or K + ), alkaline earth cations (e.g., Mg +2 , Ca +2 or Ba +2 ), the ammonium cation, as well as acid salts of organic bases, including aliphatic and aromatic substituted ammonium, and quaternary ammonium cations, such as those arising from protonation or peralkylation of triethylamine, N,N-diethylamine, N,N-dicyclohexylamine, lysine, pyridine, N,N-dimethylaminopyridine (DMAP), 1,4-diazabiclo[2.2.2]octane (DABCO), 1,5-diazabicyclo[4.3.0]non-5-ene (DBN) and 1,8-diazabicyclo[5.4.0]undec-7-ene (D
  • Base salts include alkali metal salts such as potassium and sodium salts, alkaline earth metal salts such as calcium and magnesium salts, and ammonium salts with organic bases such as dicyclohexylamine and N-methyl-D-glucamine. Additionally, basic nitrogen containing groups may be quaternized with such agents as lower alkyl halides such as methyl, ethyl, propyl, and butyl chlorides, bromides and iodides; dialkyl sulfates like dimethyl, diethyl, and dibutyl sulfate; and diamyl sulfates, long chain halides such as decyl, lauryl, myristyl and strearyl chlorides, bromides and iodides, aralkyl halides like benzyl and phenethyl bromides and others.
  • lower alkyl halides such as methyl, ethyl, propyl, and butyl chlorides
  • esters of appropriate compounds of this invention are well-tolerated, pharmaceutically acceptable esters such as alkyl esters including methyl, ethyl, propyl, isopropyl, butyl, isobutyl or pentyl esters. Additional esters such as phenyl-C 1 -C 5 alkyl may be used, although methyl ester is preferred.
  • prodrugs are well known in the art in order to enhance the properties of the parent compound; such properties include solubility, absorption, biostability and release time (see “ Pharmaceutical Dosage Form and Drug Delivery Systems ” (Sixth Edition), edited by Ansel et al., published by Williams & Wilkins, pages 27-29, (1995) which is hereby incorporated by reference).
  • Commonly used prodrugs of the disclosed oxazolyl-phenyl-2,4-diamino-pyrimidine compounds are designed to take advantage of the major drug biotransformation reactions and are also to be considered within the scope of the invention.
  • Major drug biotransformation reactions include N-dealkylation, O-dealkylation, aliphatic hydroxylation, aromatic hydroxylation, N-oxidation, S-oxidation, deamination, hydrolysis reactions, glucuronidation, sulfation and acetylation (see Goodman and Gilman's The Pharmacological Basis of Therapeutics (Ninth Edition), editor Molinoff et al., pub. by McGraw-Hill, pages 11-13, (1996), which is hereby incorporated by reference).
  • the present invention provides compounds which are capable of modulating one or more signal transduction pathways comprising, but not limited to, raf, VEGFR-2, VEGFR-3, p38, PDGFR-beta, and/or Flt-3.
  • Raf is an important signaling molecule involved in the regulation of a number of key cellular processes, including cell growth, cell survival and invasion. It is a member of the Ras/Raf/MEK/ERK pathway. This pathway is present in most tumor cells.
  • VEGFR-2, VEGFR-3, PDGFR-beta, and Flt-3 are transmembrane receptor molecules which, when stimulated by an appropriate ligand, trigger the Ras/Raf/MEK/ERK cell signaling pathway, leading to a cascade of cellular events. Each of these receptor molecules have tyrosine kinase activity.
  • VEGFR receptors are stimulated by vascular endothelial growth factors (VEGF), and are important control points in the regulation of endothelial cell development and function.
  • VEGF vascular endothelial growth factors
  • the PDGF-beta receptor regulates cell proliferation and survival in a number of cell types, including mesenchymal cells.
  • Flt-3 is a receptor for the FL ligand. It is structurally similar to c-kit, and modulates the growth of pluripotent haemopoietic cells, influencing the development of T-cells, B-cells, and dendritic cells.
  • Raf or raf-1 kinase is a family of serine/threonine kinases which comprise at least three family members, A-Raf, B-Raf, and c-raf or Raf-1. See, e.g., Dhillon and Kolch, Arch. Biochem. Biophys., 404:3-9, 2002.
  • C-raf and B-Raf are preferred targets for compounds of the present invention.
  • Activating B-Raf mutations e.g., V599E mutant
  • module it is meant that the functional activity of the pathway (or a component of it) is changed in comparison to its normal activity in the absence of the compound. This effect includes any quality or degree of modulation, including, increasing, agonizing, augmenting, enhancing, facilitating, stimulating, decreasing, blocking, inhibiting, reducing, diminishing, antagonizing, etc.
  • Tables 8 and 9 show the activity of a compound N-[4-chloro-3-(trifluoromethyl)phenyl]-N′- ⁇ 4-[2-N-methylcarbamoyl-4-pyridyloxy]phenyl ⁇ urea of the present invention in vitro (biochemical) and in vivo (cellular) assays, respectively.
  • the compound As indicated by its biochemical activity profile (Table 8), the compound is a potent inhibitor of a number of different cellular kinases. Moreover, it has the ability to affect kinase activity, cell growth, and other cellular processes in whole cells.
  • Table 9 shows the concentration of compound which inhibited target phosphorylation in whole cells by 50% (IC50).
  • activity profile is representative of an active compound of the present invention, not all active compounds are required to have the same rank order of potency for them to be useful in the present invention.
  • Assays for biochemical and cellular activity are conventional and can be carried out using any suitable format, including as described in the examples below. See, also, VEGFR-3 kinase assays (e.g., Manley et al., J. Med. Chem., 45(26):5687-93, 2002; Stahl et al., Angew Chem. Int. Ed.
  • the compounds of the present invention can also modulate one or more of the following processes, including, but not limited to, e.g., cell growth (including, e.g., differentiation, cell survival, and/or proliferation), tumor cell growth (including, e.g., differentiation, cell survival, and/or proliferation), tumor regression, endothelial cell growth (including, e.g., differentiation, cell survival, and/or proliferation), angiogenesis (blood vessel growth), lymphangiogenesis (lymphatic vessel growth), and/or hematopoiesis (e.g., T- and B-cell development, dendritic cell development, etc.).
  • cell growth including, e.g., differentiation, cell survival, and/or proliferation
  • tumor cell growth including, e.g., differentiation, cell survival, and/or proliferation
  • tumor regression including, e.g., differentiation, cell survival, and/or proliferation
  • endothelial cell growth including, e.g., differentiation, cell survival, and/or proliferation
  • angiogenesis blood vessel growth
  • kinase activity it is meant a catalytic activity in which a gamma-phosphate from adenosine triphosphate (ATP) is transferred to an amino acid residue (e.g., serine, threonine, or tyrosine) in a protein substrate.
  • ATP adenosine triphosphate
  • a compound can modulate kinase activity, e.g., inhibiting it by directly competing with ATP for the ATP-binding pocket of the kinase, by producing a conformational change in the enzyme's structure that affects its activity (e.g., by disrupting the biologically-active three-dimensional structure), etc.
  • Kinase activity can be determined routinely using conventional assay methods.
  • Kinase assays typically comprise the kinase enzyme, substrates, buffers, and components of a detection system.
  • a typical kinase assay involves the reaction of a protein kinase with a peptide substrate and an ATP, such as 32P-ATP, to produce a phosphorylated end-product (for instance, a phosphoprotein when a peptide substrate is used.
  • the resulting end-product can be detected using any suitable method.
  • radioactive ATT When radioactive ATT is utilized, a radioactively labeled phosphoprotein can be separated from the unreacted gamma-32P-ATP using an affinity membrane or gel electrophoresis, and then visualized on the gel using autoradiography or detected with a scintillation counter.
  • Non-radioactive methods can also be used. Methods can utilize an antibody which recognizes the phosphorylated substrate, e.g., an anti-phosphotyrosine antibody.
  • kinase enzyme can incubated with a substrate, in the presence of ATP and kinase buffer under conditions which are effective for the enzyme to phosphorylate the substrate.
  • the reaction mixture can be separated, e.g., electrophoretically, and then phosphorylation of the substrate can be measured, e.g., by Western blotting using an anti-phosphotyrosine antibody.
  • the antibody can be labeled with a detectable label, e.g., an enzyme, such as HRP, avidin or biotin, chemiluminescent reagents, etc.
  • detectable label e.g., an enzyme, such as HRP, avidin or biotin, chemiluminescent reagents, etc.
  • Other methods can utilize ELISA formats, affinity membrane separation, fluorescence polarization assays, luminescent assays, etc.
  • TR-FRET time-resolved fluorescence resonance energy transfer
  • the compounds of the present invention can be used to treat and/or prevent any disease or condition involving one or more cellular signal transduction pathways comprising raf, VEGFR-2, VEGFR-3, p38, PDGFR-beta, and/or Flt-3.
  • the term “treating” is used conventionally, e.g., the management or care of a subject for the purpose of combating, alleviating, reducing, relieving, improving the condition of, etc., of a disease or disorder.
  • Diseases and conditions that can be treated include any of those mentioned above and below, as well as:
  • Raf associated diseases include, e.g., cell-proliferation disorders, cancer, tumors, etc.;
  • VEGFR-2 associated diseases include, e.g., cancer, tumor growth, inflammatory disease, rheumatoid arthritis, retinopathy, psoriasis, glomerulonephritis, asthma, chronic bronchitis, atherosclerosis, transplant rejection, conditions involving angiogenesis, etc.;
  • VEGFR-3 associated diseases include, e.g., cancer, corneal disease, inflamed cornea (e.g., Hamrah, Am. J. Path., 163:57-68, 2003), corneal transplantation (Cursiefen et al., Cornea, 22:273-81, 2003), lymphatic hyperplasia, conditions involving lymphangiogenesis, etc.;
  • PDGFR-beta associated diseases include, e.g., diseases or conditions characterized by cell proliferation, cell matrix production, cell movement, and/or extracellular matrix production.
  • diseases or conditions include, e.g., tumors, malignancies, cancer, metastasis, chronic myeloid leukemia, inflammation, renal disease, diabetic nephropathy, mesangial proliferative glomerulonephritis, fibrotic conditions, atherosclerosis, restenosis, hypertension-related arterosclerosis, Venous bypass graft arterosclerosis, scleroderma, interstitial pulmonary diseases, synovial disorders, arthritis, leukemias, lymphomas, etc;
  • Flt-3 associated diseases include, e.g., immune-related disorders, blood cell disorders, conditions involving hematopoietic cell development (e.g., T-cells, B-cells, dendritic cells, cancer, anemia, HIV, acquired immune deficiency syndrome, etc.
  • immune-related disorders e.g., blood cell disorders, conditions involving hematopoietic cell development (e.g., T-cells, B-cells, dendritic cells, cancer, anemia, HIV, acquired immune deficiency syndrome, etc.
  • compounds of the present invention can be used to treat conditions and disorders disclosed in U.S. Pat. No. 6,316,479, e.g, glomerular sclerosis, interstitial nephritis, interstitial pulmonary fibrosis, atherosclerosis, wound scarring and scleroderma.
  • the compounds of this invention also have a broad therapeutic activity to treat or prevent the progression of a broad array of diseases, such as inflammatory conditions, coronary restenosis, tumor-associated angiogenesis, atherosclerosis, autoimmune diseases, inflammation, certain kidney diseases associated with proliferation of glomerular or mesangial cells, and ocular diseases associated with retinal vessel proliferation.
  • diseases such as inflammatory conditions, coronary restenosis, tumor-associated angiogenesis, atherosclerosis, autoimmune diseases, inflammation, certain kidney diseases associated with proliferation of glomerular or mesangial cells, and ocular diseases associated with retinal vessel proliferation.
  • diseases such as inflammatory conditions, coronary restenosis, tumor-associated angiogenesis, atherosclerosis, autoimmune diseases, inflammation, certain kidney diseases associated with proliferation of glomerular or mesangial cells, and ocular diseases associated with retinal vessel proliferation.
  • psoriasis hepatic cirrhosis
  • diabetes atherosclerosis
  • restenosis vascular graft restenosis
  • the present invention also provides for treating, preventing, modulating, etc., one or more of the following conditions in humans and/or other mammals: retinopathy, including diabetic retinopathy, ischemic retinal-vein occlusion, retinopathy of prematurity and age related macular degeneration; rheumatoid arthritis, psoriasis, or bullous disorder associated with subepidermal blister formation, including bullous pemphigoid, erythema multiforme, or dermatitis herpetiformis, rheumatic fever, bone resorption, postmenopausal osteoperosis, sepsis, gram negative sepsis, septic shock, endotoxic shock, toxic shock syndrome, systemic inflammatory response syndrome, inflammatory bowel disease (Crohn's disease and ulcerative colitis), Jarisch-Herxheimer reaction, asthma, adult respiratory distress syndrome, acute pulmonary fibrotic disease, pulmonary sarcoidosis, allergic respiratory disease, silicos
  • coli infection effects of enterotoxin A resulting from Staphylococcus infection, meningococcal infection, and infections from Borrelia burgdorferi, Treponema pallidum , cytomegalovirus, influenza virus, Theiler's encephalomyelitis virus, and the human immunodeficiency virus (HIV), papilloma, blastoglioma, Kaposi's sarcoma, melanoma, lung cancer, ovarian cancer, prostate cancer, squamous cell carcinoma, astrocytoma, head cancer, neck cancer, bladder cancer, breast cancer, colorectal cancer, thyroid cancer, pancreatic cancer, gastric cancer, hepatocellular carcinoma, leukemia, lymphoma, Hodgkin's disease, Burkitt's disease, arthritis, rheumatoid arthritis, diabetic retinopathy, angiogenesis, restenosis, in-stent restenosis, vascular graft restenosis, pulmonary fibro
  • hyperimmune disorders hemangioma, myocardial angiogenesis, coronary and cerebral collateral vascularization, ischemia, corneal disease, rubeosis, neovascular glaucoma, macular degeneration retinopathy of prematurity, wound healing, ulcer Helicobacter related diseases, fractures, endometriosis, a diabetic condition, cat scratch fever, thyroid hyperplasia, asthma or edema following burns, trauma, chronic lung disease, stroke, polyps, cysts, synovitis, chronic and allergic inflammation, ovarian hyperstimulation syndrome, pulmonary and cerebral edema, keloid, fibrosis, cirrhosis, carpal tunnel syndrome, adult respiratory distress syndrome, ascites, an ocular condition, a cardiovascular condition, Crow-Fukase (POEMS) disease, Crohn's disease, glomerulonophritis, osteoarthritis, multiple sclerosis, graft rejection, Lyme disease, sepsis, von Hippel Lindau disease
  • Compounds can possess more than one of the mentioned activities, and therefore can target a plurality of signal transduction pathways. Thus, these compounds can achieve therapeutic and prophylactic effects which normally are only obtained when using a combination of different compounds. For instance, the ability to inhibit both new vessel formation (e.g., associated with VEGFR-2 and -3 function) (e.g., blood and/or lymph) and cell-proliferation (e.g., associated with raf and PDGFR-beta function) using a single compound is especially beneficial in the treatment of cancer, and other cell-proliferation disorders that are facilitated by neovascularization.
  • new vessel formation e.g., associated with VEGFR-2 and -3 function
  • cell-proliferation e.g., associated with raf and PDGFR-beta function
  • the present invention relates specifically to compounds which possess at least anti-cell proliferation and anti-angiogenic (i.e., inhibits angiogenesis) activity. Any disorder or condition that would benefit from inhibiting vessel growth and cell proliferation can be treated in accordance with the present invention. Using a single compound is also advantageous because its range of activities can be more precisely defined.
  • the present invention relates to methods of treating and/or preventing diseases and conditions; and/or modulating one or more of the pathways, polypeptides, genes, diseases, conditions, etc., associated with raf, VEGFR-2, VEGFR-3, PDGFR-beta, and/or Flt-3.
  • These methods generally involve administering effective amounts of compounds of the present invention, where an effective amount is the quantity of the compound which is useful to achieve the desired result.
  • Compounds can be administered in any effective form by any effective route, as discussed in more detail below.
  • Methods include modulating tumor cell proliferation, including inhibiting cell proliferation.
  • the latter indicates that the growth and/or differentiation of tumor cells is reduced, decreased, diminished, slowed, etc.
  • the term “proliferation” includes any process which relates to cell growth and division, and includes differentiation and apoptosis.
  • raf kinases play a key role in the activation of the cytoplasmic signaling cascade involved in cell proliferation, differentiation, and apoptosis.
  • inhibiting c-raf-1 by anti-sense oligonucleotides can block cell proliferation (see above). Any amount of inhibition is considered therapeutic.
  • Any tumor or cancer can be treated, including, but not limited to, cancers having one or more mutations in raf, VEGFR-2, VEGFR-3, PDGFR-beta, Flt-3, and/or ras, as well as any upstream or downstream member of the signaling pathways of which they are a part.
  • a cancer can be treated with a compound of the present invention irrespective of the mechanism which is responsible for it.
  • cancers of any organ can be treated, including cancers of, but are not limited to, e.g., colon, pancreas, breast, prostate, bone, liver, kidney, lung, testes, skin, pancreas, stomach, colorectal cancer, renal cell carcinoma, hepatocellular carcinoma, melanoma, etc.
  • breast cancer examples include, but are not limited to, invasive ductal carcinoma, invasive lobular carcinoma, ductal carcinoma in situ, and lobular carcinoma in situ.
  • cancers of the respiratory tract include, but are not limited to, small-cell and non-small-cell lung carcinoma, as well as bronchial adenoma and pleuropulmonary blastoma.
  • brain cancers include, but are not limited to, brain stem and hypophtalmic glioma, cerebellar and cerebral astrocytoma, medulloblastoma, ependymoma, as well as neuroectodermal and pineal tumor.
  • Tumors of the male reproductive organs include, but are not limited to, prostate and testicular cancer.
  • Tumors of the female reproductive organs include, but are not limited to, endometrial, cervical, ovarian, vaginal, and vulvar cancer, as well as sarcoma of the uterus.
  • Tumors of the digestive tract include, but are not limited to, anal, colon, colorectal, esophageal, gallbladder, gastric, pancreatic, rectal, small-intestine, and salivary gland cancers.
  • Tumors of the urinary tract include; but are not limited to, bladder, penile, kidney, renal pelvis, ureter, and urethral cancers.
  • Eye cancers include, but are not limited to, intraocular melanoma and retinoblastoma.
  • liver cancers include, but are not limited to, hepatocellular carcinoma (liver cell carcinomas with or without fibrolamellar variant), cholangiocarcinoma (intrahepatic bile duct carcinoma), and mixed hepatocellular cholangiocarcinoma.
  • Skin cancers include, but are not limited to, squamous cell carcinoma, Kaposi's sarcoma, malignant melanoma, Merkel cell skin cancer, and non-melanoma skin cancer.
  • Head-and-neck cancers include, but are not limited to, laryngeal, hypopharyngeal, nasopharyngeal, and/or oropharyngeal cancers, and lip and oral cavity cancer.
  • Lymphomas include, but are not limited to, AIDS-related lymphoma, non-Hodgkin's lymphoma, cutaneous T-cell lymphoma, Hodgkin's disease, and lymphoma of the central nervous system.
  • Sarcomas include, but are not limited to, sarcoma of the soft tissue, osteosarcoma, malignant fibrous histiocytoma, lymphosarcoma, and rhabdomyosarcoma.
  • Leukemias include, but are not limited to, acute myeloid leukemia, acute lymphoblastic leukemia, chronic lymphocytic leukemia, chronic myelogenous leukemia, and hairy cell leukemia.
  • compounds of the present invention can also cause tumor regression, e.g., a decrease in the size of a tumor, or in the extent of cancer in the body.
  • the present invention also relates to methods of modulating angiogenesis and/or lymphangiogenesis in a system comprising cells, comprising administering to the system an effective amount of a compound described herein.
  • a system comprising cells can be an in vivo system, such as a tumor in a patient, isolated organs, tissues, or cells, in vitro assays systems (CAM, BCE, etc), animal models (e.g., in vivo, subcutaneous, cancer models), hosts in need of treatment (e.g., hosts suffering from diseases having angiogenic and/or lymphangiogenic component, such as cancer), etc.
  • Preferred compounds of the present invention inhibit angiogenesis and/or lymphangiogenesis, e.g., the formation of new blood vessels.
  • Inappropriate and ectopic expression of angiogenesis can be deleterious to an organism.
  • a number of pathological conditions are associated with the growth of extraneous blood vessels. These include, e.g., diabetic retinopathy, neovascular glaucoma, psoriasis, retrolental fibroplasias, angiofibroma, inflammation, etc.
  • the increased blood supply associated with cancerous and neoplastic tissue encourages growth, leading to rapid tumor enlargement and metastasis.
  • the growth of new blood and lymph vessels in a tumor provides an escape route for renegade cells, encouraging metastasis and the consequence spread of the cancer.
  • Useful systems for modulating angiogenesis include, e.g., neovascularization of tumor explants (e.g., U.S. Pat. Nos. 5,192,744; 6,024,688), chicken chorioallantoic membrane (CAM) assay (e.g., Taylor and Folkman, Nature, 297:307-312, 1982; Eliceiri et al., J. Cell Biol., 140, 1255-1263, 1998), bovine capillary endothelial (BCE) cell assay (e.g., U.S. Pat. No. 6,024,688; Polverini, P. J.
  • CAM chicken chorioallantoic membrane
  • BCE bovine capillary endothelial
  • useful systems for modulating lymphangiogenesis include, e.g., rabbit ear model (e.g., Szuba et al., FASEB J., 16(14):1985-7, 2002).
  • Modulation of angiogenesis can be determined by any suitable method.
  • the degree of tissue vascularity is typically determined by assessing the number and density of vesssels present in a given sample.
  • microvessel density MMD
  • MMD microvessel density
  • CD31 also known as platelet-endothelial cell adhesion molecule or PECAM.
  • a CD31 antibody can be employed in conventional immunohistological methods to immunostain tissue sections as described by, e.g., Penfold et al., Br. J. Oral and Maxill.
  • Vezf1 e.g., Xiang et al., Dev. Bio., 206:123-141, 1999
  • angiopoietin Tie-1
  • Tie-2 e.g., Sato et al., Nature, 376:70-74, 1995
  • levels of circulating VEGF such as VEGF-165, VEGF-C, VEGF-D, can be measured in an ELISA to determine whether it is above a threshold value that indicates angiongenic activity in the body.
  • the present invention relates to methods of screening patients to determine their susceptibility to compounds of the present invention.
  • the presenting invention relates to methods of selecting subjects having a disease for treatment with a compound of formula I, comprising, one or more of the following steps in any effective order, e.g., measuring the expression or activity of Raf, VEGFR-2, VEGFR-3, p38, PDGFR-beta, and/or Flt-3, in a sample obtained from a subject having a disease, and administering said compound of formula I to subjects who are identified as having high levels of expression or activity, where said compound is a compound of formula I of claim 1 .
  • the term “susceptibility” is used broadly to indicate, e.g., ability to respond, toxicity or other adverse effects, etc.
  • the invention relates to methods of determining whether a condition can be modulated by a compound disclosed herein, comprising measuring the expression or activity of Raf, VEGFR-2, VEGFR-3, p38, PDGFR-beta, and/or Flt-3 in cells having said condition.
  • the results can be used to determine or predict whether a subject will respond to a compound of the present invention.
  • the methods can be used to predict whether the tumor is susceptible to compounds of the present invention.
  • susceptible it is meant that tumor can be treated with it, e.g., causing tumor regression or cell death, inhibiting cell proliferation, inhibiting tumor growth, inhibiting tumor metastasis, etc.
  • Whether a condition, such as a tumor, is susceptible to a compound of the present invention can be determined routinely. For instance, cells or tissues (e.g., tumor cells, a biopsy sample, etc.) that exhibit the condition can be assayed for the presence and/or activity of Raf, VEGFR-2, VEGFR-3, p38, PDGFR-beta, and/or Flt-3. When high levels of expression and/or activity are identified, this can indicate that the subject will respond to, and benefit from, a compound of the present invention.
  • Levels of gene expression e.g., mRNA levels
  • gene amplification e.g., tyrosine kinase activity
  • gene product activity e.g., tyrosine kinase activity
  • the target genes of the present invention possess tyrosine kinase activity, and therefore kinase activity can be used to assess the cell or tissue state.
  • activity was measured by looking at the levels of substrate phosphorylated by it. This can be done quantitatively (e.g., using isotopes, spectroscopy, etc.) or semi-quantitatively as in the example where the levels were assessed visually and assigned a level of intensity from +1 to +4.
  • a cell or tissue which has a high level of phosphorylated substrate (and a high number of cells exihibiting the heightened activity) can be considered to have a high level of kinase activity, and therefore be a candidate for therapy with a compound of the present invention. More than one activity can be assessed, and the results from several targets can be utilized in deciding whether a subject's condition (e.g., a tumor) will be responsive to a compound of the present invention.
  • a subject's condition e.g., a tumor
  • High levels of target activity can be relative to a control or other standard. For instance, in the example below, high levels of activity were with reference to a cell type (stromal) in the tissue section which normally does not express substantial levels of the target gene. High levels can therefore be where cells express a statistically higher amount of measured activity or phosphoryated substrate than the standard or control used as a comparison. High levels can also be where 25% or more cells express the target activity (e.g., phospho-ERK).
  • target activity e.g., phospho-ERK
  • the method can further comprise a step of comparing the expression in a sample with a normal control, or expression in a sample obtained from normal or unaffected tissue. Comparing can be done manually, against a standard, in an electronic form (e.g., against a database), etc.
  • the normal control can be a standard sample that is provided with the assay; it can be obtained from adjacent, but unaffected, tissue from the same patient; or, it can be pre-determined values, etc.
  • Gene expression, protein expression (e.g., abundance in a cell), protein activity (e.g., kinase activity), etc. can be determined.
  • a biopsy from a cancer patient can be assayed for the presence, quantity, and/or activity of Raf, VEGFR-2, VEGFR-3, p38, PDGFR-beta, and/or Flt-3.
  • Increased expression or activity of one or more of these can indicate that the cancer can be targeted for treatment by a compound of the present invention.
  • raf activity can be monitored by its ability to initiate the cascade leading to ERK phosphorylation (i.e., raf/MEK/ERK), resulting in phospho-ERK.
  • Increased phospho-ERK levels in a cancer shows that its raf activity is elevated, suggesting the use of compounds of the present invention to treat it.
  • phospho-ERK (other markers) can also be measured in other body fluids, such as serum, blood, cerebral spinal fluid, urine, etc., such as in peripheral blood lymphocytes (PBLs).
  • PBLs peripheral blood lymphocytes
  • inhibition of ERK phosphorylation can be measured following activation with phorbol myristate acetate using antibodies as described in the examples below.
  • patients having cancer can be selected and monitored on the basis of whether the tissue is experiencing neovacularization, and how much. This can be assessed as discussed above, e.g., using immunohistochemistry for vessel markers (e.g., CD31), circulating levels of a VGFR ligand, etc.
  • vessel markers e.g., CD31
  • circulating levels of a VGFR ligand etc.
  • Patient selection and monitoring can also be made on the basis of the appearance in a body fluid (such as blood) above normal levels of the shedded ectodomains derived from the various receptors, including the extracellular portions of VEGFR-2, VEGFR-3, p38, PDGFR-beta, and Flt-3.
  • Detection methods can be carried out routinely, e.g., using antibodies which specifically bind to the extracellular domain.
  • Measuring expression includes determining or detecting the amount of the polypeptide present in a cell or shed by it, as well as measuring the underlying mRNA, where the quantity of mRNA present is considered to reflect the quantity of polypeptide manufactured by the cell. Furthermore, the genes for Raf, VEGFR-2, VEGFR-3, p38, PDGFR-beta, and/or Flt-3 can be analyzed to determine whether there is a gene defect responsible for aberrant expression or polypeptide activity.
  • NM — 004333 Homo sapiens v-raf murine sarcoma viral oncogene homolog B1 (BRAF); NM — 004119 Homo sapiens fms-related tyrosine kinase 3 (FLT3); NM — 002609 Homo sapiens platelet-derived growth factor receptor, beta polypeptide (PDGFRB); NM — 002253 Homo sapiens VEGFR2; NM — 182925 Homo sapiens fms-related tyrosine kinase 4 (FLT4); L35253 Homo sapiens p38 mitogen activated protein (MAP) kinase.
  • BRAF Homo sapiens v-raf murine sarcoma viral oncogene homolog B1
  • FLT3 Homo sapiens fms-related tyrosine kinase 3
  • PDGFRB Homo sapiens platelet
  • Polypeptide detection can be carried out by any available method, e.g., by Western blots, ELISA, dot blot, immunoprecipitation, RIA, immunohistochemistry, etc.
  • a tissue section can be prepared and labeled with a specific antibody (indirect or direct and visualized with a microscope.
  • Amount of a polypeptide can be quantitated without visualization, e.g., by preparing a lysate of a sample of interest, and then determining by ELISA or Western the amount of polypeptide per quantity of tissue.
  • Antibodies and other specific binding agents can be used. There is no limitation on how detection is performed.
  • Assays can be utilized which permit quantification and/or presence/absence detection of a target nucleic acid (e.g., genes, mRNA, etc., for raf, VEGFR, PDGFR, Flt-3, etc) in a sample. Assays can be performed at the single-cell level, or in a sample comprising many cells, where the assay is “averaging” expression over the entire collection of cells and tissue present in the sample. Any suitable assay format can be used, including, but not limited to, e.g., Southern blot analysis, Northern blot analysis, polymerase chain reaction (“PCR”) (e.g., Saiki et al., Science, 241:53, 1988; U.S. Pat. Nos.
  • PCR polymerase chain reaction
  • PCR Protocols A Guide to Methods and Applications, Innis et al., eds., Academic Press, New York, 1990
  • RT-PCR reverse transcriptase polymerase chain reaction
  • RACE rapid amplification of cDNA ends
  • LCR ligase chain reaction
  • RNA fingerprinting techniques nucleic acid sequence based amplification (“NASBA”) and other transcription based amplification systems (e.g., U.S. Pat. Nos. 5,409,818 and 5,554,527; WO 88/10315), polynucleotide arrays (e.g., U.S. Pat. Nos.
  • NASBA nucleic acid sequence based amplification
  • transcription based amplification systems e.g., U.S. Pat. Nos. 5,409,818 and 5,554,527; WO 88/10315
  • polynucleotide arrays e.g., U.S. Pat. Nos.
  • any method suitable for single cell analysis of gene or protein expression can be used, including in situ hybridization, immunocytochemistry, MACS, FACS, flow cytometry, etc.
  • expression products can be measured using antibodies, PCR, or other types of nucleic acid amplification (e.g., Brady et al., Methods Mol. & Cell. Biol. 2, 17-25, 1990; Eberwine et al., 1992, Proc. Natl. Acad. Sci., 89, 3010-3014, 1992, U.S. Pat. No. 5,723,290).
  • nucleic acid amplification e.g., Brady et al., Methods Mol. & Cell. Biol. 2, 17-25, 1990; Eberwine et al., 1992, Proc. Natl. Acad. Sci., 89, 3010-3014, 1992, U.S. Pat. No. 5,723,290.
  • Activity of raf, VEGFR-2, VEGFR-3, p38, PDGFR-beta, and Flt-3 can be assessed routinely, e.g., as described in the examples below, or using standard assays for kinase activity (see, above).
  • Measuring expression includes evaluating the all aspects of the transcriptional and translational machinery, of the gene. For instance, if a promoter defect causes, or is suspected of causing, the disorder, then a sample can be evaluated (i.e., “assessed”) by looking (e.g., sequencing or restriction mapping) at the promoter sequence in the gene, by detecting transcription products (e.g., RNA), by detecting translation product (e.g., polypeptide). Any measure of whether the gene is functional can be used, including, polypeptide, polynucleotide, and functional assays for the gene's biological activity.
  • the mRNA levels of a sample can serve as a comparison, or a gene which is known not to be affected by the disorder.
  • Methods of detecting mRNA are well known, and discussed above, e.g., but not limited to, Northern blot analysis, polymerase chain reaction (PCR), reverse transcriptase PCR, RACE PCR, etc.
  • polypeptide production is used to evaluate the gene
  • polypeptide in a normal tissue sample can be used as a comparison, or, polypeptide from a different gene whose expression is known not to be affected by the disorder.
  • Patients can also be selected for treatment if they have a particular genotype which is known to be associated with a cancer, especially genotypes which affect the Raf/Mek/Erk pathway, such as mutations in the BRAF, KRAS, or MEK genes.
  • the present invention relates to methods for selecting patients for treatment involving determining the the presence of a Raf, VEGFR-2, VEGFR-3, p38, PDGFR-beta, and/or Flt-3 gene mutation in a sample obtained from a subject, wherein said mutation is associated with a disease, and administering said compound of formula I to subjects who are identified as having said mutation.
  • the presence of the mutation can be determined conventionally, e.g., obtaining cells or a tissue sample from a subject, extracting nucleic acid from it, determining the gene sequence or structure of a target gene (using, e.g., mRNA, cDNA, genomic DNA, etc), comparing the sequence or structure of the target gene to the structure of the normal gene, whereby a difference in sequence or structure indicates a mutation in the gene in the subject.
  • Mutations can be determined using any effective method, e.g., comparing restriction maps, nucleotide sequences, amino acid sequences, RFLPs, DNAse sites, DNA methylation fingerprints (e.g., U.S. Pat. No.
  • the present invention also provides methods of assessing the efficacy of a compound of the present invention in treating a disease, comprising one or more of the following steps in any effective order, e.g., measuring the expression or activity of Raf, VEGFR-2, VEGFR-3, p38, PDGFR-beta, and/or Flt-3 in a sample obtained from said subject who has been treated with a compound of the present invention, and determining the effects of said compound on said expression or activity.
  • the measuring step can be carried out as described already.
  • biopsy samples can be removed from patients who have been treated with a compound of the present invention, and then assayed for the presence and/or activity of the mentioned signaling molecules.
  • decreased levels of phospho-ERK in the cancer tissue e.g., compared to a normal tissue or before treatment
  • Determining the effects of the compound on expression or activity includes performing a comparison step between a tissue sample and a control, or other type of standard.
  • standards include, but are not limited to, a tissue sample prior to treatment, a tissue sample from an unaffected tissue or from an unaffected region of the affected tissue (e.g., from a region of the tissue which is not transformed, cancerous, etc.), etc.
  • a standard can also be a value, or range of values, that is representative of normal levels of expression that have been established for that marker.
  • the comparison can also be made between samples collected from at least two different timepoints during the treatment regimen with a compound of the present invention.
  • samples can be collected from various times after initiation of the drug treatment, and analysis of expression and/or activity levels can be used to monitor the progress/prognosis of the subject, e.g., how the subject is responding to the drug regimen.
  • Any timepoint can be used, e.g., daily, twice a week, weekly, every two weeks, every month, yearly, a plurality of timepoints (at least 2, 3, 4, 8, 12, etc.).
  • determining the effect indicates that the result produced by the compound is analyzed and/or identified. For instance, in Example 3, data is shown that compound reduced the levels of phospho-ERK (i.e., the effect of the compound on raf activity was determined by measuring phospho-ERK). Any type of effect can be identified, e.g., where the expression and/or activity is reduced, decreased, down-regulated, inhibited, blocked, increased, up-regulated, unchanged, etc.
  • the method can be used to determine appropriate dosages and dosing regimens, e.g., how much compound to administer and at what frequency to administer it.
  • the clinician can determine the appropriate treatment protocol and whether it is achieving the desired effect, e.g., on modulating or inhibiting the signal transduction pathway. For instance, if the compound is not effective in knocking down the amounts of a marker, such as phospho-ERK, the dosage can be increased in the patient or given more frequently. Similarly, dosages and/or frequency can be reduced when it is shown that the compound is effective in knocking down the levels of phospho-ERK or other marker for the disease state. Since the compounds can be administered in combination with others treatments, e.g., radiation, chemotherapy, and other agents, the monitoring of the subject can be used to assess the combined effects of the treatment regimen on the progress of the disease.
  • others treatments e.g., radiation, chemotherapy, and other agents
  • mutations include mutations in K-RAS; mutations in the BRAF gene, such as mutations at position 599, such as V599E, and positions 461, 462, 463, 465, 468, 593, 596, 60, etc., which are associated with cancers, such as melanoma.
  • Compounds of the present invention also can be used as markers to determine the presence and quantity of raf, VEGFR-2, VEGFR-3, PDGFR-beta, and/or Flt-3.
  • Methods can involve the presence of Raf, VEGFR-2, VEGFR-3, PDGFR-beta, and/or Flt-3 in a sample comprising a biological material, comprising one or more of the following steps in any effective order, e.g., contacting said sample comprising a biological material with a compound of the present invention, and determining whether said compound binds to said material.
  • the compound can be labeled, or it can be used as a competitor to a labeled compound, such as labeled-ATP.
  • the invention also provides methods for treating, preventing, modulating, etc., diseases and conditions in mammals comprising administering a compound of this invention with another modulator of the signal transduction pathway comprising, but not limited to raf, VEGFR, PDGFR, and/or FLT-3. These can be present in the same composition or in separate formulations or dosage units. Administration can be the same or different routes, and can be simultaneous, sequential, etc.
  • the invention also relates to methods for treating, preventing, modulating, etc., diseases and conditions, comprising administering a compound of this invention with another active agent, e.g., once or more per day for up to 28 consecutive days with the concurrent or intermittent administration of another active agent over the same total time period.
  • Optional anti-hyper-proliferative agents which can be added to the composition include but are not limited to compounds listed on the cancer chemotherapy drug regimens in the 11 th Edition of the Merck Index , (1996), which is hereby incorporated by reference, such as asparaginase, bleomycin, carboplatin, carmustine, chlorambucil, cisplatin, colaspase, cyclophosphamide, cytarabine, dacarbazine, dactinomycin, daunorubicin, doxorubicin (adriamycine), epirubicin, etoposide, 5-fluorouracil, hexamethylmelamine, hydroxyurea, ifosfamide, irinotecan, leucovorin, lomustine, mechlorethamine, 6-mercaptopurine, mesna, methotrexate, mitomycin C, mitoxantrone, prednisolone, prednis
  • anti-hyper-proliferative agents suitable for use with the composition of the invention include, but are not limited to, those compounds acknowledged to be used in the treatment of neoplastic diseases in Goodman and Gilman's The Pharmacological Basis of Therapeutics (Ninth Edition), editor Molinoff et al., publ.
  • Compounds of the present invention can be administered in any form by any effective route, including, e.g., oral, parenteral, enteral, intravenous, intraperitoneal, topical, transdermal (e.g., using any standard patch), ophthalmic, nasally, local, non-oral, such as aerosal, inhalation, subcutaneous, intramuscular, buccal, sublingual, rectal, vaginal, intra-arterial, and intrathecal, etc. They can be administered alone, or in combination with any ingredient(s), active or inactive. They can be administered in any effective dosage, e.g., from about 0.1 to about 200 mg/kg of total body weight.
  • the present invention relates to a method for using the compounds described above (Compounds of Formula I), including salts and esters thereof and compositions thereof, to treat mammalian hyper-proliferative disorders.
  • This method comprises administering to a mammal in need thereof, including a human, an amount of a compound of this invention, or a pharmaceutically acceptable salt or ester thereof, which is effective to treat the disorder.
  • Hyper-proliferative disorders include but are not limited to solid tumors, such as cancers of the breast, respiratory tract, brain, reproductive organs, digestive tract, urinary tract, eye, liver, skin, head and neck, thyroid, parathyroid and their distant metastases. Those disorders also include lymphomas, sarcomas, and leukemias.
  • Synthetic transformations that may be employed in the synthesis of compounds of Formula I and in the synthesis of intermediates involved in the synthesis of compounds of Formula I are known by or accessible to one skilled in the art. Collections of synthetic transformations may be found in compilations, such as:
  • diaryl ureas of Formula I may be prepared by the use of known chemical reactions and procedures, some from starting materials which are commercially available. Nevertheless, general preparative methods are provided below to aid one skilled in the art in synthesizing these compounds, with more detailed examples being provided in the Experimental section which follows.
  • aryl amines are commonly synthesized by reduction of nitroaryls using a metal catalyst, such as Ni, Pd, or Pt, and H 2 or a hydride transfer agent, such as formate, cyclohexadiene, or a borohydride (Rylander. Hydrogenation Methods ; Academic Press: London, UK (1985)). Nitroaryls may also be directly reduced using a strong hydride source, such as LiAlH 4 (Seyden-Penne.
  • Nitroaryls are commonly formed by electrophilic aromatic nitration using HNO 3 , or an alternative NO 2 + source. Nitroaryls may be further elaborated prior to reduction. Thus, nitroaryls substituted with potential leaving groups (e.g. F, Cl, Br, etc.) may undergo substitution reactions on treatment with nucleophiles, such as thiolate (exemplified in Scheme II) or phenoxide. Nitroaryls may also undergo Ullman-type coupling reactions (Scheme II).
  • potential leaving groups e.g. F, Cl, Br, etc.
  • nucleophiles such as thiolate (exemplified in Scheme II) or phenoxide. Nitroaryls may also undergo Ullman-type coupling reactions (Scheme II).
  • Nitroaryls may also undergo transition metal mediated cross coupling reactions.
  • nitroaryl electrophiles such as nitroaryl bromides, iodides or triflates
  • palladium mediated cross coupling reactions with aryl nucleophiles, such as arylboronic acids (Suzuki reactions, exemplified below), aryltins (Stille reactions) or arylzincs (Negishi reaction) to afford the biaryl (5).
  • aryl nucleophiles such as arylboronic acids (Suzuki reactions, exemplified below), aryltins (Stille reactions) or arylzincs (Negishi reaction) to afford the biaryl (5).
  • non-symmetrical urea formation may involve reaction of an aryl isocyanate (14) with an aryl amine (13).
  • the heteroaryl isocyanate may be synthesized from a heteroaryl amine by treatment with phosgene or a phosgene equivalent, such as trichloromethyl chloroformate (diphosgene), bis(trichloromethyl) carbonate (triphosgene), or N,N′-carbonyldiimidazole (CDI).
  • the isocyanate may also be derived from a heterocyclic carboxylic acid derivative, such as an ester, an acid halide or an anhydride by a Curtius-type rearrangement.
  • reaction of acid derivative 16 with an azide source, followed by rearrangement affords the isocyanate.
  • the corresponding carboxylic acid (17) may also be subjected to Curtius-type rearrangements using diphenylphosphoryl azide (DPPA) or a similar reagent.
  • DPPA diphenylphosphoryl azide
  • ureas may be further manipulated using methods familiar to those skilled in the art.
  • the compounds may be administered orally, topically, parenterally, by inhalation or spray or rectally in dosage unit formulations.
  • administration by injection includes intravenous, intramuscular, subcutaneous and parenteral injections, as well as use of infusion techniques.
  • One or more compounds may be present in association with one or more non-toxic pharmaceutically acceptable carriers and if desired other active ingredients.
  • compositions intended for oral use may be prepared according to any suitable method known to the art for the manufacture of pharmaceutical compositions.
  • Such compositions may contain one or more agents selected from the group consisting of diluents, sweetening agents, flavoring agents, coloring agents and preserving agents in order to provide palatable preparations.
  • Tablets contain the active ingredient in admixture with non-toxic pharmaceutically acceptable excipients which are suitable for the manufacture of tablets.
  • excipients may be, for example, inert diluents, such as calcium carbonate, sodium carbonate, lactose, calcium phosphate or sodium phosphate; granulating and disintegrating agents, for example, corn starch, or alginic acid; and binding agents, for example magnesium stearate, stearic acid or talc.
  • the tablets may be uncoated or they may be coated by known techniques to delay disintegration and adsorption in the gastrointestinal tract and thereby provide a sustained action over a longer period.
  • a time delay material such as glyceryl monostearate or glyceryl distearate may be employed.
  • These compounds may also be prepared in solid, rapidly released form.
  • Formulations for oral use may also be presented as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent, for example, calcium carbonate, calcium phosphate or kaolin, or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium, for example peanut oil, liquid paraffin or olive oil.
  • an inert solid diluent for example, calcium carbonate, calcium phosphate or kaolin
  • water or an oil medium for example peanut oil, liquid paraffin or olive oil.
  • Aqueous suspensions contain the active materials in admixture with excipients suitable for the manufacture of aqueous suspensions.
  • excipients are suspending agents, for example sodium carboxymethylcellulose, methylcellulose, hydroxypropyl methylcellulose, sodium alginate, polyvinylpyrrolidone, gum tragacanth and gum acacia; dispersing or wetting agents may be a naturally occurring phosphatide, for example, lecithin, or condensation products or an alkylene oxide with fatty acids, for example polyoxyethylene stearate, or condensation products of ethylene oxide with long chain aliphatic alcohols, for example heptadecaethylene oxycetanol, or condensation products of ethylene oxide with partial esters derived from fatty acids and hexitol such as polyoxyethylene sorbitol monooleate, or condensation products of ethylene oxide with partial esters derived from fatty acids and hexitol anhydrides, for example polyethylene sorbitan monooleate.
  • the aqueous suspensions may also contain one or more preservatives, for example ethyl, or n-propyl p-hydroxybenzoate, one or more coloring agents, one or more flavoring agents, and one or more sweetening agents, such as sucrose or saccharin.
  • preservatives for example ethyl, or n-propyl p-hydroxybenzoate
  • coloring agents for example ethyl, or n-propyl p-hydroxybenzoate
  • flavoring agents for example ethyl, or n-propyl p-hydroxybenzoate
  • sweetening agents such as sucrose or saccharin.
  • Dispersible powders and granules suitable for preparation of an aqueous suspension by the addition of water provide the active ingredient in admixture with a dispersing or wetting agent, suspending agent and one or more preservatives.
  • a dispersing or wetting agent e.g., talc, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, sorbitol, sorbitol, sorbitol, sorbitol, sorbitol, sorbitol, sorbitol, sorbitol, sorbitol, sorbitol, sorbitol, mannitol, mannitol, mannitol, mannitol, mannitol, mannitol, mannitol, mannitol,
  • the compounds may also be in the form of non-aqueous liquid formulations, e.g., oily suspensions which may be formulated by suspending the active ingredients in a vegetable oil, for example arachis oil, olive oil, sesame oil or peanut oil, or in a mineral oil such as liquid paraffin.
  • oily suspensions may contain a thickening agent, for example beeswax, hard paraffin or cetyl alcohol. Sweetening agents such as those set forth above, and flavoring agents may be added to provide palatable oral preparations. These compositions may be preserved by the addition of an anti-oxidant such as ascorbic acid.
  • compositions of the invention may also be in the form of oil-in-water emulsions.
  • the oily phase may be a vegetable oil, for example olive oil or arachis oil, or a mineral oil, for example liquid paraffin or mixtures of these.
  • Suitable emulsifying agents may be naturally-occurring gums, for example gum acacia or gum tragacanth, naturally-occurring phosphatides, for example soy bean, lecithin, and esters or partial esters derived from fatty acids and hexitol anhydrides, for example sorbitan monooleate, and condensation products of the said partial esters with ethylene oxide, for example polyoxyethylene sorbitan monooleate.
  • the emulsions may also contain sweetening and flavoring agents.
  • Syrups and elixirs may be formulated with sweetening agents, for example glycerol, propylene glycol, sorbitol or sucrose. Such formulations may also contain a demulcent, a preservative and flavoring and coloring agents.
  • sweetening agents for example glycerol, propylene glycol, sorbitol or sucrose.
  • Such formulations may also contain a demulcent, a preservative and flavoring and coloring agents.
  • the compounds may also be administered in the form of suppositories for rectal administration of the drug.
  • These compositions can be prepared by mixing the drug with a suitable non-irritating excipient which is solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum to release the drug.
  • suitable non-irritating excipient include cocoa butter and polyethylene glycols.
  • the compounds of this invention may also be administered parenterally, that is, subcutaneously, intravenously, intraocularly, intrasynovially, intramuscularly, or interperitoneally, as injectable dosages of the compound in a physiologically acceptable diluent with a pharmaceutical carrier which can be a sterile liquid or mixture of liquids such as water, saline, aqueous dextrose and related sugar solutions, an alcohol such as ethanol, isopropanol, or hexadecyl alcohol, glycols such as propylene glycol or polyethylene glycol, glycerol ketals such as 2,2-dimethyl-1,1-dioxolane-4-methanol, ethers such as poly(ethylene glycol) 400, an oil, a fatty acid, a fatty acid ester or, a fatty acid glyceride, or an acetylated fatty acid glyceride, with or without the addition of a pharmaceutically acceptable surfactant such as
  • Suitable fatty acids include oleic acid, stearic acid, isostearic acid and myristic acid.
  • Suitable fatty acid esters are, for example, ethyl oleate and isopropyl myristate.
  • Suitable soaps include fatty acid alkali metal, ammonium, and triethanolamine salts and suitable detergents include cationic detergents, for example dimethyl dialkyl ammonium halides, alkyl pyridinium halides, and alkylamine acetates; anionic detergents, for example, alkyl, aryl, and olefin sulfonates, alkyl, olefin, ether, and monoglyceride sulfates, and sulfosuccinates; non-ionic detergents, for example, fatty amine oxides, fatty acid alkanolamides, and poly(oxyethylene-oxypropylene)s or ethylene oxide or propylene oxide copolymers; and amphoteric detergents, for example, alkyl-beta-aminopropionates, and 2-alkylimidazoline quarternary ammonium salts, as well as mixtures.
  • suitable detergents include cationic detergents, for example di
  • compositions of this invention will typically contain from about 0.5% to about 25% by weight of the active ingredient in solution. Preservatives and buffers may also be used advantageously. In order to minimize or eliminate irritation at the site of injection, such compositions may contain a non-ionic surfactant having a hydrophile-lipophile balance (HLB) of from about 12 to about 17. The quantity of surfactant in such formulation ranges from about 5% to about 15% by weight.
  • the surfactant can be a single component having the above HLB or can be a mixture of two or more components having the desired HLB.
  • surfactants used in parenteral formulations are the class of polyethylene sorbitan fatty acid esters, for example, sorbitan monooleate and the high molecular weight adducts of ethylene oxide with a hydrophobic base, formed by the condensation of propylene oxide with propylene glycol.
  • compositions may be in the form of sterile injectable aqueous suspensions.
  • suspensions may be formulated according to known methods using suitable dispersing or wetting agents and suspending agents such as, for example, sodium carboxymethylcellulose, methylcellulose, hydroxypropylmethyl-cellulose, sodium alginate, polyvinylpyrrolidone, gum tragacanth and gum acacia; dispersing or wetting agents which may be a naturally occurring phosphatide such as lecithin, a condensation product of an alkylene oxide with a fatty acid, for example, polyoxyethylene stearate, a condensation product of ethylene oxide with a long chain aliphatic alcohol, for example, heptadeca-ethyleneoxycetanol, a condensation product of ethylene oxide with a partial ester derived form a fatty acid and a hexitol such as polyoxyethylene sorbitol monooleate, or a condensation product of an ethylene oxide with a partial ester derived from a
  • the sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally acceptable diluent or solvent.
  • Diluents and solvents that may be employed are, for example, water, Ringer's solution, isotonic sodium chloride solutions and isotonic glucose solutions.
  • sterile fixed oils are conventionally employed as solvents or suspending media.
  • any bland, fixed oil may be employed including synthetic mono- or diglycerides.
  • fatty acids such as oleic acid can be used in the preparation of injectables.
  • transdermal patches may be used to provide continuous or discontinuous infusion of the compounds of the present invention in controlled amounts.
  • the construction and use of transdermal patches for the delivery of pharmaceutical agents is well known in the art (see, e.g., U.S. Pat. No. 5,023,252, issued Jun. 11, 1991, incorporated herein by reference).
  • patches may be constructed for continuous, pulsatile, or on demand delivery of pharmaceutical agents.
  • a solution or suspension of a compound of Formula I in a suitable volatile solvent optionally containing penetration enhancing agents can be combined with additional additives known to those skilled in the art, such as matrix materials and bacteriocides. After sterilization, the resulting mixture can be formulated following known procedures into dosage forms.
  • additional additives known to those skilled in the art, such as matrix materials and bacteriocides.
  • the resulting mixture can be formulated following known procedures into dosage forms.
  • a solution or suspension of a compound of Formula I may be formulated into a lotion or salve.
  • Suitable solvents for processing transdermal delivery systems are known to those skilled in the art, and include lower alcohols such as ethanol or isopropyl alcohol, lower ketones such as acetone, lower carboxylic acid esters such as ethyl acetate, polar ethers such as tetrahydrofuran, lower hydrocarbons such as hexane, cyclohexane or benzene, or halogenated hydrocarbons such as dichloromethane, chloroform, trichlorotrifluoroethane, or trichlorofluoroethane.
  • Suitable solvents may also include mixtures of one or more materials selected from lower alcohols, lower ketones, lower carboxylic acid esters, polar ethers, lower hydrocarbons, halogenated hydrocarbons.
  • Suitable penetration enhancing materials for transdermal delivery system include, for example, monohydroxy or polyhydroxy alcohols such as ethanol, propylene glycol or benzyl alcohol, saturated or unsaturated C 8 -C 18 fatty alcohols such as lauryl alcohol or cetyl alcohol, saturated or unsaturated C 8 -C 18 fatty acids such as stearic acid, saturated or unsaturated fatty esters with up to 24 carbons such as methyl, ethyl, propyl, isopropyl, n-butyl, sec-butyl isobutyl tertbutyl or monoglycerin esters of acetic acid, capronic acid, lauric acid, myristinic acid, stearic acid, or palmitic acid, or diesters of saturated or unsaturated dicarboxylic acids with a total of up to 24 carbons such as diisopropyl adipate, diisobutyl adipate, diisopropy
  • Additional penetration enhancing materials include phosphatidyl derivatives such as lecithin or cephalin, terpenes, amides, ketones, ureas and their derivatives, and ethers such as dimethyl isosorbid and diethyleneglycol monoethyl ether.
  • Suitable penetration enhancing formulations may also include mixtures of one or more materials selected from monohydroxy or polyhydroxy alcohols, saturated or unsaturated C 8 -C 18 fatty alcohols, saturated or unsaturated C 8 -C 18 fatty acids, saturated or unsaturated fatty esters with up to 24 carbons, diesters of saturated or unsaturated discarboxylic acids with a total of up to 24 carbons, phosphatidyl derivatives, terpenes, amides, ketones, ureas and their derivatives, and ethers.
  • Suitable binding materials for transdermal delivery systems include polyacrylates, silicones, polyurethanes, block polymers, styrenebutadiene copolymers, and natural and synthetic rubbers.
  • Cellulose ethers, derivatized polyethylenes, and silicates may also be used as matrix components. Additional additives, such as viscous resins or oils may be added to increase the viscosity of the matrix.
  • Controlled release formulations for parenteral administration include liposomal, polymeric microsphere and polymeric gel formulations which are known in the art.
  • a mechanical delivery device It may be desirable or necessary to introduce the pharmaceutical composition to the patient via a mechanical delivery device.
  • the construction and use of mechanical delivery devices for the delivery of pharmaceutical agents is well known in the art.
  • Direct techniques for, for example, administering a drug directly to the brain usually involve placement of a drug delivery catheter into the patient's ventricular system to bypass the blood-brain barrier.
  • One such implantable delivery system, used for the transport of agents to specific anatomical regions of the body is described in U.S. Pat. No. 5,011,472, issued Apr. 30, 1991.
  • compositions of the invention can also contain other conventional pharmaceutically acceptable compounding ingredients, generally referred to as carriers or diluents, as necessary or desired.
  • Conventional procedures for preparing such compositions in appropriate dosage forms can be utilized. Such ingredients and procedures include those described in the following references, each of which is incorporated herein by reference: Powell, M. F. et al, “Compendium of Excipients for Parenteral Formulations” PDA Journal of Pharmaceutical Science & Technology 1998, 52(5), 238-311; Strickley, R. G “Parenteral Formulations of Small Molecule Therapeutics Marketed in the United States (1999)-Part-1 ” PDA Journal of Pharmaceutical Science & Technology 1999, 53(6), 324-349; and Nema, S. et al, “Excipients and Their Use in Injectable Products” PDA Journal of Pharmaceutical Science & Technology 1997, 51(4), 166-171.
  • compositions containing one or more compounds of the present invention can be utilized to achieve the desired pharmacological effect by administration to a patient in need thereof.
  • a patient for the purpose of this invention, is a mammal, including a human, in need of treatment for the particular condition or disease. Therefore, the present invention includes pharmaceutical compositions which are comprised of a pharmaceutically acceptable carrier and a pharmaceutically effective amount of a compound, or salt thereof, of the present invention.
  • a pharmaceutically acceptable carrier is any carrier which is relatively non-toxic and innocuous to a patient at concentrations consistent with effective activity of the active ingredient so that any side effects ascribable to the carrier do not vitiate the beneficial effects of the active ingredient.
  • a pharmaceutically effective amount of compound is that amount which produces a result or exerts an influence on the particular condition being treated.
  • the compounds of the present invention can be administered with pharmaceutically-acceptable carriers well known in the art using any effective conventional dosage unit forms, including immediate, slow and timed release preparations, orally, parenterally, topically, nasally, ophthalmically, otically, sublingually, rectally, vaginally, and the like.
  • the compounds can be formulated into solid or liquid preparations such as capsules, pills, tablets, troches, lozenges, melts, powders, solutions, suspensions, or emulsions, and may be prepared according to methods known to the art for the manufacture of pharmaceutical compositions.
  • the solid unit dosage forms can be a capsule which can be of the ordinary hard- or soft-shelled gelatin type containing, for example, surfactants, lubricants, and inert fillers such as lactose, sucrose, calcium phosphate, and corn starch.
  • the compounds of this invention may be tableted with conventional tablet bases such as lactose, sucrose and cornstarch in combination with binders such as acacia, corn starch or gelatin, disintegrating agents intended to assist the break-up and dissolution of the tablet following administration such as potato starch, alginic acid, corn starch, and guar gum, gum tragacanth, acacia, lubricants intended to improve the flow of tablet granulation and to prevent the adhesion of tablet material to the surfaces of the tablet dies and punches, for example talc, stearic acid, or magnesium, calcium or zinc stearate, dyes, coloring agents, and flavoring agents such as peppermint, oil of wintergreen, or cherry flavoring, intended to enhance the aesthetic qualities of the tablets and make them more acceptable to the patient.
  • binders such as acacia, corn starch or gelatin
  • disintegrating agents intended to assist the break-up and dissolution of the tablet following administration such as potato starch, alginic acid, corn star
  • Suitable excipients for use in oral liquid dosage forms include dicalcium phosphate and diluents such as water and alcohols, for example, ethanol, benzyl alcohol, and polyethylene alcohols, either with or without the addition of a pharmaceutically acceptable surfactant, suspending agent or emulsifying agent.
  • Various other materials may be present as coatings or to otherwise modify the physical form of the dosage unit. For instance tablets, pills or capsules may be coated with shellac, sugar or both.
  • compositions of the invention can also contain other conventional pharmaceutically acceptable compounding ingredients, generally referred to as carriers or diluents, as necessary or desired.
  • Conventional procedures for preparing such compositions in appropriate dosage forms can be utilized. Such ingredients and procedures include those described in the following references, each of which is incorporated herein by reference: Powell, M. F. et al, “Compendium of Excipients for Parenteral Formulations” PDA Journal of Pharmaceutical Science & Technology 1998, 52(5), 238-311; Strickley, R. G “Parenteral Formulations of Small Molecule Therapeutics Marketed in the United States (1999)-Part-1 ” PDA Journal of Pharmaceutical Science & Technology 1999, 53(6), 324-349; and Nema, S. et al, “Excipients and Their Use in Injectable Products” PDA Journal of Pharmaceutical Science & Technology 1997, 51(4), 166-171.
  • compositions for its intended route of administration include:
  • acidifying agents include but are not limited to acetic acid, citric acid, fumaric acid, hydrochloric acid, nitric acid);
  • alkalinizing agents examples include but are not limited to ammonia solution, ammonium carbonate, diethanolamine, monoethanolamine, potassium hydroxide, sodium borate, sodium carbonate, sodium hydroxide, triethanolamine, trolamine;
  • adsorbents examples include but are not limited to powdered cellulose and activated charcoal
  • aerosol propellants examples include but are not limited to carbon dioxide, CCl 2 F 2 , F 2 ClC—CClF 2 and CClF 3 )
  • air displacement agents examples include but are not limited to nitrogen and argon
  • antifungal preservatives examples include but are not limited to benzoic acid, butylparaben, ethylparaben, methylparaben, propylparaben, sodium benzoate);
  • antimicrobial preservatives examples include but are not limited to benzalkonium chloride, benzethonium chloride, benzyl alcohol, cetylpyridinium chloride, chlorobutanol, phenol, phenylethyl alcohol, phenylmercuric nitrate and thimerosal;
  • antioxidants examples include but are not limited to ascorbic acid, ascorbyl palmitate, butylated hydroxyanisole, butylated hydroxytoluene, hypophosphorus acid, monothioglycerol, propyl gallate, sodium ascorbate, sodium bisulfite, sodium formaldehyde sulfoxylate, sodium metabisulfite);
  • binding materials examples include but are not limited to block polymers, natural and synthetic rubber, polyacrylates, polyurethanes, silicones, polysiloxanes and styrene-butadiene copolymers;
  • buffering agents examples include but are not limited to potassium metaphosphate, dipotassium phosphate, sodium acetate, sodium citrate anhydrous and sodium citrate dihydrate
  • carrying agents examples include but are not limited to acacia syrup, aromatic syrup, aromatic elixir, cherry syrup, cocoa syrup, orange syrup, syrup, corn oil, mineral oil, peanut oil, sesame oil, bacteriostatic sodium chloride injection and bacteriostatic water for injection
  • examples include but are not limited to acacia syrup, aromatic syrup, aromatic elixir, cherry syrup, cocoa syrup, orange syrup, syrup, corn oil, mineral oil, peanut oil, sesame oil, bacteriostatic sodium chloride injection and bacteriostatic water for injection
  • chelating agents examples include but are not limited to edetate disodium and edetic acid
  • colorants examples include but are not limited to FD&C Red No. 3, FD&C Red No. 20, FD&C Yellow No. 6, FD&C Blue No. 2, D&C Green No. 5, D&C Orange No. 5, D&C Red No. 8, caramel and ferric oxide red);
  • clarifying agents examples include but are not limited to bentonite
  • emulsifying agents examples include but are not limited to acacia, cetomacrogol, cetyl alcohol, glyceryl monostearate, lecithin, sorbitan monooleate, polyoxyethylene 50 monostearate);
  • encapsulating agents examples include but are not limited to gelatin and cellulose acetate phthalate
  • flavorants examples include but are not limited to anise oil, cinnamon oil, cocoa, menthol, orange oil, peppermint oil and vanillin);
  • humectants examples include but are not limited to glycerol, propylene glycol and sorbitol
  • levigating agents examples include but are not limited to mineral oil and glycerin
  • oils examples include but are not limited to arachis oil, mineral oil, olive oil, peanut oil, sesame oil and vegetable oil);
  • ointment bases examples include but are not limited to lanolin, hydrophilic ointment, polyethylene glycol ointment, petrolatum, hydrophilic petrolatum, white ointment, yellow ointment, and rose water ointment;
  • penetration enhancers include but are not limited to monohydroxy or polyhydroxy alcohols, mono- or polyvalent alcohols, saturated or unsaturated fatty alcohols, saturated or unsaturated fatty esters, saturated or unsaturated dicarboxylic acids, essential oils, phosphatidyl derivatives, cephalin, terpenes, amides, ethers, ketones and ureas
  • monohydroxy or polyhydroxy alcohols mono- or polyvalent alcohols
  • saturated or unsaturated fatty alcohols saturated or unsaturated fatty esters
  • saturated or unsaturated dicarboxylic acids saturated or unsaturated dicarboxylic acids
  • essential oils phosphatidyl derivatives
  • cephalin cephalin
  • terpenes amides, ethers, ketones and ureas
  • plasticizers examples include but are not limited to diethyl phthalate and glycerol
  • solvents examples include but are not limited to ethanol, corn oil, cottonseed oil, glycerol, isopropanol, mineral oil, oleic acid, peanut oil, purified water, water for injection, sterile water for injection and sterile water for irrigation);
  • stiffening agents examples include but are not limited to cetyl alcohol, cetyl esters wax, microcrystalline wax, paraffin, stearyl alcohol, white wax and yellow wax;
  • suppository bases examples include but are not limited to cocoa butter and polyethylene glycols (mixtures));
  • surfactants examples include but are not limited to benzalkonium chloride, nonoxynol 10, oxtoxynol 9, polysorbate 80, sodium lauryl sulfate and sorbitan mono-palmitate);
  • suspending agents examples include but are not limited to agar, bentonite, carbomers, carboxymethylcellulose sodium, hydroxyethyl cellulose, hydroxypropyl cellulose, hydroxypropyl methylcellulose, kaolin, methylcellulose, tragacanth and veegum);
  • sweetening agents examples include but are not limited to aspartame, dextrose, glycerol, mannitol, propylene glycol, saccharin sodium, sorbitol and sucrose;
  • tablet anti-adherents examples include but are not limited to magnesium stearate and talc
  • tablet binders examples include but are not limited to acacia, alginic acid, carboxymethylcellulose sodium, compressible sugar, ethylcellulose, gelatin, liquid glucose, methylcellulose, non-crosslinked polyvinyl pyrrolidone, and pregelatinized starch;
  • tablet and capsule diluents examples include but are not limited to dibasic calcium phosphate, kaolin, lactose, mannitol, microcrystalline cellulose, powdered cellulose, precipitated calcium carbonate, sodium carbonate, sodium phosphate, sorbitol and starch);
  • tablet coating agents examples include but are not limited to liquid glucose, hydroxyethyl cellulose, hydroxypropyl cellulose, hydroxypropyl methylcellulose, methylcellulose, ethylcellulose, cellulose acetate phthalate and shellac);
  • tablet direct compression excipients examples include but are not limited to dibasic calcium phosphate
  • tablet disintegrants examples include but are not limited to alginic acid, carboxymethylcellulose calcium, microcrystalline cellulose, polacrillin potassium, cross-linked polyvinylpyrrolidone, sodium alginate, sodium starch glycollate and starch;
  • tablet glidants examples include but are not limited to colloidal silica, corn starch and talc;
  • tablet lubricants examples include but are not limited to calcium stearate, magnesium stearate, mineral oil, stearic acid and zinc stearate);
  • tablet/capsule opaquants examples include but are not limited to titanium dioxide
  • tablet polishing agents examples include but are not limited to carnauba wax and white wax
  • thickening agents examples include but are not limited to beeswax, cetyl alcohol and paraffin
  • tonicity agents examples include but are not limited to dextrose and sodium chloride
  • viscosity increasing agents examples include but are not limited to alginic acid, bentonite, carbomers, carboxymethylcellulose sodium, methylcellulose, polyvinyl pyrrolidone, sodium alginate and tragacanth; and
  • wetting agents examples include but are not limited to heptadecaethylene oxycetanol, lecithins, sorbitol monooleate, polyoxyethylene sorbitol monooleate, and polyoxyethylene stearate).
  • the total amount of the active ingredient to be administered will generally range from about 0.001 mg/kg to about 200 mg/kg, and preferably from about 0.01 mg/kg to about 20 mg/kg body weight per day.
  • a unit dosage may contain from about 0.5 mg to about 1500 mg of active ingredient, and can be administered one or more times per day.
  • the daily oral dosage regimen will preferably be from 0.01 to 200 mg/Kg of total body weight.
  • the daily dosage for administration by injection, including intravenous, intramuscular, subcutaneous and parenteral injections, and use of infusion techniques will preferably be from 0.01 to 200 mg/Kg of total body weight.
  • the daily rectal dosage regime will preferably be from 0.01 to 200 mg/Kg of total body weight.
  • the daily vaginal dosage regime will preferably be from 0.01 to 200 mg/Kg of total body weight.
  • the daily topical dosage regime will preferably be from 0.1 to 200 mg administered between one to four times daily.
  • the transdermal concentration will preferably be that required to maintain a daily dose of from 0.01 to 200 mg/Kg.
  • the daily inhalation dosage regime will preferably be from 0.01 to 100 mg/Kg of total body weight.
  • the effective dosage of the compounds of this invention can readily be determined for treatment of each desired indication.
  • the amount of the active ingredient to be administered in the treatment of one of these conditions can vary widely according to such considerations as the particular compound and dosage unit employed, the mode of administration, the period of treatment, the age and gender of the patient treated, and the nature and extent of the condition treated.
  • the particular method of administration will depend on a variety of factors, all of which are considered routinely when administering therapeutics.
  • the specific dose level for a given patient depends on a variety of factors, including specific activity of the compound administered, age, body weight, health, sex, diet, time and route of administration, rate of excretion, etc.
  • the optimal course of treatment i.e., the mode of treatment and the daily number of doses of a compound of Formula I or a pharmaceutically acceptable salt thereof given for a defined number of days, can be ascertained by those skilled in the art using conventional treatment tests.
  • the specific dose level for any particular patient will depend upon a variety of factors, including the activity of the specific compound employed, the age, body weight, general health, sex, diet, time of administration, route of administration, and rate of excretion, drug combination and the severity of the condition undergoing therapy.
  • compositions according to the present invention can be illustrated as follows:
  • Sterile IV Solution A 5 mg/ml solution of the desired compound of this invention is made using sterile, injectable water, and the pH is adjusted if necessary. The solution is diluted for administration to 1-2 mg/ml with sterile 5% dextrose and is administered as an IV infusion over 60 minutes.
  • Lyophilized powder for IV administration A sterile preparation can be prepared with (i) 100-1000 mg of the desired compound of this invention as a lyophilized powder, (ii) 32-327 mg/ml sodium citrate, and (iii) 300-3000 mg Dextran 40.
  • the formulation is reconstituted with sterile, injectable saline or dextrose 5% to a concentration of 10 to 20 mg/ml, which is further diluted with saline or dextrose 5% to 0.2-0.4 mg/ml, and is administered either IV bolus or by IV infusion over 15-60 minutes.
  • Intramuscular suspension The following solution or suspension can be prepared, for intramuscular injection:
  • the compounds can be produced from known compounds (or from starting materials which, in turn, can be produced from known compounds), e.g., through the general preparative methods shown below.
  • the activity of a given compound to inhibit raf kinase can be routinely assayed, e.g., according to procedures disclosed below.
  • the following examples are for illustrative purposes only and are not intended, nor should they be construed to limit the invention in any way.
  • N-cyclohexyl-N′-(methylpolystyrene)carbodiimide was purchased from Calbiochem-Novabiochem Corp. 3-tert-Butylaniline, 5-tert-butyl-2-methoxyamine, 4-bromo-3-(trifluoromethyl)aniline, 4-chloro-3-(trifluoromethyl)aniline 2-methoxy-5-(trifluoromethyl)aniline, 4-tert-butyl-2-nitroaniline, 3-amino-2-naphthol, ethyl 4-isocyanatobenzoate, N-acetyl-4-chloro-2-methoxy-5-(trifluoromethyl)aniline and 4-chloro-3-(trifluoromethyl)phenyl isocyanate were purchased and used without further purification.
  • Thin-layer chromatography was performed using Whatman® pre-coated glass-backed silica gel 60A F-254 250 ⁇ m plates. Visualization of plates was effected by one or more of the following techniques: (a) ultraviolet illumination, (b) exposure to iodine vapor, (c) immersion of the plate in a 10% solution of phosphomolybdic acid in ethanol followed by heating, (d) immersion of the plate in a cerium sulfate solution followed by heating, and/or (e) immersion of the plate in an acidic ethanol solution of 2,4-dinitrophenylhydrazine followed by heating. Column chromatography (flash chromatography) was performed using 230400 mesh EM Science® silica gel.
  • Electron impact ionization was performed with electron energy of 70 eV and a trap current of 300 ⁇ A.
  • Liquid-cesium secondary ion mass spectra FAB-MS
  • FAB-MS Liquid-cesium secondary ion mass spectra
  • CI-MS Chemical ionization mass spectra
  • the direct insertion desorption chemical ionization (DCI) probe (Vaccumetrics, Inc.) was ramped from 0-1.5 amps in 10 sec and held at 10 amps until all traces of the sample disappeared ( ⁇ 1-2 min). Spectra were scanned from 50-800 amu at 2 sec per scan.
  • HPLC-electrospray mass spectra (HPLC ES-MS) were obtained using a Hewlett-Packard 1100 HPLC equipped with a quaternary pump, a variable wavelength detector, a C-18 column, and a Finnigan LCQ ion trap mass spectrometer with electrospray ionization. Spectra were scanned from 120-800 amu using a variable ion time according to the number of ions in the source.
  • GC-MS Gas chromatography-ion selective mass spectra
  • Step 3 Preparation of N-[4-chloro-3-(trifluoromethyl)phenyl]-N′- ⁇ 4-[2-carbamoyl-(4-pyridyloxy)]phenyl ⁇ urea
  • Step 1 4-Chloro-N-methyl-2-pyridinecarboxamide is first synthesized from 4-chloropyridine-2-carbonyl chloride by adding 4-chloropyridine-2-carbonyl chloride HCl salt (7.0 g, 32.95 mmol) in portions to a mixture of a 2.0 M methylamine solution in THF (100 mL) and MeOH (20 mL) at 0° C. The resulting mixture is stored at 3° C. for 4 h, then concentrated under reduced pressure. The resulting nearly dry solids are suspended in EtOAc (100 mL) and filtered. The filtrate is washed with a saturated NaCl solution (2 ⁇ 100 mL), dried Na 2 SO 4 ) and concentrated under reduced pressure to provide 4-chloro-N-methyl-2-pyridinecarboxaxnide as a yellow, crystalline solid.
  • 4-chloro-N-methyl-2-pyridinecarboxaxnide is first synthesized from 4-chloropyridine-2-carbonyl chloride by adding
  • Step 2 A solution of 4-aminophenol (9.60 g, 88.0 mmol) in anh. DMF (150 mL) is treated with potassium tert-butoxide (10.29 g, 91.7 mmol), and the reddish-brown mixture is stirred at room temp. for 2 h. The contents are treated with 4-chloro-N-methyl-2-pyridinecarboxamide (15.0 g, 87.9 mmol) from Step 1 and K 2 CO 3 (6.50 g, 47.0 mmol) and then heated at 80° C. for 8 h. The mixture is cooled to room temp. and separated between EtOAc (500 mL) and a saturated NaCl solution (500 mL).
  • Step 3 A solution of 4-chloro-3-(trifluoromethyl)phenyl isocyanate (14.60 g, 65.90 mmol) in CH 2 Cl 2 (35 mL) is added dropwise to a suspension of 4-(2-(N-methylcarbamoyl)-4-pyridyloxy)aniline from Step 2; (16.0 g, 65.77 mmol) in CH 2 Cl 2 (35 mL) at 0° C. The resulting mixture is stirred at room temp. for 22 h.
  • Step 1 4-Chloro-N-methyl-2-pyridinecarboxamide is first synthesized from 4-chloropyridine-2-carbonyl chloride by adding 4-chloropyridine-2-carbonyl chloride HCl salt (7.0 g, 32.95 mmol) in portions to a mixture of a 2.0 M methylamine solution in THF (100 mL) and MeOH (20 mL) at 0° C. The resulting mixture is stored at 3° C. for 4 h, then concentrated under reduced pressure. The resulting nearly dry solids are suspended in EtOAc (100 mL) and filtered. The filtrate is washed with a saturated NaCl solution (2 ⁇ 100 mL), dried (Na 2 SO 4 ) and concentrated under reduced pressure to provide 4-chloro-N-methyl-2-pyridinecarboxamide as a yellow, crystalline solid.
  • Step 2 A solution of 4-aminophenol (9.60 g, 88.0 mmol) in anh. DMF (150 mL) is treated with potassium tert-butoxide (10.29 g, 91.7 mmol), and the reddish-brown mixture is stirred at room temp. for 2 h. The contents are treated with 4-chloro-N-methyl-2-pyridinecarboxamide (15.0 g, 87.9 mmol) from Step 1 and K 2 CO 3 (6.50 g, 47.0 mmol) and then heated at 80° C. for 8 h. The mixture is cooled to room temp. and separated between EtOAc (500 mL) and a saturated NaCl solution (500 mL).
  • Step 3 To a solution of 2-methoxy-5-(trifluoromethyl)aniline (0.15 g) in anh CH 2 Cl 2 (15 mL) at 0° C. is added CDI (0.13 g). The resulting solution is allowed to warm to room temp. over 1 h, is stirred at room temp. for 16 h, then is treated with 4-(2-(N-methylcarbamoyl)-4-pyridyloxy)aniline (0.18 g) from Step 2. The resulting yellow solution is stirred at room temp. for 72 h, then is treated with H 2 O (125 mL). The resulting aqueous mixture is extracted with EtOAc (2 ⁇ 150 mL).
  • the c-Raf biochemical assay was performed with a c-Raf enzyme that was activated (phosphorylated) by Lck kinase.
  • Lck-activated c-Raf (Lck/c-Raf) was produced in Sf9 insect cells by co-infecting cells with baculoviruses expressing, under the control of the polyhedrin promoter, GST-c-Raf (from amino acid 302 to amino acid 648) and Lck (full-length). Both baculoviruses were used at the multiplicity of infection of 2.5 and the cells were harvested 48 hours post infection.
  • MEK-1 protein was produced in Sf9 insect cells by infecting cells with the baculovirus expressing GST-MEK-1 (full-length) fusion protein at the multiplicity of infection of 5 and harvesting the cells 48 hours post infection. Similar purification procedure was used for GST-c-Raf 302-648 and GST-MEK-1.
  • Transfected cells were suspended at 100 mg of wet cell biomass per mL in a buffer containing 10 mM sodium phosphate, 140 mM sodium chloride pH 7.3, 0.5% Triton X-100 and the protease inhibitor cocktail.
  • the cells were disrupted with Polytron homogenizer and centrifuged 30,000 g for 30 minutes. The 30,000 g supernatant was applied onto GSH-Sepharose. The resin was washed with a buffer containing 50 mM Tris, pH 8.0, 150 M NaCl and 0.01% Triton X-100.
  • the GST-tagged proteins were eluted with a solution containing 100 mM Glutathione, 50 mM Tris, pH 8.0, 150 mM NaCl and 0.01% Triton X-100.
  • the purified proteins were dialyzed into a buffer containing 20 mM Tris, pH 7.5, 150 mM NaCl and 20% Glycerol.
  • Test compounds were serially diluted in DMSO using three-fold dilutions to stock concentrations ranging typically from 50 ⁇ M to 20 nM (final concentrations in the assay range from 1 ⁇ M to 0.4 nM).
  • the c-Raf biochemical assay was performed as a radioactive filtermat assay in 96-well Costar polypropylene plates (Costar 3365). The plates were loaded with 75 ⁇ L solution containing 50 mM HEPES pH 7.5, 70 mM NaCl, 80 ng of Lck/c-Raf and 1 ⁇ g MEK-1. Subsequently, 2 ⁇ L of the serially diluted individual compounds were added to the reaction, prior to the addition of ATP.
  • the reaction was initiated with 25 ⁇ L ATP solution containing 5 ⁇ M ATP and 0.3 ⁇ Ci [33P]-ATP. The plates were sealed and incubated at 32° C. for 1 hour. The reaction was quenched with the addition of 50 ⁇ l of 4% Phosphoric Acid and harvested onto P30 filtermats (PerkinElmer) using a Wallac Tomtec Harvester. Filtermats were washed with 1% Phosphoric Acid first and deinonized H 2 O second. The filters were dried in a microwave, soaked in scintillation fluid and read in a Wallac 1205 Betaplate Counter (Wallac Inc., Atlanta, Ga., U.S.A.). The results were expressed as percent inhibition.
  • a 96-well phospho-ERK (PERK) immunoassay using laser flow cytometry platform has been established to measure inhibition of basal pERK in cell lines.
  • MDA-MB-231 cells were plated at 50,000 cells per well in 96-well microtitre plates in complete growth media.
  • test compounds were transferred to DMEM with 0.1% BSA and incubated with test compounds diluted 1:3 to a final concentration of 3 ⁇ M to 12 nM in 0.1% DMSO. Cells were incubated with test compounds for 2 h, washed, and lysed in Bio-Plex whole cell lysis buffer A.
  • Samples are diluted with buffer B 1:1 (v/v) and directly transferred to assay plate or frozen at ⁇ 80 C degrees until processed.
  • 50 ⁇ L of diluted MDA-MB-231 cell lysates were incubated with about 2000 of 5 micron Bio-Plex beads conjugated with an anti-ERK1/2 antibody overnight on a shaker at room temperature. The next day, biotinylated phospho-ERK1/2 sandwich immunoassay was performed, beads are washed 3 times during each incubation and then 50 ⁇ L of PE-strepavidin was used as a developing reagent.
  • the relative fluorescence units of pERK1/2 were detected by counting 25 beads with Bio-Plex flow cell (probe) at high sensitivity.
  • the IC 50 was calculated by taking untreated cells as maximum and no cells (beads only) as background.
  • Raf kinase phosphorylates and activates MEK (mitogen-activated protein kinase kinase), which in turn phosphorylates and activates ERK (extracellular signal-regulated kinase), which translocates to the nucleus and modifies gene expression.
  • MEK mitogen-activated protein kinase kinase
  • ERK extracellular signal-regulated kinase
  • This pathway is often aberrantly activated in tumor cells due to the presence of activated ras, mutant BRAF, or elevation of growth factor receptors.
  • a semi-quantitative immunohistochemical method was developed to detect phosphorylated ERK (pERK) in human tumor biopsies as a patient selection biomarker for determining whether a patient will be responsive to a compound of the present invention, and also to assess its efficacy.
  • the antibody used was a polyclonal antibody (rabbit) that detects the phosphorylation status of p44 and p42 (phospho-p44/42 MAPK (Thr202/Tyr204)) MAP kinases (ERK1 and ERK2). The procedure was accomplished as follows:
  • Tumor samples were obtained from patients. Samples were embedded in paraffin and sectioned.
  • Tumor section slides were deparaffinized, and slides were incubated in 950 C citrate buffer for 35 min.
  • FIG. 1 shows the semi-quantitative procedure utilized to analyze the tissue sections.
  • FIG. 2 shows data establishing that, prior to the administration of the compound, a subject having melanoma had high levels of phospho-ERK in tumor tissue as compared to stromal cells, indicating that the subject was a candidate for treatment with a compound of the present invention. The subject was then administered a 600 mg BID dose of the compound.
  • This assay was performed in 96-well opaque plates (Costar 3915) in the TR-FRET format. Reaction conditions are as follows: 10 ⁇ M ATP, 25 nM poly GT-biotin, 2 nM Eu-labelled phospho-Tyr Ab, 10 nM APC, 7 nM Flk-1 (kinase domain), 1% DMSO, 50 mM HEPES pH 7.5, 10 mM MgCl 2 , 0.1 mM EDTA, 0.015% BRIJ, 0.1 mg/mL BSA, 0.1% mercapto-ethanol). Reaction is initiated upon addition of enzyme. Final reaction volume in each well is 100 ⁇ L.
  • Plates are read at both 615 and 665 nM on a Perkin Elmer Victor V Multilabel counter at about 1.5-2.0 hours after reaction initiation. Signal is calculated as a ratio: (665 nm/615 nm)*10000 for each well.
  • This assay was formatted in a 96-well black plate (Costar 3915). The following reagents (and their sources) are used: Europium-labeled anti-phosphotyrosine antibody pY20 and streptavidin-APC; poly GT-biotin, and mouse PDGFR within DRT.
  • reaction conditions are as follows: 1 nM mouse PDGFR is combined with 20 ⁇ M ATP, 7 nM poly GT-biotin, 1 nM pY20 antibody, 5 nM streptavidin-APC, and 1% DMSO in assay buffer (50 mM HEPES pH 7.5, 10 mM MgCl 2 , 0.1 mM EDTA, 0.015% BRIJ 35, 0.1 mg/mL BSA, 0.1% mercaptoethanol). Reaction is initiated upon addition of enzyme. Final reaction volume in each well is 100 ⁇ L. After 90 minutes, the reaction is stopped by addition of 10 ⁇ L/well of 5 ⁇ M staurosporine. Plates are read at both 615 and 665 nm on a Perkin Elmer VictorV Multilabel counter at about 1 hour after the reaction is stopped. Signal is calculated as a ratio: (665 nm/615 nm)*10000 for each well.
  • 100K P3-P6 Aortic SMC were plated in each well of 12-well cluster in 1000 uL volume/well of SGM-2 using standard cell culture techniques. Next day, cells were rinsed with 1000 uL D-PBS (Gibco) once, then serum starved in 500 uL SBM (smooth muscle cell basal media) with 0.1% BSA (Sigma, Cat A9576) overnight. Compounds were diluted at a dose range from (10 uM to 1-nM in 10-fold dilution steps in DMSO. Final DMSO concentration 0.1%). Remove old media by inversion into the sink quickly then add 100 ul of each dilution to corresponding well of cells for 1 hr at 37° C.
  • Cells were then stimulated with 10 ng/mL PDGF BB ligand for 7 minutes at 37° C.
  • the media is decanted and 150 uL of isotonic lysis buffer with protease inhibitor tablet (Complete; EDTA-free) and 0.2 mM Na vanadate is added.
  • Cells are lysed for 15 min at 4° C. on shaker in cold room. Lysates are put in eppendorf tubes to which 15 uL of agarose-conjugated anti-PDGFR-b antibody is added (Santa Cruz, sc-339) and incubated at 4° C. overnight.
  • Human breast carcinoma cells (MDA MB-231, NCI) were cultured in standard growth medium (DMEM) supplemented with 10% heat-inactivated FBS at 37° C. in 5%. CO 2 (vol/vol) in a humidified incubator. Cells were plated at a density of 3000 cells per well in 90 ⁇ L growth medium in a 96 well culture dish. In order to determine T 0h CTG values, 24 hours after plating, 100 ⁇ L of CellTiter-Glo Luminescent Reagent (Promega) was added to each well and incubated at room temperature for 30 minutes. Luminescence was recorded on a Wallac Victor II instrument. The CellTiter-Glo reagent results in cell lysis and generation of a luminescent signal proportional to the amount of ATP present, which, in turn is directly proportional to the number of cells present.
  • DMEM standard growth medium
  • FBS heat-inactivated FBS
  • CO 2 vol/vol
  • T 0h CTG values 24 hours after
  • Test compounds are dissolved in 100% DMSO to prepare 10 mM stocks. Stocks were further diluted 1:400 in growth medium to yield working stocks of 25 ⁇ M test compound in 0.25% DMSO. Test compounds were serially diluted in growth medium containing 0.25% DMSO to maintain constant DMSO concentrations for all wells. 60 ⁇ L of diluted test compound were added to each culture well to give a final volume of 180 ⁇ L. The cells with and without individual test compounds were incubated for 72 hours at which time ATP dependent luminescence was measured, as described previously, to yield T 72h values. Optionally, the IC 50 values can be determined with a least squares analysis program using compound concentration versus percent inhibition.
  • an N-[4-chloro-3-(trifluoromethyl)phenyl]-N′- ⁇ 4-[2-N-methylcarbamoyl-4-pyridyloxy]phenyl ⁇ urea salt was administered to staged HCT 116 colon (mutant k-Ras), MIA PaCa-2 pancreatic (mutant k-Ras), NCI-H460 lung (mutant k-Ras) and SK-OV-3 ovarian (wt k-Ras) xenograft models. All treatments were administered p.o. on a qd ⁇ 14 schedule.
  • Dosages of 10, 30 and 100 mg/kg/dose produced dose-dependent inhibition of HCT-116 tumor growth that ranged between 45% and 68%.
  • the growth of MIA PaCa-2 tumors was inhibited 44%, 66%, and 73% at dosages of 10, 30, and 100 mg/kg/dose, respectively.
  • the growth of NCI-H460 tumors was inhibited 27% and 56% at dosages of 10 and 30 mg/kg/dose of this Raf kinase inhibitor.
  • the SK-OV-3 model was slightly more sensitive, generating 45%-81% tumor growth inhibitions at dosages of 3 to 100 mg/kg/dose.
  • the anti-tumor efficacy of longer duration was also evaluated in the HCT 116 model.
  • the compound produced net tumor stasis at dosages of 30 and 100 mg/kg/dose when treatment was extended to 30 days duration.
  • HCT 116, SK-OV-3, and MIA PaCa-2 stock tumors were maintained as serial s.c. passages of fragments in CD-1 Nu/Nu female mice (HCT 116 and SK-OV-3) or CB17 SCID female mice (MIA-PaCa-2). Treatments were administered orally and were initiated against established tumors. Tumor dimensions were measured via calipers 2-3 times per week and were converted into tumor mass by the formula [L ⁇ (W2)]2, where L and W refer to the largest and smallest dimensions, respectively.
  • the in vitro inhibitory properties of compounds were determined using a p38 kinase inhibition assay.
  • P38 activity was detected using an in vitro kinase assay run in 96-well microtiter plates.
  • Recombinant human p38 0.5 ⁇ g/mL was mixed with substrate (myelin basic protein, 5 ⁇ g/mL) in kinase buffer (25 mM Hepes, 20 mM MgCl 2 and 150 mM NaCl) and compound.
  • substrate myelin basic protein, 5 ⁇ g/mL
  • kinase buffer 25 mM Hepes, 20 mM MgCl 2 and 150 mM NaCl
  • One ⁇ Ci/well of 33 P-labeled ATP (10 ⁇ M) was added to a final volume of 100 ⁇ L.
  • the reaction was run at 32° C. for 30 min. and stopped with a 1M HCl solution.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Engineering & Computer Science (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Immunology (AREA)
  • Biomedical Technology (AREA)
  • Epidemiology (AREA)
  • Urology & Nephrology (AREA)
  • Molecular Biology (AREA)
  • Hematology (AREA)
  • Oncology (AREA)
  • Virology (AREA)
  • Biotechnology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Diabetes (AREA)
  • Food Science & Technology (AREA)
  • Analytical Chemistry (AREA)
  • Physics & Mathematics (AREA)
  • Pathology (AREA)
  • Biochemistry (AREA)
  • General Physics & Mathematics (AREA)
  • Microbiology (AREA)
  • Cell Biology (AREA)
  • Hospice & Palliative Care (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Pulmonology (AREA)
  • Cardiology (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Communicable Diseases (AREA)
US10/571,100 2003-05-20 2004-05-19 Diaryl ureas with kinase inhibiting activity Abandoned US20070020704A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US10/571,100 US20070020704A1 (en) 2003-05-20 2004-05-19 Diaryl ureas with kinase inhibiting activity

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US47173503P 2003-05-20 2003-05-20
US52039903P 2003-11-17 2003-11-17
US55606204P 2004-03-25 2004-03-25
US10/571,100 US20070020704A1 (en) 2003-05-20 2004-05-19 Diaryl ureas with kinase inhibiting activity
PCT/US2004/015655 WO2004113274A2 (fr) 2003-05-20 2004-05-19 Diaryl-urees presentant une activite d'inhibition des kinases

Publications (1)

Publication Number Publication Date
US20070020704A1 true US20070020704A1 (en) 2007-01-25

Family

ID=33545309

Family Applications (2)

Application Number Title Priority Date Filing Date
US10/848,567 Active 2030-03-17 US8796250B2 (en) 2003-05-20 2004-05-19 Diaryl ureas for diseases mediated by PDGFR
US10/571,100 Abandoned US20070020704A1 (en) 2003-05-20 2004-05-19 Diaryl ureas with kinase inhibiting activity

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US10/848,567 Active 2030-03-17 US8796250B2 (en) 2003-05-20 2004-05-19 Diaryl ureas for diseases mediated by PDGFR

Country Status (12)

Country Link
US (2) US8796250B2 (fr)
EP (2) EP1626714B1 (fr)
JP (2) JP4860474B2 (fr)
AT (2) ATE366108T1 (fr)
CA (2) CA2526636C (fr)
DE (2) DE602004011340T2 (fr)
DK (2) DK1636585T3 (fr)
ES (2) ES2305808T3 (fr)
MX (1) MXPA05012486A (fr)
PL (2) PL1636585T3 (fr)
PT (2) PT1636585E (fr)
WO (2) WO2004113274A2 (fr)

Cited By (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030181442A1 (en) * 1999-01-13 2003-09-25 Bayer Corporation omega-carboxyaryl substituted diphenyl ureas as raf kinase inhibitors
US20030207870A1 (en) * 2002-02-11 2003-11-06 Bayer Corporation Aryl ureas with angiogenisis inhibiting activity
US20050038080A1 (en) * 2003-07-23 2005-02-17 Stephen Boyer Fluoro substituted omega-carboxyaryl diphenyl urea for the treatment and prevention of diseases and conditions
US20050059703A1 (en) * 2003-05-20 2005-03-17 Scott Wilhelm Diaryl ureas for diseases mediated by PDGFR
US20070105142A1 (en) * 2005-10-31 2007-05-10 Scott Wilhelm Methods for prognosis and monitoring cancer therapy
US20080027061A1 (en) * 1999-01-13 2008-01-31 Bernd Riedl omega-Carboxy Aryl Substituted Diphenyl Ureas As p38 Kinase Inhibitors
US20080108672A1 (en) * 2002-01-11 2008-05-08 Bernd Riedl Omega-Carboxyaryl Substituted Diphenyl Ureas As Raf Kinase Inhibitors
US20080105264A1 (en) * 2003-02-21 2008-05-08 Resmed Limited Nasal assembly
US20080194580A1 (en) * 2001-04-20 2008-08-14 Jacques Dumas Inhibition Of Raf Kinase Using Quinolyl, Isoquinolyl Or Pyridyl Ureas
US20080242707A1 (en) * 2005-03-07 2008-10-02 Bayer Healthcare Ag Pharmaceutical Composition for the Treatment of Cancer
US20080308776A1 (en) * 2007-06-14 2008-12-18 Bindra Perminder S Chemiluminescent compositions and methods of making and using thereof
US20090215833A1 (en) * 2004-09-29 2009-08-27 Bayer Healthcare Ag Thermodynamically stable form of a tosylate salt
US20100075971A1 (en) * 2003-02-28 2010-03-25 Jacques Dumas Substituted pyridine derivatives useful in the treatment of cancer and other disorders
US20100173953A1 (en) * 2006-10-11 2010-07-08 Alfons Grunenberg 4-[4-(amino)-3-fluorophenoxy]-N-methylpyridine-2-carboxamide monohydrate
US20100185419A1 (en) * 2008-09-05 2010-07-22 Avila Therapeutics, Inc. Algorithm for designing irreversible inhibitors
US20100288984A1 (en) * 2009-05-12 2010-11-18 Bindra Perminder S Chemiluminescent powders and methods of making and using thereof
US20110117073A1 (en) * 2009-09-16 2011-05-19 Avila Therapeutics, Inc. Protein Kinase Conjugates and Inhibitors
US20130225641A1 (en) * 2010-05-18 2013-08-29 Banghe Pharmaceutical Co., Ltd. Anticancer compounds and preparation methods thereof
US8658066B2 (en) 2009-05-12 2014-02-25 Mega Glow, Llc Chemiluminescent powders and methods of making and using thereof
US9458107B2 (en) 2010-04-15 2016-10-04 Bayer Intellectual Property Gmbh Process for the preparation of 4-{4-[({[4 chloro-3-(trifluoromethyl)-phenyl]amino}carbonyl)amino]-3-fluorphenoxy-N-ethylpyridie-carboxamide, its salts and monohydrate
WO2018035346A1 (fr) * 2016-08-17 2018-02-22 Ichan School Of Medicine At Mount Sinal Composés inhibiteurs de kinase, compositions et méthodes de traitement du cancer
US11542492B2 (en) 2009-12-30 2023-01-03 Celgene Car Llc Ligand-directed covalent modification of protein

Families Citing this family (52)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DK2305255T3 (da) * 2001-12-03 2012-12-10 Bayer Healthcare Llc Arylurinstofforbindelser i kombination med andre cytostatiske eller cytotoksiske midler til behandling af humane cancere
ATE529406T1 (de) 2002-02-11 2011-11-15 Bayer Healthcare Llc Aryl-harnstoffe als kinase inhibitoren
MXPA05007513A (es) * 2003-01-14 2005-09-21 Cytokinetics Inc Compuestos, composiciones y metodos.
RU2006122853A (ru) * 2003-11-28 2008-01-10 Новартис АГ (CH) Производные диарилмочевины для лечения заболеваний, зависимых от протеинкиназы
US7803178B2 (en) * 2004-01-30 2010-09-28 Trivascular, Inc. Inflatable porous implants and methods for drug delivery
BRPI0512193B8 (pt) * 2004-06-17 2021-05-25 Cytokinetics Inc composto ou sal farmaceuticamente aceitável, composição farmacêutica, e, uso do composto ou sal
GB0512324D0 (en) 2005-06-16 2005-07-27 Novartis Ag Organic compounds
UA90691C2 (en) 2004-09-29 2010-05-25 Баер Шеринг Фарма Акциенгезельшафт Process for the preparation of 4-{ 4-[({ [4-chloro-3-(trifluoromethyl)phenyl]amino}carbonyl)amino]phenoxy}-n-methylpyridine-2-carboxamide
US7538223B2 (en) * 2005-08-04 2009-05-26 Cytokinetics, Inc. Compounds, compositions and methods
DE602006019262D1 (de) * 2005-08-11 2011-02-10 Siemens Healthcare Diagnostics QUANTITATIVE ASSAYS FÜR PDGFR-beta- IN KÖRPERFLÜSSIGKEITEN
US20090068146A1 (en) * 2005-10-31 2009-03-12 Scott Wilhelm Diaryl ureas and combinations
WO2007053573A2 (fr) * 2005-10-31 2007-05-10 Bayer Pharmaceuticals Corporation Traitement du cancer au moyen de sorafenib
JP5084736B2 (ja) * 2005-11-10 2012-11-28 バイエル・ファルマ・アクチェンゲゼルシャフト 肺高血圧を処置するためのジアリールウレア
KR20080066948A (ko) * 2005-11-10 2008-07-17 바이엘 헬스케어 아게 당뇨병성 신경병증을 치료하기 위한 디아릴 우레아
EP1948176B1 (fr) * 2005-11-10 2011-01-05 Bayer Schering Pharma Aktiengesellschaft Diaryle-urees permettant de traiter l'hypertension pulmonaire
MX2008006979A (es) * 2005-12-01 2009-01-14 Bayer Healthcare Llc Compuestos de urea utiles en el tratamiento contra el cancer.
JP2009524585A (ja) * 2005-12-15 2009-07-02 バイエル・ヘルスケア・アクチェンゲゼルシャフト ウイルス感染を処置するためのジアリールウレア
TW200808321A (en) 2005-12-15 2008-02-16 Cytokinetics Inc Certain chemical entities, compositions and methods
US20070208000A1 (en) * 2005-12-15 2007-09-06 Morgan Bradley P Certain chemical entities, compositions and methods
JP2009519265A (ja) * 2005-12-15 2009-05-14 バイエル・ヘルスケア・アクチェンゲゼルシャフト ウイルス感染を処置するためのジアリールウレア
US7825120B2 (en) * 2005-12-15 2010-11-02 Cytokinetics, Inc. Certain substituted ((piperazin-1-ylmethyl)benzyl)ureas
EP1959947A2 (fr) * 2005-12-15 2008-08-27 Cytokinetics, Inc. Entites chimiques, compositions et procedes
WO2007078815A2 (fr) * 2005-12-16 2007-07-12 Cytokinetics, Inc. Entites chimiques, compositions et methodes
EP1962852B1 (fr) * 2005-12-19 2017-01-25 Cytokinetics, Inc. Composes, compositions et methodes
JP5331485B2 (ja) * 2005-12-21 2013-10-30 バイエル・インテレクチュアル・プロパティ・ゲゼルシャフト・ミット・ベシュレンクテル・ハフツング 癌および他の障害の処置に有用な置換ピリミジン誘導体
WO2007139930A2 (fr) * 2006-05-26 2007-12-06 Bayer Healthcare Llc Associations de médicaments comportant des urées de diaryle substituées pour le traitement du cancer
WO2008044688A1 (fr) * 2006-10-11 2008-04-17 Daiichi Sankyo Company, Limited Dérivé de l'urée
US8680124B2 (en) 2007-01-19 2014-03-25 Bayer Healthcare Llc Treatment of cancers with acquired resistance to kit inhibitors
WO2008115263A2 (fr) * 2007-03-20 2008-09-25 Curis, Inc. Inhibiteurs de la raf kinase contenant un fragment de liaison au zinc
EP1992344A1 (fr) * 2007-05-18 2008-11-19 Institut Curie P38 alpha comme cible therapeutique pour les maladies associées á une mutation de FGFR3
CN101220024A (zh) * 2007-12-11 2008-07-16 杜晓敏 一组抑制激酶的抗癌化合物
US20110263657A1 (en) * 2008-06-25 2011-10-27 Bayer Schering Pharma Aktiengesellschaft Diaryl ureas for treating heart failure
WO2009156070A1 (fr) * 2008-06-25 2009-12-30 Bayer Schering Pharma Aktiengesellschaft Diaryle-urée pour le traitement de l'insuffisance cardiaque
CN103254126A (zh) * 2008-09-19 2013-08-21 苏州泽璟生物制药有限公司 氘代的ω-二苯基脲及衍生物以及包含该化合物的药物组合物
CN101362717B (zh) * 2008-09-28 2013-02-06 四川大学 4-(4-氨基苯胺基)-2-(甲基氨甲酰基)吡啶及其衍生物和它们的制备方法、用途
FR2943670B1 (fr) * 2009-03-24 2011-05-06 Sanofi Aventis Derives anticancereux,leur preparation et leur application en therapeutique
CN102459180B (zh) * 2009-06-09 2014-05-14 江苏迈度药物研发有限公司 作为激酶抑制剂的脲衍生物
RU2012109233A (ru) * 2009-09-03 2013-10-10 Аллерган, Инк. Соединения как модуляторы тирозинкиназы
ES2634470T3 (es) 2009-09-30 2017-09-27 Toray Industries, Inc. Derivado de 2,3-dihidro-1H-inden-2-ilurea y aplicación farmacéutica del mismo
AU2011210326B2 (en) 2010-01-29 2014-10-09 Sun Pharmaceutical Industries Limited Sorafenib dimethyl sulphoxide solvate
WO2012012404A1 (fr) 2010-07-19 2012-01-26 Bayer Healthcare Llc Associations médicamenteuses contenant une oméga-carboxyaryl diphénylurée fluorosubstituée utilisées pour le traitement et la prévention de maladies et d'affections
AP2013006834A0 (en) * 2010-10-01 2013-04-30 Bayer Ip Gmbh Substituted N-(2-arylamino)aryl sulfonamide-containing combinations
US20140235678A1 (en) 2011-06-28 2014-08-21 Bayer Healthcare Llc Topical Ophthalmological Pharmaceutical Composition containing Sorafenib
AU2013261719B2 (en) * 2012-05-18 2017-07-27 Sanofi Benzo[1,3]dioxine derivatives and their use as LPAR5 antagonists
WO2014040242A1 (fr) * 2012-09-12 2014-03-20 湖南有色凯铂生物药业有限公司 Composé 3-chloro et 3-méthoxy-n-méthyl -2-pyridine-carboxamide et son application en tant que médicament anticancéreux
EP2976081B1 (fr) * 2013-03-15 2020-06-17 Alumend, LLC Méthodes de restauration aiguë d'une compliance vasculaire
CN109069509B (zh) 2016-03-02 2020-10-16 麦迪维尔股份公司 索拉非尼或瑞戈非尼与曲沙他滨的氨基磷酸酯前药的组合治疗
CN108314703B (zh) * 2017-01-17 2022-02-01 亚飞(上海)生物医药科技有限公司 分子定点靶向和激活的激酶抑制剂的制备和用途
JP2020517600A (ja) * 2017-04-20 2020-06-18 ザ・リージエンツ・オブ・ザ・ユニバーシテイー・オブ・カリフオルニア K−Rasモジュレーター
CN109771407B (zh) * 2019-03-29 2021-11-26 成都医学院 迷迭香酸在制备抑制水痘-带状疱疹病毒的药物中的用途
CN110054584B (zh) * 2019-05-28 2022-03-11 沈阳药科大学 1-芳基-3-{4-[(吡啶-2-基甲基)硫基]苯基}脲类化合物及应用
IT202000022636A1 (it) 2020-09-25 2022-03-25 Ente Ospedaliero Specializzato In Gastroenterologia Istituto Nazionale Di Ricovero E Cura A Caratter “terapia di combinazione di sorafenib e/o regorafenib con la proteina ricombinante umana proteoglicano-4 per il trattamento dell'epatocarcinoma”

Citations (97)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US502504A (en) * 1893-08-01 Hermann thoms
US1792156A (en) * 1928-01-17 1931-02-10 Gen Aniline Works Inc 5-halogen-2-amino-1-alkyloxy and 1-aralkyloxy-benzenes and intermediate products thereof and process of preparing them
US2046375A (en) * 1931-06-04 1936-07-07 Ici Ltd p-halogen-omicron-alkoxy-aniline derivatives and process of preparing the same
US2093265A (en) * 1931-03-31 1937-09-14 Ici Ltd Process for the manufacture of diaryl ureas
US2288422A (en) * 1938-11-11 1942-06-30 Gen Aniline & Film Corp Mixed ureas
US2649476A (en) * 1950-04-29 1953-08-18 Variapat Ag Trifluoromethylated diphenyl ether sulfonic acids
US2683082A (en) * 1950-12-09 1954-07-06 Ethyl Corp Nu-aryl-nu'-(p-hydroxyphenyl) ureas as antioxidants for petroleum hydrocarbon fuels
US2722544A (en) * 1950-12-26 1955-11-01 Variapat Ag Trifluoromethyl halogenated diphenylcarbamide sulfonic acids and their preparation
US2745874A (en) * 1953-06-18 1956-05-15 Geigy Ag J R Insecticidal derivatives of diphenyl urea
US2781330A (en) * 1953-02-09 1957-02-12 Monsanto Chemicals Rubber containing urea compound as an anti-exposure cracking agent
US2797214A (en) * 1953-03-06 1957-06-25 Geigy Ag J R Tetrakisazo dyestuffs
US2867659A (en) * 1953-12-22 1959-01-06 Geigy Ag J R Polyhalogen substituted monohydroxydiphenyl urea and thiourea compounds
US2877268A (en) * 1956-12-24 1959-03-10 Monsanto Chemicals Substituted ureas
US2960488A (en) * 1958-04-25 1960-11-15 Eastman Kodak Co Poly-alpha-olefins containing substituted ureas
US2973386A (en) * 1943-01-05 1961-02-28 Harry A Weldon Purification of sym dichloro-bis (2, 4, 6-trichlorophenyl)urea
US3151023A (en) * 1961-04-21 1964-09-29 Ciba Ltd Preparations for combating phytopathogenic microorganisms
US3200035A (en) * 1962-06-01 1965-08-10 Ciba Ltd Treatment of synthetic products, especially synthetic fibers
US3230141A (en) * 1959-08-14 1966-01-18 Geigy Ag J R Method for protecting fibers against attack by insects and bacteria with diphenyl urea compositions
US3284433A (en) * 1963-07-17 1966-11-08 Merck & Co Inc 4-phenoxy-carbanilides
US3424762A (en) * 1966-03-07 1969-01-28 Robins Co Inc A H Certain 3-ureidopyrrolidines
US3424760A (en) * 1966-03-07 1969-01-28 Robins Co Inc A H 3-ureidopyrrolidines
US3424761A (en) * 1966-03-07 1969-01-28 Robins Co Inc A H 3-ureidopyrrolidines
US3639668A (en) * 1968-04-19 1972-02-01 Bayer Ag N-(2 2 4 4-tetrafluoro-1 3-benzodioxanyl)-ureas
US3646059A (en) * 1969-05-05 1972-02-29 Du Pont Plant growth regulatory ureidopyrazoles
US3668222A (en) * 1969-05-14 1972-06-06 Sandoz Ltd 11-desacetoxy-wortmannin
US3689550A (en) * 1968-03-21 1972-09-05 Ciba Geigy Ag N-hydroxyphenyl-n{40 -phenylureas
US3743498A (en) * 1967-10-31 1973-07-03 Du Pont Method of selectively controlling undesirable vegetation
US3754887A (en) * 1969-05-05 1973-08-28 Du Pont Ureidopyrazoles defoliants
US3823161A (en) * 1970-05-07 1974-07-09 Exxon Research Engineering Co Aminothiophene derivatives
US3828001A (en) * 1969-08-14 1974-08-06 May & Baker Ltd Thiophene derivatives
US3860645A (en) * 1973-05-23 1975-01-14 Givaudan Corp Bacteriostatic substituted carbanilides
US3990879A (en) * 1974-12-26 1976-11-09 Eli Lilly And Company Method of controlling aquatic weeds
US4001256A (en) * 1973-12-26 1977-01-04 The Upjohn Company Pyridylalkyl phenyl ureas and their n-oxides
US4009847A (en) * 1974-04-17 1977-03-01 E. I. Du Pont De Nemours And Company 1-Tertiary-alkyl-3-(substituted thienyl)ureas and 1-tertiary-alkyl-3-(substituted thietyl)ureas as antihypertensive agents
US4042372A (en) * 1976-11-19 1977-08-16 Eli Lilly And Company Substituted thiadiazolotriazinediones and method of preparation
US4071524A (en) * 1976-11-08 1978-01-31 Riker Laboratories, Inc. Derivatives of urea
US4103022A (en) * 1976-08-24 1978-07-25 Bayer Aktiengesellschaft Combating arthropods with 3-(2,2,4,4-tetrafluoro-benz-1,3-dioxin-6-yl)-1-(substituted benzoyl)-ureas
US4111680A (en) * 1973-07-27 1978-09-05 Shionogi & Co., Ltd. Herbicidal compositions containing 3-isoxazolylurea derivatives
US4111683A (en) * 1975-06-27 1978-09-05 Chevron Research Company N-alkyl or alkoxy-N'-substituted hydrocarbyl urea
US4173638A (en) * 1976-10-29 1979-11-06 Ishihara Sangyo Kaisha Ltd. N-Benzoyl-N'-pyridyloxy phenyl urea and insecticidal compositions thereof
US4173637A (en) * 1976-10-29 1979-11-06 Ishihara Sangyo Kaisha Ltd. N-Benzoyl-N'-pyridyloxy phenyl urea and insecticidal compositions thereof
US4183854A (en) * 1976-11-10 1980-01-15 John Wyeth & Brother Limited Thiazole compound
US4212981A (en) * 1973-07-27 1980-07-15 Shionogi & Co., Ltd. Process for preparing 3-isoxazolylurea derivatives
US4279639A (en) * 1978-11-02 1981-07-21 Toshihiko Okamoto N-(2-Substituted-4-pyridyl)ureas and thioureas as well as plant growth regulators containing same, and method for using compounds as plant growth regulators
US4358596A (en) * 1978-11-03 1982-11-09 Schering Ag Process for the preparation of 1,2,3-thiadiazol-5-yl ureas
US4405644A (en) * 1979-07-14 1983-09-20 Bayer Aktiengesellschaft Medicaments for the treatment of disorders of lipometabolism and their use
US4410697A (en) * 1980-01-25 1983-10-18 Reanal Finomvegyszergyar Process for the preparation of N-aryl-N'-(mono- or di substituted)-urea derivatives
US4437878A (en) * 1982-03-31 1984-03-20 Basf Aktiengesellschaft Dihydrothiophenecarboxylates and their use for controlling undersirable plant growth
US4468380A (en) * 1979-12-26 1984-08-28 Eli Lilly And Company Anticoccidial combinations comprising polyether antibiotics and carbanilides
US4473579A (en) * 1982-01-26 1984-09-25 American Cyanamid Company Antiatherosclerotic tetrasubstituted ureas and thioureas
US4499097A (en) * 1983-03-10 1985-02-12 American Cyanamid Company 2-(Pyridyl)imidazolyl ketones
US4511571A (en) * 1981-10-20 1985-04-16 Ciba Geigy Corporation N-(2-Pyridyloxyphenyl)-N'-benzoyl ureas, pesticidal compositions containing same and pesticidal methods of use
US4514571A (en) * 1982-05-25 1985-04-30 Ube Industries, Ltd. Process for the preparation of urea derivatives
US4526997A (en) * 1981-05-06 1985-07-02 Doherty George O P O Anticoccidial combinations comprising polyether antibiotics and carbanilides
US4540566A (en) * 1984-04-02 1985-09-10 Forest Laboratories, Inc. Prolonged release drug dosage forms based on modified low viscosity grade hydroxypropylmethylcellulose
US4546191A (en) * 1979-03-19 1985-10-08 Ishihara Sangyo Kaisha Ltd. Trifluoromethyl-2-pyridinone or pyridinthione compounds and process for the preparation of the same
US4587240A (en) * 1983-09-23 1986-05-06 National Research Development Corp. Pharmaceutical compositions
US4643849A (en) * 1982-11-12 1987-02-17 Toyama Chemical Co., Ltd. Intermediates for urea and thiourea derivatives
US4677111A (en) * 1984-10-12 1987-06-30 Ishihara Sangyo Kaisha Ltd. N-benzoyl-N'-(3-nitrophenyl) urea compounds, and antitumorous compositions containing them
US5773459A (en) * 1995-06-07 1998-06-30 Sugen, Inc. Urea- and thiourea-type compounds
US20030211466A1 (en) * 1999-12-28 2003-11-13 Ribonomics, Inc. Methods for identifying functionally related genes and drug targets
US20030216396A1 (en) * 2002-02-11 2003-11-20 Bayer Corporation Pyridine, quinoline, and isoquinoline N-oxides as kinase inhibitors
US7411055B2 (en) * 2001-05-25 2008-08-12 Immunex Corporation Attractin/mahogany-like polypeptides, polynucleotides, antibodies and methods of use thereof
US20090068146A1 (en) * 2005-10-31 2009-03-12 Scott Wilhelm Diaryl ureas and combinations
US20090093526A1 (en) * 1997-12-22 2009-04-09 Scott Miller Inhibition of p38 kinase using symmetrical and unsymmetrical diphenyl ureas
US20090118268A1 (en) * 1999-01-13 2009-05-07 Bernd Riedl OMEGA-CARBOXY ARYL SUBSTITUTED DIPHENYL UREAS AS p38 KINASE INHIBITORS
US20090176791A1 (en) * 2005-11-10 2009-07-09 Bayer Healthcare Ag Diaryl Ureas for Treating Pulmonary Hypertension
US20090192127A1 (en) * 2003-07-23 2009-07-30 Bayer Healthcare Ag Combination therapy comprising a diaryl urea compound and a p13, akt kinase or mtor inhibitors (rapamycins) for cancer treatment
US20090215833A1 (en) * 2004-09-29 2009-08-27 Bayer Healthcare Ag Thermodynamically stable form of a tosylate salt
US20090215835A1 (en) * 2005-10-31 2009-08-27 Scott Wilhelm Treatment of cancer with sorafenib
US20100035888A1 (en) * 2005-11-10 2010-02-11 Bater Healthcare AG Diaryl Urea for Treating Pulmonary Hypertension
US20100063112A1 (en) * 2006-11-09 2010-03-11 Bayer Schering Pharma Aktiengesellschaft Polymorph iii of 4-[4-(amino)-3-fluorophenoxy]-n-methylpyridine-2-carboxamide
US20100063088A1 (en) * 1997-05-23 2010-03-11 Wood Jill E Raf Kinase Inhibitors
US20100075971A1 (en) * 2003-02-28 2010-03-25 Jacques Dumas Substituted pyridine derivatives useful in the treatment of cancer and other disorders
US20100081812A1 (en) * 2005-12-21 2010-04-01 Roger Smith Substituted pyrimidine derivatives useful in the treatment of cancer and other disorders
US20100113533A1 (en) * 2006-11-14 2010-05-06 Bayer Schering Pharma Aktiengesellschaft Polymorph II of 4-[4-(Amino)-3- Fluorophenoxy]-N-Methylpyridine-2-Carboxamide
US20100125139A1 (en) * 2002-03-22 2010-05-20 Donald Bankston Process for preparing quinazoline rho-kinase inhibitors and intermediates thereof
US20100129321A1 (en) * 2005-12-15 2010-05-27 Bayer Healthcare Llc Diaryl urea for treating virus infections
US20100144749A1 (en) * 2005-11-14 2010-06-10 Scott Wilhelm Treatment of cancers with acquired resistance to kit inhibitors
US20100150863A1 (en) * 2006-12-20 2010-06-17 Bayer Pharmaceuticals Corporation Hydroxy methyl phenyl pyrazolyl urea compound useful in treatment of cancer
US20100160371A1 (en) * 1997-05-23 2010-06-24 Gerald Ranges Inhibition of p38 kinase activity by aryl ureas
US20100173954A1 (en) * 2007-01-19 2010-07-08 Bayer Healthcare Llc Treatment of cancers having resistance to chemotherapeutic agents
US20100173953A1 (en) * 2006-10-11 2010-07-08 Alfons Grunenberg 4-[4-(amino)-3-fluorophenoxy]-N-methylpyridine-2-carboxamide monohydrate
US20100267777A1 (en) * 2007-01-19 2010-10-21 Bayer Pharmaceuticals Corporation Treatment of cancers with acquired resistance to kit inhibitors
US7833698B2 (en) * 2000-01-12 2010-11-16 Ventana Medical Systems, Inc. Method for determining the response to cancer therapy
US20110015195A1 (en) * 2002-02-11 2011-01-20 Jacques Dumas Aryl ureas with angiogenisis inhibiting activity
US20110136809A1 (en) * 2004-04-30 2011-06-09 Bayer Pharmaceuticals Corporation Substituted Pyrazolyl Urea Derivatives Useful In The Treatment Of Cancer
US20110158942A1 (en) * 2005-12-15 2011-06-30 Olaf Weber Diaryl ureas for treating virus infections
US20110178137A1 (en) * 2008-06-25 2011-07-21 Bayer Schering Pharma Aktiengesellschaft Diaryl urea for treating heart failure
US20110195110A1 (en) * 2005-12-01 2011-08-11 Roger Smith Urea compounds useful in the treatment of cancer
US20110257035A1 (en) * 2008-10-21 2011-10-20 Bayer Healthcare Llc Identification of signature genes associated with hepatocellular carcinoma
US20120040925A1 (en) * 2001-12-03 2012-02-16 Carter Christopher A Aryl urea compounds in combination with other cytostatic or cytotoxic agents for treating human cancers
US20120040986A1 (en) * 2002-01-11 2012-02-16 Bernd Riedl Omega carboxyaryl substituted diphenyl ureas as raf kinase inhibitors
US20120046290A1 (en) * 1997-12-22 2012-02-23 Jacques Dumas Inhibition of p38 kinase activity using substituted heterocyclic ureas
US20120129893A1 (en) * 1997-12-22 2012-05-24 Jacques Dumas Inhibition Of Raf Kinase Using Substituted Heterocyclic Ureas
US20120142741A1 (en) * 2004-08-27 2012-06-07 Fritz Schueckler Pharmaceutical compositions for the treatment of cancer
US20130142742A1 (en) * 2008-04-02 2013-06-06 Nicolas Thiebault Methods of Inhibiting Microbial Growth Using Deoxyaldohexoses

Family Cites Families (418)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE487014C (de) 1927-10-23 1929-12-09 I G Farbenindustrie Akt Ges Verfahren zur Darstellung von Harnstoffderivaten
DE511468C (de) 1928-01-18 1930-10-30 I G Farbenindustrie Akt Ges Verfahren zur Darstellung von 5-Chlor-2-amino-1-methoxybenzol
DE523437C (de) 1928-12-25 1931-05-05 I G Farbenindustrie Akt Ges Verfahren zur Darstellung von 5-Chlor- und 5-Brom-2-amino-1-alkyloxy- und 1-aralkyloxybenzolen
GB771333A (en) 1953-12-22 1957-03-27 Geigy Ag J R Improvements relating to halogen substituted diphenyl urea and thiourea compounds and their use
GB828231A (en) 1955-10-20 1960-02-17 Geigy Ag J R Improvements relating to insecticidal compounds and their use
CH428747A (de) 1961-09-11 1967-01-31 Wander Ag Dr A Verfahren zur Herstellung neuer Harnstoff- und Thioharnstoffderivate
FR1457172A (fr) 1964-12-12 1966-10-28 Ferrania Spa Procédé pour la production d'images photographiques en couleurs et matériel photographique correspondant
FR90420E (fr) 1965-06-09 1968-02-21
US3547940A (en) 1967-10-31 1970-12-15 Du Pont Substituted ureido isoxazoles
JPS5031039A (fr) 1973-07-27 1975-03-27
US4062861A (en) 1973-07-27 1977-12-13 Shionogi & Co., Ltd. 3-Isoxazolylurea derivatives
US3931201A (en) 1974-01-22 1976-01-06 The Dow Chemical Company Substituted pyridinyloxy(thio)phenyl -acetamides, -ureas and urea derivatives
DE2817449A1 (de) 1978-04-21 1979-10-31 Bayer Ag Mittel zur regulierung des pflanzenwachstums
GB1590870A (en) 1978-05-31 1981-06-10 Shionogi & Co N-(5-t-butyl-3-isoxazolyl) alkanamide derivatives having herbicidal activity
FI800559A (fi) 1979-03-14 1980-09-15 Hoffmann La Roche Urinaemnederivat
EP0057797B1 (fr) 1981-02-03 1986-06-25 Imperial Chemical Industries Plc Procédé pour l'extraction de métaux et agents d'extraction pour métaux
US4623662A (en) 1985-05-23 1986-11-18 American Cyanamid Company Antiatherosclerotic ureas and thioureas
AU1862083A (en) 1982-09-02 1984-03-08 Duphar International Research B.V. Tumor growth inhibitors
DE3305866A1 (de) 1983-02-19 1984-08-23 Basf Ag, 6700 Ludwigshafen Thiophen-carbonester, verfahren zu ihrer herstellung und ihre verwendung zur bekaempfung unerwuenschten pflanzenwuchses
US4727077A (en) 1985-02-20 1988-02-23 Ishihara Sangyo Kaisha Ltd. Benzoyl urea compounds, process for their production, and antitumorous compositions containing them
DE3529247A1 (de) 1985-05-17 1986-11-20 Bayer Ag, 5090 Leverkusen Verwendung von thienylharnstoffen und -isoharnstoffen als leistungsfoerdernde mittel bei tieren, neue thienylharnstoffe und -isoharnstoffe und ihre herstellung
DE3540377A1 (de) 1985-11-14 1987-05-21 Bayer Ag Thienooxazinone, verfahren zu ihrer herstellung und ihre verwendung als leistungsfoerderer
DE3541631A1 (de) 1985-11-26 1987-05-27 Bayer Ag Selektiv-fungizide verwendung von thienylharnstoff-derivaten
AU594098B2 (en) 1985-12-11 1990-03-01 Ishihara Sangyo Kaisha Ltd. N-benzoyl urea compounds, antitumorous compositions containing them, and process for their preparation
EP0230400A3 (fr) 1986-01-21 1990-02-14 Ciba-Geigy Ag N-3-(5-Trifluorométhyl-pyridyl-2-oxy)phényl-N'-benzoylureds pour combattre des helminths chez le bétail
JPS62185013A (ja) 1986-02-08 1987-08-13 Green Cross Corp:The 易吸収性医薬組成物
DE3612830A1 (de) 1986-04-16 1987-10-22 Basf Ag Thiadiazolylharnstoff enthaltendes mittel zur entblaetterung von pflanzen
DE3785507T2 (de) 1986-07-31 1993-07-29 Beecham Group Plc Azabicyclische verbindungen, verfahren zu ihrer herstellung und ihre pharmazeutische verwendung.
NZ221964A (en) 1986-10-03 1990-03-27 Ishihara Sangyo Kaisha Benzoylurea compounds and insecticidal compositions
EP0264904A3 (fr) 1986-10-23 1988-08-17 Ishihara Sangyo Kaisha, Ltd. Compositions pharmaceutiques contenant des dérivés de benzoylurée
DE3636190A1 (de) 1986-10-24 1988-04-28 Bayer Ag Verfahren zur herstellung von n,n-diaryl-harnstoffen
JPH06100808B2 (ja) 1987-05-28 1994-12-12 富士写真フイルム株式会社 ハロゲン化銀カラ−写真感光材料の処理方法
DE3853662D1 (de) 1987-10-16 1995-06-01 Ciba Geigy Ag Schädlingsbekämpfungsmittel.
DE3810382A1 (de) 1988-03-26 1989-10-12 Bayer Ag 5-amino-1-phenylpyrazole, verfahren sowie 5-halogen-1-phenylpyrazole als zwischenprodukte zu deren herstellung und ihre verwendung als herbizide
AU636853B2 (en) 1988-08-23 1993-05-13 Nutrasweet Company, The Substituted aryl ureas as high potency sweeteners
JPH0278699A (ja) 1988-09-12 1990-03-19 Green Cross Corp:The ベンゾイルウレア系化合物・アルブミン複合体
FR2662692B1 (fr) 1990-05-30 1995-04-28 Novapharme Derives heterocycliques doues d'activite anticonvulsivante, procede de preparation et composition pharmaceutique.
FR2639636B1 (fr) 1988-11-30 1994-03-04 Novapharme Nouveaux composes heterocycliques a activite anticonvulsivante, procede de preparation et compositions therapeutiques les contenant
JPH02196719A (ja) 1989-01-24 1990-08-03 Green Cross Corp:The 粉末状医薬組成物
JPH02237922A (ja) 1989-01-24 1990-09-20 Green Cross Corp:The 抗ウィルス剤
EP0379915A1 (fr) 1989-01-26 1990-08-01 Bayer Ag Dérivés substitués du phénoxybenzonitrile, procédés pour leur préparation et leur utilisation comme herbicides et comme régulateurs de la croissance des plantes
JP3002204B2 (ja) 1989-03-13 2000-01-24 株式会社東芝 時系列信号認識装置
US4973675A (en) 1989-04-13 1990-11-27 University Of Tennessee Research Center Hybrid nitrosoureidoanthracyclines having antitumor activity
JPH0395153A (ja) 1989-06-15 1991-04-19 Mitsubishi Kasei Corp ジフェニル尿素誘導体
US5665543A (en) 1989-07-18 1997-09-09 Oncogene Science, Inc. Method of discovering chemicals capable of functioning as gene expression modulators
IL95860A0 (en) 1989-10-13 1991-07-18 Ciba Geigy Ag Thienylthioureas,-isothioureas and-carbodiimides
EP0425443A1 (fr) 1989-10-27 1991-05-02 Ciba-Geigy Ag Agent antiparasitaire injectable
US5177110A (en) 1989-10-27 1993-01-05 Ciba-Geigy Corporation Injectable parasiticidal composition
CA2051705A1 (fr) 1990-06-19 1991-12-20 Kiyoaki Katano Derives de la pyridine, antagonistes de l'angiotensine ii
US5210015A (en) 1990-08-06 1993-05-11 Hoffman-La Roche Inc. Homogeneous assay system using the nuclease activity of a nucleic acid polymerase
GB9017892D0 (en) 1990-08-15 1990-09-26 May & Baker Ltd New compositions of matter
PT98990A (pt) 1990-09-19 1992-08-31 American Home Prod Processo para a preparacao de esteres de acidos carboxilicos de rapamicina
KR0133550B1 (ko) 1991-01-21 1998-04-23 요시또시 가즈오 3-벤질리덴-1-카르바모일-2-피롤리돈 유사체
JP2671693B2 (ja) 1991-03-04 1997-10-29 松下電器産業株式会社 バイオセンサおよびその製造法
US5120842A (en) 1991-04-01 1992-06-09 American Home Products Corporation Silyl ethers of rapamycin
US5270458A (en) 1991-04-02 1993-12-14 The Trustees Of Princeton University Nucleic acids encoding fragments of hematopoietic stem cell receptor flk-2
US5185438A (en) 1991-04-02 1993-02-09 The Trustees Of Princeton University Nucleic acids encoding hencatoporetic stem cell receptor flk-2
US5100883A (en) 1991-04-08 1992-03-31 American Home Products Corporation Fluorinated esters of rapamycin
JPH05112573A (ja) 1991-04-17 1993-05-07 American Home Prod Corp ラパマイシンのカルバミン酸エステル類
US5118678A (en) 1991-04-17 1992-06-02 American Home Products Corporation Carbamates of rapamycin
US5721237A (en) 1991-05-10 1998-02-24 Rhone-Poulenc Rorer Pharmaceuticals Inc. Protein tyrosine kinase aryl and heteroaryl quinazoline compounds having selective inhibition of HER-2 autophosphorylation properties
US5118677A (en) 1991-05-20 1992-06-02 American Home Products Corporation Amide esters of rapamycin
US5185358A (en) 1991-06-24 1993-02-09 Warner-Lambert Co. 3-heteroatom containing urea and thiourea ACAT inhibitors
US5162360A (en) 1991-06-24 1992-11-10 Warner-Lambert Company 2-heteroatom containing urea and thiourea ACAT inhibitors
AU2552492A (en) 1991-08-23 1993-03-16 United States of America as represented by The Secretary Department of Health and Human Services, The Raf protein kinase therapeutics
US5151413A (en) 1991-11-06 1992-09-29 American Home Products Corporation Rapamycin acetals as immunosuppressant and antifungal agents
DE69231853T2 (de) 1991-11-07 2001-09-13 Nanotronics Inc Hybridisierung von mit chromophoren und fluorophoren konjugierten polynukleotiden zur erzeugung eines donor-zu-donor energietransfersystems
FR2683818B1 (fr) 1991-11-14 1993-12-31 Adir Cie Nouveaux derives de 3-sulfonylamino-2-(1h)-quinoleinone, leur procede de preparation et les compositions pharmaceutiques qui les contiennent.
US5348853A (en) 1991-12-16 1994-09-20 Biotronics Corporation Method for reducing non-specific priming in DNA amplification
JPH07504429A (ja) 1992-03-12 1995-05-18 スミスクライン・ビーチャム・パブリック・リミテッド・カンパニー 5HT↓1cアンタゴニストとしてのインドール誘導体
DE4212407A1 (de) 1992-04-13 1993-10-14 Knorr Bremse Gmbh Lüftspieldetektor für eine druckluftbetätigte Scheibenbremse
ZA935111B (en) 1992-07-17 1994-02-04 Smithkline Beecham Corp Rapamycin derivatives
US5312820A (en) 1992-07-17 1994-05-17 Merck & Co., Inc. Substituted carbamoyl and oxycarbonyl derivatives of biphenylmethylamines
ZA935112B (en) 1992-07-17 1994-02-08 Smithkline Beecham Corp Rapamycin derivatives
EP0655066A1 (fr) 1992-08-12 1995-05-31 PHARMACIA & UPJOHN COMPANY Inhibiteurs de proteine kinase et composes apparentes combines avec du taxol
US5256790A (en) 1992-08-13 1993-10-26 American Home Products Corporation 27-hydroxyrapamycin and derivatives thereof
JP2717481B2 (ja) 1992-08-25 1998-02-18 富士写真フイルム株式会社 ハロゲン化銀カラー写真感光材料
EP0672187A4 (fr) 1992-10-08 1999-11-17 Univ California Dosages pcr permettant de determiner la presence et la concentration d'une cible.
US5258389A (en) 1992-11-09 1993-11-02 Merck & Co., Inc. O-aryl, O-alkyl, O-alkenyl and O-alkynylrapamycin derivatives
US6177401B1 (en) 1992-11-13 2001-01-23 Max-Planck-Gesellschaft Zur Forderung Der Wissenschaften Use of organic compounds for the inhibition of Flk-1 mediated vasculogenesis and angiogenesis
WO1994014801A1 (fr) 1992-12-29 1994-07-07 Smithkline Beecham Plc Derives heterocycliques de l'uree antagonistes de 5ht2c et 5h¿2b?
US5512473A (en) 1993-01-29 1996-04-30 Brent; Roger Max-interacting proteins and related molecules and methods
GB9302275D0 (en) 1993-02-05 1993-03-24 Smithkline Beecham Plc Novel compounds
TW272942B (fr) 1993-02-10 1996-03-21 Takeda Pharm Industry Co Ltd
DK41193D0 (da) 1993-04-07 1993-04-07 Neurosearch As Ionkanalaabnere
AU6633694A (en) 1993-04-09 1994-11-08 Board Of Regents Of The University Of Nebraska, The Novel methods and compositions for the treatment of (ras)-activated cancer with heterotypic anti-(raf) antisense oligonucleotides
GB9308802D0 (en) 1993-04-28 1993-06-09 Smithkline Beecham Plc Treatment
DE4322806A1 (de) 1993-07-08 1995-01-12 Janich Gmbh & Co Vorrichtung zur Abdichtung zwischen zwei Anlagenteilen
IL110296A (en) 1993-07-16 1999-12-31 Smithkline Beecham Corp Imidazole compounds process for their preparation and pharmaceutical compositions containing them
US5378725A (en) 1993-07-19 1995-01-03 The Arizona Board Of Regents Inhibition of phosphatidylinositol 3-kinase with wortmannin and analogs thereof
US6514745B1 (en) 1993-07-19 2003-02-04 The Regents Of The University Of California Oncoprotein protein kinase
NZ264063A (en) 1993-08-13 1995-11-27 Nihon Nohyaku Co Ltd N-(2-phenylpyrid-3-yl)- and n-(4-phenylpyrimidin-5-yl)-n'-phenylurea derivatives and pharmaceutical compositions
US5441947A (en) 1993-08-25 1995-08-15 Eli Lilly And Company Methods of inhibiting vascular restenosis
US5468773A (en) 1993-08-25 1995-11-21 Eli Lilly And Company Methods for inhibiting bone loss and cartilage degradation using wortmannin and its analogs
US6361773B1 (en) 1993-09-17 2002-03-26 Smithkline Beecham Corporation Antibodies produced against cytokine suppressive anti-inflammatory drug binding proteins
JP3377529B2 (ja) 1993-09-17 2003-02-17 スミスクライン・ビーチャム・コーポレイション 薬物結合タンパク質
US5783664A (en) 1993-09-17 1998-07-21 Smithkline Beecham Corporation Cytokine suppressive anit-inflammatory drug binding proteins
US5869043A (en) 1993-09-17 1999-02-09 Smithkline Beecham Corporation Drug binding protein
US5547966A (en) 1993-10-07 1996-08-20 Bristol-Myers Squibb Company Aryl urea and related compounds
CA2133815A1 (fr) 1993-10-12 1995-04-13 Jeffrey Alan Dodge Inhibition de la phosphatidylinositol 3-kinase au moyen de la viridine, de la demethoxyviridine, du viridiol, du demethoxyviridiol, de la virone et de la wortmannolone et analogues de ces substances.
US5596001A (en) 1993-10-25 1997-01-21 Pfizer Inc. 4-aryl-3-(heteroarylureido)quinoline derivatves
JPH09505055A (ja) 1993-11-08 1997-05-20 スミスクライン・ビーチャム・コーポレイション サイトカイン媒介疾患治療用オキサゾール
US5538848A (en) 1994-11-16 1996-07-23 Applied Biosystems Division, Perkin-Elmer Corp. Method for detecting nucleic acid amplification using self-quenching fluorescence probe
JPH07141053A (ja) 1993-11-17 1995-06-02 Nec Niigata Ltd クロック発生回路
WO1995014023A1 (fr) 1993-11-19 1995-05-26 Abbott Laboratories Analogues semi-synthetiques de rapamycine (macrolides) utilises comme immunomodulateurs
CN1046944C (zh) 1993-12-17 1999-12-01 山道士有限公司 雷怕霉素类衍生物
US5700823A (en) 1994-01-07 1997-12-23 Sugen, Inc. Treatment of platelet derived growth factor related disorders such as cancers
CH686211A5 (de) 1994-01-27 1996-02-15 Ciba Geigy Ag Motten- und Koferschutzmittel.
US6524832B1 (en) 1994-02-04 2003-02-25 Arch Development Corporation DNA damaging agents in combination with tyrosine kinase inhibitors
US6811779B2 (en) 1994-02-10 2004-11-02 Imclone Systems Incorporated Methods for reducing tumor growth with VEGF receptor antibody combined with radiation and chemotherapy
DE4412334A1 (de) 1994-04-11 1995-10-19 Hoechst Ag Substituierte N-Heteroaroylguanidine, Verfahren zu ihrer Herstellung, ihre Verwendung als Medikament oder Diagnostikum sowie sie enthaltendes Medikament
US5871918A (en) 1996-06-20 1999-02-16 The University Of North Carolina At Chapel Hill Electrochemical detection of nucleic acid hybridization
JP2909216B2 (ja) 1994-04-29 1999-06-23 パーキン‐エルマー コーポレイション 核酸増幅生成物のリアルタイム検出装置
US5559137A (en) 1994-05-16 1996-09-24 Smithkline Beecham Corp. Compounds
US5656612A (en) 1994-05-31 1997-08-12 Isis Pharmaceuticals, Inc. Antisense oligonucleotide modulation of raf gene expression
US5744362A (en) 1994-05-31 1998-04-28 Isis Pharmaceuticals, Inc. Antisense oligonucleotide modulation of raf gene expression
US5563255A (en) * 1994-05-31 1996-10-08 Isis Pharmaceuticals, Inc. Antisense oligonucleotide modulation of raf gene expression
WO1995033458A1 (fr) 1994-06-02 1995-12-14 Smithkline Beecham Corporation Composes anti-inflammatoires
US5447957A (en) 1994-06-02 1995-09-05 Smithkline Beecham Corp. Anti-inflammatory compounds
US5470882A (en) 1994-06-02 1995-11-28 Smithkline Beecham Corp. Anti-inflammatory compounds
US5786362A (en) 1994-06-16 1998-07-28 University Of Miami Method of treating Hormone independent cancer
EP0690344A1 (fr) 1994-06-29 1996-01-03 Konica Corporation Matériau photographique à couleurs à l'halogénure d'argent sensible à la lumière
US5480906A (en) 1994-07-01 1996-01-02 Eli Lilly And Company Stereochemical Wortmannin derivatives
FR2722319B1 (fr) 1994-07-08 1996-08-14 Thomson Csf Dispositif de visualisation couleurs
US5597719A (en) 1994-07-14 1997-01-28 Onyx Pharmaceuticals, Inc. Interaction of RAF-1 and 14-3-3 proteins
PT708085E (pt) 1994-10-19 2002-11-29 Novartis Ag Eteres antivirais de isosteros de substrato de aspartato protease
JP3591938B2 (ja) 1994-10-27 2004-11-24 キヤノン株式会社 インクジェット用記録媒体及びこれを用いた画像形成方法
CA2161376C (fr) 1994-10-27 2005-01-11 Toshiaki Minami Support d'enregistrement thermique multicolore reversible
EP0788564B1 (fr) 1994-10-28 2001-05-16 Novozymes A/S Procede d'appret chimique de fibres polymeres insolubles
US5723290A (en) 1994-11-03 1998-03-03 Trustees Of The University Of Pennsylvania Methods for profiling mRNA expression in neurites
TW313568B (fr) 1994-12-20 1997-08-21 Hoffmann La Roche
US5780483A (en) 1995-02-17 1998-07-14 Smithkline Beecham Corporation IL-8 receptor antagonists
EP0809492A4 (fr) 1995-02-17 2007-01-24 Smithkline Beecham Corp Antagonistes des recepteurs d'il-8
US5814646A (en) 1995-03-02 1998-09-29 Eli Lilly And Company Inhibitors of amyloid beta-protein production
US5624937A (en) 1995-03-02 1997-04-29 Eli Lilly And Company Chemical compounds as inhibitors of amyloid beta protein production
DE19512639A1 (de) 1995-04-05 1996-10-10 Merck Patent Gmbh Benzonitrile und -fluoride
US5747498A (en) 1996-05-28 1998-05-05 Pfizer Inc. Alkynyl and azido-substituted 4-anilinoquinazolines
US5658903A (en) 1995-06-07 1997-08-19 Smithkline Beecham Corporation Imidazole compounds, compositions and use
US5710173A (en) 1995-06-07 1998-01-20 Sugen, Inc. Thienyl compounds for inhibition of cell proliferative disorders
CZ292233B6 (cs) 1995-06-09 2003-08-13 Novartis Ag Deriváty rapamycinu a jejich použití jako léčiv
US5994063A (en) 1995-06-23 1999-11-30 Metzker; Michael L. Substituted 4,4-difluoro-4-bora-3A,4A-diaza-s-indacene compounds for homogenous amplification/detection assays
GB9514265D0 (en) 1995-07-13 1995-09-13 Wellcome Found Hetrocyclic compounds
US5849761A (en) 1995-09-12 1998-12-15 Regents Of The University Of California Peripherally active anti-hyperalgesic opiates
AU712467B2 (en) 1995-09-18 1999-11-04 Sankyo Company Limited New urea derivatives having ACAT inhibitory activity, their preparation and their therapeutic and prophylactic use
SE510846C2 (sv) 1995-11-06 1999-06-28 Moelnlycke Health Care Ab Innerförpackning för i en yttre förpackning sterilförpackade bukdukar
DE69622183D1 (de) 1995-11-07 2002-08-08 Kirin Brewery Chinolinderivate und chinazolinderivate welche die autophosphorylierung des von blutplättchen abstammenden wachstumsfaktorrezeptors inhibiren und sie enthaltende pharmazeutische zusammensetzungen
EP0882715B1 (fr) 1995-12-28 2003-04-23 Kureha Kagaku Kogyo Kabushiki Kaisha Nouveaux n-(non substitue ou substitue)-4-substitue-6-(non substitue ou substitue) phenoxy-2-pyridinecarboxamides ou thiocarboxamides, leurs procedes de production, et herbicides
US5807876A (en) 1996-04-23 1998-09-15 Vertex Pharmaceuticals Incorporated Inhibitors of IMPDH enzyme
US6005008A (en) 1996-02-16 1999-12-21 Smithkline Beecham Corporation IL-8 receptor antagonists
US6011029A (en) 1996-02-26 2000-01-04 Bristol-Myers Squibb Company Inhibitors of farnesyl protein transferase
US6103692A (en) 1996-03-12 2000-08-15 The General Hospital Corporation Inhibiting protein interactions
US6211373B1 (en) 1996-03-20 2001-04-03 Smithkline Beecham Corporation Phenyl urea antagonists of the IL-8 receptor
US6262113B1 (en) 1996-03-20 2001-07-17 Smithkline Beecham Corporation IL-8 receptor antagonists
CA2202578A1 (fr) 1996-04-15 1997-10-15 Mitsuru Shiraishi Derives d'hydroxypyridine, leur production et leur utilisation
CZ298463B6 (cs) 1996-04-23 2007-10-10 Vertex Pharmaceuticals Incorporated Deriváty mocoviny pro použití jako inhibitory IMPDH enzymu a farmaceutické prostredky, které je obsahují
US6232299B1 (en) 1996-05-01 2001-05-15 Eli Lilly And Company Use of protein kinase C inhibitors to enhance the clinical efficacy of oncolytic agents and radiation therapy
JPH09301858A (ja) 1996-05-13 1997-11-25 Senju Pharmaceut Co Ltd グルコン酸クロルヘキシジン安定化水性薬剤
EP0906273B1 (fr) 1996-05-24 2002-10-16 Neurosearch A/S Derives phenyle contenant un groupe acide, leur preparation et leur utilisation comme inhibiteurs des vannes a chlorure
US5910417A (en) 1996-05-31 1999-06-08 National Jewish Center For Immunology And Respiratory Medicine Regulation of cytokine production in a hematopoietic cell
JP2000513359A (ja) 1996-06-27 2000-10-10 スミスクライン・ビーチャム・コーポレイション Il―8受容体拮抗薬
US6218539B1 (en) 1996-06-27 2001-04-17 Smithkline Beecham Corporation IL-8 receptor antagonists
KR20000022274A (ko) 1996-06-27 2000-04-25 스튜어트 알. 수터, 스티븐 베네티아너, 피터 존 기딩스 Il-8 수용체 길항제
CZ425698A3 (cs) 1996-06-27 1999-06-16 Smithkline Beecham Corporation Antagonista IL-8 receptoru
CZ59398A3 (cs) 1996-06-29 1998-07-15 Samjin Pharmaceutical Co., Ltd. Piperazinové deriváty a způsob jejich přípravy
US6117635A (en) 1996-07-16 2000-09-12 Intergen Company Nucleic acid amplification oligonucleotides with molecular energy transfer labels and methods based thereon
US6150415A (en) 1996-08-13 2000-11-21 The Regents Of The University Of California Epoxide hydrolase complexes and methods therewith
US5808080A (en) 1996-09-05 1998-09-15 Eli Lilly And Company Selective β3 adrenergic agonists
US6019793A (en) 1996-10-21 2000-02-01 Synthes Surgical prosthetic device
US5965573A (en) 1996-10-23 1999-10-12 Zymogenetics, Inc. Compositions and methods for treating bone deficit conditions
AU4988997A (en) 1996-10-23 1998-05-15 Osteoscreen, Inc. Compositions and methods for treating bone deficit conditions
ES2339306T3 (es) 1996-11-15 2010-05-18 Cytokine Pharmasciences, Inc. Guanilhidrazonas utiles en el tratamiento de las enfermedades asociadas con la activacion de las celulas t.
GB9623833D0 (en) 1996-11-16 1997-01-08 Zeneca Ltd Chemical compound
AU4966497A (en) 1996-11-18 1998-06-10 Yamanouchi Pharmaceutical Co., Ltd. Novel acylamino-substituted acylanilide derivatives or pharmaceutical composition comprising the same
FR2755967B1 (fr) 1996-11-21 1999-01-29 Pf Medicament Derives de la pyridin-2-yl-methylamine, leur procede de preparation et leur application comme medicaments
CA2278504A1 (fr) 1997-01-23 1998-07-30 Smithkline Beecham Corporation Antagonistes du recepteur de l'il-8
US5929250A (en) 1997-01-23 1999-07-27 Smithkline Beecham Corporation IL-8 receptor antagonists
ES2224365T3 (es) 1997-02-12 2005-03-01 Smithkline Beecham Corporation Antagonistas de receptores de il-8.
YU49899A (sh) 1997-04-04 2002-09-19 Pfizer Products Inc. Nikotinamidni derivati
US20020111495A1 (en) 1997-04-04 2002-08-15 Pfizer Inc. Nicotinamide acids, amides, and their mimetics active as inhibitors of PDE4 isozymes
US5958983A (en) 1997-04-10 1999-09-28 Pharmacia & Upjohn Company Polyaromatic antiviral compositions
AU7080298A (en) 1997-04-25 1998-11-24 Takeda Chemical Industries Ltd. Triazine derivatives, their production and agrochemical composition
US6210710B1 (en) 1997-04-28 2001-04-03 Hercules Incorporated Sustained release polymer blend for pharmaceutical applications
AR012634A1 (es) 1997-05-02 2000-11-08 Sugen Inc Compuesto basado en quinazolina, composicion famaceutica que lo comprende, metodo para sintetizarlo, su uso, metodos de modulacion de la funcion deserina/treonina proteinaquinasa con dicho compuesto y metodo in vitro para identificar compuestos que modulan dicha funcion
US6316479B1 (en) 1997-05-19 2001-11-13 Sugen, Inc. Isoxazole-4-carboxamide compounds active against protein tryosine kinase related disorders
AU7726898A (en) 1997-05-22 1998-12-11 G.D. Searle & Co. Pyrazole derivatives as p38 kinase inhibitors
EP0983260A2 (fr) 1997-05-22 2000-03-08 G.D. Searle & Co. PYRAZOLES 3(5)-HETEROARYL SUBSTITUEES UTILISEES COMME INHIBITEURS DE KINASE p38
US6514977B1 (en) 1997-05-22 2003-02-04 G.D. Searle & Company Substituted pyrazoles as p38 kinase inhibitors
PT1019040E (pt) 1997-05-23 2005-01-31 Bayer Pharmaceuticals Corp Inibicao da actividade de p38-quinase por meio de arilureias
CA2291065C (fr) 1997-05-23 2010-02-09 Bayer Corporation Inhibiteurs de raf kinase
US6150395A (en) 1997-05-30 2000-11-21 The Regents Of The University Of California Indole-3-carbinol (I3C) derivatives and methods
US6294350B1 (en) 1997-06-05 2001-09-25 Dalhousie University Methods for treating fibroproliferative diseases
US5985592A (en) 1997-06-05 1999-11-16 Dalhousie University Uses for pentoxifylline or functional derivatives/metabolites thereof
AU7966198A (en) 1997-06-13 1998-12-30 Smithkline Beecham Corporation Novel pyrazole and pyrazoline substituted compounds
US6159901A (en) 1997-06-27 2000-12-12 Kureha Kagaku Kogyo Kabushiki Kaisha 6-phenoxy picolinic acid alkylidene hydrazide derivative, process for producing the same and herbicide using the same
US6093742A (en) 1997-06-27 2000-07-25 Vertex Pharmaceuticals, Inc. Inhibitors of p38
US6525065B1 (en) 1997-06-30 2003-02-25 Targacept, Inc. Pharmaceutical compositions and methods for effecting dopamine release
AUPP003197A0 (en) 1997-09-03 1997-11-20 Fujisawa Pharmaceutical Co., Ltd. New heterocyclic compounds
UA71555C2 (en) 1997-10-06 2004-12-15 Zentaris Gmbh Methods for modulating function of serine/threonine protein kinases by 5-azaquinoline derivatives
AU9802198A (en) 1997-10-21 1999-05-10 Pharmacia & Upjohn Company Antiinflammatory thiadiazolyl ureas which act as lfa-1 and mac-1 inhibitors
DK1027328T3 (da) 1997-10-31 2006-11-13 Aventis Pharma Ltd Substituerede anilider
JP2001521934A (ja) 1997-11-03 2001-11-13 ベーリンガー インゲルハイム ファーマシューティカルズ インコーポレイテッド 抗炎症薬としての芳香族ヘテロ環式化合物
US6022884A (en) 1997-11-07 2000-02-08 Amgen Inc. Substituted pyridine compounds and methods of use
US6018065A (en) 1997-11-10 2000-01-25 Advanced Technology Materials, Inc. Method of fabricating iridium-based materials and structures on substrates, iridium source reagents therefor
IL135176A0 (en) 1997-11-10 2001-05-20 Bristol Myers Squibb Co Benzothiazole derivatives and pharmaceutical compositions containing the same
GB9723789D0 (en) 1997-11-12 1998-01-07 Zeneca Ltd Chemical compounds
WO1999026657A1 (fr) 1997-11-25 1999-06-03 Musc Foundation For Research Development Inhibiteurs de la monoxyde d'azote-synthase
AU1407899A (en) 1997-12-01 1999-06-16 E.I. Du Pont De Nemours And Company Fungicidal cyclic amides
US6635421B1 (en) 1997-12-09 2003-10-21 Children's Medical Center Corporation Neuropilins and use thereof in methods for diagnosis and prognosis of cancer
SK282727B6 (sk) 1997-12-19 2002-11-06 Slovakofarma, A. S. 1,3-Disubstituované močoviny - inhibítory ACAT a spôsob ich prípravy
US6174901B1 (en) 1998-12-18 2001-01-16 Amgen Inc. Substituted pyridine and pyridazine compounds and methods of use
UA67763C2 (uk) 1997-12-22 2004-07-15 Байер Фармасьютікалс Корпорейшн Арил- та гетероарилзаміщені гетероциклічні похідні сечовини, фармацевтична композиція та спосіб лікування захворювання, спричиненого кіназою raf
EP1049664B1 (fr) 1997-12-22 2005-03-16 Bayer Pharmaceuticals Corporation Inhibition de raf kinase au moyen de diphenylurees substituees symetriques et asymetriques
US20080300281A1 (en) 1997-12-22 2008-12-04 Jacques Dumas Inhibition of p38 Kinase Activity Using Aryl and Heteroaryl Substituted Heterocyclic Ureas
KR100571588B1 (ko) 1997-12-22 2006-04-17 바이엘 코포레이션 치환 헤테로시클릭 우레아를 이용한 raf 키나제의 억제
ATE346600T1 (de) 1997-12-22 2006-12-15 Bayer Pharmaceuticals Corp Inhibierung der p38 kinase-aktivität durch die verwendung von aryl- und heteroarylsubstituierten harnstoffen
US7329670B1 (en) 1997-12-22 2008-02-12 Bayer Pharmaceuticals Corporation Inhibition of RAF kinase using aryl and heteroaryl substituted heterocyclic ureas
CA2315720A1 (fr) 1997-12-22 1999-07-01 Bayer Corporation Inhibition de l'activite de p38 kinase au moyen d'urees heterocycliques substituees
EP1042305B1 (fr) 1997-12-22 2005-06-08 Bayer Pharmaceuticals Corp. INHIBITION DE LA KINASE p38 PAR DES DIPHENYL-UREES SYMETRIQUES ET DISSYMETRIQUES
WO1999032109A1 (fr) 1997-12-23 1999-07-01 Bristol-Myers Squibb Company Paclitaxels portant un substituant 7-desoxy-6-azote
AU1689999A (en) 1997-12-25 1999-07-19 Daiichi Pharmaceutical Co., Ltd. Medicinal composition for percutaneous administration
GB9800575D0 (en) 1998-01-12 1998-03-11 Glaxo Group Ltd Heterocyclic compounds
US6221913B1 (en) 1998-01-21 2001-04-24 Zymogenetics, Inc. Dialkyl ureas as calcitonin mimetics
FR2774824B1 (fr) 1998-02-09 2000-04-28 Moving Magnet Tech Actionneur lineaire ameliore
AU741632B2 (en) 1998-02-18 2001-12-06 Theryte Limited Treating cancer
MY132496A (en) 1998-05-11 2007-10-31 Vertex Pharma Inhibitors of p38
US6413773B1 (en) 1998-06-01 2002-07-02 The Regents Of The University Of California Phosphatidylinositol 3-kinase inhibitors as stimulators of endocrine differentiation
UA60365C2 (uk) 1998-06-04 2003-10-15 Пфайзер Продактс Інк. Похідні ізотіазолу, спосіб їх одержання, фармацевтична композиція та спосіб лікування гіперпроліферативного захворювання у ссавця
US6667340B1 (en) 1998-06-26 2003-12-23 Arizona Board Of Regents On Behalf Of The University Of Arizona Inhibitors of phosphatidyl myo-inositol cycle
US6352977B1 (en) 1998-07-13 2002-03-05 Aventis Pharma Limited Substituted β-alanines
US6184226B1 (en) 1998-08-28 2001-02-06 Scios Inc. Quinazoline derivatives as inhibitors of P-38 α
WO2000017175A1 (fr) 1998-09-18 2000-03-30 Vertex Pharmaceuticals Incorporated Inhibiteurs de p38
US6383734B1 (en) 1998-09-30 2002-05-07 Advanced Research And Technology Institute, Inc. Method to determine inhibition of PAK3 activation of Raf-1
US6130053A (en) 1999-08-03 2000-10-10 Cell Pathways, Inc. Method for selecting compounds for inhibition of neoplastic lesions
GB9823873D0 (en) 1998-10-30 1998-12-30 Pharmacia & Upjohn Spa 2-ureido-thiazole derivatives,process for their preparation,and their use as antitumour agents
CZ20011564A3 (cs) 1998-11-06 2002-04-17 Basf Aktiengesellschaft Léčivo pro inhibici vaskulární hyperpermeability
IL143328A0 (en) 1998-11-25 2002-04-21 Genetica Inc Methods and reagents for increasing proliferative capacity and preventing replicative senescence
US6114517A (en) 1998-12-10 2000-09-05 Isis Pharmaceuticals Inc. Methods of modulating tumor necrosis factor α-induced expression of cell adhesion molecules
US6337338B1 (en) 1998-12-15 2002-01-08 Telik, Inc. Heteroaryl-aryl ureas as IGF-1 receptor antagonists
UY25842A1 (es) 1998-12-16 2001-04-30 Smithkline Beecham Corp Antagonistas de receptores de il-8
US6331541B1 (en) 1998-12-18 2001-12-18 Soo S. Ko N-ureidoalkyl-piperidines as modulators of chemokine receptor activity
EP1140087A4 (fr) 1998-12-18 2002-04-03 Du Pont Pharm Co N-ureidoalkyl-piperidines utilisees en tant que modulateurs de l'activite des recepteurs des chimiokines
US6147107A (en) 1998-12-20 2000-11-14 Virginia Commonwealth University Specific inhibition of the P42/44 mitogen activated protein (map) kinase cascade sensitizes tumor cells
US20080269265A1 (en) 1998-12-22 2008-10-30 Scott Miller Inhibition Of Raf Kinase Using Symmetrical And Unsymmetrical Substituted Diphenyl Ureas
GB9828511D0 (en) 1998-12-24 1999-02-17 Zeneca Ltd Chemical compounds
JP4632544B2 (ja) 1998-12-25 2011-02-16 あすか製薬株式会社 アミノピラゾール誘導体
US6351764B1 (en) 1998-12-31 2002-02-26 Michael Voticky System and method for prioritizing communications messages
RU2319693C9 (ru) 1999-01-13 2008-08-20 Байер Копэрейшн Производные мочевины (варианты), фармацевтическая композиция (варианты) и способ лечения заболевания, связанного с ростом раковых клеток (варианты)
US20020065296A1 (en) * 1999-01-13 2002-05-30 Bayer Corporation Heteroaryl ureas containing nitrogen hetero-atoms as p38 kinase inhibitors
US8124630B2 (en) 1999-01-13 2012-02-28 Bayer Healthcare Llc ω-carboxyaryl substituted diphenyl ureas as raf kinase inhibitors
EP1140840B1 (fr) 1999-01-13 2006-03-22 Bayer Pharmaceuticals Corp. Diphenylurees a substituants -g(v)-carboxyaryles, inhibitrices de kinase raf
US7928239B2 (en) 1999-01-13 2011-04-19 Bayer Healthcare Llc Inhibition of RAF kinase using quinolyl, isoquinolyl or pyridyl ureas
TWI284642B (en) 1999-01-18 2007-08-01 Hoffmann La Roche Novel heterocyclic sulfonamides
UA73492C2 (en) 1999-01-19 2005-08-15 Aromatic heterocyclic compounds as antiinflammatory agents
PL206627B1 (pl) 1999-01-22 2010-09-30 Kirin Pharma Kk Pochodna chinazoliny, jej zastosowanie i kompozycja farmaceutyczna
EP1150977B1 (fr) 1999-02-12 2004-08-25 SmithKline Beecham plc Derives de phenyluree et de phenylthiouree utilises comme antagonistes des recepteurs de l'orexine
CA2360934A1 (fr) 1999-02-22 2000-08-31 Lin-Hua Zhang Derives polycyclo heterocycliques utilises en tant qu'agents anti-inflammatoires
NZ514711A (en) 1999-03-12 2004-02-27 Boehringer Ingelheim Pharma Compounds useful as anti-inflammatory agents
MXPA01009077A (es) 1999-03-12 2002-03-27 Boehringer Ingelheim Pharma Composiciones heterociclicos aromaticos como agentes anti-inflamatorios.
ES2405316T3 (es) 1999-03-19 2013-05-30 Vertex Pharmaceuticals Incorporated Inhibidores de la enzima IMPDH
WO2000057877A1 (fr) 1999-03-26 2000-10-05 Euro-Celtique S.A. Pyrazoles, imidazoles, oxazoles, thiazoles et pyrroles aryle substitues et leurs utilisations
US6140124A (en) 1999-04-06 2000-10-31 Isis Pharmaceuticals Inc. Antisense modulation of P38 mitogen activated protein kinase expression
US6316462B1 (en) 1999-04-09 2001-11-13 Schering Corporation Methods of inducing cancer cell death and tumor regression
CA2370805C (fr) 1999-05-05 2012-07-03 Aventis Pharma Limited Urees en tant que modulateurs de l'adhesion cellulaire
PL365999A1 (en) 1999-05-14 2005-01-24 Imclone Systems Incorporated Treatment of refractory human tumors with epidermal growth factor receptor antagonists
HN2000000051A (es) 1999-05-19 2001-02-02 Pfizer Prod Inc Derivados heterociclicos utiles como agentes anticancerosos
TWI234557B (en) 1999-05-26 2005-06-21 Telik Inc Novel naphthalene ureas as glucose uptake enhancers
DE19927835A1 (de) 1999-06-18 2000-12-21 Clariant Gmbh Verwendung von verbesserten Cyanpigmenten in elektrophotographischen Tonern und Entwicklern, Pulverlacken und Ink-Jet-Tinten
ES2253233T3 (es) 1999-07-09 2006-06-01 Boehringer Ingelheim Pharmaceuticals Inc. Procedimiento para sintesis de compuestos de urea heteroaril sustituidos.
ES2251395T3 (es) 1999-07-26 2006-05-01 Banyu Pharmaceutical Co., Ltd. Derivados de biarilureas.
DE60027011T2 (de) 1999-07-28 2006-12-07 Kirin Beer K.K. Harnstoffderivate als CCR-3 Rezeptor-Inhibitoren
US6387900B1 (en) 1999-08-12 2002-05-14 Pharmacia & Upjohn S.P.A. 3(5)-ureido-pyrazole derivatives process for their preparation and their use as antitumor agents
US6372933B1 (en) 1999-08-26 2002-04-16 Smithkline Beecham Corporation Process for preparing certain phenyl urea compounds
US6420378B1 (en) 1999-10-15 2002-07-16 Supergen, Inc. Inhibition of abnormal cell proliferation with camptothecin and combinations including the same
WO2001054727A1 (fr) 1999-10-15 2001-08-02 The Van Kampen Group, Inc. Traitement de tumeurs cutanees, verrues et infections virales utilisant du p. acnes tue par la chaleur
US6794393B1 (en) 1999-10-19 2004-09-21 Merck & Co., Inc. Tyrosine kinase inhibitors
ES2292488T3 (es) 1999-11-16 2008-03-16 Boehringer Ingelheim Pharmaceuticals Inc. Derivados de la urea, como agentes antiinflamatorios.
CN1262541C (zh) 1999-12-29 2006-07-05 惠氏公司 三环蛋白激酶抑制剂
US6525046B1 (en) 2000-01-18 2003-02-25 Boehringer Ingelheim Pharmaceuticals, Inc. Aromatic heterocyclic compounds as antiinflammatory agents
US7740841B1 (en) 2000-01-28 2010-06-22 Sunnybrook Health Science Center Therapeutic method for reducing angiogenesis
MXPA02007632A (es) 2000-02-07 2004-08-23 Abbott Gmbh & Co Kg Derivados de 2-benzotiazolil urea y su uso como inhibidores de proteina cinasa.
US6608052B2 (en) 2000-02-16 2003-08-19 Boehringer Ingelheim Pharmaceuticals, Inc. Compounds useful as anti-inflammatory agents
US6658473B1 (en) 2000-02-25 2003-12-02 Sun Microsystems, Inc. Method and apparatus for distributing load in a computer environment
WO2001064235A1 (fr) 2000-03-02 2001-09-07 Ludwig Institute For Cancer Research Procedes de traitement, d'examen et de detection de cancers exprimant le facteur d de croissance endotheliale vasculaire
WO2001066099A2 (fr) 2000-03-06 2001-09-13 Astrazeneca Ab Therapie
GB0005357D0 (en) 2000-03-06 2000-04-26 Smithkline Beecham Plc Compounds
US6667300B2 (en) 2000-04-25 2003-12-23 Icos Corporation Inhibitors of human phosphatidylinositol 3-kinase delta
US6403588B1 (en) 2000-04-27 2002-06-11 Yamanouchi Pharmaceutical Co., Ltd. Imidazopyridine derivatives
US6390102B1 (en) 2000-07-14 2002-05-21 General Electric Company Silicone compositions for personal care products and method for making
EP1311277A4 (fr) 2000-07-18 2004-08-25 Joslin Diabetes Center Inc Procede de modulation de fibrose
MXPA02012909A (es) 2000-07-24 2004-05-05 Boehringer Ingelheim Pharma Formulaciones mejoradas de dosis oral de 1-(5-ter-butil -2-p-tiolil2h -pirazol-3 -il)-3-(4-2 (2-morfolin-4-il -etoxi) -naftalen -1-il) -urea.
GT200100147A (es) 2000-07-31 2002-06-25 Derivados de imidazol
US20020165349A1 (en) 2000-08-04 2002-11-07 Kirsch Wolff M. Iron regulating protein -2 (IRP-2) as a diagnostic for neurodegenerative disease
MXPA03001256A (es) 2000-08-11 2004-12-06 Vertex Pharma Inhibidores de p38.
US20020173507A1 (en) 2000-08-15 2002-11-21 Vincent Santora Urea compounds and methods of uses
KR20040066185A (ko) 2000-08-17 2004-07-23 샤단호칭키타사토겐큐쇼 신규 슈도에리쓰로마이신 유도체
YU14703A (sh) 2000-08-31 2006-05-25 Pfizer Products Inc. Derivati pirazola i njihova upotreba kao inhibitora protein kinaze
WO2002025286A2 (fr) 2000-09-20 2002-03-28 The President And Fellows Of Harvard College Procedes de detection du cancer de la prostate
AUPR034000A0 (en) 2000-09-25 2000-10-19 Fujisawa Pharmaceutical Co., Ltd. Aminoalcohol derivatives
ATE399766T1 (de) 2000-10-20 2008-07-15 Eisai R&D Man Co Ltd Stickstoff enthaltende aromatische heterozyklen
JP3768800B2 (ja) 2000-10-31 2006-04-19 キヤノン株式会社 画像形成装置
AU2002223500A1 (en) 2000-11-17 2002-05-27 Novo-Nordisk A/S Glucagon antagonists/inverse agonists
CN1525889A (zh) 2000-11-27 2004-09-01 ����ˡ�A�������� 分类物品的系统、方法及程序
EA005859B1 (ru) 2000-11-28 2005-06-30 Пфайзер Продактс Инк. Соли изотиазол-4-карбоксамида и их применение в качестве антигиперпролиферативных агентов
AU2002232439A1 (en) 2000-11-29 2002-06-11 Glaxo Group Limited Benzimidazole derivatives useful as tie-2 and/or vegfr-2 inhibitors
EP1488797B1 (fr) 2000-12-08 2008-04-23 Kyowa Hakko Kogyo Co., Ltd. Preparations orales ayant une action de soulagement des demangeaisons ou un effet antiprurigineux
ES2275621T3 (es) 2000-12-21 2007-06-16 Glaxo Group Limited Antibioticos macrolidos.
KR100362390B1 (ko) 2000-12-29 2002-11-23 삼성전자 주식회사 화상형성장치의 현상기-토너 카트리지 조립체
US7235576B1 (en) * 2001-01-12 2007-06-26 Bayer Pharmaceuticals Corporation Omega-carboxyaryl substituted diphenyl ureas as raf kinase inhibitors
EP1360173A2 (fr) 2001-01-25 2003-11-12 Guilford Pharmaceuticals Inc. Composes de liaison de la cyclophiline carboxylique trisubstituee et leur utilisation
FR2820136A1 (fr) 2001-01-26 2002-08-02 Aventis Pharma Sa Nouveaux derives de l'uree, leur procede de preparation, leur application a titre de medicaments, compositions pharmaceutiques et nouvelle utilisation
WO2002059081A2 (fr) 2001-01-26 2002-08-01 Kirin Beer Kabushiki Kaisha Derives d'uree en tant qu'inhibiteurs du recepteur ccr-3
US6670364B2 (en) 2001-01-31 2003-12-30 Telik, Inc. Antagonists of MCP-1 function and methods of use thereof
PT2269603E (pt) 2001-02-19 2015-09-09 Novartis Ag Tratamento de tumores da mama com um derivado de rapamicina em combinação com exemestano
UA76977C2 (en) 2001-03-02 2006-10-16 Icos Corp Aryl- and heteroaryl substituted chk1 inhibitors and their use as radiosensitizers and chemosensitizers
DE60221104T2 (de) 2001-03-07 2008-03-13 TELIK, INC., Palo Alto Substituierte diarylharnstoffe als stimulatoren der fas-vermittelten apoptose
US6525091B2 (en) 2001-03-07 2003-02-25 Telik, Inc. Substituted diarylureas as stimulators for Fas-mediated apoptosis
MY142915A (en) 2001-03-23 2011-01-31 Bayer Healthcare Llc Rho-kinase inhibitors
ATE507839T1 (de) 2001-04-02 2011-05-15 Genentech Inc Kombinationstherapie
EP1381592A1 (fr) 2001-04-13 2004-01-21 Boehringer Ingelheim Pharmaceuticals Inc. Composes benzo-fusionnes a disubstitution en positions 1,4
EP1381594A1 (fr) 2001-04-13 2004-01-21 Boehringer Ingelheim Pharmaceuticals Inc. Composes d'uree utilises en tant qu'agents anti-inflammatoires
ATE355272T1 (de) * 2001-04-20 2006-03-15 Bayer Pharmaceuticals Corp Inhibierung der raf-kinase durch chinolin-, isochinolin- oder pyridin-harnstoffe
US7371763B2 (en) 2001-04-20 2008-05-13 Bayer Pharmaceuticals Corporation Inhibition of raf kinase using quinolyl, isoquinolyl or pyridyl ureas
US20030207914A1 (en) 2001-04-20 2003-11-06 Bayer Corporation Inhibition of raf kinase using quinolyl, isoquinolyl or pyridyl ureas
JP2004538266A (ja) 2001-04-27 2004-12-24 バーテックス ファーマシューティカルズ インコーポレイテッド ピラゾール由来のキナーゼインヒビター
EP1392661A1 (fr) 2001-05-16 2004-03-03 Boehringer Ingelheim Pharmaceuticals Inc. Derives de diaryluree utilisables en tant qu'agents anti-inflammatoires
WO2003004523A1 (fr) 2001-07-02 2003-01-16 Bayer Aktiengesellschaft Regulation de la kinase humaine crik a interaction avec rho/rac
ES2284887T3 (es) 2001-07-11 2007-11-16 Boehringer Ingelheim Pharmaceuticals Inc. Metodos para tratar enfermedades transmitidas por citocinas.
CA2466762A1 (fr) 2001-12-04 2003-06-12 Onyx Pharmaceuticals, Inc. Inhibiteurs de la voie raf-mek-erk pour traiter le cancer
BRPI0214840B8 (pt) 2001-12-21 2021-05-25 The Wellcome Trust métodos para a detecção de mutações oncogênicas humanas, métodos automatizados para detectar uma mutação e método e ensaio para identificar um ou mais compostos tendo atividade anti-proliferativa
AU2002356323A1 (en) 2001-12-24 2003-07-30 Catalyst Biomedica Limited Cancer-specific mutants of b-raf genes and uses thereof
US20040096855A1 (en) 2001-12-24 2004-05-20 Michael Stratton Genes
US20030207872A1 (en) 2002-01-11 2003-11-06 Bayer Corporation Omega-carboxyaryl substituted diphenyl ureas as raf kinase inhibitors
WO2003059373A2 (fr) 2002-01-16 2003-07-24 Ramot At Tel Aviv University Ltd. Procedes et compositions destines a stimuler ou a inhiber la fecondation
US20050026868A1 (en) 2003-07-11 2005-02-03 Metcalf Chester A. Phosphorus-containing macrocycles
US6998391B2 (en) 2002-02-07 2006-02-14 Supergen.Inc. Method for treating diseases associated with abnormal kinase activity
WO2003068223A1 (fr) 2002-02-11 2003-08-21 Bayer Corporation Urees aryliques a kinase de raf et activite inhibitrice d'angiogenese
ATE529406T1 (de) 2002-02-11 2011-11-15 Bayer Healthcare Llc Aryl-harnstoffe als kinase inhibitoren
NZ535985A (en) 2002-03-29 2007-04-27 Chiron Corp Substituted benzazoles and use thereof as RAF kinase inhibitors
US8299108B2 (en) 2002-03-29 2012-10-30 Novartis Ag Substituted benzazoles and methods of their use as inhibitors of raf kinase
DE10221524A1 (de) 2002-05-14 2003-12-04 Kmb Produktions Ag Felben Verfahren und Vorrichtung zum Herstellen von Verzehrgütern
AU2003233576A1 (en) 2002-05-17 2003-12-02 Sugen, Inc. Novel biomarkers of tyrosine kinase inhibitor exposure and activity in mammals
PE20040522A1 (es) 2002-05-29 2004-09-28 Novartis Ag Derivados de diarilurea dependientes de la cinasa de proteina
EP1545523A1 (fr) 2002-07-03 2005-06-29 Astex Technology Limited 3-(hetero) arylmethoxy pyridines et leurs analogues en tant qu'inhibiteurs de la p38 map kinase
ES2311120T3 (es) 2002-08-27 2009-02-01 Merck Patent Gmbh Derivados de glicinamida como inhibidores de quinasa-raf.
ATE435199T1 (de) 2002-09-05 2009-07-15 Neurosearch As Diarylharnstoffderivate und deren verwendung als chloridkanalblocker
BR0315580A (pt) 2002-10-24 2005-08-30 Merck Patent Gmbh Derivados de metileno uréia
CA2504933C (fr) 2002-11-06 2012-10-16 Dana-Farber Cancer Institute, Inc. Procedes et compositions de traitement du cancer au moyen d'inhibiteurs de la proteasome
PL376169A1 (en) 2002-11-15 2005-12-27 Novartis Ag Drug delivery system
SE0203654D0 (sv) 2002-12-09 2002-12-09 Astrazeneca Ab New compounds
US20030232400A1 (en) 2002-12-20 2003-12-18 Susan Radka Methods of screening subjects for expression of soluble receptors of vascular endothelial growth factor (VEGF) for use in managing treatment and determining prognostic outcome
US7202257B2 (en) 2003-12-24 2007-04-10 Deciphera Pharmaceuticals, Llc Anti-inflammatory medicaments
CN1839126A (zh) 2003-02-28 2006-09-27 拜耳制药公司 用于治疗癌症和其它病症的新的氰基吡啶衍生物
CA2516627A1 (fr) 2003-02-28 2004-09-16 Bayer Pharmaceuticals Corporation Derives de pyridine substitues utilises pour traiter des cancers et d'autres troubles
JP5229853B2 (ja) * 2003-02-28 2013-07-03 ニッポネックス インコーポレイテッド 癌その他の疾患の治療に有用な新規な二環尿素誘導体
US7531553B2 (en) 2003-03-21 2009-05-12 Amgen Inc. Heterocyclic compounds and methods of use
CN1764645A (zh) 2003-03-24 2006-04-26 默克专利有限公司 作为raf-激酶抑制剂的草酰胺衍生物
WO2004084889A1 (fr) 2003-03-28 2004-10-07 Pfizer Inc. Utilisation de l'inhibiteur de la proteinekinase c pour supprimer l'excitation postsynaptique lente soutenue (sspe) de neurones enteriques
US6896863B2 (en) 2003-04-01 2005-05-24 E. I. Du Pont De Nemours And Company Sodium cyanide process
DK1636585T3 (da) 2003-05-20 2008-05-26 Bayer Pharmaceuticals Corp Diarylurinstoffer med kinasehæmmende aktivitet
US20040259926A1 (en) 2003-06-05 2004-12-23 Bruendl Michelle M. 3-Aryloxy and 3-heteroaryloxy substituted benzo[b]thiophenes as therapeutic agents
WO2004108713A1 (fr) 2003-06-05 2004-12-16 Warner-Lambert Company Llc Benzothiophenes substitutes par cycloalkyle et heterocycloalkyle, utiles comme agents therapeutiques
WO2005002673A1 (fr) 2003-07-03 2005-01-13 Astex Therapeutics Limited Inhibiteurs de raf kinase
WO2005005389A2 (fr) 2003-07-07 2005-01-20 Merck Patent Gmbh Derives de malonamide
MXPA06000117A (es) 2003-07-08 2006-04-27 Novartis Ag Uso de rapamicina y derivados de rapamicina para el tratamiento de perdida osea.
ATE538787T1 (de) 2003-07-11 2012-01-15 Merck Patent Gmbh Benzimidazol-derivative als raf-kinase-hemmer
WO2005004863A1 (fr) 2003-07-11 2005-01-20 Merck Patent Gmbh Benzimidazole carboxamides utilises en tant qu'inhibiteurs de la kinase raf
US20060234931A1 (en) 2003-07-17 2006-10-19 Biggs William H Iii Treatment of diseases with kinase inhibitors
AR045134A1 (es) 2003-07-29 2005-10-19 Smithkline Beecham Plc Compuesto de 1h - imidazo [4,5-c] piridin-ilo, composicion farmaceutica que lo comprende, proceso para prepararla, su uso para preparar dicha composicion farmaceutica, combinacion farmaceutica, uso de la combinacion farmaceutica para la preparacion de un medicamento, procedimientos para preparar dic
DE10334663A1 (de) 2003-07-30 2005-03-10 Merck Patent Gmbh Harnstoffderivate
EP1668360A4 (fr) 2003-08-15 2007-08-08 Univ Pittsburgh Analyse multifactorielle pour la detection du cancer
US20080085902A1 (en) 2003-09-23 2008-04-10 Guido Bold Combination Of A Vegf Receptor Inhibitor Or With A Chemotherapeutic Agent
DE10344223A1 (de) 2003-09-24 2005-04-21 Merck Patent Gmbh 1,3-Benzoxazolylderivate als Kinase-Inhibitoren
EP1678123A1 (fr) 2003-10-15 2006-07-12 Bayer HealthCare AG Derives de tetrahydro-naphthalene et d'uree
EP1682126B1 (fr) 2003-10-16 2009-07-01 Novartis Vaccines and Diagnostics, Inc. Benzazoles substitues et leur utilisation en tant qu'inhibiteurs de la raf kinase
CA2542329A1 (fr) 2003-10-16 2005-04-28 Chiron Corporation Quinoxalines, quinolines, isoquinolines et quinazolines 2,6-bisubstituees servant d'inhibiteurs a la kinase raf pour le traitement du cancer
DE10349587A1 (de) 2003-10-24 2005-05-25 Merck Patent Gmbh Benzimidazolylderivate
DE10352979A1 (de) 2003-11-13 2005-06-09 Merck Patent Gmbh Pyridopyrimidinone
US20050171172A1 (en) 2003-11-13 2005-08-04 Ambit Biosciences Corporation Amide derivatives as PDGFR modulators
DE10354060A1 (de) 2003-11-19 2005-06-02 Merck Patent Gmbh Pyrrolderivate
TW200531979A (en) 2003-12-05 2005-10-01 Compound Therapeutics Inc Inhibitors of type 2 vascular endothelial growth factor receptors
TR200808208T1 (tr) 2003-12-09 2008-12-22 The Government Of The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Bir immün tepkinin bastırılması veya proliferatif bir hastalığın tedavisi
AU2004299174A1 (en) 2003-12-10 2005-06-30 Merck Patent Gmbh Diacylhydrazine derivatives
CA2548394A1 (fr) 2003-12-12 2005-06-30 Bayer Pharmaceuticals Corporation Procede de prediction et de pronostic du cancer, et surveillance de traitement anticancereux
CN1972925A (zh) 2004-01-30 2007-05-30 默克专利有限公司 双芳基脲类衍生物
US20050267060A1 (en) 2004-03-19 2005-12-01 The Penn State Research Foundation Combinatorial methods and compositions for treatment of melanoma
MY191349A (en) 2004-08-27 2022-06-17 Bayer Pharmaceuticals Corp New pharmaceutical compositions for the treatment of hyper-proliferative disorders
US7745446B2 (en) 2004-09-06 2010-06-29 Bayer Schering Pharma Aktiengesellschaft Pyrazolo[1,5-c]pyrimidines
UA90691C2 (en) 2004-09-29 2010-05-25 Баер Шеринг Фарма Акциенгезельшафт Process for the preparation of 4-{ 4-[({ [4-chloro-3-(trifluoromethyl)phenyl]amino}carbonyl)amino]phenoxy}-n-methylpyridine-2-carboxamide
PL1858877T3 (pl) 2005-01-14 2014-08-29 Gilead Connecticut Inc 1,3 podstawione diarylem moczniki jako modulatory aktywności kinazy
CN101132779B (zh) 2005-03-07 2016-03-16 拜尔健康护理有限责任公司 用于治疗癌症的包含ω-羧芳基取代的二苯基脲的药物组合物
DE102005015253A1 (de) 2005-04-04 2006-10-05 Merck Patent Gmbh Pyrazolderivate
US20090306020A1 (en) 2005-05-27 2009-12-10 Bayer Healthcare Ag Combination therapy comprising diaryl ureas for treating diseases
CA2609389A1 (fr) 2005-05-27 2006-11-30 Bayer Healthcare Ag Traitement de combinaison comprenant un compose diaryluree et des inhibiteurs de pi3- ou akt-kinase ou de mtor (rapamycines) pour le traitement du cancer
WO2007015947A2 (fr) 2005-07-29 2007-02-08 Bayer Healthcare Llc Methodes et trousses pour la prediction du succes therapeutique, de la survie sans recidive et globale dans des therapies du cancer
DE602006019262D1 (de) 2005-08-11 2011-02-10 Siemens Healthcare Diagnostics QUANTITATIVE ASSAYS FÜR PDGFR-beta- IN KÖRPERFLÜSSIGKEITEN
SI1928872T1 (sl) 2005-09-21 2012-06-29 4Sc Ag Novi sulfonilpiroli kot inhibitorji HDAC
BRPI0616040A2 (pt) 2005-09-21 2011-06-07 Nycomed Gmbh cloridrato de sulfonilpirról como inibidor de histona desacetilases
WO2007047955A2 (fr) 2005-10-21 2007-04-26 Bayer Healthcare Llc Methodes de prevision et de pronostic du cancer, et surveillance d'une therapie anticancereuse
US8329408B2 (en) 2005-10-31 2012-12-11 Bayer Healthcare Llc Methods for prognosis and monitoring cancer therapy
CN101351709A (zh) 2005-11-02 2009-01-21 拜尔健康护理有限责任公司 癌症的预测和预后以及监控癌症治疗的方法
WO2007056012A2 (fr) 2005-11-02 2007-05-18 Bayer Healthcare Llc Methodes pour la prevision et le pronostic du cancer, et pour la surveillance de therapie anticancereuse
BRPI0618597A2 (pt) 2005-11-14 2011-09-06 Bayer Healthcare Llc métodos de predição e prognóstico de cáncer e método de monitoramento de terapia contra o cáncer
WO2007059155A1 (fr) 2005-11-14 2007-05-24 Bayer Pharmaceuticals Corporation Traitement de cancers a resistance a des agents chimiotherapeutiques
US20090227637A1 (en) 2005-12-15 2009-09-10 Olaf Weber Diaryl ureas for treating virus infections
WO2007087575A2 (fr) 2006-01-24 2007-08-02 University Of Chicago Compositions et procedes destines au traitement de l'hypertension pulmonaire
BRPI0710073A2 (pt) 2006-02-22 2011-08-02 4Sc Ag indolopiridinas
KR101415354B1 (ko) 2006-02-22 2014-07-04 4에스체 악티엔게젤샤프트 Eg5 키네신 조절제로서의 인돌로피리딘
US7908091B2 (en) 2006-03-17 2011-03-15 Prometheus Laboratories Inc. Methods of predicting and monitoring tyrosine kinase inhibitor therapy
CA2648106A1 (fr) 2006-03-31 2007-11-01 Bayer Healthcare Llc Procedes de prevision et de pronostic de cancer, et surveillance d'une therapie anticancereuse
WO2007139930A2 (fr) 2006-05-26 2007-12-06 Bayer Healthcare Llc Associations de médicaments comportant des urées de diaryle substituées pour le traitement du cancer
CA2667720A1 (fr) 2006-11-09 2008-05-15 Abbott Gmbh & Co. Kg. Forme de dosage pharmaceutique pour l'administration orale d'un inhibiteur de la tyrosine kinase
EP2111401B1 (fr) 2006-12-20 2011-02-16 Bayer HealthCare, LLC 4- {4-[({3- tert- butyl-1- [3- (hydroxymethyl) phenyl]- 1h- pyrazol- 5- yl } carbamoyl) -amino]- 3- fluorophenoxy} - n- methylpyridine- 2- carboxamide ainsi que des promédicaments et des sels de ceux-ci utilisés pour traiter un cancer
CA2698795C (fr) 2007-09-10 2016-04-19 Cipla Limited Procede de preparation d'un inhibiteur de la kinase raf et intermediaires utilisables dans ledit procede
WO2009054004A2 (fr) 2007-10-22 2009-04-30 Natco Pharma Limited Nouveau procédé pour la préparation du sorafénib
JP5432468B2 (ja) 2008-04-10 2014-03-05 光洋サーモシステム株式会社 横型炉装置
EP2341777B1 (fr) 2008-11-07 2015-01-21 NovaBay Pharmaceuticals, Inc. Compositions antimicrobiennes d'oxazolidinone, d'hydantoïne et d'imidazolidinone
CN102334367B (zh) 2009-04-16 2014-04-30 上海贝尔股份有限公司 多基站mimo系统中选择对象终端的方法和装置
JP5821626B2 (ja) 2011-12-27 2015-11-24 富士ゼロックス株式会社 画像形成装置及びプログラム

Patent Citations (99)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US502504A (en) * 1893-08-01 Hermann thoms
US1792156A (en) * 1928-01-17 1931-02-10 Gen Aniline Works Inc 5-halogen-2-amino-1-alkyloxy and 1-aralkyloxy-benzenes and intermediate products thereof and process of preparing them
US2093265A (en) * 1931-03-31 1937-09-14 Ici Ltd Process for the manufacture of diaryl ureas
US2046375A (en) * 1931-06-04 1936-07-07 Ici Ltd p-halogen-omicron-alkoxy-aniline derivatives and process of preparing the same
US2288422A (en) * 1938-11-11 1942-06-30 Gen Aniline & Film Corp Mixed ureas
US2973386A (en) * 1943-01-05 1961-02-28 Harry A Weldon Purification of sym dichloro-bis (2, 4, 6-trichlorophenyl)urea
US2649476A (en) * 1950-04-29 1953-08-18 Variapat Ag Trifluoromethylated diphenyl ether sulfonic acids
US2683082A (en) * 1950-12-09 1954-07-06 Ethyl Corp Nu-aryl-nu'-(p-hydroxyphenyl) ureas as antioxidants for petroleum hydrocarbon fuels
US2722544A (en) * 1950-12-26 1955-11-01 Variapat Ag Trifluoromethyl halogenated diphenylcarbamide sulfonic acids and their preparation
US2781330A (en) * 1953-02-09 1957-02-12 Monsanto Chemicals Rubber containing urea compound as an anti-exposure cracking agent
US2797214A (en) * 1953-03-06 1957-06-25 Geigy Ag J R Tetrakisazo dyestuffs
US2745874A (en) * 1953-06-18 1956-05-15 Geigy Ag J R Insecticidal derivatives of diphenyl urea
US2867659A (en) * 1953-12-22 1959-01-06 Geigy Ag J R Polyhalogen substituted monohydroxydiphenyl urea and thiourea compounds
US2877268A (en) * 1956-12-24 1959-03-10 Monsanto Chemicals Substituted ureas
US2960488A (en) * 1958-04-25 1960-11-15 Eastman Kodak Co Poly-alpha-olefins containing substituted ureas
US3230141A (en) * 1959-08-14 1966-01-18 Geigy Ag J R Method for protecting fibers against attack by insects and bacteria with diphenyl urea compositions
US3151023A (en) * 1961-04-21 1964-09-29 Ciba Ltd Preparations for combating phytopathogenic microorganisms
US3200035A (en) * 1962-06-01 1965-08-10 Ciba Ltd Treatment of synthetic products, especially synthetic fibers
US3284433A (en) * 1963-07-17 1966-11-08 Merck & Co Inc 4-phenoxy-carbanilides
US3424760A (en) * 1966-03-07 1969-01-28 Robins Co Inc A H 3-ureidopyrrolidines
US3424762A (en) * 1966-03-07 1969-01-28 Robins Co Inc A H Certain 3-ureidopyrrolidines
US3424761A (en) * 1966-03-07 1969-01-28 Robins Co Inc A H 3-ureidopyrrolidines
US3743498A (en) * 1967-10-31 1973-07-03 Du Pont Method of selectively controlling undesirable vegetation
US3689550A (en) * 1968-03-21 1972-09-05 Ciba Geigy Ag N-hydroxyphenyl-n{40 -phenylureas
US3639668A (en) * 1968-04-19 1972-02-01 Bayer Ag N-(2 2 4 4-tetrafluoro-1 3-benzodioxanyl)-ureas
US3646059A (en) * 1969-05-05 1972-02-29 Du Pont Plant growth regulatory ureidopyrazoles
US3754887A (en) * 1969-05-05 1973-08-28 Du Pont Ureidopyrazoles defoliants
US3668222A (en) * 1969-05-14 1972-06-06 Sandoz Ltd 11-desacetoxy-wortmannin
US3828001A (en) * 1969-08-14 1974-08-06 May & Baker Ltd Thiophene derivatives
US3823161A (en) * 1970-05-07 1974-07-09 Exxon Research Engineering Co Aminothiophene derivatives
US3860645A (en) * 1973-05-23 1975-01-14 Givaudan Corp Bacteriostatic substituted carbanilides
US4111680A (en) * 1973-07-27 1978-09-05 Shionogi & Co., Ltd. Herbicidal compositions containing 3-isoxazolylurea derivatives
US4212981A (en) * 1973-07-27 1980-07-15 Shionogi & Co., Ltd. Process for preparing 3-isoxazolylurea derivatives
US4116671A (en) * 1973-07-27 1978-09-26 Shionogi & Co., Ltd. 3-Isoxazolylcarbamate derivatives
US4001256A (en) * 1973-12-26 1977-01-04 The Upjohn Company Pyridylalkyl phenyl ureas and their n-oxides
US4009847A (en) * 1974-04-17 1977-03-01 E. I. Du Pont De Nemours And Company 1-Tertiary-alkyl-3-(substituted thienyl)ureas and 1-tertiary-alkyl-3-(substituted thietyl)ureas as antihypertensive agents
US3990879A (en) * 1974-12-26 1976-11-09 Eli Lilly And Company Method of controlling aquatic weeds
US4111683A (en) * 1975-06-27 1978-09-05 Chevron Research Company N-alkyl or alkoxy-N'-substituted hydrocarbyl urea
US4103022A (en) * 1976-08-24 1978-07-25 Bayer Aktiengesellschaft Combating arthropods with 3-(2,2,4,4-tetrafluoro-benz-1,3-dioxin-6-yl)-1-(substituted benzoyl)-ureas
US4173638A (en) * 1976-10-29 1979-11-06 Ishihara Sangyo Kaisha Ltd. N-Benzoyl-N'-pyridyloxy phenyl urea and insecticidal compositions thereof
US4173637A (en) * 1976-10-29 1979-11-06 Ishihara Sangyo Kaisha Ltd. N-Benzoyl-N'-pyridyloxy phenyl urea and insecticidal compositions thereof
US4071524A (en) * 1976-11-08 1978-01-31 Riker Laboratories, Inc. Derivatives of urea
US4183854A (en) * 1976-11-10 1980-01-15 John Wyeth & Brother Limited Thiazole compound
US4042372A (en) * 1976-11-19 1977-08-16 Eli Lilly And Company Substituted thiadiazolotriazinediones and method of preparation
US4279639A (en) * 1978-11-02 1981-07-21 Toshihiko Okamoto N-(2-Substituted-4-pyridyl)ureas and thioureas as well as plant growth regulators containing same, and method for using compounds as plant growth regulators
US4358596A (en) * 1978-11-03 1982-11-09 Schering Ag Process for the preparation of 1,2,3-thiadiazol-5-yl ureas
US4546191A (en) * 1979-03-19 1985-10-08 Ishihara Sangyo Kaisha Ltd. Trifluoromethyl-2-pyridinone or pyridinthione compounds and process for the preparation of the same
US4405644A (en) * 1979-07-14 1983-09-20 Bayer Aktiengesellschaft Medicaments for the treatment of disorders of lipometabolism and their use
US4468380A (en) * 1979-12-26 1984-08-28 Eli Lilly And Company Anticoccidial combinations comprising polyether antibiotics and carbanilides
US4410697A (en) * 1980-01-25 1983-10-18 Reanal Finomvegyszergyar Process for the preparation of N-aryl-N'-(mono- or di substituted)-urea derivatives
US4526997A (en) * 1981-05-06 1985-07-02 Doherty George O P O Anticoccidial combinations comprising polyether antibiotics and carbanilides
US4511571A (en) * 1981-10-20 1985-04-16 Ciba Geigy Corporation N-(2-Pyridyloxyphenyl)-N'-benzoyl ureas, pesticidal compositions containing same and pesticidal methods of use
US4473579A (en) * 1982-01-26 1984-09-25 American Cyanamid Company Antiatherosclerotic tetrasubstituted ureas and thioureas
US4437878A (en) * 1982-03-31 1984-03-20 Basf Aktiengesellschaft Dihydrothiophenecarboxylates and their use for controlling undersirable plant growth
US4514571A (en) * 1982-05-25 1985-04-30 Ube Industries, Ltd. Process for the preparation of urea derivatives
US4643849A (en) * 1982-11-12 1987-02-17 Toyama Chemical Co., Ltd. Intermediates for urea and thiourea derivatives
US4499097A (en) * 1983-03-10 1985-02-12 American Cyanamid Company 2-(Pyridyl)imidazolyl ketones
US4587240A (en) * 1983-09-23 1986-05-06 National Research Development Corp. Pharmaceutical compositions
US4540566A (en) * 1984-04-02 1985-09-10 Forest Laboratories, Inc. Prolonged release drug dosage forms based on modified low viscosity grade hydroxypropylmethylcellulose
US4677111A (en) * 1984-10-12 1987-06-30 Ishihara Sangyo Kaisha Ltd. N-benzoyl-N'-(3-nitrophenyl) urea compounds, and antitumorous compositions containing them
US5773459A (en) * 1995-06-07 1998-06-30 Sugen, Inc. Urea- and thiourea-type compounds
US20100063088A1 (en) * 1997-05-23 2010-03-11 Wood Jill E Raf Kinase Inhibitors
US20100160371A1 (en) * 1997-05-23 2010-06-24 Gerald Ranges Inhibition of p38 kinase activity by aryl ureas
US20120129893A1 (en) * 1997-12-22 2012-05-24 Jacques Dumas Inhibition Of Raf Kinase Using Substituted Heterocyclic Ureas
US20120046290A1 (en) * 1997-12-22 2012-02-23 Jacques Dumas Inhibition of p38 kinase activity using substituted heterocyclic ureas
US20090093526A1 (en) * 1997-12-22 2009-04-09 Scott Miller Inhibition of p38 kinase using symmetrical and unsymmetrical diphenyl ureas
US20090118268A1 (en) * 1999-01-13 2009-05-07 Bernd Riedl OMEGA-CARBOXY ARYL SUBSTITUTED DIPHENYL UREAS AS p38 KINASE INHIBITORS
US20030211466A1 (en) * 1999-12-28 2003-11-13 Ribonomics, Inc. Methods for identifying functionally related genes and drug targets
US7833698B2 (en) * 2000-01-12 2010-11-16 Ventana Medical Systems, Inc. Method for determining the response to cancer therapy
US7411055B2 (en) * 2001-05-25 2008-08-12 Immunex Corporation Attractin/mahogany-like polypeptides, polynucleotides, antibodies and methods of use thereof
US20120040925A1 (en) * 2001-12-03 2012-02-16 Carter Christopher A Aryl urea compounds in combination with other cytostatic or cytotoxic agents for treating human cancers
US20120040986A1 (en) * 2002-01-11 2012-02-16 Bernd Riedl Omega carboxyaryl substituted diphenyl ureas as raf kinase inhibitors
US20110015195A1 (en) * 2002-02-11 2011-01-20 Jacques Dumas Aryl ureas with angiogenisis inhibiting activity
US20030216396A1 (en) * 2002-02-11 2003-11-20 Bayer Corporation Pyridine, quinoline, and isoquinoline N-oxides as kinase inhibitors
US20100125139A1 (en) * 2002-03-22 2010-05-20 Donald Bankston Process for preparing quinazoline rho-kinase inhibitors and intermediates thereof
US20100075971A1 (en) * 2003-02-28 2010-03-25 Jacques Dumas Substituted pyridine derivatives useful in the treatment of cancer and other disorders
US20090192127A1 (en) * 2003-07-23 2009-07-30 Bayer Healthcare Ag Combination therapy comprising a diaryl urea compound and a p13, akt kinase or mtor inhibitors (rapamycins) for cancer treatment
US20110136809A1 (en) * 2004-04-30 2011-06-09 Bayer Pharmaceuticals Corporation Substituted Pyrazolyl Urea Derivatives Useful In The Treatment Of Cancer
US20120142741A1 (en) * 2004-08-27 2012-06-07 Fritz Schueckler Pharmaceutical compositions for the treatment of cancer
US20090215833A1 (en) * 2004-09-29 2009-08-27 Bayer Healthcare Ag Thermodynamically stable form of a tosylate salt
US20090215835A1 (en) * 2005-10-31 2009-08-27 Scott Wilhelm Treatment of cancer with sorafenib
US20090068146A1 (en) * 2005-10-31 2009-03-12 Scott Wilhelm Diaryl ureas and combinations
US20090176791A1 (en) * 2005-11-10 2009-07-09 Bayer Healthcare Ag Diaryl Ureas for Treating Pulmonary Hypertension
US20100035888A1 (en) * 2005-11-10 2010-02-11 Bater Healthcare AG Diaryl Urea for Treating Pulmonary Hypertension
US20100144749A1 (en) * 2005-11-14 2010-06-10 Scott Wilhelm Treatment of cancers with acquired resistance to kit inhibitors
US20110195110A1 (en) * 2005-12-01 2011-08-11 Roger Smith Urea compounds useful in the treatment of cancer
US20120009150A1 (en) * 2005-12-15 2012-01-12 Olaf Weber Diaryl ureas for treating virus infections
US20110158942A1 (en) * 2005-12-15 2011-06-30 Olaf Weber Diaryl ureas for treating virus infections
US20100129321A1 (en) * 2005-12-15 2010-05-27 Bayer Healthcare Llc Diaryl urea for treating virus infections
US20100081812A1 (en) * 2005-12-21 2010-04-01 Roger Smith Substituted pyrimidine derivatives useful in the treatment of cancer and other disorders
US20100173953A1 (en) * 2006-10-11 2010-07-08 Alfons Grunenberg 4-[4-(amino)-3-fluorophenoxy]-N-methylpyridine-2-carboxamide monohydrate
US20100063112A1 (en) * 2006-11-09 2010-03-11 Bayer Schering Pharma Aktiengesellschaft Polymorph iii of 4-[4-(amino)-3-fluorophenoxy]-n-methylpyridine-2-carboxamide
US20100113533A1 (en) * 2006-11-14 2010-05-06 Bayer Schering Pharma Aktiengesellschaft Polymorph II of 4-[4-(Amino)-3- Fluorophenoxy]-N-Methylpyridine-2-Carboxamide
US20100150863A1 (en) * 2006-12-20 2010-06-17 Bayer Pharmaceuticals Corporation Hydroxy methyl phenyl pyrazolyl urea compound useful in treatment of cancer
US20100267777A1 (en) * 2007-01-19 2010-10-21 Bayer Pharmaceuticals Corporation Treatment of cancers with acquired resistance to kit inhibitors
US20100173954A1 (en) * 2007-01-19 2010-07-08 Bayer Healthcare Llc Treatment of cancers having resistance to chemotherapeutic agents
US20130142742A1 (en) * 2008-04-02 2013-06-06 Nicolas Thiebault Methods of Inhibiting Microbial Growth Using Deoxyaldohexoses
US20110178137A1 (en) * 2008-06-25 2011-07-21 Bayer Schering Pharma Aktiengesellschaft Diaryl urea for treating heart failure
US20110257035A1 (en) * 2008-10-21 2011-10-20 Bayer Healthcare Llc Identification of signature genes associated with hepatocellular carcinoma

Non-Patent Citations (8)

* Cited by examiner, † Cited by third party
Title
Chen et al, Molecular & Cellular Proteomics, 2002, Vol. 1, pp. 304-313 *
Greenbuam et al, Genome Biology, 2003, Vol. 4, article 117, 8-pages. *
Holder et al, Annals of Surgery, 1998, Vol. 272, pp. 764-771 *
Ide et al (Lung Cancer, 2010, vol. 67, pp. 248-250) *
Kurahara et al (Journal of Surgical Oncology, 2010, Vol. 102, pp. 809-815) *
Nagahashi et al (World Journal of Gastroenterology, 2010, Vol. 16, pp. 4003-4012) *
Thelen et al (Ann Surg Oncol, 2009, Vol. 16, pp. 1222-1230) *
Zhang et al (Human Pathology, 2011, Vol. 42, pp. 1275-1282). *

Cited By (44)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030181442A1 (en) * 1999-01-13 2003-09-25 Bayer Corporation omega-carboxyaryl substituted diphenyl ureas as raf kinase inhibitors
US7897623B2 (en) 1999-01-13 2011-03-01 Bayer Healthcare Llc ω-carboxyl aryl substituted diphenyl ureas as p38 kinase inhibitors
US8124630B2 (en) 1999-01-13 2012-02-28 Bayer Healthcare Llc ω-carboxyaryl substituted diphenyl ureas as raf kinase inhibitors
US8841330B2 (en) 1999-01-13 2014-09-23 Bayer Healthcare Llc Omega-carboxyaryl substituted diphenyl ureas as raf kinase inhibitors
US20080027061A1 (en) * 1999-01-13 2008-01-31 Bernd Riedl omega-Carboxy Aryl Substituted Diphenyl Ureas As p38 Kinase Inhibitors
US20080194580A1 (en) * 2001-04-20 2008-08-14 Jacques Dumas Inhibition Of Raf Kinase Using Quinolyl, Isoquinolyl Or Pyridyl Ureas
US20080108672A1 (en) * 2002-01-11 2008-05-08 Bernd Riedl Omega-Carboxyaryl Substituted Diphenyl Ureas As Raf Kinase Inhibitors
US8618141B2 (en) 2002-02-11 2013-12-31 Bayer Healthcare Llc Aryl ureas with angiogenesis inhibiting activity
US7838541B2 (en) 2002-02-11 2010-11-23 Bayer Healthcare, Llc Aryl ureas with angiogenesis inhibiting activity
US20080227828A1 (en) * 2002-02-11 2008-09-18 Jacques Dumas Aryl Ureas With Angiogenisis Inhibiting Activity
US8242147B2 (en) 2002-02-11 2012-08-14 Bayer Healthcare Llc Aryl ureas with angiogenisis inhibiting activity
US20030207870A1 (en) * 2002-02-11 2003-11-06 Bayer Corporation Aryl ureas with angiogenisis inhibiting activity
US20110015195A1 (en) * 2002-02-11 2011-01-20 Jacques Dumas Aryl ureas with angiogenisis inhibiting activity
US20080105264A1 (en) * 2003-02-21 2008-05-08 Resmed Limited Nasal assembly
US20100075971A1 (en) * 2003-02-28 2010-03-25 Jacques Dumas Substituted pyridine derivatives useful in the treatment of cancer and other disorders
US8076488B2 (en) 2003-02-28 2011-12-13 Bayer Healthcare Llc Bicyclic urea derivatives useful in the treatment of cancer and other disorders
US8796250B2 (en) 2003-05-20 2014-08-05 Bayer Healthcare Llc Diaryl ureas for diseases mediated by PDGFR
US20050059703A1 (en) * 2003-05-20 2005-03-17 Scott Wilhelm Diaryl ureas for diseases mediated by PDGFR
US8637553B2 (en) 2003-07-23 2014-01-28 Bayer Healthcare Llc Fluoro substituted omega-carboxyaryl diphenyl urea for the treatment and prevention of diseases and conditions
US20050038080A1 (en) * 2003-07-23 2005-02-17 Stephen Boyer Fluoro substituted omega-carboxyaryl diphenyl urea for the treatment and prevention of diseases and conditions
US8877933B2 (en) 2004-09-29 2014-11-04 Bayer Intellectual Property Gmbh Thermodynamically stable form of a tosylate salt
US20090215833A1 (en) * 2004-09-29 2009-08-27 Bayer Healthcare Ag Thermodynamically stable form of a tosylate salt
US20080242707A1 (en) * 2005-03-07 2008-10-02 Bayer Healthcare Ag Pharmaceutical Composition for the Treatment of Cancer
US9737488B2 (en) 2005-03-07 2017-08-22 Bayer Healthcare Llc Pharmaceutical composition for the treatment of cancer
US20070105142A1 (en) * 2005-10-31 2007-05-10 Scott Wilhelm Methods for prognosis and monitoring cancer therapy
US8329408B2 (en) * 2005-10-31 2012-12-11 Bayer Healthcare Llc Methods for prognosis and monitoring cancer therapy
US9957232B2 (en) 2006-10-11 2018-05-01 Bayer Healthcare Llc 4-[4-({[4-chloro-3-(trifluoromethyl)phenyl]carbamoyl}amino)-3-fluorophenoxy]-N-methylpyridine-2-carboxamide monohydrate
US20100173953A1 (en) * 2006-10-11 2010-07-08 Alfons Grunenberg 4-[4-(amino)-3-fluorophenoxy]-N-methylpyridine-2-carboxamide monohydrate
US20080308776A1 (en) * 2007-06-14 2008-12-18 Bindra Perminder S Chemiluminescent compositions and methods of making and using thereof
US7674406B2 (en) * 2007-06-14 2010-03-09 Bindra Perminder S Chemiluminescent compositions and methods of making and using thereof
US20100185419A1 (en) * 2008-09-05 2010-07-22 Avila Therapeutics, Inc. Algorithm for designing irreversible inhibitors
US8658066B2 (en) 2009-05-12 2014-02-25 Mega Glow, Llc Chemiluminescent powders and methods of making and using thereof
US7964119B2 (en) 2009-05-12 2011-06-21 Mega Glow, LLC. Chemiluminescent powders and methods of making and using thereof
US20100288984A1 (en) * 2009-05-12 2010-11-18 Bindra Perminder S Chemiluminescent powders and methods of making and using thereof
US20110117073A1 (en) * 2009-09-16 2011-05-19 Avila Therapeutics, Inc. Protein Kinase Conjugates and Inhibitors
US9556426B2 (en) 2009-09-16 2017-01-31 Celgene Avilomics Research, Inc. Protein kinase conjugates and inhibitors
US10662195B2 (en) 2009-09-16 2020-05-26 Celgene Car Llc Protein kinase conjugates and inhibitors
US11542492B2 (en) 2009-12-30 2023-01-03 Celgene Car Llc Ligand-directed covalent modification of protein
US9458107B2 (en) 2010-04-15 2016-10-04 Bayer Intellectual Property Gmbh Process for the preparation of 4-{4-[({[4 chloro-3-(trifluoromethyl)-phenyl]amino}carbonyl)amino]-3-fluorphenoxy-N-ethylpyridie-carboxamide, its salts and monohydrate
US10822305B2 (en) 2010-04-15 2020-11-03 Bayer Healthcare Llc Process for the preparation of 4-(4-amino-3-fluorophenoxy)-N-methylpyyridine-2-carboxamide
US20130225641A1 (en) * 2010-05-18 2013-08-29 Banghe Pharmaceutical Co., Ltd. Anticancer compounds and preparation methods thereof
WO2018035346A1 (fr) * 2016-08-17 2018-02-22 Ichan School Of Medicine At Mount Sinal Composés inhibiteurs de kinase, compositions et méthodes de traitement du cancer
US10519113B2 (en) 2016-08-17 2019-12-31 Icahn School Of Medicine At Mount Sinai Kinase inhibitor compounds, compositions, and methods of treating cancer
US11066370B2 (en) 2016-08-17 2021-07-20 Icahn School Of Medicine At Mount Sinai Kinase inhibitor compounds, compositions, and methods of treating cancer

Also Published As

Publication number Publication date
ES2288694T3 (es) 2008-01-16
EP1636585A2 (fr) 2006-03-22
CA2526636A1 (fr) 2005-01-06
WO2005000284A2 (fr) 2005-01-06
PT1626714E (pt) 2007-08-24
WO2004113274A2 (fr) 2004-12-29
DE602004011340T2 (de) 2008-11-06
CA2526617C (fr) 2015-04-28
JP2007511203A (ja) 2007-05-10
CA2526636E (fr) 2005-01-06
WO2004113274A3 (fr) 2005-03-03
PT1636585E (pt) 2008-03-27
DK1636585T3 (da) 2008-05-26
ES2305808T3 (es) 2008-11-01
CA2526636C (fr) 2012-10-02
DE602004011340D1 (de) 2008-03-06
ATE366108T1 (de) 2007-07-15
US8796250B2 (en) 2014-08-05
PL1636585T3 (pl) 2008-10-31
EP1636585B2 (fr) 2012-06-13
MXPA05012486A (es) 2006-07-03
US20050059703A1 (en) 2005-03-17
ATE384264T1 (de) 2008-02-15
JP4860474B2 (ja) 2012-01-25
PL1626714T3 (pl) 2007-12-31
DE602004007382T2 (de) 2008-04-17
DE602004007382D1 (de) 2007-08-16
EP1626714B1 (fr) 2007-07-04
WO2005000284A3 (fr) 2005-03-10
EP1626714A2 (fr) 2006-02-22
CA2526617A1 (fr) 2004-12-29
DK1626714T3 (da) 2007-10-15
EP1636585B1 (fr) 2008-01-16
JP2006528986A (ja) 2006-12-28

Similar Documents

Publication Publication Date Title
EP1636585B1 (fr) Diaryl-urees presentant une activite d'inhibition des kinases
EP2111401B1 (fr) 4- {4-[({3- tert- butyl-1- [3- (hydroxymethyl) phenyl]- 1h- pyrazol- 5- yl } carbamoyl) -amino]- 3- fluorophenoxy} - n- methylpyridine- 2- carboxamide ainsi que des promédicaments et des sels de ceux-ci utilisés pour traiter un cancer
EP1663978B1 (fr) Omega-carboxyaryldiphenyluree fluoro-subtituee pour le traitement et la prevention de maladies et d'etats pathologiques
EP1599467B1 (fr) Nouveaux derives de cyanopyridine utiles dans le traitement du cancer et d'autres troubles
US20110195110A1 (en) Urea compounds useful in the treatment of cancer
EP2134677B1 (fr) 4-{4-[({3-tert-butyl-1-[3-(hydroxyméthyl)phényl]-1h-pyrazol-5-yl}carbamoyl)-amino]-3-chlorophénoxy}-n-méthylpyridine-2-carboxamide en tant que inhibiteur de la kinase vegfr pour le traitement du cancer
EP1973897B1 (fr) Dérivés substitués de la pyrimidine utilisables dans le traitement du cancer et d'autres maladies
MXPA05012491A (en) Diaryl ureas with kinase inhibiting activity
AU2013200394B2 (en) Fluoro substituted omega-carboxyaryl diphenyl urea for the treatment and prevention of diseases and conditions
ZA200600609B (en) Fluoro substituted omega-carboxyaryl diphenyl urea for the treatment and prevention of diseases and conditions
MXPA06000860A (en) Fluoro substituted omega-carboxyaryl diphenyl urea for the treatment and prevention of diseases and conditions

Legal Events

Date Code Title Description
AS Assignment

Owner name: BAYER PHARMACEUTICALS CORPORATION, CONNECTICUT

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:WHILHELM, SCOTT;DUMAS, JAXQUES;LADOCUEUR, GAETAN;AND OTHERS;REEL/FRAME:018170/0725;SIGNING DATES FROM 20060314 TO 20060323

AS Assignment

Owner name: BAYER HEALTHCARE LLC, NEW YORK

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:BAYER PHARMACEUTICALS CORPORATION;REEL/FRAME:021893/0806

Effective date: 20071113

AS Assignment

Owner name: BAYER HEALTHCARE LLC, NEW YORK

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:BAYER PHARMACEUTICALS CORPORATION;REEL/FRAME:023031/0963

Effective date: 20071219

Owner name: BAYER HEALTHCARE LLC,NEW YORK

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:BAYER PHARMACEUTICALS CORPORATION;REEL/FRAME:023031/0963

Effective date: 20071219

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION