WO2005030140A2 - C-met modulators and methods of use - Google Patents

C-met modulators and methods of use Download PDF

Info

Publication number
WO2005030140A2
WO2005030140A2 PCT/US2004/031523 US2004031523W WO2005030140A2 WO 2005030140 A2 WO2005030140 A2 WO 2005030140A2 US 2004031523 W US2004031523 W US 2004031523W WO 2005030140 A2 WO2005030140 A2 WO 2005030140A2
Authority
WO
WIPO (PCT)
Prior art keywords
optionally substituted
phenyl
fluoro
methoxy
alkyl
Prior art date
Application number
PCT/US2004/031523
Other languages
French (fr)
Other versions
WO2005030140A3 (en
Inventor
Lynne Canne Bannen
Diva Sze-Ming Chan
Jeff Chen
Lisa Esther Dalrymple
Timothy Patrick Forsyth
Tai Phat Huynh
Vasu Jammalamadaka
Richard George Khoury
James William Leahy
Morrison B. Mac
Grace Mann
Larry W. Mann
John M. Nuss
Jason Jevious Parks
Craig Stacy Takeuchi
Yong Wang
Wei Xu
Original Assignee
Exelixis, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=34397024&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=WO2005030140(A2) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Priority to EP04789057A priority Critical patent/EP1673085B1/en
Application filed by Exelixis, Inc. filed Critical Exelixis, Inc.
Priority to AU2004275842A priority patent/AU2004275842B2/en
Priority to EP17190013.7A priority patent/EP3329918B1/en
Priority to CA2537812A priority patent/CA2537812C/en
Priority to JP2006528265A priority patent/JP4638436B2/en
Priority to AT04789057T priority patent/ATE532782T1/en
Publication of WO2005030140A2 publication Critical patent/WO2005030140A2/en
Publication of WO2005030140A3 publication Critical patent/WO2005030140A3/en
Priority to US11/586,751 priority patent/US9174947B2/en
Priority to US11/753,503 priority patent/US8476298B2/en
Priority to US11/753,514 priority patent/US8067436B2/en
Priority to US11/753,462 priority patent/US8178532B2/en
Priority to US12/393,806 priority patent/US7579473B2/en
Priority to AU2010204461A priority patent/AU2010204461B2/en
Priority to AU2010204459A priority patent/AU2010204459B2/en
Priority to US12/911,442 priority patent/US20110077233A1/en
Priority to US13/249,815 priority patent/US8497284B2/en
Priority to US13/427,093 priority patent/US20120184523A1/en
Priority to US13/888,592 priority patent/US20140155378A9/en
Priority to US13/928,977 priority patent/US20140155396A1/en
Priority to US14/847,801 priority patent/US20150376133A1/en
Priority to US15/066,872 priority patent/US20160185725A1/en
Priority to US15/688,310 priority patent/US11124482B2/en
Priority to US16/921,542 priority patent/US20200331860A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D215/00Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems
    • C07D215/02Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom
    • C07D215/16Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D215/20Oxygen atoms
    • C07D215/22Oxygen atoms attached in position 2 or 4
    • C07D215/233Oxygen atoms attached in position 2 or 4 only one oxygen atom which is attached in position 4
    • 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
    • A61K31/47Quinolines; Isoquinolines
    • 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
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/4709Non-condensed quinolines and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic 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
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/472Non-condensed isoquinolines, e.g. papaverine
    • A61K31/4725Non-condensed isoquinolines, e.g. papaverine containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/496Non-condensed piperazines containing further heterocyclic rings, e.g. rifampin, thiothixene
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/50Pyridazines; Hydrogenated pyridazines
    • A61K31/501Pyridazines; Hydrogenated pyridazines not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/517Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with carbocyclic ring systems, e.g. quinazoline, perimidine
    • 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
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/02Drugs 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
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • 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
    • 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
    • A61P37/06Immunosuppressants, 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
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D215/00Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems
    • C07D215/02Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom
    • C07D215/16Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D215/20Oxygen atoms
    • C07D215/22Oxygen atoms attached in position 2 or 4
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D215/00Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems
    • C07D215/02Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom
    • C07D215/16Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D215/36Sulfur atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D215/00Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems
    • C07D215/02Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom
    • C07D215/16Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D215/38Nitrogen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D215/00Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems
    • C07D215/02Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom
    • C07D215/16Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D215/38Nitrogen atoms
    • C07D215/42Nitrogen atoms attached in position 4
    • C07D215/46Nitrogen atoms attached in position 4 with hydrocarbon radicals, substituted by nitrogen atoms, attached to said nitrogen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/70Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings condensed with carbocyclic rings or ring systems
    • C07D239/72Quinazolines; Hydrogenated quinazolines
    • C07D239/86Quinazolines; Hydrogenated quinazolines with hetero atoms directly attached in position 4
    • C07D239/88Oxygen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/70Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings condensed with carbocyclic rings or ring systems
    • C07D239/72Quinazolines; Hydrogenated quinazolines
    • C07D239/86Quinazolines; Hydrogenated quinazolines with hetero atoms directly attached in position 4
    • C07D239/94Nitrogen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D295/00Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms
    • C07D295/04Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms with substituted hydrocarbon radicals attached to ring nitrogen atoms
    • C07D295/14Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms with substituted hydrocarbon radicals attached to ring nitrogen atoms substituted by carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals
    • C07D295/145Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms with substituted hydrocarbon radicals attached to ring nitrogen atoms substituted by carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals with the ring nitrogen atoms and the carbon atoms with three bonds to hetero atoms attached to the same carbon chain, which is not interrupted by carbocyclic rings
    • C07D295/15Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms with substituted hydrocarbon radicals attached to ring nitrogen atoms substituted by carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals with the ring nitrogen atoms and the carbon atoms with three bonds to hetero atoms attached to the same carbon chain, which is not interrupted by carbocyclic rings to an acyclic saturated chain
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/14Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/12Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
    • C07D417/12Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/48Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving transferase
    • C12Q1/485Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving transferase involving kinase
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2500/00Screening for compounds of potential therapeutic value
    • G01N2500/04Screening involving studying the effect of compounds C directly on molecule A (e.g. C are potential ligands for a receptor A, or potential substrates for an enzyme A)
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • Protein kinases are enzymes that catalyze the phosphorylation of proteins, in particular, hydroxy groups on tyrosine, serine and threonine residues of proteins.
  • the consequences of this seemingly simple activity are staggering; cell differentiation and proliferation; i.e., virtually all aspects of cell life in one-way or another depend on protein kinase activity.
  • abnormal protein kinase activity has been related to a host of disorders, ranging from relatively non-life threatening diseases such as psoriasis to extremely virulent diseases such as glioblastoma (brain cancer).
  • the non-receptor type of tyrosine kinases is also comprised of numerous subfamilies, including Src, Frk, Btk, Csk, Abl, Zap70, Fes/Fps, Fak, Jak, Ack, and LLMK. Each of these subfamilies is further sub-divided into varying receptors.
  • the Src subfamily is one of the largest and includes Src, Yes, Fyn, Lyn, Lck, Blk, Hck, Fgr, and Yrk.
  • the Src subfamily of enzymes has been linked to oncogenesis.
  • Tumor growth progression requires the recruitment of new blood vessels into the tumor from preexisting vessels as well as invasion, adhesion and proliferation of malignant cells. Accordingly, c-Met overexpression has been demonstrated on a wide variety of tumor types including breast, colon, renal, lung, squamous cell myeloid leukemia, hemangiomas, melanomas, astrocytomas, and glioblastomas. Additionally activating mutations in the kinase domain of c-Met have been identified in hereditary and sporadic renal papilloma and squamous cell carcinoma.
  • EGF EGF
  • VEGF vascular endothelial growth factor
  • ephrin signal transduction will prevent cell proliferation and angiogenesis, two key cellular processes needed for tumor growth and survival (Matter A. Drug Disc. Technol. 2001 6, 1005-1024).
  • EGF and VEGF receptors are previously described targets for small molecule inhibition.
  • KDR and flt-4 are both VEGF receptors
  • c-Kit binds the ligand stem cell factor (SCF), and triggers its multiple signal transduction pathways including Src family kinases, phosphatidyl-inositol 3 kinase, the Ras-Raf-Map kinase cascade, and phospholipase C (Broudy et al Blood 1999 94: 1979- 1986; Lennartsson et al Oncogene 1999 18: 5546-5553 ; Timokhina et al EMBO J 1998 17;6250-6262; Chian et al Blood 2001 98(5)1365-1373; Blume-Jensen et al Curr Biol
  • Flt-3 is normally expressed on hematopoietic progenitor cells and a subset of mature myeloid and lymphoid cells, where it modulates cell survival and proliferation.
  • Flt-3 is constitutively activated via mutation, either in the juxtamembrane region or in the activation loop of the kinase domain, in a large proportion of patients with AML (Reilly Leuk Lymphoma 2003 44: 1-7). Also, mutations in flt-3 are significantly correlated with poor prognosis in AML patients (Sawyers Cancer Cell 2002 1: 413-415).
  • G is a group -B-L-T, wherein
  • D is selected from -O-, -S(O) 0 . 2 -, and -NR 15 -;
  • X , X , and optionally X represent the atoms of a saturated bridged ring system, said saturated bridged ring system comprising up to four annular heteroatoms represented by any of X 1 , X 2 , and X 3 ; wherein, each X 1 is independently selected from -C(R 6 )R 7 -, -O-, -S(O) 0-2 -, and -NR 8 -; each X 2 is independently an optionally substituted bridgehead methine or a bridgehead nitrogen; each X 3 is independently selected from -C(R 6 )R 7 -, -O-, -S(O) 0-2 -, and -NR 8 -;
  • R and R when taken together with a common carbon to which they are attached, form a optionally substituted three- to seven-membered spirocyclyl, said optionally substituted three- to seven-membered spirocyclyl optionally containing at least one additional annular heteroatom selected from N, O, S, and P;
  • R 8 is selected from -R 3 , Y, -SO 2 NR 3 R 4 , -CO 2 R 4 , -C(O)NR 3 R 3 , -SO 2 R 4 , and -C(O)R 3 ;
  • the compound is according to paragraph [0038], wherein R 1 is -OH or -OC 1-6 alkyl.
  • R 1 is -OH or -OC 1-6 alkyl.
  • R 25 is selected from halogen, trihalomethyl, oxo, -CN, -NO 2 , -NH 2 , -OR 3 , -NR 3 R 4 , -S(O) 0-2 R 3 , -SO 2 NR 3 R 3 , -CO 2 R 3 , -C(O)NR 3 R 3 , -N(R 3 )SO 2 R 3 , -N(R 3 )C(O)R 3 , -N(R 3 )CO 2 R 3 , -C(O)R 3 , optionally substituted aryl, optionally substituted arylalkyl,
  • R 25 is selected from halogen, trihalomethyl, oxo, -CN, -NO 2 , -NH 2 , -OR 3 , -NR 3 R 4 , -S(O)o -2 R 3 , -SO 2 NR 3 R 3 , -CO 2 R 3 , -C(O)NR 3 R 3 , -N(R 3 )SO 2 R 3 , -N(R 3 )C(O)R 3 , -N(R 3 )CO 2 R 3 , -C(O)R 3 , optionally substituted aryl, optionally substituted arylalkyl, heteroarylalkyl, and optionally substituted lower alkyl; two of R 25 , together with the carbon or carbons
  • the compound is according to paragraph [0060], wherein one of R 10 and R 11 is -OR 12 , wherein R 12 is selected from -H, -C(O)R 3 , and optionally substituted lower alkyl; and R 9 and the other of R 10 and R 11 are both -H.
  • the compound is according to paragraph [0061], wherein Y is either -CH 2 - or absent.
  • the compound is according to paragraph [0064], wherein Y is either -CH 2 - or absent.
  • the compound is according to paragraph [0071], wherein U is either -CR 6 R 7 - or absent.
  • B is selected from:
  • R 2 is selected from -H, halogen, trihalomethyl, -CN, -NH 2 , -NO 2 , -OR 3 , -NR 3 R 3 , -S(O) 0-2 R 3 , -SO 2 NR 3 R 3 , -CO 2 R 3 , -C(O)NR 3 R 3 , -N(R 3 )SO 2 R 3 , -N(R 3 )C(O)R 3 , -N(R 3 )CO 2 R 3 , -C(O)R 3 , and optionally substituted lower alkyl; q is 0 to 2; each R 3 is independently selected from -H, optionally substituted lower alkyl, optionally substituted aryl, optionally substituted arylalkyl, and optionally substituted heteroarylalkyl; two R 3 , together with the nitrogen to which they are attached, form a four- to seven- membered heteroalicyclic, said four- to seven-membere
  • R 8 is selected from R 3 , -SO 2 NR 3 R 3 , -CO 2 R 3 , -C(O)NR 3 R 3 , -SO 2 R 3 , and -C(O)R 3 ;
  • R 9 , R 10 , and R 11 are each independently selected from -H, and -OR 12 ; or
  • R 20 is selected from -H, halogen, trihalomethyl, -CN, -NO 2 , -NH 2 , -OR 3 , -NR 3 R 3 , -S(O) 0-2 R 3 , -SO 2 NR 3 R 3 , -CO 2 R 3 , -C(O)NR 3 R 3 , -N(R 3 )SO 2 R 3 , -N(R 3 )C(O)R 3 , -N(R 3 )CO 2 R 3 , -C(O)R 3 , and optionally substituted lower alkyl;
  • R 2 , R 3 , R 5 , R 20 , R 25 and R 60 are as defined above.
  • R 70 is selected from -H, halogen, -OR 3 , -S(O) 0-2 R 3 , -NO 2 , -NH 2 , -NR 3 R 4 , and optionally substituted C ⁇ -6 alkyl;
  • Ar is either a five- or six-membered arylene or a five- or six-membered heteroarylene containing between one and three heteroatoms;
  • G is either an optionally substituted cycloalkyl or an optionally substituted heteroalicyclic; each R 2 is independently selected from halogen, trihalomethyl, -CN, -NO 2 , -NH 2 , -OR 3 , -NR 3 R 4 , -S(O)o -2 R 3 , -SO 2 NR 3 R 3 , -CO 2 R 3 , -C(O)NR 3 R 3 , -N(R 3 )SO 2 R 3 , -N(R 3 )C(O)R 3 , -N(R 3 )CO 2 R 3 , -C(O)R 3 , and optionally substituted C 1-6 alkyl; each R 3 is independently -H or R 4 ; each R 4 is independently selected from optionally substituted C 1-6 alkyl, optionally substituted aryl, optionally substituted aryl optionally substituted heterocyclyl, and optionally substituted heterocyclyl -ealkyl; or
  • R 3 and R 4 when taken together with a common nitrogen to which they are attached, form an optionally substituted five- to seven-membered heterocyclyl, said optionally substituted five- to seven-membered heterocyclyl optionally containing at least one additional annular heteroatom selected from N, O, S, and P;
  • R 5 is -H or optionally substituted C h alky!; each D is independently selected from -O-, -S(O)o -2 -, and -NR 5 -; each R 50 is independently either R 3 , or according to formula XII;
  • X 1 , X 2 , and optionally X 3 represent the atoms of a saturated bridged ring system, said saturated bridged ring system comprising up to four annular heteroatoms represented by any of X 1 , X 2 , and X 3 ; wherein, each X 1 is independently selected from -C(R 6 )R 7 -, -O-, -S(O) 0-2 -, and -NR 8 -; each X 2 is independently an optionally substituted bridgehead methine or a bridgehead nitrogen; each X 3 is independently selected from -C(R 6 )R 7 -, -O-, -S(O) 0-2 -, and -NR 8 -;
  • R 6 and R 7 are each independently selected from -H, halogen, trihalomethyl, -CN, -NH 2 , -NO 2 , -OR 3 , -NR 3 R 4 , -S(O) 0-2 R 4 , -SO 2 NR 3 R 4 , -CO 2 R 3 , -C(O)NR 3 R 4 , -N(R 3 )SO 2 R 4 , -N(R 3 )C(O)R 3 , -NCO 2 R 3 , -C(O)R 3 , optionally substituted C ⁇ -6 alkyl, optionally substituted aryl, optionally substituted aryl C ⁇ -6 alkyl, optionally substituted heterocyclyl, optionally substituted heterocyclyl C 1-6 alkyl, and a bond to either Y or D; or
  • R 6 and R 7 when taken together are oxo;
  • R 8 is selected from -R 3 , Y, -SO 2 NR 3 R 4 , -CO 2 R 4 , -C(O)NR 3 R 3 , -SO 2 R 4 , and -C(O)R 3 ; and each R 30 is independently selected from halogen, trihalomethyl, -CN, -NO 2 , -NH 2 , -OR 3 , -NR 3 R 4 , -S(O) 0-2 R 3 , -SO 2 NR 3 R 3 , -CO 2 R 3 , -C(O)NR 3 R 3 , -N(R 3 )SO 2 R 3 , -N(R 3 )C(O)R 3 , -N(R 3 )CO 2 R 3 , -C(O)R 3 , and optionally substituted Ci- ⁇ -ukyl.
  • the compound is according to paragraph [0091], wherein R 3a is C 1-6 alkyl.
  • the compound is according to paragraph [0092], wherein Z is -O-.
  • the compound is according to paragraph [0093], wherein G is selected from cyclopropyl, aziradine, cyclobutyl, and azetidine, each optionally substituted with between zero and four of R 30 .
  • J is a five- to ten-membered ring, optionally substituted with between zero and five of R 20 ; each R 20 is independently selected from -H, halogen, trihalomethyl, -CN, -NO 2 , -NH 2 , -OR 3 , -NR 3 R 4 , -S(O) 0-2 R 3 , -SO 2 NR 3 R 3 , -CO 2 R 3 , -C(O)NR 3 R 3 , -N(R 3 )SO 2 R 3 , -N(R 3 )C(O)R 3 , -N(R 3 )CO 2 R 3 , -C(O)R 3 , optionally substituted C 1-6 alkyl, optionally substituted aryl, optionally substituted aryl C 1-6 alkyl, optionally substituted heterocyclyl, and optionally substituted heterocyclyl C 1-6 alkyl; two of R 20 , together with the atom or atoms to
  • each R 60 is independently selected from halogen, trihalomethyl, -CN, -NO 2 , -NH 2 , -OR 3 , -NR 3 R 4 , -S(O)o -2 R 3 , -SO 2 NR 3 R 3 , -CO 2 R 3 , -C(O)NR 3 R 3 , -N(R 3 )SO 2 R 3 , -N(R 3 )C(O)R 3 , -N(R 3 )CO 2 R 3 , -C(O)R 3 , optionally substituted C ⁇ .
  • each methylene in any of the above formulae, other than those in a depicted ring, is independently optionally substituted with R 25 ;
  • R 25 is selected from halogen, trihalomethyl, oxo, -CN, -NO 2 , -NH 2 , -OR 3 , -NR 3 R 4 , -S(O) 0 - 2 R 3 , -SO 2 NR 3 R 3 , -CO 2 R 3 , -C(O)NR 3 R 3 , -N(R 3 )SO 2 R 3 , -N(R 3 )C(O)R 3 , -N(R 3 )CO 2 R 3 , -C(O)R 3 , optionally substituted aryl, optionally substituted aryl C 1-6 alkyl, heteroaryl C 1-6 alkyl, and optionally substituted C 1-6 alkyl; or two of R 25 , together with the carbon or carbons to which they are attached, can combine to form a three- to seven-membered alicyclic or heteroalicyclic;
  • R is equivalent to R as defined above;
  • R 4 and R 5 are as defined above.
  • the compound is according to paragraph [0098], wherein is C ⁇ -6 alkyl optionally substituted with a group selected from optionally substituted amino, an optionally substituted alkylamino, optionally substituted dialkylamino, and optionally substituted heteroalicylic.
  • the compound is according to paragraph [0099], wherein the heteroalicyclic portion of said optionally substituted heteroalicyclic of R 50 is selected from the group consisting of piperidine, piperazine, morpholine, thiomorpholine, thiomorpholine 1 -oxide, thiomorpholine 1,1 -dioxide, 2-oxo-morpholine, pyrrolidine, and azepine.
  • the compound is according to paragraph [0102], wherein Y is selected from -CH 2 CH 2 CH 2 CH 2 -, -CH 2 CH 2 CH 2 -, -CH 2 CH 2 -, -CH 2 -, and absent.
  • the compound is according to paragraph [0103], wherein n is 0 and the saturated bridged ring system according to formula XII has a geometry selected from the group consisting of [4.4.0], [4.3.0], [4.2.0], [4.1.0], [3.3.0], [3.2.0], and [3.1.0].
  • the compound is according to paragraph [0104], wherein said saturated bridged ring system contains at least one annular nitrogen or at least one annular oxygen.
  • U 1 is selected from -O-, -S(O) 0-2 -, -NR 8 -, -CR 6 R 7 -, and absent; and e is 0 or 1.
  • the compound is according to paragraph [0107], wherein Y is -CH 2 -.
  • the compound is according to paragraph [0108], wherein U 1 is -NR 8 -, wherein R 8 is selected from -H, optionally substituted lower alkyl, -CO 2 R 3 , -C(O)NR 3 R 3 , -SO 2 R 3 , and -C(O)R 3 .
  • the compound is according to paragraph [0108], wherein U 1 is -O-.
  • the compound is according to paragraph [0108], wherein U 1 is absent.
  • the compound is according to paragraph [0103], wherein Y is selected from -CH 2 CH 2 -, -CH 2 -, and absent.
  • R 9 , R 10 , and R 11 are each independently selected from -H, and -OR 12 ; or
  • R 9 is selected from -H, and -OR 12 , and R 10 and R 11 , when taken together, are either an optionally substituted alkylidene or an oxo;
  • the compound is according to paragraph [0113], wherein one of R 10 and R 11 is -OR 12 , wherein R 12 is selected from -H, -C(O)R 3 , and optionally substituted lower alkyl; and R 9 and the other of R 10 and R 11 are both -H.
  • the compound is according to paragraph [0114], wherein Y is either -CH 2 - or absent.
  • the compound is according to paragraph [0117], wherein Y is either -CH 2 - or absent.
  • the compound is according to paragraph [0118], wherein R 8 is methyl or ethyl.
  • the compound is according to paragraph [0119], wherein at least one of R 2 is halogen.
  • the compound is according to paragraph [0106], wherein said saturated bridged ring system is of formula XVIII.
  • the compound is according to paragraph [0121], wherein Y is -CH 2 -.
  • the compound is according to paragraph [0122], wherein R 8 is methyl or ethyl.
  • U 2 is selected from -O-, -S(O) 0-2 -, -NR 8 -, -CR 6 R 7 -, and absent.
  • the compound is according to paragraph [0124], wherein R 3 of formula XIX is selected from -H and optionally substituted alkyl. [0126] In another example, the compound is according to paragraph [0125], wherein U 2 is either -CR 6 R 7 - or absent.
  • the compound is according to paragraph [0127], wherein Y is -CH 2 -.
  • the compound is according to any of paragraphs [0099] - [0130], wherein R is selected from C 1-6 alkyl, perfluoro C ⁇ -6 alkyl, and halogen.
  • the compound is according to paragraph [0131], wherein R 2 is selected from perfluoro C 1-3 alkyl and halogen.
  • the compound is according to paragraph [0086], selected from Table 2.
  • Another aspect of the invention is a pharmaceutical composition
  • a pharmaceutical composition comprising a compound according to any one of paragraphs [0033]-[0120] and a pharmaceutically acceptable carrier.
  • Another aspect of the invention is a metabolite of the compound or the pharmaceutical composition according to any one of paragraphs [0022]-[0122].
  • Another aspect of the invention is the method according to paragraph [0123], wherein modulating the in vivo activity of the kinase comprises inhibition of said kinase.
  • Another aspect of the invention is the method according to paragraph [0124], wherein the kinase is at least one of c-Met, KDR, c-Kit, flt-3, and flt-4.
  • Another aspect of the invention is the method according to paragraph [0125], wherein the kinase is c-Met.
  • Another aspect of the invention is a method of treating diseases or disorders associated with uncontrolled, abnormal, and/or unwanted cellular activities, the method comprising administering, to a mammal in need thereof, a therapeutically effective amount of the compound or the pharmaceutical composition as described in any one of paragraphs [0033H0121].
  • Another aspect of the invention is a method of screening for a modulator of a kinase, said kinase selected from c-Met, KDR, c-Kit, flt-3, and flt-4, the method comprising combining a compound according to any one of paragraphs [0033]-[0120], and at least one candidate agent and determining the effect of the candidate agent on the activity of said kinase.
  • Another aspect of the invention is a method of inhibiting proliferative activity in a cell, the method comprising administering an effective amount of a composition comprising a compound according any one of paragraphs [0033]-[0120] to a cell or a plurality of cells.
  • Z is selected from -S(O)o -2 -, -O-, and -NR 5 -; each R 5 is independently selected from -H, optionally substituted C 1-6 alkyl, optionally substituted aryl, and optionally substituted aryl C ⁇ - 6 alkyl;
  • Ar is either a five- to ten-membered arylene or a five- to ten-membered heteroarylene containing between one and three heteroatoms;
  • R 2 is selected from -H, halogen, trihalomethyl, -CN, -NO 2 , -NH 2 , -OR 3 , -NR 3 R 3 , -S(O) 0-2 R 3 , -SO 2 NR 3 R 3 , -CO 2 R 3 , -C(O)NR 3 R 3 , -N(R 3 )SO 2 R 3 , -N(R 3 )C(O)R 3 , -N(R 3 )CO 2 R 3 , -C(O)R 3 , and optionally substituted C ⁇ -6 alkyl; each R 3 is independently selected from -H, -Si(R 5 )(R 5 )R 5 , optionally substituted lower alkyl, optionally substituted aryl, optionally substituted arylalkyl, and optionally substituted heteroarylalkyl; two R 3 , together with the nitrogen to which they are attached, form a four- to seven- membered
  • Q is a five- to ten-membered ring system, optionally substituted with between zero and four of R 20 ; each R 20 is independently selected from -H, halogen, trihalomethyl, -CN, -NO 2 , -NH 2 , -OR 3 , -NR 3 R 3 , -S(O) 0-2 R 3 , -SO 2 NR 3 R 3 , -CO 2 R 3 , -C(O)NR 3 R 3 , -N(R 3 )SO 2 R 3 , -N(R 3 )C(O)R 3 , -N(R 3 )CO 2 R 3 , -C(O)R 3 , optionally substituted C ⁇ -6 alkyl, optionally substituted aryl, optionally substituted aryl C ⁇ -6 alkyl, optionally substituted heterocyclyl, and optionally substituted heterocyclyl -eal yl; two of R 20 , together with the atom or atom
  • D is selected from -O-, -S(O) 0-2 -, and -NR 15 -;
  • R is either R , or according to formula XXIV;
  • R and R are each independently selected from -H, halogen, trihalomethyl, -CN, -NH 2 , -NO 2 , -OR 3 , -NR 3 R 3 , -S(O)o -2 R 3 , -SO 2 NR 3 R 3 , -CO 2 R 3 , -C(O)NR 3 R 3 , -N(R 3 )SO 2 R 3 , -N(R 3 )C(O)R 3 , -NCO 2 R 3 , -C(O)R 3 , optionally substituted C 1-6 alkyl, optionally substituted aryl, optionally substituted aryl Ci- ⁇ alkyl, optionally substituted heterocyclyl, optionally substituted heterocyclyl a and a bond to either Y or D; or R 6 and R 7 , when taken together are oxo; or , when taken together with a common carbon to which they are attached, form a optionally substituted three- to seven-member
  • R 8 is selected from -R 3 , Y, -SO 2 NR 3 R 3 , -CO 2 R 3 , -C(O)NR 3 R 3 , -SO 2 R 3 , and -C(O)R 3 ;
  • two R 13 together with the atom or atoms to which they are attached, can combine to form a heteroalicyclic optionally substituted with between one and four of R 60 , said heteroalicyclic comprising up to four annular heteroatoms, and said heteroalicyclic optionally comprising an aryl or heteroaryl fused thereto, in which case said aryl or heteroaryl is optionally substituted with an additional one to four of R 60 ;
  • R 14 is selected from -H, -NO 2 , -NH 2 , -N(R 3 )R 3 , -CN, -OR 3 , optionally substituted C 1-6 alkyl, optionally substituted heteroalicyclyl Ci- ⁇ alkyl, optionally substituted aryl, optionally substituted aryl C ⁇ alkyl and optionally substituted heteroalicyclic;
  • P 2 is selected from -H, a metal, and a group removed in-situ when combining XXII and XXIII to make XXI.
  • Ar is para- phenylene as defined by the substitution pattern of -Z- and -B-L-T about said phenylene.
  • Z is either -O- or -NR 5 -
  • R 25 is selected from halogen, trihalomethyl, oxo, -CN, -NO 2 , -NH 2 , -OR 3 , -NR 3 R 3 , -S(O) 0-2 R 3 , -SO 2 NR 3 R 3 , -CO 2 R 3 , -C(O)NR 3 R 3 , -N(R 3 )SO 2 R 3 , -N(R 3 )C(O)R 3 , -N(R 3 )CO 2 R 3 , -C(O)R 3 , optionally substituted aryl, optionally substituted aryl C 1-6 alkyl, heteroaryl C 1-6 alkyl, and optionally substituted C 1-6 alkyl; two of R 25 , together with the
  • R is defined as above, and P is a five- to seven-membered ring, including the two shared carbons of the aromatic ring to which P is fused, P optionally containing between one and three heteroatoms.
  • R 70 is selected from -H, -NO 2 , -NH 2 , and -NR 3 R 3 ; provided when Z is -N(R 5 )- that R 5 is selected from -H, C ⁇ -3 alkyl, and aryl C 1-3 alkyl;
  • P 1 is selected from halogen, optionally substituted alkyl-S(O)o-2-, optionally substituted arylsulfonate, optionally substituted alkylsulfonate, a group containing boron, an azide, a group containing phosphorus, and a metal; and P 2 is selected from -H and a metal.
  • the process is according to paragraph [0147], wherein R 2 is selected from C ⁇ -6 alkyl, perfluoro C ⁇ -6 alkyl, and halogen.
  • the process is according to paragraph [0148], wherein XXIIa and XXIIIa are heated together, optionally with a base, optionally with microwave radiation, to form XXIa.
  • the process is according to paragraph [0149], wherein the base is selected from an organic base, an inorganic base, and a combination of an organic base and an inorganic base.
  • the process is according to paragraph [0150], wherein the base is selected from 2,6-lutidine, 4-N,N-dimethylaminopyridine, and a metal carbonate.
  • the process is according to paragraph [0152], wherein the solvent is an organic solvent.
  • the process is according to paragraph [0163], wherem the non- aromatic solvent is N,N-dimethylacetamide.
  • the process is according to paragraph [0154], wherein one molar equivalent of XXIIIa is combined with more than one but less than two molar equivalents of XXIIa.
  • the process is according to paragraph [0172], wherein XXIIa is combined with XXIIIa and said base in an aromatic solvent to form a mixture, and said mixture is heated to between about 100°C and 200°C for between about ten and twenty hours to form XXIa.
  • the process is according to any of paragraphs [0149] - [0177], wherein a compound of formula XXIIb is substituted for the compound of formula XXIIa, and either a compound of formula XXIIIb or a compound of formula XXIIIc is substituted for the compound of formula XXIIIa, in order to make a compound of formula XXIb or a compound of formula XXIc, respectively,
  • the symbol "-" means a single bond
  • " ⁇ ” means a triple bond
  • the symbol "» ⁇ /V” refers to a group on a double-bond as occupying either position on the terminus of a double bond to which the symbol is attached; that is, the geometry, E- or Z-, of the double bond is ambiguous.
  • the " ⁇ ⁇ " symbol will be used at the end of the bond which was theoretically cleaved in order to separate the group from its parent structural formula.
  • the "R” group may reside on either the 5-membered or the 6-membered ring of the fused ring system.
  • the two "R's" may reside on any two atoms of the ring system, again assuming each replaces a depicted, implied, or expressly defined hydrogen on the ring.
  • alkyl groups are those of C 20 or below.
  • Cycloalkyl is a subset of alkyl and includes cyclic hydrocarbon groups of from three to thirteen carbon atoms. Examples of cycloalkyl groups include c- propyl, c-butyl, c-pentyl, norbomyl, adamantyl and the like.
  • alkyl refers to alkanyl, alkenyl, and alkynyl residues (and combinations thereof); it is intended to include cyclohexylmethyl, vinyl, allyl, isoprenyl, and the like.
  • alkyl residue having a specific number of carbons all geometric isomers having that number of carbons are intended to be encompassed; thus, for example, either “butyl” or “C 4 alkyl” is meant to include ⁇ -butyl, -fee-butyl, isobutyl, t-butyl, isobutenyl and but-2- yne radicals; and for example, "propyl” or “C 3 alkyl” each include ⁇ -propyl, propenyl, and isopropyl.
  • Alkylene refers to straight or branched chain divalent radical consisting solely of carbon and hydrogen atoms, containing no unsaturation and having from one to ten carbon atoms, for example, methylene, ethylene, propylene, ⁇ -butylene and the like. Alkylene is a subset of alkyl, referring to the same residues as alkyl, but having two points of attachment and, specifically, fully saturated. Examples of alkylene include ethylene (-CH 2 CH2-), propylene (-CH 2 CH 2 CH 2 -), dimethylpropylene (-CH 2 C(CH 3 ) 2 CH 2 -), and cyclohexylpropylene (-CH 2 CH 2 CH(C 6 H ⁇ 3 )).
  • Alkylidene refers to a straight or branched chain unsaturated divalent radical consisting solely of carbon and hydrogen atoms, having from two to ten carbon atoms, for example, ethylidene, propylidene, n-butylidene, and the like. Alkylidene is a subset of alkyl, referring to the same residues as alkyl, but having two points of attachment and, specifically, double bond unsaturation. The unsaturation present includes at least one double bond.
  • Alkylidyne refers to a straight or branched chain unsaturated divalent radical consisting solely of carbon and hydrogen atoms having from two to ten carbon atoms, for example, propylid-2-ynyl, /z-butylid-1-ynyl, and the like. Alkylidyne is a subset of alkyl, referring to the same residues as alkyl, but having two points of attachment and, specifically, triple bond unsaturation. The unsaturation present includes at least one triple bond.
  • alkylene when optionally substituted, may contain alkyl substitution which itself contains unsaturation.
  • 2-(2-phenylethynyl-but-3-enyl)-naphthalene (IUPAC name) contains an n-butylid-3-ynyl radical with a vinyl substituent at the 2-position of said radical.
  • Alkoxy refers to the group -O-alkyl, for example including from one to eight carbon atoms of a straight, branched, cyclic configuration, unsaturated chains, and combinations thereof attached to the parent structure through an oxygen atom. Examples include methoxy, ethoxy, propoxy, isopropoxy, cyclopropyloxy, cyclohexyloxy and the like. Lower-alkoxy refers to groups containing one to six carbons.
  • Substituted alkoxy refers to the group -O-(substituted alkyl), the substitution on the alkyl group generally containing more than only carbon (as defined by alkoxy).
  • One exemplary substituted alkoxy group is "polyalkoxy” or -O-optionally substituted alkylene-optionally substituted alkoxy, and includes groups such as -OCH 2 CH 2 OCH 3 , and glycol ethers such as polyethyleneglycol and -O(CH 2 CH 2 O) x CH 3 , where x is an integer of between about two and about twenty, in another example, between about two and about ten, and in a further example between about two and about five.
  • Another exemplary substituted alkoxy group is hydroxyalkoxy or -OCH 2 (CH 2 ) y OH, where y is for example an integer of between about one and about ten, in another example y is an integer of between about one and about four.
  • Acyl refers to groups of from one to ten carbon atoms of a straight, branched, cyclic configuration, saturated, unsaturated and aromatic and combinations thereof, attached to the parent structure through a carbonyl functionality. One or more carbons in the acyl residue may be replaced by nitrogen, oxygen or sulfur as long as the point of attachment to the parent remains at the carbonyl. Examples include acetyl, benzoyl, propionyl, isobutyryl, t-butoxycarbonyl, benzyloxycarbonyl and the like. Lower-acyl refers to groups containing one to six carbons.
  • ⁇ -Amino Acids refer to naturally occurring and commercially available amino acids and optical isomers thereof. Typical natural and commercially available ⁇ -amino acids are glycine, alanine, serine, homoserine, threonine, valine, norvaline, leucine, isoleucine, norleucine, aspartic acid, glutamic acid, lysine, omithine, histidine, arginine, cysteine, homocysteine, methionine, phenylalanine, homophenylalanine, phenylglycine, ortho-tyrosine, meta-tyrosine, para-tyrosine, tryptophan, glutamine, asparagine, proline and hydroxyproline.
  • a "side chain of an ⁇ -amino acid” refers to the radical found on the ⁇ -carbon of an ⁇ -amino acid as defined above, for example, hydrogen (for glycine), methyl (for alanine), benzyl (for phenylalanine), and the like.
  • Amino refers to the group -NH 2 .
  • Substituted amino refers to the group -N(H)R or -N(R)R where each R is independently selected from the group: optionally substituted alkyl, optionally substituted alkoxy, optionally substituted aryl, optionally substituted heterocyclyl, acyl, carboxy, alkoxycarbonyl, sulfanyl, sulfinyl and sulfonyl, for example, diethylamino, methylsulfonylamino, furanyl-oxy-sulfonamino.
  • Aryl refers to aromatic six- to fourteen-membered carbocyclic ring, for example, benzene, naphthalene, indane, tetralin, fluorene and the like, univalent radicals.
  • univalent radicals the aforementioned ring examples are named, phenyl, naphthyl, indanyl, tetralinyl, and fluorenyl.
  • Arylene genetically refers to any aryl that has at least two groups attached thereto.
  • phenylene refers to a divalent phenyl ring radical. A phenylene, thus may have more than two groups attached, but is defined by a minimum of two non-hydrogen groups attached thereto.
  • Arylalkyl refers to a residue in which an aryl moiety is attached to a parent structure via one of an alkylene, alkylidene, or alkylidyne radical. Examples include benzyl, phenethyl, phenylvinyl, phenylallyl and the like.
  • Exo-alkenyl refers to a double bond that emanates from an annular carbon, and is not within the ring system, for example the double bond depicted in the formula below.
  • fused-polycyclic or "fused ring system” refers to a polycyclic ring system that contains bridged or fused rings; that is, where two rings have more than one shared atom in their ring structures.
  • fused-polycyclics and fused ring systems are not necessarily all aromatic ring systems.
  • fused-polycyclics share a vicinal set of atoms, for example naphthalene or 1,2,3,4-tetrahydro-naphthalene.
  • a spiro ring system is not a fused-polycyclic by this definition, but fused polycyclic ring systems of the invention may themselves have spiro rings attached thereto via a single ring atom of the fused-polycyclic.
  • Heteroatom refers to O, S, N, or P.
  • the group -S(O)o -2 - refers to -S- (sulfide), -S(O)- (sulfoxide), and -SO 2 - (sulfone).
  • nitrogens particularly but not exclusively, those defined as annular aromatic nitrogens, are meant to include their corresponding N-oxide form, although not explicitly defined as such in a particular example.
  • annular nitrogen atoms may be optionally quaternized; and the ring radical may be partially or fully saturated or aromatic.
  • heterocyclyl radicals include, but are not limited to, azetidinyl, acridinyl, benzodioxolyl, benzodioxanyl, benzofuranyl, carbazoyl, cinnolinyl, dioxolanyl, indolizinyl, naphthyridinyl, perhydroazepinyl, phenazinyl, phenothiazinyl, phenoxazinyl, phthalazinyl, pteridinyl, purinyl, quinazolinyl, quinoxalinyl, quinolinyl, isoquinolinyl, tetrazoyl, tetrahydroisoquinolyl, piperidinyl, piperazinyl, 2-oxopiperazinyl, 2-oxopiperidinyl, 2-oxopyrrolidinyl, 2-oxoazepinyl, azepiny
  • Heterocyclylalkyl refers to a residue in which a heterocyclyl is attached to a parent structure via one of an alkylene, alkylidene, or alkylidyne radical. Examples include (4-methylpiperazin-l-yl) methyl, (morpholin-4-yl) methyl, (pyridine-4-yl) methyl, 2-(o ⁇ azolin-2-yl) ethyl, 4-(4-methylpiperazin-l-yl)-2-butenyl, and the like. Both the heterocyclyl, and the corresponding alkylene, alkylidene, or alkylidyne radical portion of a heterocyclylalkyl group may be optionally substituted.
  • “Lower heterocyclylalkyl” refers to a heterocyclylalkyl where the “alkyl” portion of the group has one to six carbons.
  • “Heteroalicyclylalkyl” refers specifically to a heterocyclylalkyl where the heterocyclyl portion of the group is non-aromatic; and “heteroarylalkyl” refers specifically to a heterocyclylalkyl where the heterocyclyl portion of the group is aromatic
  • Such terms may be described in more than one way, for example, “lower heterocyclylalkyl” and “heterocyclyl C ⁇ -6 alkyl” are equivalent terms.
  • Optionally substituted refers to all subsequent modifiers in a term, for example in the term “optionally substituted arylC 1-8 alkyl,” optional substitution may occur on both the “C 1-8 alkyl” portion and the “aryl” portion of the molecule; and for example, optionally substituted alkyl includes optionally substituted cycloalkyl groups, which in turn are defined as including optionally substituted alkyl groups, potentially ad infinitum. A list of exemplary optional substitution are listed below in the definition of "substituted.”
  • saturated bridged ring system refers to a bicyclic or polycyclic ring system that is not aromatic. Such a system may contain isolated or conjugated unsaturation, but not aromatic or heteroaromatic rings in its core structure (but may have aromatic substitution thereon).
  • a ring atom of a saturated bridged ring system (rings B and B'), but not a bridgehead atom, can be a shared atom between the saturated bridged ring system and a spirocyclyl (ring A) attached thereto.
  • a spirocyclyl can be carbocyclic or heteroalicyclic.
  • Suitable leaving group is defined as the term would be understood by one of ordinary skill in the art; that is, a carbon with such a group attached, upon reaction wherein a new bond is to be formed, loses such a group upon formation of the new bond.
  • the invention pertains particularly with respect convergent synthesis, to reactions where such a, leaving group is bonded to a reaction partner that is aromatic, undergoes a bond- forming reaction and remains aromatic.
  • a typical example of such a reaction is a nucleophilic aromatic substitution reaction, as would be understood by one of ordinary skill in the art.
  • the invention is not limited to such mechanistic restrictions; for example, reactions where there is, for example, an insertion reaction (for example by a transition metal) into the bond between the aromatic reaction partner and its leaving group followed by reductive coupling can also be used within the scope of the invention.
  • suitable leaving groups include halogens, optionally substituted aryl or alkyl sulfonates, phosphonates, azides, RS(O)o -2 - where R is, for example optionally substituted alkyl, optionally substituted aryl, or optionally substituted heteroaryl.
  • Sulfanyl refers to the groups: -S-(optionally substituted alkyl), -S-(optionally substituted aryl), and -S-(optionally substituted heterocyclyl).
  • Sulfinyl refers to the groups: -S(O)-H, -S(O)-(optionally substituted alkyl), -S(O)-optionally substituted aryl), and -S(O)-(optionally substituted heterocyclyl).
  • Sulfonyl refers to the groups: -S(O 2 )-H, -S(O 2 )-(optionally substituted alkyl), -S(O 2 )-optionally substituted aryl), -S(O 2 )-(optionally substituted heterocyclyl), -S(O 2 )-(optionally substituted alkoxy), -S(O 2 )-o ⁇ tionally substituted aryloxy), and -S(O 2 )-(optionally substituted heterocyclyloxy).
  • Yield for each of the reactions described herein is expressed as a 'percentage of the theoretical yield.
  • Some of the compounds of the invention may have imino, amino, oxo or hydroxy substituents off aromatic heterocyclyl systems.
  • imino, amino, oxo or hydroxy substituents may exist in their corresponding tautomeric form, i.e., amino, imino, hydroxy or oxo, respectively.
  • the compounds of the invention may have asymmetric carbon atoms, oxidized sulfur atoms or quaternized nitrogen atoms in their structure.
  • the compounds of the invention and their pharmaceutically acceptable salts may exist as single stereoisomers, racemates, and as mixtures of enantiomers and diastereomers.
  • the compounds may also exist as geometric isomers. All such single stereoisomers, racemates and mixtures thereof, and geometric isomers are intended to be within the scope of this invention.
  • a particular group with its bonding structure is denoted as being bonded to two partners; that is, a divalent radical, for example, -OCH 2 -, then it is understood that either of the two partners may be bound to the particular group at one end, and the other partner is necessarily bound to the other end of the particular group, unless stated explicitly otherwise.
  • divalent radicals are not to be construed as limited to the depicted orientation, for example "-OCH 2 -" is meant to mean not only "-OCH 2 -" as drawn, but also "-CH 2 O-.”
  • optically active (R)- and (S)- isomers may be prepared using chiral synthons or chiral reagents, or resolved using conventional techniques.
  • Enantiomers may be resolved by methods known to one of ordinary skill in the art, for example by: formation of diastereoisomeric salts or complexes which may be separated, for example, by crystallization; via formation of diastereoisomeric derivatives which may be separated, for example, by crystallization, selective reaction of one enantiomer with an enantiomer-specific reagent, for example enzymatic oxidation or reduction, followed by separation of the modified and unmodified enantiomers; or gas-liquid or liquid chromatography in a chiral environment, for example on a chiral support, such as silica with a bound chiral ligand or in the presence of a chiral solvent.
  • enantiomer may be synthesized by asymmetric synthesis using optically active reagents, substrates, catalysts or solvents, or by converting on enantiomer to the other by asymmetric transformation.
  • enantiomers enriched in a particular enantiomer, the major component enantiomer may be further enriched (with concomitant loss in yield) by recrystallization.
  • Patient for the purposes of the present invention includes humans and other animals, particularly mammals, and other organisms. Thus the methods are applicable to both human therapy and veterinary applications. In a preferred embodiment the patient is a mammal, and in a most preferred embodiment the patient is human.
  • Kinase-dependent diseases or conditions refer to pathologic conditions that depend on the activity of one or more protein kinases.
  • Kinases either directly or indirectly participate in the signal transduction pathways of a variety of cellular activities including proliferation, adhesion, migration, differentiation and invasion.
  • Diseases associated with kinase activities include tumor growth, the pathologic neovascularization that supports solid tumor growth, and associated with other diseases where excessive local vascularization is involved such as ocular diseases (diabetic retinopathy, age-related macular degeneration, and the like) and inflammation (psoriasis, rheumatoid arthritis, and the like).
  • phosphatases can also play a role in "kinase-dependent diseases or conditions" as cognates of kinases; that is, kinases phosphorylate and phosphatases dephosphorylate, for example protein substrates. Therefore compounds of the invention, while modulating kinase activity as described herein, may also modulate, either directly or indirectly, phosphatase activity. This additional modulation, if present, may be synergistic (or not) to activity of compounds of the invention toward a related or otherwise interdependent kinase or kinase family. In any case, as stated previously, the compounds of the invention are useful for treating diseases characterized in part by abnormal levels of cell proliferation (i.e. tumor growth), programmed cell death (apoptosis), cell migration and invasion and angiogenesis associated with tumor growth.
  • abnormal levels of cell proliferation i.e. tumor growth
  • apoptosis programmed cell death
  • “Therapeutically effective amount” is an amount of a compound of the invention, that when administered to a patient, ameliorates a symptom of the disease.
  • the amount of a compound of the invention which constitutes a “therapeutically effective amount” will vary depending on the compound, the disease state and its severity, the age of the patient to be treated, and the like.
  • the therapeutically effective amount can be determined routinely by one of ordinary skill in the art having regard to his own knowledge and to this disclosure.
  • “Pharmaceutically acceptable acid addition salt” refers to those salts that retain the biological effectiveness of the free bases and that are not biologically or otherwise undesirable, formed with inorganic acids such as hydrochloric acid, hydrobromie acid, sulfuric acid, nitric acid, phosphoric acid, and the like, as well as organic acids such as acetic acid, trifluoroacetic acid, propionic acid, glycolic acid, pyruvic acid, oxalic acid, maleic acid, malonic acid, succinic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, salicylic acid and the like.
  • inorganic acids such as hydrochloric acid, hydrobromie acid, sulfuric acid, nitric acid, phosphoric acid, and the like
  • organic acids such as acetic acid, trifluoro
  • “Pharmaceutically acceptable base addition salts” include those derived from inorganic bases such as sodium, potassium, lithium, ammonium, calcium, magnesium, iron, zinc, copper, manganese, aluminum salts and the like. Exemplary salts are the ammonium, potassium, sodium, calcium, and magnesium salts.
  • Salts derived from pharmaceutically acceptable organic non-toxic bases include, but are not limited to, salts of primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines and basic ion exchange resins, such as isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, ethanolamine, 2-dimethylaminoethanol, 2-diethylaminoethanol, dicyclohexylamine, lysine, arginine, histidine, caffeine, procaine, hydrabamine, choline, betaine, ethylenediamine, glucosamine, methylglucamine, theobromine, purines, piperazine, piperidine, N-ethylpiperidine, polyamine resins, and the like.
  • salts of primary, secondary, and tertiary amines substituted amines including naturally occurring substituted amines, cyclic amines
  • Prodrug refers to compounds that are transformed (typically rapidly) in vivo to yield the parent compound of the above formulae, for example, by hydrolysis in blood.
  • Common examples include, but are not limited to, ester and amide forms of a compound having an active form bearing a carboxylic acid moiety.
  • Examples of pharmaceutically acceptable esters of the compounds of this invention include, but are not limited to, alkyl esters (for example with between about one and about six carbons) wherein the alkyl group is a straight or branched chain. Acceptable esters also include cycloalkyl esters and arylalkyl esters such as, but not limited to benzyl.
  • Examples of pharmaceutically acceptable amides of the compounds of this invention include, but are not limited to, primary amides, and secondary and tertiary alkyl amides (for example with between about one and about six carbons).
  • Amides and esters of the compounds of the present invention may be prepared according to conventional methods. A thorough discussion of prodrugs is provided in T. Higuchi and V. Stella, "Pro-drugs as Novel Delivery Systems," Vol 14 of the A.C.S. Symposium Series, and in Bioreversible Carriers in Drug Design, ed. Edward B. Roche, American Pharmaceutical Association and Pergamon Press, 1987, both of which are incorporated herein by reference for all purposes.
  • Methodabolite refers to the break-down or end product of a compound or its salt produced by metabolism or biotransformation in the animal or human body; for example, biotransformation to a more polar molecule such as by oxidation, reduction, or hydrolysis, or to a conjugate (see Goodman and Gilman, "The Pharmacological Basis of Therapeutics” ⁇ .sup.th Ed., Pergamon Press, Gilman et al. (eds), 1990 for a discussion of biotransformation).
  • the metabolite of a compound of the invention or its salt may be the biologically active form of the compound in the body.
  • a prodrug may be used such that the biologically active form, a metabolite, is released in vivo.
  • a biologically active metabolite is discovered serendipitously, that is, no prodrug design per se was undertaken.
  • An assay for activity of a metabolite of a compound of the present invention is known to one of skill in the art in light of the present disclosure.
  • the compounds of the present invention can exist in unsolvated as well as solvated forms with pharmaceutically acceptable solvents such as water, ethanol, and the like.
  • pharmaceutically acceptable solvents such as water, ethanol, and the like.
  • the solvated forms are considered equivalent to the unsolvated forms for the purposes of the present invention.
  • the present invention cover compounds made either using standard organic synthetic techniques, including combinatorial chemistry or by biological methods, such as bacterial digestion, metabolism, enzymatic conversion, and the like.
  • Such suitable x-ray quality crystals can be used as part of a method of identifying a candidate agent capable of binding to and modulating the activity of kinases.
  • Such methods may be characterized by the following aspects: a) introducing into a suitable computer program, information defining a ligand binding domain of a kinase in a conformation (e.g.
  • Such methods may further entail: employing a candidate agent, so-determined to fit spatially into the ligand binding domain, in a biological activity assay for kinase modulation, and determining whether said candidate agent modulates kinase activity in the assay. Such methods may also include administering the candidate agent, determined to modulate kinase activity, to a mammal suffering from a condition treatable by kinase modulation, such as those described above.
  • compounds of the invention can be used in a method of evaluating the ability of a test agent to associate with a molecule or molecular complex comprising a ligand binding domain of a kinase.
  • a method may be characterized by the following aspects: a) creating a computer model of a kinase binding pocket using stracture coordinates obtained from suitable x-ray quality crystals of the kinase, b) employing computational algorithms to perform a fitting operation between the test agent and the computer model of the binding pocket, and c) analyzing the results of the fitting operation to quantify the association between the test agent and the computer model of the binding pocket.
  • Administration of the compounds of the invention, or their pharmaceutically acceptable salts, in pure form or in an appropriate pharmaceutical composition can be carried out via any of the accepted modes of administration or agents for serving si ilar utilities.
  • administration can be, for example, orally, nasally, parenterally (intravenous, intramuscular, or subcutaneous), topically, transdermally, intravaginally, intravesically, intracistemally, or rectally, in the form of solid, semi-solid, lyophilized powder, or liquid dosage forms, such, as for example, tablets, suppositories, pills, soft elastic and hard gelatin capsules, powders, solutions, suspensions, or aerosols, or the like, preferably in unit dosage forms suitable for simple administration of precise dosages.
  • compositions will include a conventional pharmaceutical carrier or excipient and a compound of the invention as the/an active agent, and, in addition, may include other medicinal agents, pharmaceutical agents, carriers, adjuvants, etc.
  • Compositions of the invention may be used in combination with anticancer or other agents that are generally administered to a patient being treated for cancer.
  • Adjuvants include preserving, wetting, suspending, sweetening, flavoring, perfuming, emulsifying, and dispensing agents. Prevention of the action of microorganisms can be ensured by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, and the like. It may also be desirable to include isotonic agents, for example sugars, sodium chloride, and the like. Prolonged absorption of the injectable pharmaceutical form can be brought about by the use of agents delaying absorption, for example, aluminum monostearate and gelatin.
  • compositions suitable for parenteral injection may comprise physiologically acceptable sterile aqueous or nonaqueous solutions, dispersions, suspensions or emulsions, and sterile powders for reconstitution into sterile injectable solutions or dispersions.
  • suitable aqueous and nonaqueous carriers, diluents, solvents or vehicles include water, ethanol, polyols (propyleneglycol, polyethyleneglycol, glycerol, and the like), suitable mixtures thereof, vegetable oils (such as olive oil) and injectable organic esters such as ethyl oleate.
  • Proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersions and by the use of surfactants.
  • One preferable route of administration is oral, using a convenient daily dosage regimen that can be adjusted according to the degree of severity of the disease-state to be treated.
  • Solid dosage forms for oral administration include capsules, tablets, pills, powders, and granules.
  • the active compound is admixed with at least one inert customary excipient (or carrier) such as sodium citrate or dicalcium phosphate or
  • fillers or extenders as for example, starches, lactose, sucrose, glucose, mannitol, and silicic acid
  • binders as for example, cellulose derivatives, starch, alignates, gelatin, polyvinylpyrrolidone, sucrose, and gum acacia
  • humectants as for example, glycerol
  • disintegrating agents as for example, agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, croscarmellose sodium, complex silicates, and sodium carbonate
  • solution retarders as for example paraffin
  • absorption accelerators as for example,
  • the dosage forms may also comprise buffering agents.
  • Solid dosage forms as described above can be prepared with coatings and shells, such as enteric coatings and others well known in the art. They may contain pacifying agents, and can also be of such composition that they release the active compound or compounds in a certain part of the intestinal tract in a delayed manner. Examples of embedded compositions that can be used are polymeric substances and waxes. The active compounds can also be in microencapsulated form, if appropriate, with one or more of the above-mentioned excipients.
  • Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, solutions, suspensions, syrups, and elixirs. Such dosage forms are prepared, for example, by dissolving, dispersing, etc., a compound(s) of the invention, or a pharmaceutically acceptable salt thereof, and optional pharmaceutical adjuvants in a carrier, such as, for example, water, saline, aqueous dextrose, glycerol, ethanol and the like; solubilizmg agents and emulsifiers, as for example, ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propyleneglycol, 1,3- butyleneglycol, dimethylformamide; oils, in particular, cottonseed oil, groundnut oil, corn germ oil, olive oil, castor oil and sesame oil, glycerol, tetrahydrofurfuryl
  • Suspensions in addition to the active compounds, may contain suspending agents, as for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar and tragacanth, or mixtures of these substances, and the like.
  • suspending agents as for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar and tragacanth, or mixtures of these substances, and the like.
  • compositions for rectal administrations are, for example, suppositories that can be prepared by mixing the compounds of the present invention with for example suitable non- irritating excipients or carriers such as cocoa butter, polyethyleneglycol or a suppository wax, which are solid at ordinary temperatures but liquid at body temperature and therefore, melt while in a suitable body cavity and release the active component therein.
  • suitable non- irritating excipients or carriers such as cocoa butter, polyethyleneglycol or a suppository wax, which are solid at ordinary temperatures but liquid at body temperature and therefore, melt while in a suitable body cavity and release the active component therein.
  • composition to be administered will, in any event, contain a therapeutically effective amount of a compound of the invention, or a pharmaceutically acceptable salt thereof, for treatment of a disease-state in accordance with the teachings of this invention.
  • the compounds of the invention are administered in a therapeutically effective amount which will vary depending upon a variety of factors including the activity of the specific compound employed, the metabolic stability and length of action of the compound, the age, body weight, general health, sex, diet, mode and time of administration, rate of excretion, drug combination, the severity of the particular disease-states, and the host undergoing therapy.
  • the compounds of the present invention can be administered to a patient at dosage levels in the range of about 0.1 to about 1,000 mg per day. For a normal human adult having a body weight of about 70 kilograms, a dosage in the range of about 0.01 to about 100 mg per kilogram of body weight per day is an example. The specific dosage used, however, can vary.
  • the dosage can depend on a number of factors including the requirements of the patient, the severity of the condition being treated, and the pharmacological activity of the compound being used.
  • the determination of optimum dosages for a particular patient is well known to one of ordinary skill in the art.
  • assays may be used for this purpose, including labeled in vitro protein-protein binding assays, electrophoretic mobility shift assays, immunoassays for protein binding, functional assays (phosphorylation assays, etc.) and the like.
  • a labeled agent for example a compound of the invention in which at least one atom has been replaced by a detectable isotope
  • washing off excess reagent for example a compound of the invention in which at least one atom has been replaced by a detectable isotope
  • Various blocking and washing steps may be utilized as is known in the art.
  • c-Met, KDR, c-Kit, flt-3, or flt-4 protein may be labeled at tyrosine positions using 125 I, or with fluorophores.
  • more than one component may be labeled with different labels; using I for the proteins, for example, and a fluorophor for the candidate agents.
  • the compounds of the invention may also be used as competitors to screen for additional drug candidates.
  • "Candidate bioactive agent” or “drug candidate” or grammatical equivalents as used herein describe any molecule, e.g., protein, oligopeptide, small organic molecule, polysaccharide, polynucleotide, etc., to be tested for bioactivity. They may be capable of directly or indirectly altering the cellular proliferation phenotype or the expression of a cellular proliferation sequence, including both nucleic acid sequences and protein sequences. In other cases, alteration of cellular proliferation protein binding and/or activity is screened. In the case where protein binding or activity is screened, some embodiments exclude molecules already known to bind to that particular protein.
  • Candidate agents can encompass numerous chemical classes, though typically they are organic molecules having a molecular weight of more than about 100 daltons and less than about 2,500 daltons.
  • Candidate agents comprise functional groups necessary for structural interaction with proteins, particularly hydrogen bonding and lipophilic binding, and typically include at least an amine, carbonyl, hydroxyl, ether, or carboxyl group, for example at least two of the functional chemical groups.
  • the candidate agents often comprise cyclical carbon or heterocyclyl structures and/or aromatic or polyaromatic structures substituted with one or more of the above functional groups.
  • Candidate agents are also found among biomolecules including peptides, saccharides, fatty acids, steroids, purines, pyrimidmes, derivatives, structural analogs, or combinations thereof.
  • Candidate agents are obtained from a wide variety of sources including libraries of synthetic or natural compounds. For example, numerous means are available for random and directed synthesis of a wide variety of organic compounds and biomolecules, including expression of randomized oligonucleotides. Alternatively, libraries of natural compounds in the form of bacterial, fungal, plant and animal extracts are available or readily produced. Additionally, natural or synthetically produced libraries and compounds are readily modified through conventional chemical, physical and biochemical means. Known pharmacological agents may be subjected to directed or random chemical modifications, such as acylation, alkylation, esterification, amidification to produce structural analogs.
  • the binding of the candidate agent is determined through the use of competitive binding assays.
  • the competitor is a binding moiety known to bind to c-Met, KDR, c-Kit, flt-3, or flt-4, such as an antibody, peptide, binding partner, ligand, etc.
  • the candidate agent is labeled. Either the candidate agent, or the competitor, or both, is added first to for example c-Met, KDR, c-Kit, flt-3, or flt-4 for a time sufficient to allow binding, if present. Incubations may be performed at any temperature that facilitates optimal activity, typically between 4°C and 40°C
  • Incubation periods are selected for optimum activity, but may also be optimized to facilitate rapid high throughput screening. Typically between 0.1 and 1 hour will be sufficient. Excess reagent is generally removed or washed away. The second component is then added, and the presence or absence of the labeled component is followed, to indicate binding.
  • the candidate agent is added first, with incubation and washing, followed by the competitor.
  • the absence of binding by the competitor may indicate the candidate agent is bound to c-Met, KDR, c-Kit, flt-3, or flt-4 with a higher affinity.
  • the candidate agent is labeled, the presence of the label on the support, coupled with a lack of competitor binding, may indicate the candidate agent is capable of binding to c-Met, KDR, c-Kit, flt-3, or flt-4.
  • c-Met, KDR, c-Kit, flt-3, or flt-4 it may be of value to identify the binding site of c-Met, KDR, c-Kit, flt-3, or flt-4. This can be done in a variety of ways. In one embodiment, once c-Met, KDR, c-Kit, flt-3, or flt-4 has been identified as binding to the candidate agent, the c-Met, KDR, c-Kit, flt-3, or flt-4 is fragmented or modified and the assays repeated to identify the necessary components for binding.
  • differential screening may be used to identify drag candidates that bind to native c-Met, KDR, c-Kit, flt-3, or flt-4, but cannot bind to modified c-Met, KDR, c-Kit, flt-3, or flt-4.
  • Positive controls and negative controls may be used in the assays. For example, all control and test samples are performed in at least triplicate to obtain statistically significant results. Incubation of samples is for a time sufficient for the binding of the agent to the protein. Following incubation, samples are washed free of non-specifically bound material and the amount of bound, generally labeled agent determined. For example, where a radiolabel is employed, the samples may be counted in a scintillation counter to determine the amount of bound compound.
  • a variety of other reagents may be included in the screening assays. These include reagents like salts, neutral proteins, e.g., albumin, detergents, etc which may be used to facilitate optimal protein-protein binding and/or reduce non-specific or background interactions. Also reagents that otherwise improve the efficiency of the assay, such as protease inhibitors, nuclease inhibitors, anti-microbial agents, etc., may be used. The mixture of components may be added in any order that provides for the requisite binding.
  • Schemes 1 and 2 depict general synthetic routes for compounds of the invention and are not intended to be limiting. More specifically, Scheme 1 depicts synthesis of quinazoline compounds, and Scheme 2 depicts synthesis of quinoline compounds. Specific examples are described subsequently to these general synthetic descriptions so as to allow one skilled in the art to make and use either quinazolines or quinolines of the invention.
  • Aromatic ring nitration and reduction of the corresponding nitro group are carried out in a regio- and chemoselective manner by methods well known in the art to give anthranilate derivative 3.
  • Formation of quinazolin-4-one 4 is carried out by methods well known in the art, for example by heating 3 in formamide solution in the presence of ammonium formate or for example by heating directly with formamidine hydrochloride.
  • Introduction of 4-position functionality groups is carried out by methods known in the art. For example, quinazolin-4-one 4 is converted to an intermediate quinazoline 5, where "L" represents a leaving group, e.g. chlorine. Quinazoline 5 is then converted to 6 by reaction with a range of nucleophiles, e.g.
  • group "Z” is either left “as is” or converted at some subsequent stage to a derivative thereof.
  • Z is -NH-
  • the hydrogen on the nitrogen may optionally be replaced with an alkyl group, or when Z is sulfur, then that sulfur atom may be oxidized to, for example, a sulfone.
  • Stracture 6 may represent compounds of the invention or, for example when E 1 serves as a protecting group, E 1 may be removed to provide phenol 7.
  • Introduction of a group E 2 is carried out by methods well established in the art; for example alkylation with an appropriately derivatized alkyl halide (or mesylate or the like) to give 8 which also represents compounds of the invention.
  • Scheme 2 shows a general route used to make exemplary quinolines of the invention.
  • compound 9 contains an alkyl group, R 1 , a protecting group, P.
  • the arrangement of the protected and alkylated phenolic oxygens may vary from the pattern depicted in compound 9.
  • Compound 9 is nitrated to provide compound 10.
  • the nitro group of compound 10 is reduced to give aniline 11.
  • Compound 11 is treated, for example, with ethyl formate under basic conditions followed by acidification and isolation to form 4-hydroxy quinoline 12.
  • Quinoline 12 may be converted to compounds of the invention in a number of ways.
  • the 4-oxygen is used as a nucleophile in a nucleophilic aromatic substitution reaction to form quinoline-aryl-ether 13.
  • compound 13 is further derivatized, via removal of protecting group P, to afford compound 14.
  • the 7-hydroxy of compound 14 is alkylated, for example with electrophile E, to provide a compound of the invention.
  • compounds 12, 13, and 14 may also be compounds of the invention according to formula I.
  • the 4-hydroxy quinoline compound 12 are converted to a corresponding 4-nitrogen or 4-sulfur quinoline using chemistry known in the art to make compounds of the invention, or alternatively the corresponding 4-nitrogen or 4-sulfur quinolines are made via routes analogous to that depicted in Schemes 1 and 2.
  • Schemes 1 and 2 are illustrative of quinolines and quinazolines having oxygen substitution at their respective 6- and 7-positions; the invention is not so limited, but rather is intended to encompass quinolines and quinazolines not necessarily having substitution, oxygen or otherwise, at their respective 6- or 7-positions.
  • a benzoic ester 16 for example, where R is typically but not necessarily a methyl radical and R 1 is typically but not necessarily one or more alkoxy or hydroxy groups.
  • R 1 within Scheme 3 is a hydroxyl which is converted (or protected )via one or more steps to a group important to the activity of the compounds as described as kinase modulators (in the case that -OH itself is desired in the final compound, then deprotection affords the -OH, vide supra).
  • this group is complete once the synthesis of XXII is complete.
  • Scheme 4 shows a general route used to make compounds of formula XXIII.
  • aromatic compound 19, where "X” is a leaving group, such as fluorine and "E” is an electron withdrawing group such as nitro is converted to 20 by reaction with a range of nucleophiles, e.g. amines, alcohols, and thiols (where "Z” is oxygen, nitrogen (substituted or not), or sulfur).
  • nucleophiles e.g. amines, alcohols, and thiols (where "Z” is oxygen, nitrogen (substituted or not), or sulfur.
  • R represents a removable group, for example benzyl.
  • group "E” is either left “as is” or converted at some subsequent stage to a derivative thereof.
  • E is converted to B', a precursor to B in accord with formula XXI, to make 21.
  • B' could might be an amino group, made via reduction of the nitro group.
  • Structure 21 may be further derivitized by synthesis of -B-L-T in accord with formula XXI.
  • scheme 4 this is depicted as a serial process whereby L', a precursor to L, is introduced to give 22, followed by introduction of T' (a precursor to T) to give 23.
  • T' a precursor to T
  • -L-T is preformed and appended to B.
  • Compound 23 is converted to XXIII via conversion of T' to T and introduction of P 2 (for example, when R is benzyl, removal of the benzyl after completion of -B-L-T).
  • one aspect of the invention encompasses combination of XXII and XXIII to make compounds of formula XXI. Because of the diversity and complexity of compounds described for kinase modulation (vide supra), methods of the invention provide advantages to serial synthesis.
  • reaction mixture was then cooled to 0° C and 37% solution of formaldehyde in water was added (0.2 g, 7.8 mmol, 2.0 eq). While keeping the temperature at 0°C Na(OAc) 3 BH was added (4.4g, 20.7 mmol, 3.0 eq). After 1 hour the pH was adjusted to 10 and the aqueous was extracted 2 x DCM (100 ml). Removal of the DCM resulted in a white solid.
  • reaction mixture was stirred at room temperature for 1-2 hrs, then diluted with EtOAc and washed with sat'd NaHCO 3 (3x), H 2 O (lx), sat'd NaCl (lx), dried (Na 2 SO ), and concentrated in vacuo to give crude 5-[4-(4-amino-2-fluoro-phenoxy)-6- methoxy-quinazolin-7-yloxymethyl]-hexahydro-cyclopenta[c]pyrrole-2-carboxylic acid benzyl ester (5.6g, -100%) which was used in the next reaction without further purification.
  • reaction mixture was stirred at room temperature for 1 hr, diluted with Et 2 O (1000ml) and the resulting solids filtered, washed with Et 2 O, and dried under high vacuum to give the l- ⁇ 3- fluoro-4-[6-methoxy-7-(octahydro-cyclopenta[c]pyrrol-5-ylmethoxy)-quinazolin-4- yloxy] -phenyl ⁇ -3 -phenylacetyl-thiourea, dihydrobromide salt (3.4g, 100%).
  • Dimethoxy-quinazolin-4-yl)-(2-fluoro-4-nitro-phenyl)-amine (673mg, 1.95mmol) was dissolved in a combination of DMF (20ml) and MeOH (20ml), to which was added 10% Pd/C (227mg). The mixture was shaken under an atmosphere of H 2 on a Parr hydrogenator at 40psi for 3hrs. The reaction mixture was filtered through celite and the filtrate concentrated in vacuo. The resulting residue was triturated in EtOAc/Et 2 O.
  • the Pd/C (10% by weight) (0.090 g, 20% by weight) was then added.
  • a balloon filled with H 2 was connected to the flask after the nitrogen was vacuumed out.
  • the reaction mixture was stirred at room temperature for 4 hours.
  • the palladium was filtered out through Celite, and the filtrated was collected and concentrated via rotary evaporation.
  • the resulting oil-like product was taken up into 5 mL of water and 1 mL of acetonitrile and lyophilized to yield 6-(6,7-dimethoxy-quinolin-4-yloxy)-pyridin-3-ylamine as a light brown solid (0.411 g, 98.1%).
  • N-r4-(6,7-Dimethoxy-quinolin-4-yloxy)-3-fluoro-phenvn-N'-phenethyl-oxalamide To a solution of 4-(6,7-dimethoxy-quinolin-4-yloxy)-3-fluoro-phenylamine (263 mg, 0.83 mmol) and Et 3 N (0.223 ml, 1.67 mmol) in CH 2 C1 2 (10 mL) was added dropwise a solution of ethyl oxalyl chloride in CH 2 C1 2 (1 mL). The stirring was continued for 0.5 h at it.
  • N- ⁇ 3-Fluoro-4-[6-methoxy-7-(l-methyl-piperidin-4-ylmethoxy)-quinolin-4- yloxy1-phenyl ⁇ -N'-phenethyl-oxalamide To a flask containing 7-benzyloxy-4-(2-fluoro-4- nitro-phenoxy)-6-methoxy-quinoline (850 mg, 2.0 mmol) was added 20 mL of 30% HBr in AcOH. The resulted solution was stirred for 4 h at rt; at this time, a large amount of precipitate formed. The crude product was filtered, washed with Et 2 O and dried in air, giving 4-(2-fluoro-4-nitro-phenoxy)-6-methoxy-7-hydroxyquinoline (609 mg, 92% yield).
  • Trifluoromethanesulfonic acid 7-benzyloxy-6-methoxy-quinolin-4-yl ester To a dry 2L RBF containing 7-benzyloxy-6-methoxyquinolin-4-ol (75.3 g, 267 mmol) was added DCM (1 L), 4-dimethylaminopyridine (3.28 g, 26.8 mmol) and 2,6-lutidine (62 mL, 534 mmol). The mixture was cooled to -20°C by controlled addition of dry ice to an acetone bath. Trifluoromethanesulfonyl chloride (37 mL, 350 mmol) was added dropwise to the cooled solution with magnetic stirring over 25 minutes.
  • Trifluoromethanesulfonyl chloride 14 mL, 132 mmol was added dropwise to the solution. After addition was complete, the mixture was stirred ice bath for 2 to 3 hrs. On LC/MS indicating the reaction completion, the reaction mixture was concentrated in vacuo and placed under high vacuum to remove residual 2,6-lutidine. To the resulting brown solids was added methanol (250 mL). The resulting slurry was stirred for 30 min before adding water (1 L). The solids were isolated by filtration, followed by a water wash.
  • Ethyl [(4-benzyloxy-3-fluorophenyl)amino1(oxo)acetate Ethyl oxalyl chloride (44 mL, 390 mmol) was added to a solution of 4-benzyloxy-3-fluoroaniline (44 g, 180 mmol) in diisopropylethylamine (69 mL, 400 mmol) and stirred at room temperature for 15 min. The mixture was extracted with dichloromethane and washed with water and brine.
  • N-(4-Benzyloxy-3-flourophenyl)-N'-(2-phenylethyl)ethanedi amide Phenethylamine (33 mL, 520 mmol) was added to ethyl [(4-benzyloxy-3- fluorophenyl)amino](oxo)acetate (81 g, 260 mmol) and the mixture was sonicated at room temperature for 30 min. The resulting solid was filtered, washed with water and dried to give N-(4-benzyloxy-3-flourophenyl)-N'-(2-phenylethyl)ethanediamide (100 g, 99 %).
  • N-(3-Fluoro-4-hyckoxyphenyl)-N'-(2-phenylethyl)ethanediamide A mixture of N- (4-benzyloxy-3-flourophenyl)-N , -(2-phenylethyl)ethanediamide (40 g, 100 mmol) and 38% hydrobromie acid in acetic acid (250 mL) was stirred at room temperature overnight. The resulting solid was filtered, washed with water and dried to give N-(3-fluoro-4- hydroxyphenyl)-N'-(2-phenylethyl)ethanediamide as a slightly yellow solid (30.6 g, 99 %yield).

Abstract

The present invention provides compounds for modulating protein kinase enzymatic activity for modulating cellular activities such as proliferation, differentiation, programmed cell death, migration and chemoinvasion. More specifically, the invention provides quinazolines and quinolines which inhibit, regulate and/or modulate kinase receptor, particularly c-Met, KDR, c-Kit, flt-3 and flt-4, signal transduction pathways related to the changes in cellular activities as mentioned above, compositions which contain these compounds, and methods of using them to treat kinase-dependent diseases and conditions. The present invention also provides methods for making compounds as mentioned above, and compositions which contain these compounds.

Description

c-Met Modulators and Method of Use
CROSS-REFERENCE TO RELATED APPLICATIONS
[0001] This application claims priority to U.S. provisional patent applications 60/506,181 filed on September 26, 2003, entitled "c-Met Modulators and Method of Use," naming Bannen, Lynne et. al as inventors, 60/535,377 filed on January 9, 2004, entitled "c-Met Modulators and Method of Use," naming Bannen, Lynne et. al as inventors and 60/577,384 filed on June 4, 2004, entitled "Synthesis of Quinoline and Quinazoline Kinase Modulators," naming Bannen, Lynne et. al as inventors; each of which is hereby incorporated by reference in its entirety for all purposes.
FIELD OF THE INVENTION
[0002] This invention relates to compounds for modulating protein kinase enzymatic activity for modulating cellular activities such as proliferation, differentiation, programmed cell death, migration and chemoinvasion. Even more specifically, the invention relates to quinazolines and quinolines which inhibit, regulate and/or modulate kinase receptor signal transduction pathways related to the changes in cellular activities as mentioned above, compositions which contain these compounds, methods of using them to treat kinase-dependent diseases and conditions, synthesis of the compounds as well as processes for formulating the compounds for pharmaceutical purposes.
BACKGROUND OF THE INVENTION [0003] improvements in the specificity of agents used to treat cancer is of considerable interest because of the therapeutic benefits which would be realized if the side effects associated with the administration of these agents could be reduced. Traditionally, dramatic improvements in the treatment of cancer are associated with identification of therapeutic agents acting through novel mechanisms.
[0004] Protein kinases are enzymes that catalyze the phosphorylation of proteins, in particular, hydroxy groups on tyrosine, serine and threonine residues of proteins. The consequences of this seemingly simple activity are staggering; cell differentiation and proliferation; i.e., virtually all aspects of cell life in one-way or another depend on protein kinase activity. Furthermore, abnormal protein kinase activity has been related to a host of disorders, ranging from relatively non-life threatening diseases such as psoriasis to extremely virulent diseases such as glioblastoma (brain cancer).
[0005] Protein kinases can be categorized as receptor type or non-receptor type. Receptor- type tyrosine kinases have an extracellular, a transmembrane, and an intracellular portion, while non-receptor type tyrosine kinases are wholly intracellular.
[0006] Receptor-type tyrosine kinases are comprised of a large number of transmembrane receptors with diverse biological activity. In fact, about 20 different subfamilies of receptor-type tyrosine kinases have been identified. One tyrosine kinase subfamily, designated the HER subfamily, is comprised of EGFR (HER1), HER2, HER3, and HER4. Ligands of this subfamily of receptors identified so far include epithelial growth factor, TGF-alpha, amphiregulin, HB-EGF, betacellulin and heregulin. Another subfamily of these receptor-type tyrosine kinases is the insulin subfamily, which includes L S-R, IGF- LR, and IR-R. The PDGF subfamily includes the PDGF-alpha and beta receptors, CSFLR, c-Kit and FLK-JJ. Then there is the FLK family, which is comprised of the kinase insert domain receptor (KDR), fetal liver kinase-1 (FLK-1), fetal liver kinase-4 (FLK-4) and the fms-like tyrosine kinase-1 (flt-1). The PDGF and FLK families are usually considered together due to the similarities of the two groups. For a detailed discussion of the receptor- type tyrosine kinases, see Plowman et al., DN&P 7(6): 334-339, 1994, which is hereby incorporated by reference.
[0007] The non-receptor type of tyrosine kinases is also comprised of numerous subfamilies, including Src, Frk, Btk, Csk, Abl, Zap70, Fes/Fps, Fak, Jak, Ack, and LLMK. Each of these subfamilies is further sub-divided into varying receptors. For example, the Src subfamily is one of the largest and includes Src, Yes, Fyn, Lyn, Lck, Blk, Hck, Fgr, and Yrk. The Src subfamily of enzymes has been linked to oncogenesis. For a more detailed discussion of the non-receptor type of tyrosine kinases, see Bolen, Oncogene, 8:2025-2031 (1993), which is hereby incorporated by reference.
[0008] Since protein kinases and their ligands play critical roles in various cellular activities, deregulation of protein kinase enzymatic activity can lead to altered cellular properties, such as uncontrolled cell growth associated with cancer. In addition to oncological indications, altered kinase signaling is implicated in numerous other pathological diseases. These include, but are not limited to: immunological disorders, cardiovascular diseases, inflammatory diseases, and degenerative diseases. Therefore, both receptor and non-receptor protein kinases are attractive targets for small molecule drug discovery.
[0009] One particularly attractive goal for therapeutic use of kinase modulation relates to oncological indications. For example, modulation of protein kinase activity for the treatment of cancer has been demonstrated successfully with the FDA approval of Gleevec® (imatinib mesylate, produced by Novartis Pharmaceutical Corporation of East Hanover, NJ) for the treatment of Chronic Myeloid Leukemia (CML) and gastrointestinal stroma cancers (GIST). Gleevec is a c-Kit and Abl kinase inhibitor.
[0010] Modulation (particularly inhibition) of cell proliferation and angiogenesis, two key cellular processes needed for tumor growth and survival (Matter A. Drug Disc Technol 2001 6, 1005-1024), is an attractive goal for development of small-molecule drugs. Anti- angiogenic therapy represents a potentially important approach for the treatment of solid tumors and other diseases associated with dysregulated vascularization, including ischemic coronary artery disease, diabetic retinopathy, psoriasis and rheumatoid arthritis. As well, cell antiproliferative agents are desirable to slow or stop the growth of tumors.
[0011] One particularly attractive target for small-molecule modulation, with respect to antiangiogenic and antiproliferative activity is c-Met. The kinase, c-Met, is the prototypic member of a subfamily of heterodimeric receptor tyrosine kinases (RTKs) which include Met, Ron and Sea. Expression of c-Met occurs in a wide variety of cell types including epithelial, endothelial and mesenchymal cells where activation of the receptor induces cell migration, invasion, proliferation and other biological activities associated with "invasive cell growth." As such, signal transduction through c-Met receptor activation is responsible for many of the characteristics of tumor cells.
[0012] The endogenous ligand for c-Met is the hepatocyte growth factor (HGF), a potent inducer of angiogenisis, also known as "scatter factor" (SF). Binding of HGF to c-Met induces activation of the receptor via autophosphorylation resulting in an increase of receptor dependent signaling, which promotes cell growth and invasion. Anti-HGF antibodies or HGF antagonists have been shown to inhibit tumor metastasis in vivo (See: Maulik et al Cytokine & Growth Factor Reviews 2002 13, 41-59).
[0013] Tumor growth progression requires the recruitment of new blood vessels into the tumor from preexisting vessels as well as invasion, adhesion and proliferation of malignant cells. Accordingly, c-Met overexpression has been demonstrated on a wide variety of tumor types including breast, colon, renal, lung, squamous cell myeloid leukemia, hemangiomas, melanomas, astrocytomas, and glioblastomas. Additionally activating mutations in the kinase domain of c-Met have been identified in hereditary and sporadic renal papilloma and squamous cell carcinoma. (See: Maulik et al Cytokine & growth Factor reviews 2002 13, 41-59; Longati et al Curr Drug Targets 2001, 2, 41-55; Funakoshi et al Clinica Chimica Acta 2003 1-23). Thus modulation of c-Met is desirable as a means to treat cancer and cancer-related disease.
[0014] The Eph receptors comprise the largest family of receptor tyrosine kinases and are divided into two groups, EphA and EphB, based on their sequence homology. The ligands for the Eph receptors are ephrin, which are membrane anchored. Ephrin A ligands bind preferentially to EphA receptors whilst ephrin B ligands bind to EphB receptors. Binding of ephrin to Eph receptors causes receptor autophosphorylation and typically requires a cell-cell interaction since both receptor and ligand are membrane bound.
[0015] Overexpression of Eph receptors has been linked to increased cell proliferation in a variety of tumors (Zhou R 1998 Pharmacol Ther. 77, 151-181; Kiyokawa E, Takai S, Tanaka M et al 1994 Cancer Res 54, 3645-3650; Takai N Miyazaki T, Fujisawa K, Nasu K and Miyakawa. 2001 Oncology reports 8, 567-573). The family of Eph receptor tyrosine kinases and their ephrin ligands play important roles in a variety of processes during embryonic development and also in pathological angiogenesis and potentially metastasis. Therefore modulation of Eph receptor kinase activity should provide means to treat or prevent disease states associated with abnormal cell proliferation such as those described above.
[0016] Inhibition of EGF, VEGF and ephrin signal transduction will prevent cell proliferation and angiogenesis, two key cellular processes needed for tumor growth and survival (Matter A. Drug Disc. Technol. 2001 6, 1005-1024). EGF and VEGF receptors are previously described targets for small molecule inhibition. KDR and flt-4 are both VEGF receptors
[0017] One particularly attractive target for small-molecule modulation is c-Kit. The proto-oncogene c-Kit was first identified as the oncogenic component of the acutely transforming Hardy-Zuckerman 4-feline sarcoma virus (Besmer et al Nature 1986 320:415-421). c-Kit (also called stem cell factor receptor or steel factor receptor) is a type 3 receptor tyrosine kinase (RTK) belonging to the platelet-derived growth factor receptor subfamily. c-Kit binds the ligand stem cell factor (SCF), and triggers its multiple signal transduction pathways including Src family kinases, phosphatidyl-inositol 3 kinase, the Ras-Raf-Map kinase cascade, and phospholipase C (Broudy et al Blood 1999 94: 1979- 1986; Lennartsson et al Oncogene 1999 18: 5546-5553 ; Timokhina et al EMBO J 1998 17;6250-6262; Chian et al Blood 2001 98(5)1365-1373; Blume-Jensen et al Curr Biol
1998 8:779-782; Kissel et al EMBO J 2000 19:1312-1326; Lennartsson et al. Oncogene
1999 18: 5546-5553; Sue et al Blood, 199892:1242-1149; Lev etal EMBO J 1991 10:647- 654). c-Kit is required for normal hematopoiesis, melanonogenesis, and gametogenesis. c-Kit is expressed in mast cells, immature myeloid cells, melanocytes, epithelial breast cells and the interstitial cells of Cajal (ICC). In mast cells, it is required not only for the differentiation, maturation, chemotaxis, and haptotaxis but also for the promotion of survival and proliferation.
[0018] Mutations in c-Kit have been implicated in human disease. Mutations in the juxtamembrane domain are found in many human gastrointestinal stromal tumors, and mutations in the kinase domain are found in mastocytosis, germ cell tumors, acute myeloid leukemia (AML), NK lymphoma, and other hematologic disorders (Hirota et al Science 1998 279:577-580; Singer et al J Clin Oncol 2002 203898-3905; Longley et al Proc Natl Aca Sci USA 1999: 1609-1614; Tian et al Am J Pathol 1999 154: 1643-1647; Beghini et al Blood 2000 95:726-727; Hongyo et al Cancer Res 2000 60:2345-2347). These mutations result in ligand-independent tyrosine kinase activity, autophosphorylation of c- Kit, uncontrolled cell proliferation, and stimulation of downstream signaling pathways. Overexpression of c-Kit and c-Kit ligand have also been described in other tumors including small-cell lung cancer, neuroblastomas, gynecological tumors, and colon carcinoma, which might result in autocrine or paracrine c-Kit activation.
[0019] The overexpression of c-Kit has also been implicated in the development of neoplasia associated with neurofibromatosis type 1 (NF1). Mutations in the tumor suppressor gene NFI lead to a deficiency in neurofibromin, a GTPase-activating protein for Ras. This deficiency results in abnormal proliferation of Schwann cells in the peripheral nervous system, and predisposes affected individuals to peripheral nerve sheath tumors (neurofibromas), astrocytomas (optic pathway gliomas), learning disabilities, seizures, strokes, macrocephaly, vascular abnormalities, and juvenile myelomonocytic leukemia (Lynch & Gutmann Neurol Clin 200220:841-865). Genetic experiments in mice demonstrate that haploinsufficiency at NFI partially rescues some of the phenotypes associated with mutations in the gene for c-Kit, indicating that these genes function along a common developmental pathway (Ingram, et al. J. Exp Med 2000 191:181-187). Also, c- Kit is expressed in schwannoma cells from NFI patients, but not in normal schwann cells (Ryan et al. J Neurosci Res 1994 37:415-432). These data indicate that elevated c-Kit expression and sensitivity to stem cell factor may play important roles in the development of proliferative disorders associated with NF-1. Therefore, c-Kit inhibitors may be effective chemotherapeutic agents for treating patients with NF-1.
[0020] GISTs are the most common mesenchymal tumors of the gastrointestinal tract, and they are generally resistant to chemotherapy and radiation therapy. However, recent results with the c-Kit/BCR-Abl inhibitor STI571 indicate that targeting c-Kit may be an effective therapeutic strategy for this disease (Eisenberg & Mehren Expert Opin Pharmacother 2003 4:869-874). Malignant mast cell disease often suggests an extremely poor prognosis, and no reliable effective chemotherapeutic agents have been identified (Marone et al Leuk Res 2001 25:583-594). Systemic mast cell disorders have been treated with interferon-alpha, although the effectiveness of this therapy has been variable (Lehmann & Lammle Ann Hematol 1999 78:483-484; Butterfield Br J Dermatol 1998 138: 489-495). Therefore, activated c-Kit might serve as a therapeutic target in GISTs and mast cell disease, as well as other disorders associated with activated c-Kit.
[0021] Flt-3 is normally expressed on hematopoietic progenitor cells and a subset of mature myeloid and lymphoid cells, where it modulates cell survival and proliferation. Flt-3 is constitutively activated via mutation, either in the juxtamembrane region or in the activation loop of the kinase domain, in a large proportion of patients with AML (Reilly Leuk Lymphoma 2003 44: 1-7). Also, mutations in flt-3 are significantly correlated with poor prognosis in AML patients (Sawyers Cancer Cell 2002 1: 413-415).
[0022] Accordingly, the identification of small-molecule compounds that specifically inhibit, regulate and/or modulate the signal transduction of kinases, particularly including c-Met, KDR, c-Kit, flt-3, and flt-4, is desirable as a means to treat or prevent disease states associated with abnormal cell proliferation and angiogenesis, and is an object of this invention.
[0023] Quinolines and quinazolines bearing substitution, for example at the two, four, six and seven positions of their fused ring system have been shown to be particularly attractive targets for kinase inhibition by a number of groups. Conventional quinoline and quinazoline kinase inhibitors typically have fairly simple substitution about the quinoline or quinazoline fused ten-membered ring system, but recently more complex molecules are being disclosed. For example, we have previously disclosed, in U.S. provisional patent applications 60/506,181 and 60/535,377 which are both incorporated by reference herein in their entirety for all purposes, that certain quinolines and quinazolines are particularly well suited as kinase modulators, more particularly inhibitors of for example c-Met, KDR, c-Kit, flt-3, and flt-4. These molecules in some cases are particularly complex and although they can be made via conventional methods, more efficient routes are desirable, especially in a pharmaceutical setting.
[0024] Conventional methods of making quinolines and quinazolines with the aforementioned substitution patterns usually involve linear construction of a quinoline or quinazoline template upon which relatively simple substitutions are appended. With the advent of more complex substitution about such quinolines and quinazolines (vide supra), for example side chains containing cyclic and bicyclic systems with multiple functional groups, conventional methods of synthesis become problematic due to the linear or serial reactions used. Indeed, as such molecules become more complex and the utility of such complex groups is realized, the quinoline and quinazoline ring system becomes more of a sub-structure than a main structure of such inhibitors. Thus it is desirable to find more efficient methods of synthesis, particularly convergent syntheses which are an object of this invention.
SUMMARY OF THE INVENTION
[0025] In one aspect, the present invention provides compounds for modulating kinase activity and methods of treating diseases mediated by kinase activity utilizing the compounds and pharmaceutical compositions thereof. Diseases mediated by kinase activity include, but are not limited to, diseases characterized in part by migration, invasion, proliferation and other biological activities associated with invasive cell growth. In particular to this invention is modulation, even more particularly inhibition, of c-Met, KDR, c-Kit, flt-3, and flt-4.
[0026] In another aspect, the invention provides methods of screening for modulators of c- Met, KDR, c-Kit, flt-3, and flt-4 activity. The methods comprise combining a composition of the invention, a kinase, e.g. c-Met, KDR, c-Kit, flt-3, or flt-4, and at least one candidate agent and determining the effect of the candidate agent on the c-Met, KDR, c-Kit, flt-3, or flt-4, activity.
[0027] In yet another aspect, the invention also provides pharmaceutical kits comprising one or more containers filled with one or more of the ingredients of pharmaceutical compounds and/or compositions of the present invention, including, one or more kinase, e.g. c-Met, KDR, c-Kit, flt-3, or flt-4, enzyme activity modulators as described herein. Such kits can also include, for example, other compounds and/or compositions (e.g., diluents, permeation enhancers, lubricants, and the like), a device(s) for administering the compounds and/or compositions, and written instructions in a form prescribed by a governmental agency regulating the manufacture, use or sale of pharmaceuticals or biological products, which instructions can also reflects approval by the agency of manufacture, use or sale for human administration.
[0028] In another aspect, the invention also provides a diagnostic agent comprising a compound of the invention and, optionally, pharmaceutically acceptable adjuvants and excipients.
[0029] In still yet another aspect, the present invention provides processes for making compounds, and pharmaceutical compositions thereof, for modulating kinase activity and treating diseases mediated by kinase activity. In particular to this invention are methods for making quinolines and quinazolines used for modulation of kinase activity, even more particularly inhibition of kinase activity, and yet even more particularly inhibition of c- Met, KDR, c-Kit, flt-3, and flt-4.
[0030] These and other features and advantages of the present invention will be described in more detail below with reference to the associated drawings.
DETAILED DESCRD?TION OF THE INVENTION
[0031] The compositions of the invention are used to treat diseases associated with abnormal and or unregulated cellular activities. Disease states which can be treated by the methods and compositions provided herein include, but are not limited to, cancer (further discussed below), immunological disorders such as rheumatoid arthritis, graft-host diseases, multiple sclerosis, psoriasis; cardiovascular diseases such as artheroscrosis, myocardioinfarction, ischemia, stroke and restenosis; other inflammatory and degenerative diseases such as interbowel diseases, osteoarthritus, macular degeneration, diabetic retinopathy.
[0032] It is appreciated that in some cases the cells may not be in a hyper- or hypo- proliferative and/or migratory state (abnormal state) and still require treatment. For example, during wound healing, the cells may be proliferating "normally", but proliferation and migration enhancement may be desired. Alternatively, reduction in "normal" cell proliferation and/or migration rate may be desired. 33] Thus, in one aspect the present invention comprises a compound for modulating kinase activity according to Formula I,
Figure imgf000010_0001
or a pharmaceutically acceptable salt, hydrate, or prodrug thereof, wherein,
R1 is selected from -H, halogen, -OR3, -NO2, -NH2, -NR3R4, and optionally substituted lower alkyl;
A1 is selected from =N-, =C(H)-, and =C(CN)-;
Z is selected from -S(O)0-2-, -O-, and -NR5-;
Ar is either a group of formula II, or of formula III,
Figure imgf000010_0002
II III wherein,
R2 is selected from -H, halogen, trihalomethyl, -CN, -NO2, -NH2, -OR3, -NR3R4, -S(O)0-2R3, -SO2NR3R3, -CO2R3, -C(O)NR3R3, -N(R3)SO2R3, -N(R3)C(O)R3, -N(R3)CO2R3, -C(O)R3, and optionally substituted lower alkyl; q is 0 to 4;
G is a group -B-L-T, wherein
B is selected from absent, -N(R13)-, -N(SO2R13)-, -O-, -S(O)0-2-, and -C(=O)-;
L is selected from absent, -C(=S)N(R13)-, -C(=NR14)N(R13)-, -SO2N(R13)-, -SO2-, -C(=O)N(R13)-, -N(R13)-, -C(=O)C1-2alkylN(R13)-, -N(R13)C1-2alkylC(=O)-, -C(=O)C0-1alkylC(=O)N(R13)-, -C0-4alkylene-, -C(=O)C0-1alkylC(=O)OR3-, -C(=NR14)C0-1alkylC(=O)-, -C(=O)-, -C(=O)C0-1alkylC(=O)-, and an optionally substituted four to six-membered heterocyclyl containing between one and three annular heteroatoms including at least one nitrogen; and
T is selected from -H, -R13, -C0-4alkyl, -C0-4alkylQ, -OC0-4alkylQ, -C0- alkylOQ, -N(R13)C0-4alkylQ, -SO2C0-4alkylQ, -C(=O)C0-4alkylQ, -C0-4alkylN(R13)Q, and -C(=O)N(R13)Co-4alkylQ, wherein each of the aforementioned Co- alkyl is optionally substituted;
J is selected from -S(O)o-2-, -O-, and -NR15-;
R3 is -H or R4;
R4 is selected from optionally substituted lower alkyl, optionally substituted aryl, optionally substituted lower arylalkyl, optionally substituted heterocyclyl, and optionally substituted lower heterocyclylalkyl; or
R3 and R4, when taken together with a common nitrogen to which they are attached, form an optionally substituted five- to seven-membered heterocyclyl, said optionally substituted five- to seven-me ibered heterocyclyl optionally containing at least one additional annular heteroatom selected from N, O, S, and P;
A2 and A3 are each independently selected from =N-, =C(R2)-;
R5 is -H or optionally substituted lower alkyl;
D is selected from -O-, -S(O)0.2-, and -NR15-;
R50 is either R3, or according to formula IV;
Figure imgf000011_0001
IV wherein X , X , and optionally X , represent the atoms of a saturated bridged ring system, said saturated bridged ring system comprising up to four annular heteroatoms represented by any of X1, X2, and X3; wherein, each X1 is independently selected from -C(R6)R7-, -O-, -S(O)0-2-, and -NR8-; each X2 is independently an optionally substituted bridgehead methine or a bridgehead nitrogen; each X3 is independently selected from -C(R6)R7-, -O-, -S(O)0-2-, and -NR8-;
Y is either: an optionally substituted lower alkylene linker, between D and either 1) any annular atom of the saturated bridged ring system, except X when X is a bridgehead nitrogen, or 2) any heteroatom, represented by any of R6 or R7; provided there are at least two carbon atoms between D and any annular heteroatom of the saturated bridged ring system or any heteroatom represented by any of R6 or R7; or Y is absent, when Y is absent, said saturated bridged ring system, is directly attached to D via an annular carbon of said saturated bridged ring system, unless D is -SO2-, in which case said saturated bridged ring system, is directly attached to D via an any annular atom of said saturated bridged ring system; m and p are each independently 1-4; n is 0-2, when n = 0, then there is a single bond between the two bridgehead X2 's;
R6 and R7 are each independently selected from -H, halogen, trihalomethyl, -CN, -NH2, -NO2, -OR3, -NR3R4, -S(O)o-2R4, -SO2NR3R4, -CO2R3, -C(O)NR3R4, -N(R3)SO2R4, -N(R3)C(O)R3, -NCO2R3, -C(O)R3, optionally substituted lower alkyl, optionally substituted aryl, optionally substituted lower arylalkyl, optionally substituted heterocyclyl, optionally substituted lower heterocyclylalkyl, and a bond to either Y or D; or
R and R , when taken together are oxo; or
R and R , when taken together with a common carbon to which they are attached, form a optionally substituted three- to seven-membered spirocyclyl, said optionally substituted three- to seven-membered spirocyclyl optionally containing at least one additional annular heteroatom selected from N, O, S, and P;
R8 is selected from -R3, Y, -SO2NR3R4, -CO2R4, -C(O)NR3R3, -SO2R4, and -C(O)R3;
R13 is selected from -H, -C(=O)R3, -C(=O)OR3, -C(=O)SR3, -SO2R4, -C(=O)N(R3)R3, and optionally substituted lower alkyl, 1 * two R , together with the atom or atoms to which they are attached, can combine to form a heteroalicyclic optionally substituted with between one and four of R60, said heteroalicyclic can have up to four annular heteroatoms, and said heteroalicyclic can have an aryl or heteroaryl fused thereto, in which case said aryl or heteroaryl is optionally substituted with an additional one to four of R60;
R14 is selected from -H, -NO2, -NH2, -N(R3)R4, -CN, -OR3, optionally substituted lower alkyl, optionally substituted heteroalicyclylalkyl, optionally substituted aryl, optionally substituted arylalkyl and optionally substituted heteroalicyclic;
R15 is a group -M^M2, wherein M1 is selected from absent, -C(=S)N(R13)-, -C(=NR14)N(R13)-, -SO2N(R13)-, -SO2-, -C(=O)N(R13)-, -C(=O)C(=O)N(R13)-, -Co. 4alkylene-, -C(=O)-, and an optionally substituted four to six-membered hetercyclyl annular containing between one and three heteratoms including at least one nitrogen; and M2 is selected from -H, -C0.6alkyl, alkoxy, -C(=O)C0- alkylQ, -C0- alkylQ, -OC0-4alkylQ-, -N(R13)C0-4alkylQ-, and -C(=O)N(R13)C0.4alkylQ; and
Q is a five- to ten-membered ring system, optionally substituted with between zero and four of R20;
R20 is selected from -H, halogen, trihalomethyl, -CN, -NO2, -NH2, -OR3, -NR3R4, -S(O)0-2R3, -SO2NR3R3, -CO2R3, -C(O)NR3R3, -N(R3)SO2R3, -N(R3)C(O)R3, -N(R3)CO2R3, -C(O)R3, and optionally substituted lower alkyl;
R60 is selected from -H, halogen, trihalomethyl, -CN, -NO2, -NH2, -OR3, -NR3R4, -S(O)o-2R3, -SO2NR3R3, -CO2R3, -C(O)NR3R3, -N(R3)SO2R3, -N(R3)C(O)R3, -N(R3)CO2R3, -C(O)R3, optionally substituted lower alkyl, optionally substituted aryl, optionally substituted heteroarylalkyl, and optionally substituted arylalkyl; two of R60, when attached to a non-aromatic carbon, can be oxo; with the proviso, only when Ar is according to formula II, if Y is a Cι-6 alkylene; Z is -NH- or -N(CH3)-; R1 is a C1-6alkyl optionally substituted in the 2-position by -OH or a CL 4alkoxy group; R is -H or halogen; n = 0; and the atoms, X , of one bridge of the saturated bridged ring system, when combined with both bridgehead atoms, X , of the saturated bridged ring system, represent:
1 9
1) either a pyrrolidine or a piperidine, and any atom, X or X , of either of said pyrrolidine or said piperidine is attached to Y, then the other bridge of said saturated bridged ring system cannot be any one of -OC(O)CH2-, -CH2OC(O)-, -OC(O)CH2CH2-, -CH2OC(O)CH2-, -CH2CH2OC(O)-, -OC(O)CH2NH-, -OC(O)CH2N(Ci-4alkyl)-, and -OC(O)CH2O-; or
2) either a piperazine or a 4-(C1- alkyl)-piperazine, and any atom, X1 or X2, of either of said piperazine or said 4-(C1-4alkyl)-piperazine is attached to Y, then the other bridge of said saturated bridged ring system, only when attached via the 2- and the 3-position of either of said piperazine or said 4-(C1-4alkyl)-piperazine, cannot be one of -CH2OC(O)CH -, -CH2CH2OC(O)-, and either of the two aforementioned bridges optionally substituted by one or two C1-2alkyl groups; or
3) a piperazine, and any atom, X1 or X2, of said piperazine is attached to Y, then the other bridge of said saturated bridged ring system, only when attached via the 3- and the 4-position of said piperazine, cannot be one of -C(O)OCH2CH2-, -CH2OC(O)CH2-, and either of the two aforementioned bridges optionally substituted by one or two Cι-2alkyl groups, and only when either of the two aforementioned bridges are attached to the 3-position of said piperazine via their left-hand end as depicted above; or
4) a 2-oxomorpholine, said 2-oxomorpholine attached to Y via its 4-position, then the other bridge of said saturated bridged ring system, only when attached via the 5- and the 6-position of said 2-oxomorpholine, cannot be one of -(CH2)g-, -CH2WCH2-, -CH2WCH2CH2-, and -CH2CH2WCH2-, wherein W is -O-, -S(O)0-2-, -NH-, or -N(C1-4alkyl)- wherein g is 2, 3, or 4; and with the proviso that when Z is-O-, Ar is according to formula II, and the portion of G directly attached to Ar is selected from:
Figure imgf000014_0001
Figure imgf000015_0001
then R must be of formula IV; and with the proviso that when Ar is phenylene or substituted phenylene, Z is -S(O)o- - or
-O-, then the portion of G directly attached to Ar cannot contain
Figure imgf000015_0002
, when R70 is selected from -H, C1-4alkyl, and Cι-4alkoxyl.
[0034] In one example, the compound is according to paragraph [0033], wherein Z is either -O- or -NR5-.
[0035] In another example, the compound is according to paragraph [0034], wherein G is selected from the following:
Figure imgf000015_0003
Figure imgf000016_0001
Figure imgf000017_0001
wherein wherein Q, R , and R are as defined above; each E is selected from -O-, -N(R13)-, -CH2-, and -S(O)0-2-; M is selected from -O-, -N(R13)-, -CH2-, and -C(=O)N(R13)-; each V is independently either =N- or =C(H)-; each methylene in any of the above formulae is independently optionally substituted with R25; and R25 is selected from halogen, trihalomethyl, -CN, -NO2, -NH2, -OR3, -NR3R4, -S(O)0-2R3, -SO2NR3R3, -CO2R3, -C(O)NR3R3, -N(R3)SO2R3, -N(R3)C(O)R3, -N(R3)CO2R3, -C(O)R3, optionally substituted aryl, optionally substituted arylalkyl, heteroarylalkyl, and optionally substituted lower alkyl; two of R25, together with the carbon or carbons to which they are attached, can combine to form a three- to seven-membered alicyclic or heteroalicyclic, two of R25 on a single carbon can be oxo.
[0036] In another example, the compound is according to paragraph [0035], wherein Ar is according to one of formula Ila, lib, and Ilia.
Figure imgf000017_0002
Ila Ilb Hla
[0037] In another example, the compound is according to paragraph [0036], wherein D is -O- and R1 is -OR3.
[0038] In another example, the compound is according to paragraph [0037], wherein -O-R50 and R1 are interchangeably located at the 6-position and 7-position of the quinazoline or quinoline according to formula I.
[0039] In another example, the compound is according to paragraph [0038], wherein R1 is -OH or -OC1-6alkyl. [0040] In another example, the compound is according to paragraph [0039], wherein A1 is =N- or =C(H)-.
[0041] In another example, the compound is according to paragraph [0040], wherein G is selected from:
Figure imgf000018_0001
wherein Q, R20, R13, E, and R60 are as defined above; each methylene in any of the above formulae, other than those in a depicted ring, is independently optionally substituted with R25; and R25 is selected from halogen, trihalomethyl, oxo, -CN, -NO2, -NH2, -OR3, -NR3R4, -S(O)0-2R3, -SO2NR3R3, -CO2R3, -C(O)NR3R3, -N(R3)SO2R3, -N(R3)C(O)R3, -N(R3)CO2R3, -C(O)R3, optionally substituted aryl, optionally substituted arylalkyl,
9^ heteroarylalkyl, and optionally substituted lower alkyl; two of R , together with the carbon or carbons to which they are attached, can combine to form a three- to seven- membered alicyclic or heteroalicyclic.
[0042] In another example, the compound is according to paragraph [0041], wherein Q is selected from:
Figure imgf000018_0002
wherein R20 is defined as above, and P is a five- to seven-membered ring, including the two shared carbons of the aromatic ring to which P is fused, P optionally containing between one and three heteroatoms.
[0043] In another example, the compound is according to paragraph [0042], wherein Ar is according to formula Ha, and G is selected from:
Figure imgf000019_0001
wherein Q, R20, R13, E, and R60 are as defined above, and each methylene in any of the above formulae, other than those in a depicted ring, is independently optionally substituted with R25; and R25 is selected from halogen, trihalomethyl, oxo, -CN, -NO2, -NH2, -OR3, -NR3R4, -S(O)o-2R3, -SO2NR3R3, -CO2R3, -C(O)NR3R3, -N(R3)SO2R3, -N(R3)C(O)R3, -N(R3)CO2R3, -C(O)R3, optionally substituted aryl, optionally substituted arylalkyl, heteroarylalkyl, and optionally substituted lower alkyl; two of R25, together with the carbon or carbons to which they are attached, can combine to form a three- to seven- membered alicyclic or heteroalicyclic.
[0044] In another example, the compound is according to paragraph [0042], wherein Ar is according to formula lib, and G is selected from:
Figure imgf000019_0002
Figure imgf000020_0001
wherein Q, R20, R13, E, and R60 are as defined above, and each methylene in any of the above formulae, other than those depicted in a ring, is independently optionally substituted with R25; and R25 is selected from halogen, trihalomethyl, oxo, -CN, -NO2, -NH2, -OR3, -NR3R4, -S(O)0-2R3, -SO2NR3R3, -CO2R3, -C(O)NR3R3, -N(R3)SO2R3, -N(R3)C(O)R3, -N(R3)CO2R3, -C(O)R3, optionally substituted aryl, optionally substituted arylalkyl, heteroarylalkyl, and optionally substituted lower alkyl; two of R25, together with the carbon or carbons to which they are attached, can combine to form a three- to seven- membered alicyclic or heteroalicyclic.
[0045] In another example, the compound is according to paragraph [0044], wherein the methylene between the two carbonyls of the depicted formulae is di-substituted with either optionally substituted lower alkyl, or an optionally substituted spirocycle.
[0046] In another example, the compound is according to either [0043] or paragraph [0044], wherein R50 is a heteroalicylic or a C1-6alkyl-heteroalicylic.
[0047] In another example, the compound is according to paragraph [0046], wherein at least one of R2 is halogen.
[0048] In another example, the compound is according to paragraph [0046], wherein R50 is according to formula IV.
[0049] In another example, the compound is according to paragraph [0048], wherein the saturated bridged ring system according to formula IV has a geometry selected from the group consisting of [4.4.0], [4.3.0], [4.2.0], [4.1.0], [3.3.0], [3.2.0], [3.1.0], [3.3.3], [3.3.2], [3.3.1], [3.2.2], [3.2.1], [2.2.2], and [2.2.1].
[0050] In another example, the compound is according to paragraph [0049], wherein Y is selected from -CH2CH2CH2CH2-, -CH2CH2CH2-, -CH2CH2-, -CH2-, and absent.
[0051] In another example, the compound is according to paragraph [0050], wherein n is 0 and the saturated bridged ring system according to formula IV has a geometry selected from the group consisting of [4.4.0], [4.3.0], [4.2.0], [4.1.0], [3.3.0], [3.2.0], and [3.1.0]. [0052] In another example, the compound is according to paragraph [0051], wherein said saturated bridged ring system contains at least one annular nitrogen or at least one annular oxygen.
[0053] In another example, the compound is according to paragraph [0052], wherein said saturated bridged ring system contains -NR8-, wherein R8 is selected from -H, optionally substituted lower alkyl, -CO2R3, -C(O)NR3R3, -SO2R3, and -C(O)R3.
[0054] In another example, the compound is according to paragraph [0052], wherein said saturated bridged ring system is of formula V,
Figure imgf000021_0001
v wherein U1 is selected from -O-, -S(O)0-2-, -NR8-, -CR6R7-, and absent; and e is 0 or 1.
[0055] In another example, the compound is according to paragraph [0054], wherein Y is -CH2-.
[0056] In another example, the compound is according to paragraph [0055], wherein U1 is -NR8-, wherein R8 is selected from -H, optionally substituted lower alkyl, -CO2R3, -C(O)NR3R3, -SO2R3, and -C(O)R3.
[0057] In another example, the compound is according to paragraph [0055], wherein U1 is -O-.
[0058] In another example, the compound is according to paragraph [0055], wherein U1 is absent.
[0059] In another example, the compound is according to paragraph [0052], wherein Y is selected from -CH2CH2-, -CH2-, and absent. (
[0060] In another example, the compound is according to paragraph [0059], wherein said saturated bridged ring system is of formula VI,
Figure imgf000021_0002
VI wherein R9, R10, and R11 are each independently selected from -H, and -OR12; or
R9 is selected from -H, and -OR12, and R10 and R11, when taken together, are either an optionally substituted alkylidene or an oxo;
R12 is selected from -H, -C(O)R3, optionally substituted lower alkylidyne, optionally substituted lower arylalkylidyne, optionally substituted lower heterocyclylalkylidyne, optionally substituted lower alkylidene, optionally substituted lower alkylidenearyl, optionally substituted lower alkylideneheterocyclyl, optionally substituted lower alkyl, optionally substituted lower alkylaryl, optionally substituted aryl, optionally substituted lower heterocyclylalkyl, and optionally substituted heterocyclyl; or two R12's, when taken together, form 1) a corresponding spirocyclic ketal when said two R12's stem from R10 and R11, or 2) a corresponding cyclic ketal when said two R12's stem from R9 and one of R10 and R11.
[0061] In another example, the compound is according to paragraph [0060], wherein one of R10 and R11 is -OR12, wherein R12 is selected from -H, -C(O)R3, and optionally substituted lower alkyl; and R9 and the other of R10 and R11 are both -H.
[0062] In another example, the compound is according to paragraph [0061], wherein Y is either -CH2- or absent.
[0063] In another example, the compound is according to paragraph [0062], wherein R9 is an alkyl group containing at least one fluorine substitution thereon.
[0064] In another example, the compound is according to paragraph [0053], wherein said saturated bridged ring system is of formula VII.
Figure imgf000022_0001
VII
[0065] In another example, the compound is according to paragraph [0064], wherein Y is either -CH2- or absent.
[0066] In another example, the compound is according to paragraph [0065], wherein R8 is methyl or ethyl. [0067] In another example, the compound is according to paragraph [0053], wherein said saturated bridged ring system is of formula VIII.
Figure imgf000023_0001
VIII
[0068] In another example, the compound is according to paragraph [0067], wherein Y is -CH2-.
[0069] In another example, the compound is according to paragraph [0068], wherein R8 is methyl or ethyl.
[0070] In another example, the compound is according to paragraph [0052], wherein said saturated bridged ring system is of formula IX
Figure imgf000023_0002
IX wherein U2 is selected from -O-, -S(O)0-2-, -NR8-, -CR6R7-, and absent.
[0071] In another example, the compound is according to paragraph [0070], wherein R of formula IX is selected from -H and optionally substituted alkyl.
[0072] In another example, the compound is according to paragraph [0071], wherein U is either -CR6R7- or absent.
[0073] In another example, the compound is according to paragraph [0072], wherein U2 is either -CH2- or absent.
[0074] In another example, the compound is according to paragraph [0073], wherein Y is -CH2-.
[0075] In another example, the compound is according to paragraph [0053], wherein said saturated bridged ring system is according to formula X.
Figure imgf000023_0003
[0076] In another example, the compound is according to paragraph [0075], wherein R is methyl or ethyl.
[0077] In another example, the compound is according to paragraph [0033], selected from Table 1.
Table 1
Figure imgf000024_0001
Table 1
Figure imgf000025_0001
Table 1
Figure imgf000026_0001
Table 1
Figure imgf000027_0001
Table 1
Figure imgf000028_0001
Table 1
Figure imgf000029_0001
Table 1
Figure imgf000030_0001
Table 1
Figure imgf000031_0001
Table 1
Figure imgf000032_0001
Table 1
Figure imgf000033_0001
Table 1
Figure imgf000034_0001
Table 1
Figure imgf000035_0001
Table 1
Figure imgf000036_0001
Table 1
Figure imgf000037_0001
Table 1
Figure imgf000038_0001
Table 1
Figure imgf000039_0001
Table 1
Figure imgf000040_0001
Table 1
Figure imgf000041_0001
Table 1
Figure imgf000042_0001
Table 1
Figure imgf000043_0001
Table 1
Figure imgf000044_0001
Table 1
Figure imgf000045_0001
Table 1
Figure imgf000046_0001
Table 1
Figure imgf000047_0001
Table 1
Figure imgf000048_0001
Table 1
Entry Name Structure
N- { 3-Fluoro-4-[6-methoxy-7 -
(piperidin-4-ylmethoxy)-
142 quinolin-4-yloxy] -phenyl } -2- oxo-2-(3-phenyl-pyrrolidin-l- yl)-acetamide
N-{3-Fluoro-4-[6-methoxy-7-
(piperidin-4-ylmethoxy)-
143 quinolin-4-yloxy] -phenyl } -2- oxo-2-(2-phenyl-morpholin-4- yl)-acetamide
N-(2-Dimethylamino-2- phenyl-ethyl)-N'-{3-fluoro-4-
144 [6-methoxy-7-(piperidin-4- ylmethoxy)-quinolin-4- yloxy] -phenyl } -oxalamide
N-{3-Fluoro-4-[6-methoxy-7- (piperidin-4-ylmethoxy)-
145 quinolin-4-yloxy]-phenyl } -N'-
(2-oxo-2-phenyl-ethyl)- oxalamide
Figure imgf000049_0001
Table 1
Figure imgf000050_0001
Table 1
Entry Name Structure
N-{3-Fluoro-4-[6-methoxy-7-
(piperidin-4-ylmethoxy)-
150 quinolin-4-yloxy] -phenyl } -N'-
[2-(2-methoxy-phenyl)-ethyl] - oxalamide
N- { 3-Fluoro-4-[6-methoxy-7-
(piperidin-4-ylmethoxy)-
151 quinolin-4-yloxy] -phenyl }-N'-
(2-ρyridin-3-yl-ethyl)- oxalamide
N-Benzyl-N'-{3-fluoro-4-[6-
152 methoxy-7 -(piperidin-4- ylmethoxy)-quinolin-4- yloxy] -phenyl } -oxalamide
N-[2-(2,5-Dimethoxy- phenyl)-ethyl]-N'-{3-fluoro-4-
153 [6-methoxy-7-(piperidin-4- ylmethoxy)-quinolin-4- yloxy]-phenyl } -oxalamide
Figure imgf000051_0001
Table 1
Entry Name Structure
N- { 3-Fluoro-4-[6-methoxy-7 -
(piperidin-4-ylmethoxy)-
154 quinolin-4-yloxy] -phenyl } -N'-
[2-(2-trifluoromethyl-phenyl)- ethyl]-oxalamide
N-[2-(2-Ethoxy-phenyl)- ethyl]-N'-{3-fluoro-4-[6-
155 methoxy-7-(piperidin-4- ylmethoxy)-quinolin-4- yloxy] -phenyl } -oxalamide
N-[2-(2,4-Dimethyl-phenyl)- ethyl]-N'-{3-fluoro-4-[6-
156 methoxy-7-(piperidin-4- ylmethoxy)-quinolin-4- yloxy] -phenyl } -oxalamide
N- { 3-Fluoro-4-[6-methoxy-7-
(piperidin-4-ylmethoxy)-
157 quinolin-4-yloxy] -phenyl } -N-
(1S -ρhenyl-2-p-tolyl-ethyl)- oxalamide
Figure imgf000052_0001
Table 1
Figure imgf000053_0001
Table 1
Entry Name Structure
N-{3-Fluoro-4-[6-methoxy-7-
(piperidin-4-ylmethoxy)-
162 quinolin-4-yloxy] -phenyl } -N'-
[2-(3 -methoxy-phenyl)-ethyl] - oxalamide
N-(l,2-Diphenyl-ethyl)-N'-{3- fluoro-4- [6-methoxy-7-
163 (piperidin-4-ylmethoxy)- quinolin-4-yloxy] -phenyl } - oxalamide
N-[2-(2,4-Dichloro-ρhenyl)- ethyl]-N'-{3-fluoro-4-[6-
164 methoxy-7-(piperidin-4- ylmethoxy)-quinolin-4- yloxy]-phenyl } -oxalamide
N-[2-(3,4-Dimethoxy- phenyl)-ethyl] -N'- { 3 -fluoro-4-
165 [6-methoxy-7-(piperidin-4- ylmethoxy)-quinolin-4- yloxy] -phenyl} -oxalamide
Figure imgf000054_0001
Table 1
Entry Name Structure
N-[2-(4-Ethyl-phenyl)-ethyl]-
N'-{ 3-fluoro-4-[6-methoxy-7-
166 (piperidin-4-ylmethoxy)- quinolin-4-yloxy] -phenyl } - oxalamide
N-[2-(4-Ethoxy-ρhenyl)- ethyl]-N'-{3-fluσro-4-[6-
167 methoxy-7-(piperidin-4- ylmethoxy)-quinolin-4- yloxy] -phenyl } -oxalamide
N-[2-(4-Ethoxy-3-methoxy- phenyl)-ethyl]-N'-{3-fluoro-4-
168 [6-methoxy-7-(piperidin-4- ylmethoxy)-quinolin-4- yloxy] -phenyl } -oxalamide
Figure imgf000055_0001
Table 1
Entry Name Structure
N-{3-Fluoro-4-[6-methoxy-7-
(piperidin-4-ylmethoxy)-
169 quinolin-4-yloxy] -phenyl }-N'-
[2-(4-phenoxy-phenyl)-ethyl]- oxalamide
N-[2-(3-Ethoxy-4-methoxy- phenyl)-ethyl]-N'-{3-fluoro-4-
170 [6-methoxy-7-(piperidin-4- ylmethoxy)-quinolin-4- yloxy]-phenyl }-oxalamide
N-{3-Fluoro-4-[6-methoxy-7-
(piperidin-4-ylmethoxy)-
171 quinolin-4-yloxy] -phenyl } -N'-
(2-pyridin-2-yl-ethyl)- oxalamide
N-{3-Fluoro-4-[6-methoxy-7-
(piperidin-4-ylmethoxy)-
172 quinolin-4-yloxy] -phenyl } -N'-
(2-pyridin-4-yl-ethyl)- oxalamide
Figure imgf000056_0001
Table 1
Figure imgf000057_0001
Table 1
Entry Name Structure
N-{ 3-Fluoro-4-[6-methoxy-7-
(piperidin-4-ylmethoxy)-
177 quinolin-4-yloxy] -phenyl }-N'- indan- 1-yl-oxalamide
N-{3-Fluoro-4-[6-methoxy-7-
(l-methyl-piperidm-4-
178 ylmethoxy)-quinolin-4- yloxy] -phenyl } -N'-isobutyl- oxalamide
N-{3-Fluoro-4-[6-methoxy-7- ( 1 -methyl-piperidin-4-
179 ylmethoxy)-quinolin-4- yloxy] -phenyl } -N'-(3 -methyl- butyl)-oxalamide
N-{3-Fluoro-4-[6-methoxy-7-
( 1 -methyl-piperidin-4-
180 ylmethoxy)-quinolin-4- yloxy]-phenyl}-N'-(2R- phenyl-propyl)-oxalamide
Figure imgf000058_0001
Table 1
Figure imgf000059_0001
Table 1
Entry Name Structure
N-[2-(3-Bromo-phenyl)- ethyl]-N'-{3-fluoro-4-[6-
185 methoxy-7-(piperidin-4- ylmethoxy)-quinolin-4- yloxy] -phenyl } -oxalamide
N-[2-(2,6-Dichloro-phenyl)- ethyl]-N'-{3-fluoro-4-[6-
186 methoxy-7-(piperidin-4- ylmethoxy)-quinolin-4- yloxy]-phenyl } -oxalamide
N-[2-(2,4-Dichloro-phenyl)- ethyl]-N'-{3-fluoro-4-[6-
187 methoxy-7-(piρeridin-4- ylmethoxy)-quinolin-4- yloxy] -phenyl } -oxalamide
N-(2-Benzo[l,3]dioxol-5-yl- ethyl)-N'-{3-fluoro-4-[6-
188 methoxy-7-(piperidin-4- ylmethoxy)-quinolin-4- yloxy]-phenyl } -oxalamide
Figure imgf000060_0001
Table 1
Figure imgf000061_0001
Table 1
Figure imgf000062_0001
Table 1
Entry Name Structure
N-[2-(3-Chloro-4-ρropoxy- phenyl)-ethyl]-N'-{ 3-fluoro-4-
196 [6-methoxy-7-(piperidin-4- ylmethoxy)-quinolin-4- yloxy] -phenyl } -oxalamide
N-[2-(4-Butoxy-3-chloro- phenyl)-ethyl]-N'-{3-fluoro-4-
197 [6-methoxy-7-(piperidin-4- ylmethoxy)-quinolin-4- yloxy] -phenyl } -oxalamide
N-[2-(4-tert-Butyl-ρhenyl)- ethyl]-N:-{3-fluoro-4-[6-
198 methoxy-7-(piperidin-4- ylmethoxy)-quinolin-4- yloxy]-phenyl } -oxalamide
Figure imgf000063_0001
Table 1
Entry Name Structure
N- { 3-Fluoro-4-[6-methoxy-7- (piperidin-4-ylmethoxy)-
199 quinolin-4-yloxy] -phenyl } -N'-
[2-(4-sulfamoyl-phenyl)- ethyl] -oxalamide
N-{3-Fluoro-4-[6-methoxy-7- (piperidin-4-ylmethoxy)-
200 quinolin-4-yloxy]-phenyl } -N'- [2-(4-hydroxy-3-methoxy- phenyl)-ethyl] -oxalamide
N-{3-Fluoro-4-[6-methoxy-7- (piperidin-4-ylmethoxy)-
201 quinolin-4-yloxy] -phenyl } -N'- [2-(3 -hydroxy-4-methoxy- phenyl)-ethyl] -oxalamide
Figure imgf000064_0001
Table 1
Entry Name Structure
N-(2,4-Dichloro-benzyl)-N*-
{ 3-fluoro-4-[6-methoxy-7-
202 (piperidin-4-ylmethoxy)- quinolin-4-yloxy] -phenyl } - oxalamide
N-{3-Fluoro-4-[6-methoxy-7-
(piperidin-4-ylmethoxy)-
203 quinolin-4-yloxy] -phenyl } -N'-
(4-fluoro-2-trifluoromethyl- benzyl)-oxalamide
N- { 3-Fluoro-4-[6-methoxy-7-
(piperidin-4-ylmethoxy)-
204 quinolin-4-yloxy] -phenyl } -N'-
( 1 -p-tolyl-ethyl)-oxalamide
Figure imgf000065_0001
Table 1
Entry Name Structure
N-{3-Fluoro-4-[6-methoxy-7-
(piperidin-4-ylmethoxy)-
205 quinolin-4-yloxy] -phenyl }-N
(3-fluoro-4-trifluoromethyl- benzyl)-oxalamide
N-(3-Chloro-4-fluoro-benzyl)-
N'-{3-fluoro-4-[6-methoxy-7-
206 (piperidin-4-ylmethoxy)- quinolin-4-yloxy] -phenyl } - oxalamide
N-{3-Fluoro-4-[6-methoxy-7-
(piperidin-4-ylmethoxy)-
207 quinolin-4-yloxy] -phenyl }-N'-
[ 1 -(3 -methoxy-phenyl)-ethyl] - oxalamide
Figure imgf000066_0001
Table 1
Entry Name Structure
N- { 3-Fluoro-4-[6-methoxy-7- (piperidin-4-ylmethoxy)-
208 quinolin-4-yloxy] -phenyl } -N'-
(l-naρhthalen-2-yl-ethyl)- oxalamide
N-(4-Chloro-3- trifluoromethyl-benzyl)-N'-
{ 3-fluoro-4-[6-methoxy-7-
209 (piperidin-4-ylmethoxy)- quinolin-4-yloxy] -phenyl } - oxalamide
N- { 3-Fluoro-4- [6-methoxy-7-
(piperidin-4-ylmethoxy)-
210 quinolin-4-yloxy] -phenyl }-N'-
(1 -p-tolyl-ethyl)-oxalamide
Figure imgf000067_0001
Table 1
Figure imgf000068_0001
Table 1
Entry Name Structure
N-{3-Fluoro-4-[6-methoxy-7-
(piperidin-4-ylmethoxy)-
214 quinolin-4-yloxy]-phenyl } -N'-
(4-fluoro-3-trifluoromethyl- benzyl)-oxalamide
N-(3,5-Dichloro-benzyl)-N'-
{ 3-fluoro-4-[6-methoxy-7-
215 (piperidin-4-ylmethoxy)- quinolin-4-yloxy] -phenyl }- oxalamide
N-{3-Fluoro-4-[6-methoxy-7-
(piperidin-4-ylmethoxy)-
216 quinolin-4-yloxy]-phenyl } -N'-
(lR,2,3,4-tetrahydro- naphthalen- 1 -yl)-oxalamide
N-{3-Fluoro-4-[6-methoxy-7-
(piperidin-4-ylmethoxy)-
217 quinolin-4-yloxy] -phenyl }-N'-
(lS,2,3,4-tetrahydro- naphthalen-l-yl)-oxalamide
Figure imgf000069_0001
Table 1
Entry Name Structure
N-Cyclopentyl-N'- { 3 -fluoro-
4-[6-methoxy-7-(piperidin-4-
218 ylmethoxy)-quinolin-4- yloxy]-phenyl } -oxalamide
N- [ 1 -(4-Bromo-phenyl)- ethyl]-N*-{3-fluoro-4-[6-
219 methoxy-7 -(piperidin-4- ylmethoxy)-quinolin-4- yloxy] -phenyl } -oxalamide
N-(2-Fluoro-benzyl)-N'- { 3- fluoro-4- [6-methoxy-7-
220 (piperidin-4-ylmethoxy)- quinolin-4-yloxy] -phenyl } - oxalamide
N-[2-(3,4-Dichloro-phenyl)- ethyl]-N'-{3-fluoro-4-[6-
221 methoxy-7-(piperidin-4- ylmethoxy)-quinolin-4- yloxy] -phenyl } -oxalamide
Figure imgf000070_0001
Table 1
Entry Name Structure
N-(4-Fluoro-benzyl)-N*- { 3- fluoro-4-[6-methoxy-7-
222 (piperidin-4-ylmethoxy)- quinolin-4-yloxy] -phenyl } - oxalamide
N-(2,3'Difluoro-benzyl)-N'-
{ 3 -fluoro-4- [6-methoxy-7-
223 (piperidin-4-ylmethoxy)- quinolin-4-yloxy] -phenyl } - oxalamide
N-{3-Fluoro-4-[6-methoxy-7-
(piperidin-4-ylmethoxy)-
224 quinolin-4-yloxy]-phenyl } -N'- (2-phenoxy-ethyl)-oxalamide
N-(2,2-Diphenyl-ethyl)-N'-{ 3- fluoro-4- [6-methoxy-7-
225 (piperidin-4-ylmethoxy)- quinolin-4-yloxy] -phenyl } - oxalamide
Figure imgf000071_0001
Table 1
Entry Name Structure
N-{3-Fluoro-4-[6-methoxy-7-
(piperidin-4-yrmethoxy)-
226 quinolin-4-yloxy]-phenyl}-N'-
[2-(4-methoxy-phenyl)-ethyl] - oxalamide
N- { 3-Fluoro-4-[6-methoxy-7 -
(piperidin-4-ylmethoxy)-
227 quinolin-4-yloxy] -phenyl } -N'-
(2-phenyl-propyl)-oxalamide
N-[2-(4-Bromo-phenyl)- ethyl]-N'-{3-fluoro-4-[6-
228 methoxy-7-(piperidin-4- ylmethoxy)-quinolin-4- yloxy] -phenyl } -oxalamide
N-{4-[7-(l-Ethyl-piperidin-4- ylmethoxy)-6-methoxy-
229 quinolin-4-yloxy] -3 -fluorophenyl } -2-oxo-2-(2-phenyl- morpholin-4-yl)-acetamide
Figure imgf000072_0001
Table 1
Figure imgf000073_0001
Table 1
Entry Name Structure
N-{ 3-Fluoro-4-[6-methoxy-7-
(piperidin-4-ylmethoxy)-
234 quinolin-4-yloxy] -phenyl } -N'-
(4-methoxy-benzyl)- oxalamide
N-{ 3-Fluoro-4-[6-methoxy-7-
(piperidin-4-ylmethoxy)-
235 quinolin-4-yloxy] -phenyl } -N'-
(4-trifluoromethyl-benzyl)- oxalamide
N-{3-Fluoro-4-[6-methoxy-7-
(piperidin-4-ylmethoxy)-
236 quinolin-4-yloxy]-phenyl } -N'-
(3-methoxy-benzyl)- oxalamide
Figure imgf000074_0001
Table 1
Entry Name Structure
N-{ 3-Fluoro-4-[6-methoxy-7-
(piperidin-4-ylmethoxy)-
237 quinolin-4-yloxy] -phenyl }-N'-
(3-trifluoromethyl-benzyι)- oxalamide
N- { 3-Fluoro-4- [6-methoxy-7-
(piperidin-4-ylmethoxy)-
238 quinolin-4-yloxy]-phenyl } -N'-
(3 -trifluoromethoxy-benzyl)- oxalamide
N-{3-Fluoro-4-[6-methoxy-7-
(piperidin-4-ylmethoxy)-
239 quinolin-4-yloxy] -phenyl }-N'-
(2-methoxy-benzyl)- oxalamide
N- { 3-Fluoro-4-[6-methoxy-7-
(piperidin-4-ylmethoxy)-
240 quinolin-4-yloxy]-phenyl } -N'-
(2-trifluoromethyl-benzyl)- oxalamide
Figure imgf000075_0001
Table 1
Entry Name Structure
N-(3-Chloro-benzyl)-N'-{ 3- fluoro-4- [6-methoxy-7-
241 (piperidin-4-ylmethoxy)- quinolin-4-yloxy] -phenyl } - oxalamide
N- { 3-Fluoro-4-[6-methoxy-7-
(piperidin-4-ylmethoxy)-
242 quinolin-4-yloxy] -phenyl }-N'-
(2-trifluoromethoxy-benzyl)- oxalamide
N-(2-Chloro-benzyl)-N'-{ 3- fluoro-4-[6-methoxy-7-
243 (piperidin-4-ylmethoxy)- quinolin-4-yloxy] -phenyl } - oxalamide
Figure imgf000076_0001
Table 1
Entry Name Structure
N-{ 3-Fluoro-4-[6-methoxy-7-
(piperidin-4-ylmethoxy)-
244 quinolin-4-yloxy] -phenyl } -N'-
(4-trifluoromethoxy-benzyl)- oxalamide
N-{3-Fluoro-4-[6-methoxy-7-
( 1 -methyl-piperidin-4-
245 ylmethoxy)-quinolin-4- yloxy]-phenyl }-N'-(4- methoxy-benzyl)-oxalamide
N-{3-Fluoro-4-[6-methoxy-7-
( 1 -methyl-piperidin-4- ylmethoxy)-quinolin-4-
246 yloxy]-phenyl }-N'-(4- trifluoromethyl-benzyl)- oxalamide
Figure imgf000077_0001
Table 1
Figure imgf000078_0001
Table 1
Entry Name Structure
N-[5-Chloro-6-(6,7- dimethoxy-quinolin-4-yloxy)-
251 pyridin-3-yl]-N'-(4-fluoro- phenyl)-N'-methyl- malonamide
N-{3-Fluoro-4-[6-methoxy-7-
(piperidin-4-ylmethoxy)-
252 quinolin-4-yloxy] -phenyl }-N'-
(lR-phenyl-propyl)- oxalamide
N- { 3-Fluoro-4- [6-methoxy-7-
(piperidin-4-ylmethoxy)-
253 quinolin-4-yloxy] -phenyl } -N'-
( lR-phenyl-propyl)- oxalamide
Figure imgf000079_0001
Table 1
Entry Name Structure
N-(3,4-Difluoro-benzyl)-N'-
{ 3-fluoro-4-[6-methoxy-7-
254 (piperidin-4-ylmethoxy)- quinolin-4-yloxy] -phenyl }- oxalamide
N-(2,6-Difluoro-benzyl)-N'-
{ 3-fluoro-4-[6-methoxy-7-
255 (piperidin-4-ylmethoxy)- quinolin-4-yloxy] -phenyl } - oxalamide
N- { 3-Fluoro-4-[6-methoxy-7-
( 1 -methyl-piperidin-4- ylmethoxy)-quinolin-4-
256 yloxy]-phenyl }-N'-[2-(4- fluoro-phenyl)-ethyl]- oxalamide
N-{ 3-Fluoro-4-[6-methoxy-7-
( 1 -methyl-piperidin-4-
257 ylmethoxy)-quinolin-4- yloxy] -phenyl } -N'-phenyl- oxalamide
Figure imgf000080_0001
Table 1
Figure imgf000081_0001
Table 1
Entry Name Structure
N-(3,3-Dimethyl-butyl)-N'-
{ 3 -fluoro-4- [6-methoxy-7-
262 (piperidin-4-ylmethoxy)- quinolin-4-yloxy]-phenyl } - oxalamide
N-{5-Chloro-6-[6-methoxy-7-
(3 -piperidin- 1 -yl-propoxy)-
263 quinolin-4-yloxy]-pyridin-3- yl }-N'-(4-fluoro-phenyl)- malonamide
N-{ 5-Chloro-6-[6-methoxy-7-
(3-morpholin-4-yl-propoxy)-
264 quinolin-4-yloxy] -pyridin-3 - yl } -N'-(4-fluoro-phenyl)- malonamide
Figure imgf000082_0001
Table 1
Entry Name Structure
N-{5-Chloro-6-[7-(3- diethylamino-propoxy)-6-
265 methoxy-quinolin-4-yloxy] - pyridin-3-yl}-N'-(4-fluoro- phenyl)-malonamide
N-(4-Chloro-benzyl)-N'- { 3- fluoro-4-[6-methoxy-7-
266 (piperidin-4-ylmethoxy)- quinolin-4-yloxy] -phenyl } - oxalamide
N-(3 ,5-Dimethoxy-benzyl)-
N'-{3-fluoro-4-[6-methoxy-7-
267 (piperidin-4-ylmethoxy)- quinolin-4-yloxy] -phenyl } - oxalamide
Figure imgf000083_0001
Table 1
Figure imgf000084_0001
Table 1
Figure imgf000085_0001
Table 1
Figure imgf000086_0001
Table 1
Entry Name Structure
N-{3-Fluoro-4-[6-methoxy-7-
(piperidin-4-ylmethoxy)-
279 quinolin-4-yloxy] -phenyl } -N'-
(2-fluoro-5-trifluoromethyl- benzyl)-oxalamide
N-{3-Fluoro-4-[6-methoxy-7-
(piperidin-4-ylmethoxy)-
280 quinolin-4-yloxy] -phenyl }-N'-
[ 1 -(4-fluoro-phenyl)-ethyl] - oxalamide
N-(lS-Benzyl-2-oxo-2- pyrrolidin-l-yl-ethyl)-N'- { 3- fluoro-4- [6-methoxy-7-
281 (piperidin-4-ylmethoxy)- quinolin-4-yloxy] -phenyl } - oxalamide
N- { 3-Fluoro-4-[6-methoxy-7- (octahydro-
282 cyclopenta[c]pyrrol-5- ylmethoxy)-quinazolin-4- yloxy] -phenyl } -N'-phenethyl- oxalamide
Figure imgf000087_0001
Table 1
Entry Name Structure
N-[2-(4-Amino-phenyl)- ethyl]-N'-{3-fluoro-4-[6-
283 methoxy-7-(piperidin-4- ylmethoxy)-quinolin-4- yloxy] -phenyl } -oxalamide
2-(4-Benzyl-piperidin-l-yl)- N- { 3-fluoro-4-[6-methoxy-7-
284 (piperidin-4-ylmethoxy)- quinolin-4-yloxy]-phenyl }-2- oxo-acetamide
N-[4-(6,7-Dimethoxy-
285 quinolin-4-yloxy)-phenyl]-N- (4-fluoro-phenyl)-malonamide
Figure imgf000088_0001
Table 1
Figure imgf000089_0001
Table 1
Figure imgf000090_0001
Table 1
Entry Name Structure
N-{3-Fluoro-4-[6-methoxy-7-
(piperidin-4-ylmethoxy)-
295 quinolin-4-yloxy] -phenyl } -2- oxo-2-pyrrolidin- 1 -yl- acetamide
N-Ethyl-N'-{3-fluoro-4-[6- methoxy-7-(piperidin-4-
296 ylmethoxy)-quinolin-4- yloxy] -phenyl } -N-methyl- oxalamide
Figure imgf000091_0001
[0078] In another aspect, the invention comprises a compound for modulating kinase activity of formula A-B-C, or a pharmaceutically acceptable salt, hydrate, or prodrug thereof, wherein, A is selected from:
Figure imgf000091_0002
Figure imgf000092_0001
B is selected from:
Figure imgf000092_0002
and, C is selected from:
Figure imgf000092_0003
Figure imgf000093_0001
wherein R2 is selected from -H, halogen, trihalomethyl, -CN, -NH2, -NO2, -OR3, -NR3R3, -S(O)0-2R3, -SO2NR3R3, -CO2R3, -C(O)NR3R3, -N(R3)SO2R3, -N(R3)C(O)R3, -N(R3)CO2R3, -C(O)R3, and optionally substituted lower alkyl; q is 0 to 2; each R3 is independently selected from -H, optionally substituted lower alkyl, optionally substituted aryl, optionally substituted arylalkyl, and optionally substituted heteroarylalkyl; two R3, together with the nitrogen to which they are attached, form a four- to seven- membered heteroalicyclic, said four- to seven-membered heteroalicyclic optionally containing one additional heteroatom; when one said additional heteroatom is a nitrogen, then said nitrogen is optionally substituted with a group selected from -H, trihalomethyl, -SO2R5, -SO2NR5R5, -CO2R5, -C(O)NR5R5, -C(O)R5, and optionally substituted lower alkyl; each R35 is independently selected from -H, -C(=O)R3, -C(=O)OR3, -C(=O)SR3, -SO2R3,
-C(=O)N(R > 3 )TR> 3 , and optionally substituted lower alkyl; two R35, together with the nitrogen to which they are attached, can combine to form a heteroalicyclic optionally substituted with between one and four of R60, said heteroalicyclic may have an additional annular heteroatom, and said heteroalicyclic may have an aryl fused thereto, said aryl optionally substituted with an additional one to four of R60;
A1 is selected from =N-, =C(H)-, and =C(CN)-;
A2 is either =N- or =C(H)-;
R5 is -H or optionally substituted lower alkyl;
R8 is selected from R3, -SO2NR3R3, -CO2R3, -C(O)NR3R3, -SO2R3, and -C(O)R3;
R9, R10, and R11 are each independently selected from -H, and -OR12; or
R9 is selected from -H, and -OR12, and R10 and R11, when taken together, are either an optionally substituted alkylidene or an oxo; and
R12 is selected from -H, -C(O)R3, optionally substituted lower alkylidyne, optionally substituted lower arylalkylidyne, optionally substituted lower heterocyclylalkylidyne, optionally substituted lower alkylidene, optionally substituted lower alkylidenearyl, optionally substituted lower alkylideneheterocyclyl, optionally substituted lower alkyl, optionally substituted lower alkylaryl, optionally substituted aryl, optionally substituted lower heterocyclylalkyl, and optionally substituted heterocyclyl; or two R12's, when taken together, form 1) a corresponding spirocyclic ketal when said two R12's stem from R10 and R11, or 2) a corresponding cyclic ketal when said two R12's stem from R9 and one of R10 and R11;
E1 is selected from -O-, -CH2-, -N(R5)-, and -S(O)0-2-;
Q is a five- to ten-membered ring system, optionally substituted with between zero and four of R20;
R20 is selected from -H, halogen, trihalomethyl, -CN, -NO2, -NH2, -OR3, -NR3R3, -S(O)0-2R3, -SO2NR3R3, -CO2R3, -C(O)NR3R3, -N(R3)SO2R3, -N(R3)C(O)R3, -N(R3)CO2R3, -C(O)R3, and optionally substituted lower alkyl;
R60 is selected from -H, halogen, trihalomethyl, -CN, -NO2, -NH2, -OR3, -NR3R3, -S(O)o-2R3, -SO2NR3R3, -CO2R3, -C(O)NR3R3, -N(R3)SO2R3, -N(R3)C(O)R3, -N(R3)CO2R , -C(O)R3, optionally substituted lower alkyl, optionally substituted aryl, optionally substituted heteroarylalkyl, and optionally substituted arylalkyl; two of R , when attached to a non-aromatic carbon, can be oxo; each methylene in any of the above formulae is independently optionally substituted with R25; each R25 is independently selected from halogen, trihalomethyl, -CN, -NO2, -NH2, -OR3, -NR3R3, -S(O)0-2R3, -SO2NR3R3, -CO2R3, -C(O)NR3R3, -N(R3)SO2R3, -N(R3)C(O)R3, -N(R3)CO2R3, -C(O)R3, optionally substituted aryl, optionally substituted arylalkyl, heteroarylalkyl, and optionally substituted lower alkyl; two of R25, together with the carbon or carbons to which they are attached, can combine to form a three- to seven- membered alicyclic or heteroalicyclic, two of R25 on a single carbon can be oxo; with the proviso that when B is selected from:
Figure imgf000095_0001
and C contains
Figure imgf000095_0002
and the remaining portion of C contains one of:
Figure imgf000095_0003
directly attached to
Figure imgf000096_0001
then A must be one of:
Figure imgf000096_0002
and with the proviso that when C contains
Figure imgf000096_0003
and B is selected from:
Figure imgf000096_0004
then the portion of C directly attached to
Figure imgf000097_0001
cannot contain
Figure imgf000097_0002
when R 0 is selected from _H; l.4alkyl, and C1-4alkoxyl.
[0079] In another example the compound is according to paragraph [0078], wherein Q is selected from phenyl, napthyl, 1,2,3,4-tetrahydronaphthyl, indanyl, benzodioxanyl, benzofuranyl, phenazinyl, phenothiazinyl, phenoxazinyl, tetrahydroisoquinolyl, pyrrolyl, pyrazolyl, pyrazolidinyl, imidazolyl, imidazolinyl, imidazolidinyl, tetrahydropyridinyl, pyridinyl, pyrazinyl, pyrimidinyl, pyridazinyl, oxazolyl, oxazolinyl, oxazolidinyl, triazolyl, isoxazolyl, isoxazolidinyl, thiazolyl, thiazolinyl, thiazolidinyl, isothiazolyl, isothiazolidinyl, indolyl, isoindolyl, indolinyl, isoindolinyl, octahydroindolyl, octahydroisoindolyl, quinolyl, isoquinolyl, benzimidazolyl, thiadiazolyl, benzopyranyl, benzothiazolyl, benzoxazolyl, furyl, thienyl, benzothieliyl, and oxadiazolyl; each optionally substituted with between one and four of R20; wherein each R20 is independently selected from -H, halogen, trihalomethyl, -CN, -NO2, -NH2, -OR3, -NR3R3, -CO2R3, -C(O)NR3R3, -N(R3)SO2R3, -N(R3)C(O)R3, -N(R3)CO2R3, -C(O)R3, and optionally substituted lower alkyl.
[0080] In another example the compound is according to paragraph [0079], wherein B is either of the following:
Figure imgf000097_0003
wherein A1 is either =N- or =C(H)-. [0081] In another example the compound is according to paragraph [0080], wherein B is
Figure imgf000097_0004
82] In another example the compound is according to paragraph [0081], wherein C is selected from:
Figure imgf000098_0001
Figure imgf000099_0001
wherein R2, R3, R5, R20, R25 and R60 are as defined above.
[0083] In another example the compound is according to paragraph [0082], R2 is selected from halogen, trihalomethyl, -CN, -NO2, -OR3, -NR3R3, -CO2R3, -C(O)NR3R3, -N(R3)C(O)R3, -N(R3)CO2R3, -C(O)R3, and optionally substituted lower alkyl
[0084] In another example the compound is according to paragraph [0083], wherein R2 is halogen.
[0085] In another example the compound is according to paragraph [0084], wherem R2 is either fluorine or chlorine.
[0086] In another aspect, the invention comprises a compound for modulating kinase activity according to Formula XI,
Figure imgf000099_0002
or a pharmaceutically acceptable salt, hydrate, or prodrug thereof, wherein, each R1 is independently selected from halogen, -OR3, -NO2, -NH2, -NR3R4, -D-R50 and optionally substituted C1-6alkyl;
R70 is selected from -H, halogen, -OR3, -S(O)0-2R3, -NO2, -NH2, -NR3R4, and optionally substituted Cι-6alkyl;
Q is selected from =N-, =C(H)-, and =C(CN)-;
Z is selected from -S(O)o-2-, -O-, and -NR5-;
Ar is either a five- or six-membered arylene or a five- or six-membered heteroarylene containing between one and three heteroatoms;
G is either an optionally substituted cycloalkyl or an optionally substituted heteroalicyclic; each R2 is independently selected from halogen, trihalomethyl, -CN, -NO2, -NH2, -OR3, -NR3R4, -S(O)o-2R3, -SO2NR3R3, -CO2R3, -C(O)NR3R3, -N(R3)SO2R3, -N(R3)C(O)R3, -N(R3)CO2R3, -C(O)R3, and optionally substituted C1-6alkyl; each R3 is independently -H or R4; each R4 is independently selected from optionally substituted C1-6alkyl, optionally substituted aryl, optionally substituted aryl
Figure imgf000100_0001
optionally substituted heterocyclyl, and optionally substituted heterocyclyl -ealkyl; or
R3 and R4, when taken together with a common nitrogen to which they are attached, form an optionally substituted five- to seven-membered heterocyclyl, said optionally substituted five- to seven-membered heterocyclyl optionally containing at least one additional annular heteroatom selected from N, O, S, and P;
R5 is -H or optionally substituted Chalky!; each D is independently selected from -O-, -S(O)o-2-, and -NR5-; each R50 is independently either R3, or according to formula XII;
Figure imgf000100_0002
XII wherein X1, X2, and optionally X3, represent the atoms of a saturated bridged ring system, said saturated bridged ring system comprising up to four annular heteroatoms represented by any of X1, X2, and X3; wherein, each X1 is independently selected from -C(R6)R7-, -O-, -S(O)0-2-, and -NR8-; each X2 is independently an optionally substituted bridgehead methine or a bridgehead nitrogen; each X3 is independently selected from -C(R6)R7-, -O-, -S(O)0-2-, and -NR8-;
Y is either: an optionally substituted lower alkylene linker, between D and either 1) any annular atom of the saturated bridged ring system, except X when X is a bridgehead nitrogen, or 2) any heteroatom, represented by any of R or R ; provided there are at least two carbon atoms between D and any annular heteroatom of the saturated bridged ring system or any heteroatom represented by any of R6 or R7; or Y is absent, when Y is absent, said saturated bridged ring system, is directly attached to D via an annular carbon of said saturated bridged ring system, unless D is -SO2-, in which case said saturated bridged ring system, is directly attached to D via an any annular atom of said saturated bridged ring system; m and p are each independently one to four; n is zero to two, when n equals zero, then there is a single bond between the two bridgehead X2 's;
R6 and R7 are each independently selected from -H, halogen, trihalomethyl, -CN, -NH2, -NO2, -OR3, -NR3R4, -S(O)0-2R4, -SO2NR3R4, -CO2R3, -C(O)NR3R4, -N(R3)SO2R4, -N(R3)C(O)R3, -NCO2R3, -C(O)R3, optionally substituted Cι-6alkyl, optionally substituted aryl, optionally substituted aryl Cι-6alkyl, optionally substituted heterocyclyl, optionally substituted heterocyclyl C1-6alkyl, and a bond to either Y or D; or
R6 and R7, when taken together are oxo; or
R6 and R7, when taken together with a common carbon to which they are attached, form a optionally substituted three- to seven-membered spirocyclyl, said optionally substituted three- to seven-membered spirocyclyl optionally containing at least one additional annular heteroatom selected from N, O, S, and P;
R8 is selected from -R3, Y, -SO2NR3R4, -CO2R4, -C(O)NR3R3, -SO2R4, and -C(O)R3; and each R30 is independently selected from halogen, trihalomethyl, -CN, -NO2, -NH2, -OR3, -NR3R4, -S(O)0-2R3, -SO2NR3R3, -CO2R3, -C(O)NR3R3, -N(R3)SO2R3, -N(R3)C(O)R3, -N(R3)CO2R3, -C(O)R3, and optionally substituted Ci-β-ukyl.
[0087] In one example, the compound is according to paragraph [0086], wherein Z is either -O- or -NR5-.
[0088] In another example, the compound is according to paragraph [0087], wherein at least one of R1 is -D-R50.
[0089] In another example, the compound is according to paragraph [0088], wherein D is -O- and at least one other R1 is -OR3.
[0090] In another example, the compound is according to paragraph [0089], of formula XHIa or XHIb:
Figure imgf000102_0001
Xllla Xlllb wherein Q1 is either =N- or =C(H)-.
[0091] In another example, the compound is according to paragraph [0090], wherein R50 is selected from Cι-6alkyl optionally substituted with at least one of optionally substituted amino, optionally substituted C1-6alkyl amino, optionally substituted C^dia-kyl amino, optionally substituted heteroalicylic, and a group of formula XII.
[0092] In another example, the compound is according to paragraph [0091], wherein R3a is C1-6alkyl.
[0093] In another example, the compound is according to paragraph [0092], wherein Z is -O-.
[0094] In another example, the compound is according to paragraph [0093], wherein G is selected from cyclopropyl, aziradine, cyclobutyl, and azetidine, each optionally substituted with between zero and four of R30. [0095] In another example, the compound is according to paragraph [0094], wherein Q is either =N- or =C(H)-.
[0096] In another example, the compound is according to paragraph [0095], wherein R2 is selected from -H, halogen, C1-6 alkyl and perfluoro C1-6 alkyl.
[0097] In another example, the compound is according to paragraph [0096], wherein -N(R3b)R4 is selected from the following:
Figure imgf000103_0001
wherein J, is a five- to ten-membered ring, optionally substituted with between zero and five of R20; each R20 is independently selected from -H, halogen, trihalomethyl, -CN, -NO2, -NH2, -OR3, -NR3R4, -S(O)0-2R3, -SO2NR3R3, -CO2R3, -C(O)NR3R3, -N(R3)SO2R3, -N(R3)C(O)R3, -N(R3)CO2R3, -C(O)R3, optionally substituted C1-6alkyl, optionally substituted aryl, optionally substituted aryl C1-6alkyl, optionally substituted heterocyclyl, and optionally substituted heterocyclyl C1-6alkyl; two of R20, together with the atom or atoms to which they are attached, combine to form an optionally substituted three- to seven-membered heteroalicyclic, said optionally substituted three- to seven-membered heteroalicyclic either spiro- to J or fused to J;
E is selected from -O-, -N(R5)-, -CH2-, and -S(O)0-2-; each R60 is independently selected from halogen, trihalomethyl, -CN, -NO2, -NH2, -OR3, -NR3R4, -S(O)o-2R3, -SO2NR3R3, -CO2R3, -C(O)NR3R3, -N(R3)SO2R3, -N(R3)C(O)R3, -N(R3)CO2R3, -C(O)R3, optionally substituted Cι.6alkyl, optionally substituted aryl, optionally substituted heteroaryl C1-6alkyl, and optionally substituted aryl Cι-6alkyl; each methylene in any of the above formulae, other than those in a depicted ring, is independently optionally substituted with R25; and
R25 is selected from halogen, trihalomethyl, oxo, -CN, -NO2, -NH2, -OR3, -NR3R4, -S(O)0-2R3, -SO2NR3R3, -CO2R3, -C(O)NR3R3, -N(R3)SO2R3, -N(R3)C(O)R3, -N(R3)CO2R3, -C(O)R3, optionally substituted aryl, optionally substituted aryl C1-6alkyl, heteroaryl C1-6alkyl, and optionally substituted C1-6alkyl; or two of R25, together with the carbon or carbons to which they are attached, can combine to form a three- to seven-membered alicyclic or heteroalicyclic;
R is equivalent to R as defined above; and
R4 and R5 are as defined above.
[0098] In another example, the compound is according to paragraph [0097], of formula XlVa or XIVb:
Figure imgf000104_0001
[0099] In another example, the compound is according to paragraph [0098], wherein is Cι-6alkyl optionally substituted with a group selected from optionally substituted amino, an optionally substituted alkylamino, optionally substituted dialkylamino, and optionally substituted heteroalicylic.
[0100] In another example, the compound is according to paragraph [0099], wherein the heteroalicyclic portion of said optionally substituted heteroalicyclic of R50 is selected from the group consisting of piperidine, piperazine, morpholine, thiomorpholine, thiomorpholine 1 -oxide, thiomorpholine 1,1 -dioxide, 2-oxo-morpholine, pyrrolidine, and azepine.
[0101] In another example, the compound is according to paragraph [0099], wherein R50 is according to formula XII. [0102] In another example, the compound is according to paragraph [0101], wherein the saturated bridged ring system according to formula XII has a geometry selected from the group consisting of [4.4.0], [4.3.0], [4.2.0], [4.1.0], [3.3.0], [3.2.0], [3.1.0], [3.3.3], [3.3.2], [3.3.1], [3.2.2], [3.2.1], [2.2.2], and [2.2.1].
[0103] In another example, the compound is according to paragraph [0102], wherein Y is selected from -CH2CH2CH2CH2-, -CH2CH2CH2-, -CH2CH2-, -CH2-, and absent.
[0104] In another example, the compound is according to paragraph [0103], wherein n is 0 and the saturated bridged ring system according to formula XII has a geometry selected from the group consisting of [4.4.0], [4.3.0], [4.2.0], [4.1.0], [3.3.0], [3.2.0], and [3.1.0].
[0105] In another example, the compound is according to paragraph [0104], wherein said saturated bridged ring system contains at least one annular nitrogen or at least one annular oxygen.
[0106] In another example, the compound is according to paragraph [0105], wherein said saturated bridged ring system contains -NR8-, wherein R8 is selected from -H, optionally substituted C1-6alkyl, -CO2R3, -C(O)NR3R3, -SO2R3, and -C(O)R3.
[0107] In another example, the compound is according to paragraph [0105], wherem said saturated bridged ring system is of formula XV,
Figure imgf000105_0001
XV wherein U1 is selected from -O-, -S(O)0-2-, -NR8-, -CR6R7-, and absent; and e is 0 or 1.
[0108] In another example, the compound is according to paragraph [0107], wherein Y is -CH2-.
[0109] In another example, the compound is according to paragraph [0108], wherein U1 is -NR8-, wherein R8 is selected from -H, optionally substituted lower alkyl, -CO2R3, -C(O)NR3R3, -SO2R3, and -C(O)R3.
[0110] In another example, the compound is according to paragraph [0108], wherein U1 is -O-. [0111] In another example, the compound is according to paragraph [0108], wherein U1 is absent.
[0112] In another example, the compound is according to paragraph [0103], wherein Y is selected from -CH2CH2-, -CH2-, and absent.
[0113] In another example, the compound is according to paragraph [0112], wherein said saturated bridged ring system is of formula XVI,
Figure imgf000106_0001
XVI wherein R9, R10, and R11 are each independently selected from -H, and -OR12; or
R9 is selected from -H, and -OR12, and R10 and R11, when taken together, are either an optionally substituted alkylidene or an oxo;
R12 is selected from -H, -C(O)R3, optionally substituted lower alkylidyne, optionally substituted lower arylalkylidyne, optionally substituted lower heterocyclylalkylidyne, optionally substituted lower alkylidene, optionally substituted lower alkylidenearyl, optionally substituted lower alkylideneheterocyclyl, optionally substituted lower alkyl, optionally substituted lower alkylaryl, optionally substituted aryl, optionally substituted lower heterocyclylalkyl, and optionally substituted heterocyclyl; or two R12's, when taken together, form 1) a corresponding spirocyclic ketal when said two R12's stem from R10 and R11, or 2) a corresponding cyclic ketal when said two R12's stem from R9 and one of R10 and R11.
[0114] In another example, the compound is according to paragraph [0113], wherein one of R10 and R11 is -OR12, wherein R12 is selected from -H, -C(O)R3, and optionally substituted lower alkyl; and R9 and the other of R10 and R11 are both -H.
[0115] In another example, the compound is according to paragraph [0114], wherein Y is either -CH2- or absent.
[0116] In another example, the compound is according to paragraph [0113], wherein R9 is an alkyl group containing at least one fluorine substitution thereon. [0117] In another example, the compound is according to paragraph [0106], wherein said saturated bridged ring system is of formula XVII.
Figure imgf000107_0001
xvπ
[0118] In another example, the compound is according to paragraph [0117], wherein Y is either -CH2- or absent.
[0119] In another example, the compound is according to paragraph [0118], wherein R8 is methyl or ethyl.
[0120] In another example, the compound is according to paragraph [0119], wherein at least one of R2 is halogen.
[0121] In another example, the compound is according to paragraph [0106], wherein said saturated bridged ring system is of formula XVIII.
Figure imgf000107_0002
XVIII
[0122] In another example, the compound is according to paragraph [0121], wherein Y is -CH2-.
[0123] In another example, the compound is according to paragraph [0122], wherein R8 is methyl or ethyl.
[0124] In another example, the compound is according to paragraph [0105], wherein said saturated bridged ring system is of formula XIX
Figure imgf000107_0003
XIX wherem U2 is selected from -O-, -S(O)0-2-, -NR8-, -CR6R7-, and absent.
[0125] In another example, the compound is according to paragraph [0124], wherein R3 of formula XIX is selected from -H and optionally substituted alkyl. [0126] In another example, the compound is according to paragraph [0125], wherein U2 is either -CR6R7- or absent.
[0127] In another example, the compound is according to paragraph [0126], wherein U2 is either -CH2- or absent.
[0128] In another example, the compound is according to paragraph [0127], wherein Y is -CH2-.
[0129] In another example, the compound is according to paragraph [0106], wherein said saturated bridged ring system is according to formula XX.
Figure imgf000108_0001
XX
[0130] In another example, the compound is according to paragraph [0129], wherein R8 is methyl or ethyl.
[0131] In another example, the compound is according to any of paragraphs [0099] - [0130], wherein R is selected from C1-6 alkyl, perfluoro Cι-6 alkyl, and halogen.
[0132] In another example, the compound is according to paragraph [0131], wherein R2 is selected from perfluoro C1-3 alkyl and halogen.
[0133] In another example, the compound is according to any of paragraphs [0099] - [0130], wherein R20 is selected from halogen, -CN, -NO2, -NH2, -OR3, -NR3R4, -N(R3)SO2R3, -N(R3)C(O)R3, -N(R3)CO2R3, optionally substituted heterocyclyl, and optionally substituted heterocyclyl Cι-6alkyl, and (two of R20) together with the atom or atoms to which they are attached, an optionally substituted three- to six-membered heteroalicyclic, said optionally substituted three- to six-membered heteroalicyclic fused to the phenyl as in XlVa or XlVb.
[0134] In another example, the compound is according to paragraph [0133], wherein R20 is selected from halogen, -NR3R4, optionally substituted heterocyclyl, and optionally substituted heterocyclyl C1-6alkyl, and (two of R20) together with the atom or atoms to which they are attached, an optionally substituted five- to six-membered heteroalicyclic, said optionally substituted five- to six-membered heteroalicyclic fused to the phenyl as in XlVa or XlVb. [0135] In another example, the compound is according to paragraph [0134], wherein R2 is selected from C1-6 alkyl, perfluoro C1-6 alkyl, and halogen.
[0136] In another example, the compound is according to paragraph [0135], wherein R2 is selected from perfluoro C1-3 alkyl and halogen.
[0137] In another example, the compound is according to paragraph [0086], selected from Table 2.
Table 2
Figure imgf000109_0001
Table 2
Figure imgf000110_0001
Table 2
Figure imgf000111_0001
no Table 2
Figure imgf000112_0001
Table 2
Figure imgf000113_0001
Table 2
Figure imgf000114_0001
Table 2
Figure imgf000115_0001
Table 2
Figure imgf000116_0001
Table 2
Figure imgf000117_0001
Table 2
Figure imgf000118_0001
111 Table 2
Figure imgf000119_0001
Table 2
Figure imgf000120_0001
Table 2
Figure imgf000121_0001
Table 2
Figure imgf000122_0001
Table 2
Figure imgf000123_0001
Table 2
Figure imgf000124_0001
Table 2
Figure imgf000125_0001
Table 2
Figure imgf000126_0001
Table 2
Figure imgf000127_0001
Table 2
Figure imgf000128_0001
Table 2
Figure imgf000129_0001
Table 2
Figure imgf000130_0001
Table 2
Figure imgf000131_0001
[0176] Another aspect of the invention is a pharmaceutical composition comprising a compound according to any one of paragraphs [0033]-[0120] and a pharmaceutically acceptable carrier.
[0177] Another aspect of the invention is a metabolite of the compound or the pharmaceutical composition according to any one of paragraphs [0022]-[0122].
[0178] Another aspect of the invention is a method of modulating the in vivo activity of a kinase, the method comprising administering to a subject an effective amount of the compound or the pharmaceutical composition according to any of paragraphs [0033]- [0121].
[0179] Another aspect of the invention is the method according to paragraph [0123], wherein modulating the in vivo activity of the kinase comprises inhibition of said kinase.
[0180] Another aspect of the invention is the method according to paragraph [0124], wherein the kinase is at least one of c-Met, KDR, c-Kit, flt-3, and flt-4.
[0181] Another aspect of the invention is the method according to paragraph [0125], wherein the kinase is c-Met.
[0182] Another aspect of the invention is a method of treating diseases or disorders associated with uncontrolled, abnormal, and/or unwanted cellular activities, the method comprising administering, to a mammal in need thereof, a therapeutically effective amount of the compound or the pharmaceutical composition as described in any one of paragraphs [0033H0121].
[0183] Another aspect of the invention is a method of screening for a modulator of a kinase, said kinase selected from c-Met, KDR, c-Kit, flt-3, and flt-4, the method comprising combining a compound according to any one of paragraphs [0033]-[0120], and at least one candidate agent and determining the effect of the candidate agent on the activity of said kinase.
[0184] Another aspect of the invention is a method of inhibiting proliferative activity in a cell, the method comprising administering an effective amount of a composition comprising a compound according any one of paragraphs [0033]-[0120] to a cell or a plurality of cells.
[0185] As mentioned, although improved quinolines and quinazolines of the invention can be made via conventional serial methods, due to their complex structure, more efficient routes are desirable, particularly convergent syntheses. Thus, the present invention also comprises a process for preparing a compound of Formula XXI,
Figure imgf000133_0001
XI comprising reaction of a compound of Formula XXII, with a compound of Formula XXIII
Figure imgf000133_0002
Figure imgf000133_0003
XXIII wherein, each R1 is independently selected from halogen, -OR3, -NO2, -NH2, -NR3R3, -D-R50 and optionally substituted C1-6alkyl;
R70 is selected from -H, halogen, -OR3, -S(O)0-2R3, -NO2, -NH2, -NR3R3, and optionally substituted Cι-6alkyl;
J is selected from =N-, =C(H)-, =C(halogen)-, and =C(CN)-;
Z is selected from -S(O)o-2-, -O-, and -NR5-; each R5 is independently selected from -H, optionally substituted C1-6alkyl, optionally substituted aryl, and optionally substituted aryl Cι-6alkyl;
Ar is either a five- to ten-membered arylene or a five- to ten-membered heteroarylene containing between one and three heteroatoms;
R2 is selected from -H, halogen, trihalomethyl, -CN, -NO2, -NH2, -OR3, -NR3R3, -S(O)0-2R3, -SO2NR3R3, -CO2R3, -C(O)NR3R3, -N(R3)SO2R3, -N(R3)C(O)R3, -N(R3)CO2R3, -C(O)R3, and optionally substituted Cι-6alkyl; each R3 is independently selected from -H, -Si(R5)(R5)R5, optionally substituted lower alkyl, optionally substituted aryl, optionally substituted arylalkyl, and optionally substituted heteroarylalkyl; two R3, together with the nitrogen to which they are attached, form a four- to seven- membered heteroalicyclic, said four- to seven-membered heteroalicyclic optionally containing one additional heteroatom; when one said additional heteroatom is a nitrogen, then said nitrogen is optionally substituted with a group selected from -H, trihalomethyl, -SO2R5, -SO2NR5R5, ^CO2R5, -C(O)NR5R5, -C(O)R5, and optionally substituted lower alkyl;
B is selected from absent, -N(R13)-, -N(SO2R13)-, -O-, -S(O)0-2-, and -C(=O)-;
L is selected from absent, -C(=S)N(R13)-, -C(=NR14)N(R13)-, -SO2N(R13)-, -SO2-, -C(=O)N(R13)-, -N(R13)-, -C(=O)C1-2alkylN(R13)-, -N(R13)C1-2alkylC(=O)-, -C(=O)C0-ialkylC(=O)N(R13)-, -C(=O)-, -C0-4alkylene-, -C(=O)C0-ιalkylC(=O)OR3-, -C(=NR14)C0-1alkylC(=O , -C(=O)C0-ιalkylC(=O)-, and an optionally substituted four- to six-membered heterocyclyl containing between one and three annular heteroatoms and comprising at least one nitrogen;
T is selected from -H, -R13, -C0-4alkyl, -C0-4alkylQ, -OC0-4alkylQ, -C0-4alkylOQ, -N(R13)C0- alkylQ, -SO2C0.4alkylQ, -C(=O)C0-4alkylQ, -C0-4alkylN(R13)Q, and -C(=O)N(R13)Co-4alkylQ, wherein each of the aforementioned C0- alkyl is optionally substituted;
Q is a five- to ten-membered ring system, optionally substituted with between zero and four of R20; each R20 is independently selected from -H, halogen, trihalomethyl, -CN, -NO2, -NH2, -OR3, -NR3R3, -S(O)0-2R3, -SO2NR3R3, -CO2R3, -C(O)NR3R3, -N(R3)SO2R3, -N(R3)C(O)R3, -N(R3)CO2R3, -C(O)R3, optionally substituted Cι-6alkyl, optionally substituted aryl, optionally substituted aryl Cι-6alkyl, optionally substituted heterocyclyl, and optionally substituted heterocyclyl -eal yl; two of R20, together with the atom or atoms to which they are attached, combine to form an optionally substituted three- to seven-membered heteroalicyclic, said optionally substituted three- to seven-membered heteroalicyclic either spiro- to Q or fused to Q;
D is selected from -O-, -S(O)0-2-, and -NR15-;
R is either R , or according to formula XXIV;
Figure imgf000135_0001
XXIV wherein X1, X2, and optionally X3, represent the atoms of a saturated bridged ring system, said saturated bridged ring system comprising up to four annular heteroatoms represented by any of X1, X2, and X3; wherein, each X1 is independently selected from -C(R6)R7-, -O-, -S(O)0-2-, and -NR8-; each X is independently an optionally substituted bridgehead methine or a bridgehead nitrogen; each X3 is independently selected from -C(R6)R7-, -O-, -S(O)0-2-, and -NR8-;
Y is either: an optionally substituted Ci-βalkylene linker, between D and either 1) any annular atom of the saturated bridged ring system, except X2 when X2 is a bridgehead nitrogen, or 2) any heteroatom, represented by any of R6 or R7; provided there are at least two carbon atoms between D and any annular heteroatom of the saturated bridged ring system or any heteroatom represented by any of R6 or R7; or Y is absent, when Y is absent, said saturated bridged ring system, is directly attached to D via an annular carbon of said saturated bridged ring system, unless D is -SO -, in which case said saturated bridged ring system, is directly attached to D via an any annular atom of said saturated bridged ring system; m and p are each independently one to four; n is zero to two, when n is zero, then there is a single bond between the two bridgehead
X2's;
R and R are each independently selected from -H, halogen, trihalomethyl, -CN, -NH2, -NO2, -OR3, -NR3R3, -S(O)o-2R3, -SO2NR3R3, -CO2R3, -C(O)NR3R3, -N(R3)SO2R3, -N(R3)C(O)R3, -NCO2R3, -C(O)R3, optionally substituted C1-6alkyl, optionally substituted aryl, optionally substituted aryl Ci-βalkyl, optionally substituted heterocyclyl, optionally substituted heterocyclyl a
Figure imgf000135_0002
and a bond to either Y or D; or R6 and R7, when taken together are oxo; or
Figure imgf000136_0001
, when taken together with a common carbon to which they are attached, form a optionally substituted three- to seven-membered spirocyclyl, said optionally substituted three- to seven-membered spirocyclyl optionally containing at least one additional annular heteroatom selected from N, O, S, and P;
R8 is selected from -R3, Y, -SO2NR3R3, -CO2R3, -C(O)NR3R3, -SO2R3, and -C(O)R3;
R13 is selected from -H, -C(=O)R3, -C(=O)OR3, -C(=O)SR3, -SO2R3, -C(=O)N(R3)R3, and optionally substituted C1-6alkyl; two R13, together with the atom or atoms to which they are attached, can combine to form a heteroalicyclic optionally substituted with between one and four of R60, said heteroalicyclic comprising up to four annular heteroatoms, and said heteroalicyclic optionally comprising an aryl or heteroaryl fused thereto, in which case said aryl or heteroaryl is optionally substituted with an additional one to four of R60;
R14 is selected from -H, -NO2, -NH2, -N(R3)R3, -CN, -OR3, optionally substituted C1-6alkyl, optionally substituted heteroalicyclyl Ci-βalkyl, optionally substituted aryl, optionally substituted aryl C^alkyl and optionally substituted heteroalicyclic;
R15 is a group -M^M2, wherein M1 is selected from absent, -C(=S)N(R13)-, -C(=NR14)N(R13)-, -SO2N(R13)-, -SO2-, -C(=O)N(R13)-, -C(=O)C(=O)N(R13)-, -C0-4alkylene-, -C(=O)-, and an optionally substituted four to six-membered heterocyclyl containing between one and three heteroatoms but comprising at least one nitrogen; and M2 is selected from -H, -C0-6alkyl, alkoxy, -C(=O)C0-4alkylQ, -C0-4alkylQ, -OC0-4alkylQ-, -N(R13)C0- alkylQ-, and -C(=O)N(R13)C0-4alkylQ;
R60 is selected from -H, halogen, trihalomethyl, -CN, -NO2, -NH2, -OR3, -NR3R3, -S(O)0-2R3, -SO2NR3R3, -CO2R3, -C(O)NR3R3, -N(R3)SO2R3, -N(R3)C(O)R3, -N(R3)CO2R3, -C(O)R3, optionally substituted Cι-6alkyl, optionally substituted aryl, optionally substituted heteroaryl C1-6alkyl, and optionally substituted aryl C1- alkyl; two of R60, when attached to a non-aromatic carbon, can be oxo;
P1 is a suitable leaving group; and
P2 is selected from -H, a metal, and a group removed in-situ when combining XXII and XXIII to make XXI. [0131] In one example, the process is according to paragraph [0130], wherein Ar is para- phenylene as defined by the substitution pattern of -Z- and -B-L-T about said phenylene. [0132] In another example, the process is according to paragraph [0131], wherein Z is either -O- or -NR5-
[0133] In another example, the process is according to paragraph [0132], wherein -B-L-T is selected from the following:
Figure imgf000137_0001
Figure imgf000138_0001
wherein Q, R , and R are as defined above; each E is selected from -O-, -N(R )-, -CH2, and -S(O)0-2-; M is selected from -O-, -N(R13)-, -CH2-, and -C(=O)N(R13)-; each V is independently either =N- or =C(H)-; each methylene in any of the above formulae is independently optionally substituted with R25; and R25 is selected from halogen, trihalomethyl, -CN, -NO2, -NH2, -OR3, -NR3R3, -S(O)0-2R3, -SO2NR3R3, -CO2R3, -C(O)NR3R3, -N(R3)SO2R3, -N(R3)C(O)R3, -N(R3)CO2R3, -C(O)R3, optionally substituted aryl, optionally substituted aryl C1-6alkyl, heteroaryl Cι-6alkyl, and optionally substituted Ci-βalkyl; two of R25, together with the carbon or carbons to which they are attached, can combine to form an optionally substituted three- to seven-membered alicyclic or heteroalicyclic; two of R on a single carbon can be oxo.
[0134] In another example, the process is according to paragraph [0133], wherein there is one of R1 that is -D-R50 and another of R1 that is -OR3a.
[0135] In another example, the process is according to paragraph [0134], wherein D is -O-.
[0136] In another example, the process is according to paragraph [0135], wherein -O-R50 and -OR3a are interchangeably located at the 6-position and 7-position of the quinazoline or quinoline according to Formula XXI.
[0137] In another example, the process is according to paragraph [0136], wherein -OR3a is selected from -OH, -OSi(R5)(R5)R5, and optionally substituted -OC1-6alkyl.
[0138] In another example, the process is according to paragraph [0137], wherein J is =N- or =C(H)-.
[0139] In another example, the process is according to paragraph [0138], wherein -B-L-T is selected from:
Figure imgf000139_0001
Figure imgf000140_0001
wherein Q, R , R , E, and R are as defined above; each methylene in any of the above formulae, other than those in a depicted ring, is independently optionally substituted with R25; and R25 is selected from halogen, trihalomethyl, oxo, -CN, -NO2, -NH2, -OR3, -NR3R3, -S(O)0-2R3, -SO2NR3R3, -CO2R3, -C(O)NR3R3, -N(R3)SO2R3, -N(R3)C(O)R3, -N(R3)CO2R3, -C(O)R3, optionally substituted aryl, optionally substituted aryl C1-6alkyl, heteroaryl C1-6alkyl, and optionally substituted C1-6alkyl; two of R25, together with the carbon or carbons to which they are attached, can combine to form a three- to seven- membered optionally substituted alicyclic or heteroalicyclic.
[0140] In another example, the process is according to paragraph [0139], wherein Q is selected from the following three formula:
Figure imgf000140_0002
wherein R is defined as above, and P is a five- to seven-membered ring, including the two shared carbons of the aromatic ring to which P is fused, P optionally containing between one and three heteroatoms.
[0141] In another example, the process is according to paragraph [0140], wherein -B-L-T is either of formula XXV or formula XXVI,
Figure imgf000140_0003
wherein R ,20 is defined as above; G is either an optionally substituted cycloalkyl or an optionally substituted heteroalicyclic; each R 30 is independently selected from halogen, trihalomethyl, -CN, -NO2, -NH2, -OR3, -NR3R3, -S(O)0-2R3, -SO2NR3R3, -CO2R3, -C(O)NR3R3, -N(R3)SO2R3, -N(R )C(O)R3, -N(R3)CO2R3, -C(O)R3, and optionally substituted Cι-6alkyl; and R3a and R3b are each independently selected from -H and optionally substituted Cι-6alkyl.
[0142] In another example, the process is according to paragraph [0141], wherem a compound of formula XXIIa is combined with a compound of formula XXIIIa to make a compound of formula XXIa,
Figure imgf000141_0001
XXIa wherein -B-L-T, Z, J, R50, and R2 are as defined above; R70 is selected from -H, -NO2, -NH2, and -NR3R3; provided when Z is -N(R5)- that R5 is selected from -H, Cι-3alkyl, and aryl C1-3alkyl; P1 is selected from halogen, optionally substituted alkyl-S(O)o-2-, optionally substituted arylsulfonate, optionally substituted alkylsulfonate, a group containing boron, an azide, a group containing phosphorus, and a metal; and P2 is selected from -H and a metal.
[0143] In another example, the process is according to paragraph [0142], wherein P2 is selected from -H, lithium, sodium, potassium, cesium, copper, palladium, and titanium.
[0144] In another example, the process is according to paragraph [0143], wherein Z is -O-.
[0145] In another example, the process is according to paragraph [0144], wherein P1 is selected from chlorine, bromine, a toluene sulfonate, and trifluoromethansulfonate.
[0146] In another example, the process is according to paragraph [0145], wherein R70 is -H.
[0147] In another example, the process is according to paragraph [0146], wherein J is =C(H)-.
[0148] In another example, the process is according to paragraph [0147], wherein R2 is selected from Cι-6 alkyl, perfluoro Cι-6 alkyl, and halogen. [0149] In another example, the process is according to paragraph [0148], wherein XXIIa and XXIIIa are heated together, optionally with a base, optionally with microwave radiation, to form XXIa.
[0150] In another example, the process is according to paragraph [0149], wherein the base is selected from an organic base, an inorganic base, and a combination of an organic base and an inorganic base.
[0151] In another example, the process is according to paragraph [0150], wherein the base is selected from 2,6-lutidine, 4-N,N-dimethylaminopyridine, and a metal carbonate.
[0152] In another example, the process is according to paragraph [0151], wherein XXIIa and XXIIIa are heated together in a solvent with said base, at between about 40°C and 200°C for between about one hour and twenty-four hours to form XXIa.
[0153] In another example, the process is according to paragraph [0152], wherein the solvent is an organic solvent.
[0154] In another example, the process is according to paragraph [0153], wherein one molar equivalent of XXIIa is combined with between about one quarter and four molar equivalents of XXIIIa.
[0155] In another example, the process is according to paragraph [0154], wherein one molar equivalent of XXIIa is combined with more than one but less than two molar equivalents of XXIIIa.
[0156] In another example, the process is according to paragraph [0155], wherein XXIIa is combined with XXIIIa and said base in an aromatic solvent to form a mixture, and said mixture is heated to between about 100°C and 200°C for between about one and ten hours to orm la.
[0157] In another example, the process is according to paragraph [0156], wherein the aromatic solvent is an optionally substituted benzene.
[0158] In another example, the process is according to paragraph [0157], wherein the aromatic solvent is bromobenzene.
[0159] In another example, the process is according to paragraph [0158], wherein the base is 4-N,N-dimethylaminopyridine. [0160] In another example, the process is according to paragraph [0159], wherein said mixture is heated to reflux for between about three and seven hours.
[0161] In another example, the process is according to paragraph [0160], wherein said mixture is heated to reflux for between about four and six hours.
[0162] In another example, the process is according to paragraph [0155], wherein XXIIa is combined with XXIIIa and said base in a non-aromatic solvent to form a mixture, and said mixture is heated to between about 40°C and 100°C for between about one and twenty hours to form XXIa.
[0163] In another example, the process is according to paragraph [0162], wherein the non- aromatic solvent comprises a functional group selected from an amide, and ether, a nitrile, a halide, an ester, an amine, and a ketone.
[0164] In another example, the process is according to paragraph [0163], wherem the non- aromatic solvent is N,N-dimethylacetamide.
[0165] In another example, the process is according to paragraph [0164], wherein the base is potassium carbonate.
[0166] In another example, the process is according to paragraph [0165], wherein said mixture is heated to about 50°C between about ten and twenty hours.
[0167] In another example, the process is according to paragraph [0166], wherein the aromatic solvent is an optionally substituted pyridine.
[0168] In another example, the process is according to paragraph [0167], wherein the aromatic solvent is 2,6-lutidine.
[0169] In another example, the process is according to paragraph [0168], wherein the base is 2,6-lutidine.
[0170] In another example, the process is according to paragraph [0169], wherein said mixture is heated to reflux for between about three and seven hours.
[0171] In another example, the process is according to paragraph [0170], wherein said mixture is heated to reflux for between about four and six hours.
[0172] In another example, the process is according to paragraph [0154], wherein one molar equivalent of XXIIIa is combined with more than one but less than two molar equivalents of XXIIa. [0173] In another example, the process is according to paragraph [0172], wherein XXIIa is combined with XXIIIa and said base in an aromatic solvent to form a mixture, and said mixture is heated to between about 100°C and 200°C for between about ten and twenty hours to form XXIa.
[0174] In another example, the process is according to paragraph [0173], wherein the aromatic solvent is an optionally substituted pyridine.
[0175] In another example, the process is according to paragraph [0174], wherein the aromatic solvent is 2,6-lutidine.
[0176] In another example, the process is according to paragraph [0175], wherein the base is 2,6-lutidine.
[0177] In another example, the process is according to paragraph [0176], wherein said mixture is heated to between about 150°C and 200°C for between about fifteen and twenty hours.
[0178] In another example, the process is according to any of paragraphs [0149] - [0177], wherein a compound of formula XXIIb is substituted for the compound of formula XXIIa, and either a compound of formula XXIIIb or a compound of formula XXIIIc is substituted for the compound of formula XXIIIa, in order to make a compound of formula XXIb or a compound of formula XXIc, respectively,
Figure imgf000144_0001
XXIIb
Figure imgf000144_0002
XXIIIb
Figure imgf000144_0003
Figure imgf000145_0001
wherein J, R50, R20 and R2 are as defined above.
[0179] In another example, the process is according to paragraph [0178], wherein R2, if present, is halogen.
[0180] In another example, the process is according to paragraph [0179], wherein R2, if present, is fluorine.
[0181] In another example, the process is according to paragraph [0180], wherein R2, if present, is up to two fluorines ortho to the oxygen of the phenylene to which R2 is attached.
[0182] In another example, the process is according to paragraph [0130], used to make a compound listed in either Table 1 or Table 2.
[0183] In another example the process is according to any of paragraphs [0130] - [0182], further comprising converting said compound to a pharmaceutically acceptable salt, hydrate, or prodrug thereof.
Definitions
[0184] As used in the present specification, the following words and phrases are generally intended to have the meanings as set forth below, except to the extent that the context in which they are used indicates otherwise or they are expressly defined to mean something different.
[0185] The symbol "-" means a single bond, "=" means a double bond, "≡" means a triple bond. The symbol "»ΛΛ/V" refers to a group on a double-bond as occupying either position on the terminus of a double bond to which the symbol is attached; that is, the geometry, E- or Z-, of the double bond is ambiguous. When a group is depicted removed from its parent formula, the "^^" symbol will be used at the end of the bond which was theoretically cleaved in order to separate the group from its parent structural formula.
[0186] When chemical structures are depicted or described, unless explicitly stated otherwise, all carbons are assumed to have hydrogen substitution to conform to a valence of four. For example, in the structure on the left-hand side of the schematic below there are nine hydrogens implied. The nine hydrogens are depicted in the right-hand structure. Sometimes a particular atom in a structure is described in textual formula as having a hydrogen or hydrogens as substitution (expressly defined hydrogen), for example, -CH2CH2-. It is understood by one of ordinary skill in the art that the aforementioned descriptive techniques are common in the chemical arts to provide brevity and simplicity to description of otherwise complex structures.
Figure imgf000146_0001
[0187] In this application, some ring structures are depicted genetically and will be described textually. For example, in the schematic below, if in the structure on the left, ring A is used to describe a "spirocyclyl," then if ring A is cyclopropyl, there are at most four hydrogens on ring A (when "R" can also be -H). In another example, as depicted on the right side of the schematic below, if ring B is used to describe a "phenylene" then there can be at most four hydrogens on ring B (assuming depicted cleaved bonds are not C-H bonds).
Figure imgf000146_0002
[0188] If a group "R" is depicted as "floating" on a ring system, as for example in the formula:
Figure imgf000147_0001
then, unless otherwise defined, a substituent "R" may reside on any atom of the ring system, assuming replacement of a depicted, implied, or expressly defined hydrogen from one of the ring atoms, so long as a stable structure is formed.
[0189] If a group "R" is depicted as floating on a fused ring system, as for example in the formulae:
Figure imgf000147_0002
then, unless otherwise defined, a substituent "R" may reside on any atom of the fused ring system, assuming replacement of a depicted (for example the -NH- in the formula above), implied (for example as in the formula above, where the hydrogens are not shown but understood to be present), or expressly defined hydrogen (for example where in the formula above, "X" equals =CH-) from one of the ring atoms, so long as a stable structure is formed. In the example depicted, the "R" group may reside on either the 5-membered or the 6-membered ring of the fused ring system. In the formula depicted above, when y is 2 for example, then the two "R's" may reside on any two atoms of the ring system, again assuming each replaces a depicted, implied, or expressly defined hydrogen on the ring.
[0190] When there are more than one such depicted "floating" groups, as for example in the formulae:
Figure imgf000147_0003
where there are two groups, namely, the "R" and the bond indicating attachment to a parent structure; then, unless otherwise defined, the "floating" groups may reside on any atoms of the ring system, again assuming each replaces a depicted, implied, or expressly defined hydrogen on the ring.
[0191] When a group "R" is depicted as existing on a ring system containing saturated carbons, as for example in the formula:
Figure imgf000148_0001
where, in this example, "y" can be more than one, assuming each replaces a currently depicted, implied, or expressly defined hydrogen on the ring; then, unless otherwise defined, where the resulting structure is stable, two "R's" may reside on the same carbon. A simple example is when R is a methyl group; there can exist a geminal dimethyl on a carbon of the depicted ring (an "annular" carbon). In another example, two R's on the same carbon, including that carbon, may form a ring, thus creating a spirocyclic ring (a "spirocyclyl" group) structure with the depicted ring as for example in the formula:
Figure imgf000148_0002
[0192] "Alkyl" is intended to include linear, branched, or cyclic hydrocarbon structures and combinations thereof, inclusively. For example, "C8 alkyl" may refer to an /i-octyl, wo-octyl, cyclohexylethyl, and the like. Lower alkyl refers to alkyl groups of from one to six carbon atoms. Examples of lower alkyl groups include methyl, ethyl, propyl, isopropyl, butyl, s-butyl, t-butyl, isobutyl, pentyl, hexyl and the like. Higher alkyl refers to alkyl groups containing more that eight carbon atoms. Exemplary alkyl groups are those of C20 or below. Cycloalkyl is a subset of alkyl and includes cyclic hydrocarbon groups of from three to thirteen carbon atoms. Examples of cycloalkyl groups include c- propyl, c-butyl, c-pentyl, norbomyl, adamantyl and the like. In this application, alkyl refers to alkanyl, alkenyl, and alkynyl residues (and combinations thereof); it is intended to include cyclohexylmethyl, vinyl, allyl, isoprenyl, and the like. Thus when an alkyl residue having a specific number of carbons is named, all geometric isomers having that number of carbons are intended to be encompassed; thus, for example, either "butyl" or "C4 alkyl" is meant to include π-butyl, -fee-butyl, isobutyl, t-butyl, isobutenyl and but-2- yne radicals; and for example, "propyl" or "C3 alkyl" each include π-propyl, propenyl, and isopropyl.
[0193] "Alkylene" refers to straight or branched chain divalent radical consisting solely of carbon and hydrogen atoms, containing no unsaturation and having from one to ten carbon atoms, for example, methylene, ethylene, propylene, π-butylene and the like. Alkylene is a subset of alkyl, referring to the same residues as alkyl, but having two points of attachment and, specifically, fully saturated. Examples of alkylene include ethylene (-CH2CH2-), propylene (-CH2CH2CH2-), dimethylpropylene (-CH2C(CH3)2CH2-), and cyclohexylpropylene (-CH2CH2CH(C63)).
[0194] "Alkylidene" refers to a straight or branched chain unsaturated divalent radical consisting solely of carbon and hydrogen atoms, having from two to ten carbon atoms, for example, ethylidene, propylidene, n-butylidene, and the like. Alkylidene is a subset of alkyl, referring to the same residues as alkyl, but having two points of attachment and, specifically, double bond unsaturation. The unsaturation present includes at least one double bond.
[0195] "Alkylidyne" refers to a straight or branched chain unsaturated divalent radical consisting solely of carbon and hydrogen atoms having from two to ten carbon atoms, for example, propylid-2-ynyl, /z-butylid-1-ynyl, and the like. Alkylidyne is a subset of alkyl, referring to the same residues as alkyl, but having two points of attachment and, specifically, triple bond unsaturation. The unsaturation present includes at least one triple bond.
[0196] Any of the above radicals, "alkylene," "alkylidene" and "alkylidyne," when optionally substituted, may contain alkyl substitution which itself contains unsaturation. For example, 2-(2-phenylethynyl-but-3-enyl)-naphthalene (IUPAC name) contains an n-butylid-3-ynyl radical with a vinyl substituent at the 2-position of said radical.
[0197] "Alkoxy" or "alkoxyl" refers to the group -O-alkyl, for example including from one to eight carbon atoms of a straight, branched, cyclic configuration, unsaturated chains, and combinations thereof attached to the parent structure through an oxygen atom. Examples include methoxy, ethoxy, propoxy, isopropoxy, cyclopropyloxy, cyclohexyloxy and the like. Lower-alkoxy refers to groups containing one to six carbons.
[0198] "Substituted alkoxy" refers to the group -O-(substituted alkyl), the substitution on the alkyl group generally containing more than only carbon (as defined by alkoxy). One exemplary substituted alkoxy group is "polyalkoxy" or -O-optionally substituted alkylene-optionally substituted alkoxy, and includes groups such as -OCH2CH2OCH3, and glycol ethers such as polyethyleneglycol and -O(CH2CH2O)xCH3, where x is an integer of between about two and about twenty, in another example, between about two and about ten, and in a further example between about two and about five. Another exemplary substituted alkoxy group is hydroxyalkoxy or -OCH2(CH2)yOH, where y is for example an integer of between about one and about ten, in another example y is an integer of between about one and about four.
[0199] "Acyl" refers to groups of from one to ten carbon atoms of a straight, branched, cyclic configuration, saturated, unsaturated and aromatic and combinations thereof, attached to the parent structure through a carbonyl functionality. One or more carbons in the acyl residue may be replaced by nitrogen, oxygen or sulfur as long as the point of attachment to the parent remains at the carbonyl. Examples include acetyl, benzoyl, propionyl, isobutyryl, t-butoxycarbonyl, benzyloxycarbonyl and the like. Lower-acyl refers to groups containing one to six carbons.
[0200] "α-Amino Acids" refer to naturally occurring and commercially available amino acids and optical isomers thereof. Typical natural and commercially available α-amino acids are glycine, alanine, serine, homoserine, threonine, valine, norvaline, leucine, isoleucine, norleucine, aspartic acid, glutamic acid, lysine, omithine, histidine, arginine, cysteine, homocysteine, methionine, phenylalanine, homophenylalanine, phenylglycine, ortho-tyrosine, meta-tyrosine, para-tyrosine, tryptophan, glutamine, asparagine, proline and hydroxyproline. A "side chain of an α-amino acid" refers to the radical found on the α-carbon of an α-amino acid as defined above, for example, hydrogen (for glycine), methyl (for alanine), benzyl (for phenylalanine), and the like.
[0201] "Amino" refers to the group -NH2. "Substituted amino," refers to the group -N(H)R or -N(R)R where each R is independently selected from the group: optionally substituted alkyl, optionally substituted alkoxy, optionally substituted aryl, optionally substituted heterocyclyl, acyl, carboxy, alkoxycarbonyl, sulfanyl, sulfinyl and sulfonyl, for example, diethylamino, methylsulfonylamino, furanyl-oxy-sulfonamino.
[0202] "Aryl" refers to aromatic six- to fourteen-membered carbocyclic ring, for example, benzene, naphthalene, indane, tetralin, fluorene and the like, univalent radicals. As univalent radicals, the aforementioned ring examples are named, phenyl, naphthyl, indanyl, tetralinyl, and fluorenyl.
[0203] "Arylene" genetically refers to any aryl that has at least two groups attached thereto. For a more specific example, "phenylene" refers to a divalent phenyl ring radical. A phenylene, thus may have more than two groups attached, but is defined by a minimum of two non-hydrogen groups attached thereto. [0204] "Arylalkyl" refers to a residue in which an aryl moiety is attached to a parent structure via one of an alkylene, alkylidene, or alkylidyne radical. Examples include benzyl, phenethyl, phenylvinyl, phenylallyl and the like. Both the aryl, and the corresponding alkylene, alkylidene, or alkylidyne radical portion of an arylalkyl group may be optionally substituted. "Lower arylalkyl" refers to an arylalkyl where the "alkyl" portion of the group has one to six carbons; this can also be refered to as C1-6 arylalkyl.
[0205] "Exo-alkenyl" refers to a double bond that emanates from an annular carbon, and is not within the ring system, for example the double bond depicted in the formula below.
Figure imgf000151_0001
[0206] In some examples, as appreciated by one of ordinary skill in the art, two adjacent groups on an aromatic system may be fused together to form a ring structure. The fused ring structure may contain heteroatoms and may be optionally substituted with one or more groups. It should additionally be noted that saturated carbons of such fused groups (i.e. saturated ring structures) can contain two substitution groups.
[0207] "Fused-polycyclic" or "fused ring system" refers to a polycyclic ring system that contains bridged or fused rings; that is, where two rings have more than one shared atom in their ring structures. In this application, fused-polycyclics and fused ring systems are not necessarily all aromatic ring systems. Typically, but not necessarily, fused-polycyclics share a vicinal set of atoms, for example naphthalene or 1,2,3,4-tetrahydro-naphthalene. A spiro ring system is not a fused-polycyclic by this definition, but fused polycyclic ring systems of the invention may themselves have spiro rings attached thereto via a single ring atom of the fused-polycyclic.
[0208] "Halogen" or "halo" refers to fluorine, chlorine, bromine or iodine. "Haloalkyl" and "haloaryl" refer genetically to alkyl and aryl radicals that are substituted with one or more halogens, respectively. Thus, "dihaloaryl," "dihaloalkyl," "trihaloaryl" etc. refer to aryl and alkyl substituted with a plurality of halogens, but not necessarily a plurality of the same halogen; thus 4-chloro-3 -fluorophenyl is within the scope of dihaloaryl.
[0209] "Heteroarylene" genetically refers to any heteroaryl that has at least two groups attached thereto. For a more specific example, "pyridylene" refers to a divalent pyridyl ring radical. A pyridylene, thus may have more than two groups attached, but is defined by a minimum of two non-hydrogen groups attached thereto.
[0210] "Heteroatom" refers to O, S, N, or P.
[0211] "Heterocyclyl" refers to a stable three- to fifteen-membered ring radical that consists of carbon atoms and from one to five heteroatoms selected from the group consisting of nitrogen, phosphorus, oxygen and sulfur. For purposes of this invention, the heterocyclyl radical may be a monocyclic, bicyclic or tricyclic ring system, which may include fused or bridged ring systems as well as spirocyclic systems; and the nitrogen, phosphorus, carbon or sulfur atoms in the heterocyclyl radical may be optionally oxidized to various oxidation states. In a specific example, the group -S(O)o-2-, refers to -S- (sulfide), -S(O)- (sulfoxide), and -SO2- (sulfone). For convenience, nitrogens, particularly but not exclusively, those defined as annular aromatic nitrogens, are meant to include their corresponding N-oxide form, although not explicitly defined as such in a particular example. Thus, for a compound of the invention having, for example, a pyridyl ring; the corresponding pyridyl-N-oxide is meant to be included as another compound of the invention. In addition, annular nitrogen atoms may be optionally quaternized; and the ring radical may be partially or fully saturated or aromatic. Examples of heterocyclyl radicals include, but are not limited to, azetidinyl, acridinyl, benzodioxolyl, benzodioxanyl, benzofuranyl, carbazoyl, cinnolinyl, dioxolanyl, indolizinyl, naphthyridinyl, perhydroazepinyl, phenazinyl, phenothiazinyl, phenoxazinyl, phthalazinyl, pteridinyl, purinyl, quinazolinyl, quinoxalinyl, quinolinyl, isoquinolinyl, tetrazoyl, tetrahydroisoquinolyl, piperidinyl, piperazinyl, 2-oxopiperazinyl, 2-oxopiperidinyl, 2-oxopyrrolidinyl, 2-oxoazepinyl, azepinyl, pyrrolyl, 4-piperidonyl, pyrrolidinyl, pyrazolyl, pyrazolidinyl, imidazolyl, imidazolinyl, imidazolidinyl, dihydropyridinyl, tetrahydropyridinyl, pyridinyl, pyrazinyl, pyrimidinyl, pyridazinyl, oxazolyl, oxazolinyl, oxazolidinyl, triazolyl, isoxazolyl, isoxazolidinyl, morpholinyl, thiazolyl, thiazolinyl, thiazolidinyl, isothiazolyl, quinuclidinyl, isothiazolidinyl, indolyl, isoindolyl, indolinyl, isoindolinyl, octahydroindolyl, octahydroisoindolyl, quinolyl, isoquinolyl, decahydroisoquinolyl, benzimidazolyl, thiadiazolyl, benzopyranyl, benzothiazolyl, benzoxazolyl, furyl, tetrahydrofuryl, tetrahydropyranyl, thienyl, benzothieliyl, thiamorpholinyl, thiamorpholinyl sulfoxide, thiamorpholinyl sulfone, dioxaphospholanyl, and oxadiazolyl. [0212] "Heteroalicyclic" refers specifically to a non-aromatic heterocyclyl radical. A heteroalicyclic may contain unsaturation, but is not aromatic.
[0213] "Heteroaryl" refers specifically to an aromatic heterocyclyl radical.
[0214] "Heterocyclylalkyl" refers to a residue in which a heterocyclyl is attached to a parent structure via one of an alkylene, alkylidene, or alkylidyne radical. Examples include (4-methylpiperazin-l-yl) methyl, (morpholin-4-yl) methyl, (pyridine-4-yl) methyl, 2-(oχazolin-2-yl) ethyl, 4-(4-methylpiperazin-l-yl)-2-butenyl, and the like. Both the heterocyclyl, and the corresponding alkylene, alkylidene, or alkylidyne radical portion of a heterocyclylalkyl group may be optionally substituted. "Lower heterocyclylalkyl" refers to a heterocyclylalkyl where the "alkyl" portion of the group has one to six carbons. "Heteroalicyclylalkyl" refers specifically to a heterocyclylalkyl where the heterocyclyl portion of the group is non-aromatic; and "heteroarylalkyl" refers specifically to a heterocyclylalkyl where the heterocyclyl portion of the group is aromatic Such terms may be described in more than one way, for example, "lower heterocyclylalkyl" and "heterocyclyl Cι-6alkyl" are equivalent terms.
[0215] "Optional" or "optionally" means that the subsequently described event or circumstance may or may not occur, and that the description includes instances where said event or circumstance occurs and instances in which it does not. One of ordinary skill in the art would understand that, with respect to any molecule described as containing one or more optional substituents, that only sterically practical and/or synthetically feasible compounds are meant to be included. "Optionally substituted" refers to all subsequent modifiers in a term, for example in the term "optionally substituted arylC1-8 alkyl," optional substitution may occur on both the "C1-8 alkyl" portion and the "aryl" portion of the molecule; and for example, optionally substituted alkyl includes optionally substituted cycloalkyl groups, which in turn are defined as including optionally substituted alkyl groups, potentially ad infinitum. A list of exemplary optional substitution are listed below in the definition of "substituted."
[0216] "Saturated bridged ring system" refers to a bicyclic or polycyclic ring system that is not aromatic. Such a system may contain isolated or conjugated unsaturation, but not aromatic or heteroaromatic rings in its core structure (but may have aromatic substitution thereon). For example, hexahydro-furo[3,2-b]furan, 2,3,3a,4,7,7a-hexahydro-lH-indene, 7-aza-bicyclo[2.2.1]heptane, and l,2,3,4,4a,5,8,8a-octahydro-naphthalene are all included in the class "saturated bridged ring system." [0217] "Spirocyclyl" or "spirocyclic ring" refers to a ring originating from a particular annular carbon of another ring. For example, as depicted below, a ring atom of a saturated bridged ring system (rings B and B'), but not a bridgehead atom, can be a shared atom between the saturated bridged ring system and a spirocyclyl (ring A) attached thereto. A spirocyclyl can be carbocyclic or heteroalicyclic.
Figure imgf000154_0001
[0218] "Substituted" alkyl, aryl, and heterocyclyl, refer respectively to alkyl, aryl, and heterocyclyl, wherein one or more (for example up to about five, in another example, up to about three) hydrogen atoms are replaced by a substituent independently selected from: optionally substituted alkyl (for example, fluoromethyl), optionally substituted aryl (for example, 4-hydroxyphenyl), optionally substituted arylalkyl (for example, 1-phenyl-ethyl), optionally substituted heterocyclylalkyl (for example, l-pyridin-3-yl-ethyl), optionally substituted heterocyclyl (for example, 5-chloro-pyridin-3-yl or l-methyl-piperidin-4-yl), optionally substituted alkoxy, alkylenedioxy (for example methylenedioxy), optionally substituted amino (for example, alkylamino and dialkylamino), optionally substituted amidino, optionally substituted aryloxy (for example, phenoxy), optionally substituted arylalkyloxy (for example, benzyloxy), carboxy (-CO2H), carboalkoxy (that is, acyloxy or -OC(=O)R), carboxyalkyl (that is, esters or -CO2R), carboxamido, benzyloxycarbonylamino (CBZ-amino), cyano, acyl, halogen, hydroxy, nitro, sulfanyl, sulfinyl, sulfonyl, thiol, halogen, hydroxy, oxo, carbamyl, acylamino, and sulfonamido.
[0219] "Suitable leaving group" is defined as the term would be understood by one of ordinary skill in the art; that is, a carbon with such a group attached, upon reaction wherein a new bond is to be formed, loses such a group upon formation of the new bond. The invention pertains particularly with respect convergent synthesis, to reactions where such a, leaving group is bonded to a reaction partner that is aromatic, undergoes a bond- forming reaction and remains aromatic. A typical example of such a reaction is a nucleophilic aromatic substitution reaction, as would be understood by one of ordinary skill in the art. However, the invention is not limited to such mechanistic restrictions; for example, reactions where there is, for example, an insertion reaction (for example by a transition metal) into the bond between the aromatic reaction partner and its leaving group followed by reductive coupling can also be used within the scope of the invention. Examples of suitable leaving groups include halogens, optionally substituted aryl or alkyl sulfonates, phosphonates, azides, RS(O)o-2- where R is, for example optionally substituted alkyl, optionally substituted aryl, or optionally substituted heteroaryl.
[0220] "Sulfanyl" refers to the groups: -S-(optionally substituted alkyl), -S-(optionally substituted aryl), and -S-(optionally substituted heterocyclyl).
[0221] "Sulfinyl" refers to the groups: -S(O)-H, -S(O)-(optionally substituted alkyl), -S(O)-optionally substituted aryl), and -S(O)-(optionally substituted heterocyclyl).
[0222] "Sulfonyl" refers to the groups: -S(O2)-H, -S(O2)-(optionally substituted alkyl), -S(O2)-optionally substituted aryl), -S(O2)-(optionally substituted heterocyclyl), -S(O2)-(optionally substituted alkoxy), -S(O2)-oρtionally substituted aryloxy), and -S(O2)-(optionally substituted heterocyclyloxy).
[0223] "Yield" for each of the reactions described herein is expressed as a 'percentage of the theoretical yield.
[0224] Some of the compounds of the invention may have imino, amino, oxo or hydroxy substituents off aromatic heterocyclyl systems. For purposes of this disclosure, it is understood that such imino, amino, oxo or hydroxy substituents may exist in their corresponding tautomeric form, i.e., amino, imino, hydroxy or oxo, respectively.
[0225] Compounds of the invention are named according to systematic application of the nomenclature rules agreed upon by the International Union of Pure and Applied Chemistry (IUPAC), International Union of Biochemistry and Molecular Biology (IUBMB), and the Chemical Abstracts Service (CAS).
[0226] The compounds of the invention, or their pharmaceutically acceptable salts, may have asymmetric carbon atoms, oxidized sulfur atoms or quaternized nitrogen atoms in their structure.
[0227] The compounds of the invention and their pharmaceutically acceptable salts may exist as single stereoisomers, racemates, and as mixtures of enantiomers and diastereomers. The compounds may also exist as geometric isomers. All such single stereoisomers, racemates and mixtures thereof, and geometric isomers are intended to be within the scope of this invention. [0228] It is assumed that when considering generic descriptions of compounds of the invention for the purpose of constructing a compound, such construction results in the creation of a stable structure. That is, one of ordinary skill in the art would recognize that there can theoretically be some constructs which would not normally be considered as stable compounds (that is, sterically practical and/or synthetically feasible, supra).
[0229] When a particular group with its bonding structure is denoted as being bonded to two partners; that is, a divalent radical, for example, -OCH2-, then it is understood that either of the two partners may be bound to the particular group at one end, and the other partner is necessarily bound to the other end of the particular group, unless stated explicitly otherwise. Stated another way, divalent radicals are not to be construed as limited to the depicted orientation, for example "-OCH2-" is meant to mean not only "-OCH2-" as drawn, but also "-CH2O-."
[0230] Methods for the preparation and/or separation and isolation of single stereoisomers from racemic mixtures or non-racemic mixtures of stereoisomers are well known in the art. For example, optically active (R)- and (S)- isomers may be prepared using chiral synthons or chiral reagents, or resolved using conventional techniques. Enantiomers (R- and S-isomers) may be resolved by methods known to one of ordinary skill in the art, for example by: formation of diastereoisomeric salts or complexes which may be separated, for example, by crystallization; via formation of diastereoisomeric derivatives which may be separated, for example, by crystallization, selective reaction of one enantiomer with an enantiomer-specific reagent, for example enzymatic oxidation or reduction, followed by separation of the modified and unmodified enantiomers; or gas-liquid or liquid chromatography in a chiral environment, for example on a chiral support, such as silica with a bound chiral ligand or in the presence of a chiral solvent. It will be appreciated that where a desired enantiomer is converted into another chemical entity by one of the separation procedures described above, a further step may be required to liberate the desired enantiomeric form. Alternatively, specific enantiomer may be synthesized by asymmetric synthesis using optically active reagents, substrates, catalysts or solvents, or by converting on enantiomer to the other by asymmetric transformation. For a mixture of enantiomers, enriched in a particular enantiomer, the major component enantiomer may be further enriched (with concomitant loss in yield) by recrystallization.
[0231] "Patient" for the purposes of the present invention includes humans and other animals, particularly mammals, and other organisms. Thus the methods are applicable to both human therapy and veterinary applications. In a preferred embodiment the patient is a mammal, and in a most preferred embodiment the patient is human.
[0232] "Kinase-dependent diseases or conditions" refer to pathologic conditions that depend on the activity of one or more protein kinases. Kinases either directly or indirectly participate in the signal transduction pathways of a variety of cellular activities including proliferation, adhesion, migration, differentiation and invasion. Diseases associated with kinase activities include tumor growth, the pathologic neovascularization that supports solid tumor growth, and associated with other diseases where excessive local vascularization is involved such as ocular diseases (diabetic retinopathy, age-related macular degeneration, and the like) and inflammation (psoriasis, rheumatoid arthritis, and the like).
[0233] While not wishing to be bound to theory, phosphatases can also play a role in "kinase-dependent diseases or conditions" as cognates of kinases; that is, kinases phosphorylate and phosphatases dephosphorylate, for example protein substrates. Therefore compounds of the invention, while modulating kinase activity as described herein, may also modulate, either directly or indirectly, phosphatase activity. This additional modulation, if present, may be synergistic (or not) to activity of compounds of the invention toward a related or otherwise interdependent kinase or kinase family. In any case, as stated previously, the compounds of the invention are useful for treating diseases characterized in part by abnormal levels of cell proliferation (i.e. tumor growth), programmed cell death (apoptosis), cell migration and invasion and angiogenesis associated with tumor growth.
[0234] "Therapeutically effective amount" is an amount of a compound of the invention, that when administered to a patient, ameliorates a symptom of the disease. The amount of a compound of the invention which constitutes a "therapeutically effective amount" will vary depending on the compound, the disease state and its severity, the age of the patient to be treated, and the like. The therapeutically effective amount can be determined routinely by one of ordinary skill in the art having regard to his own knowledge and to this disclosure.
[0235] "Cancer" refers to cellular-proliferative disease states, including but not limited to: Cardiac: sarcoma (angiosarcoma, fibrosarcoma, rhabdomyosarcoma, liposarcoma), myxoma, rhabdomyoma, fibroma, lipoma and teratoma; Lung: bronchogenic carcinoma (squamous cell, undifferentiated small cell, undifferentiated large cell, adenocarcinoma), alveolar (bronchiolar) carcinoma, bronchial adenoma, sarcoma, lymphoma, chondromatous hanlartoma, inesothelioma; Gastrointestinal: esophagus (squamous cell carcinoma, adenocarcinoma, leiomyosarcoma, lymphoma), stomach (carcinoma, lymphoma, leiomyosarcoma), pancreas (ductal adenocarcinoma, insulinorna, glucagonoma, gastrinoma, carcinoid tumors, vipoma), small bowel (adenocarcinoma, lymphoma, carcinoid tumors, Karposi's sarcoma, leiomyoma, hemangioma, lipoma, neurofibroma, fibroma), large bowel (adenocarcinoma, tubular adenoma, villous adenoma, hamartoma, leiomyoma); Genitourinary tract: kidney (adenocarcinoma, Wilm's tumor~ [neplrroblastoma], lymphoma, leukemia), bladder and urethra (squamous cell carcinoma, transitional cell carcinoma, adenocarcinoma), prostate (adenocarcinoma, sarcoma), testis (seminoma, teratoma, embryonal carcinoma, teratocarcinoma, choriocarcinoma, sarcoma, interstitial cell carcinoma, fibroma, fibroadenoma, adenomatoid tumors, lipoma); Liver: hepatoma (hepatocellular carcinoma), cholangiocarcinoma, hepatoblastoma, angiosarcoma, hepatocellular adenoma, hemangioma; Bone: osteogenic sarcoma (osteosarcoma), fibrosarcoma, malignant fibrous histiocytoma, chondrosarcoma, Ewing's sarcoma, malignant lymphoma (reticulum cell sarcoma), multiple myeloma, malignant giant cell tumor chordoma, osteochronfroma (osteocartilaginous exostoses), benign chondroma, chondroblastoma, chondromyxofibroma, osteoid osteoma and giant cell tumors; Nervous system: skull (osteoma, hemangioma, granuloma, xanthoma, osteitis defornians), meninges (meningioma, meningiosarcoma, gliomatosis), brain (astrocytoma, medulloblastoma, glioma, ependymoma, germinoma [pinealoma], glioblastoma multiform, oligodendroglioma, schwannoma, retinoblastoma, congenital tumors), spinal cord neurofibroma, meningioma, glioma, sarcoma); Gynecological: uterus (endometrial carcinoma), cervix (cervical carcinoma, pre-tumor cervical dysplasia), ovaries (ovarian carcinoma [serous cystadenocarcinoma, mucinous cystadenocarcinoma, unclassified carcinoma], granulosa-thecal cell tumors, SertoliLeydig cell tumors, dysgerminoma, malignant teratoma), vulva (squamous cell carcinoma, intraepithelial carcinoma, adenocarcinoma, fibrosarcoma, melanoma), vagina (clear cell carcinoma, squamous cell carcinoma, botryoid sarcoma (embryonal rhabdomyosarcoma], fallopian tubes (carcinoma); Hematologic: blood (myeloid leukemia [acute and chronic], acute lymphoblastic leukemia, chronic lymphocytic leukemia, myeloproliferative diseases, multiple myeloma, myelodysplastic syndrome), Hodgkin's disease, non-Hodgkin's lymphoma [malignant lymphoma]; Skin: malignant melanoma, basal cell carcinoma, squamous cell carcinoma, Karposi's sarcoma, moles dysplastic nevi, lipoma, angiortia, dermatofibroma, keloids, psoriasis; and Adrenal lands: neuroblastoma. Thus, the term "cancerous cell" as provided herein, includes a cell afflicted by any one of the above-identified conditions.
[0236] "Pharmaceutically acceptable acid addition salt" refers to those salts that retain the biological effectiveness of the free bases and that are not biologically or otherwise undesirable, formed with inorganic acids such as hydrochloric acid, hydrobromie acid, sulfuric acid, nitric acid, phosphoric acid, and the like, as well as organic acids such as acetic acid, trifluoroacetic acid, propionic acid, glycolic acid, pyruvic acid, oxalic acid, maleic acid, malonic acid, succinic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, salicylic acid and the like.
[0237] "Pharmaceutically acceptable base addition salts" include those derived from inorganic bases such as sodium, potassium, lithium, ammonium, calcium, magnesium, iron, zinc, copper, manganese, aluminum salts and the like. Exemplary salts are the ammonium, potassium, sodium, calcium, and magnesium salts. Salts derived from pharmaceutically acceptable organic non-toxic bases include, but are not limited to, salts of primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines and basic ion exchange resins, such as isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, ethanolamine, 2-dimethylaminoethanol, 2-diethylaminoethanol, dicyclohexylamine, lysine, arginine, histidine, caffeine, procaine, hydrabamine, choline, betaine, ethylenediamine, glucosamine, methylglucamine, theobromine, purines, piperazine, piperidine, N-ethylpiperidine, polyamine resins, and the like. Exemplary organic bases are isopropylamine, diethylamine, ethanolamine, trimethylamine, dicyclohexylamine, choline, and caffeine. (See, for example, S. M. Berge, et al., "Pharmaceutical Salts," J. Pharm. Sci., 1977;66:1-19 which is incorporated herein by reference.)
[0238] "Prodrug" refers to compounds that are transformed (typically rapidly) in vivo to yield the parent compound of the above formulae, for example, by hydrolysis in blood. Common examples include, but are not limited to, ester and amide forms of a compound having an active form bearing a carboxylic acid moiety. Examples of pharmaceutically acceptable esters of the compounds of this invention include, but are not limited to, alkyl esters (for example with between about one and about six carbons) wherein the alkyl group is a straight or branched chain. Acceptable esters also include cycloalkyl esters and arylalkyl esters such as, but not limited to benzyl. Examples of pharmaceutically acceptable amides of the compounds of this invention include, but are not limited to, primary amides, and secondary and tertiary alkyl amides (for example with between about one and about six carbons). Amides and esters of the compounds of the present invention may be prepared according to conventional methods. A thorough discussion of prodrugs is provided in T. Higuchi and V. Stella, "Pro-drugs as Novel Delivery Systems," Vol 14 of the A.C.S. Symposium Series, and in Bioreversible Carriers in Drug Design, ed. Edward B. Roche, American Pharmaceutical Association and Pergamon Press, 1987, both of which are incorporated herein by reference for all purposes.
[0239] "Metabolite" refers to the break-down or end product of a compound or its salt produced by metabolism or biotransformation in the animal or human body; for example, biotransformation to a more polar molecule such as by oxidation, reduction, or hydrolysis, or to a conjugate (see Goodman and Gilman, "The Pharmacological Basis of Therapeutics" δ.sup.th Ed., Pergamon Press, Gilman et al. (eds), 1990 for a discussion of biotransformation). As used herein, the metabolite of a compound of the invention or its salt may be the biologically active form of the compound in the body. In one example, a prodrug may be used such that the biologically active form, a metabolite, is released in vivo. In another example, a biologically active metabolite is discovered serendipitously, that is, no prodrug design per se was undertaken. An assay for activity of a metabolite of a compound of the present invention is known to one of skill in the art in light of the present disclosure.
[0240] In addition, the compounds of the present invention can exist in unsolvated as well as solvated forms with pharmaceutically acceptable solvents such as water, ethanol, and the like. In general, the solvated forms are considered equivalent to the unsolvated forms for the purposes of the present invention.
[0241] In addition, it is intended that the present invention cover compounds made either using standard organic synthetic techniques, including combinatorial chemistry or by biological methods, such as bacterial digestion, metabolism, enzymatic conversion, and the like.
[0242] "Treating" or "treatment" as used herein covers the treatment of a disease-state in a human, which disease-state is characterized by abnormal cellular proliferation, and invasion and includes at least one of: (i) preventing the disease-state from occurring in a human, in particular, when such human is predisposed to the disease-state but has not yet been diagnosed as having it; (ii) inhibiting the disease-state, i.e., arresting its development; and (iii) relieving the disease-state, i.e., causing regression of the disease-state. As is known in the art, adjustments for systemic versus localized delivery, age, body weight, general health, sex, diet, time of administration, drug interaction and the severity of the condition may be necessary, and will be ascertainable with routine experimentation by one of ordinary skill in the art.
[0243] One of ordinary skill in the art would understand that certain crystallized, protein- ligand complexes, in particular c-Met, c-Kit, KDR, flt-3, or flt-4-ligand complexes, and their corresponding x-ray structure coordinates can be used to reveal new structural information useful for understanding the biological activity of kinases as described herein. As well, the key structural features of the aforementioned proteins, particularly, the shape of the ligand binding site, are useful in methods for designing or identifying selective modulators of kinases and. in solving the structures of other proteins with similar features. Such protein-ligand complexes, having compounds of the invention as their ligand component, are an aspect of the invention.
[0244] As well, one of ordinary skill in the art would appreciate that such suitable x-ray quality crystals can be used as part of a method of identifying a candidate agent capable of binding to and modulating the activity of kinases. Such methods may be characterized by the following aspects: a) introducing into a suitable computer program, information defining a ligand binding domain of a kinase in a conformation (e.g. as defined by x-ray structure coordinates obtained from suitable x-ray quality crystals as described above) wherein the computer program creates a model of the three dimensional structures of the ligand binding domain, b) introducing a model of the three dimensional structure of a candidate agent in the computer program, c) superimposing the model of the candidate agent on the model of the ligand binding domain, and d) assessing whether the candidate agent model fits spatially into the ligand binding domain. Aspects a-d are not necessarily carried out in the aforementioned order. Such methods may further entail: performing rational drug design with the model of the three-dimensional structure, and selecting a potential candidate agent in conjunction with computer modeling.
[0245] Additionally, one skilled in the art would appreciate that such methods may further entail: employing a candidate agent, so-determined to fit spatially into the ligand binding domain, in a biological activity assay for kinase modulation, and determining whether said candidate agent modulates kinase activity in the assay. Such methods may also include administering the candidate agent, determined to modulate kinase activity, to a mammal suffering from a condition treatable by kinase modulation, such as those described above.
[0246] Also, one skilled in the art would appreciate that compounds of the invention can be used in a method of evaluating the ability of a test agent to associate with a molecule or molecular complex comprising a ligand binding domain of a kinase. Such a method may be characterized by the following aspects: a) creating a computer model of a kinase binding pocket using stracture coordinates obtained from suitable x-ray quality crystals of the kinase, b) employing computational algorithms to perform a fitting operation between the test agent and the computer model of the binding pocket, and c) analyzing the results of the fitting operation to quantify the association between the test agent and the computer model of the binding pocket.
General Adniinistration
[0247] Administration of the compounds of the invention, or their pharmaceutically acceptable salts, in pure form or in an appropriate pharmaceutical composition, can be carried out via any of the accepted modes of administration or agents for serving si ilar utilities. Thus, administration can be, for example, orally, nasally, parenterally (intravenous, intramuscular, or subcutaneous), topically, transdermally, intravaginally, intravesically, intracistemally, or rectally, in the form of solid, semi-solid, lyophilized powder, or liquid dosage forms, such, as for example, tablets, suppositories, pills, soft elastic and hard gelatin capsules, powders, solutions, suspensions, or aerosols, or the like, preferably in unit dosage forms suitable for simple administration of precise dosages.
[0248] The compositions will include a conventional pharmaceutical carrier or excipient and a compound of the invention as the/an active agent, and, in addition, may include other medicinal agents, pharmaceutical agents, carriers, adjuvants, etc. Compositions of the invention may be used in combination with anticancer or other agents that are generally administered to a patient being treated for cancer. Adjuvants include preserving, wetting, suspending, sweetening, flavoring, perfuming, emulsifying, and dispensing agents. Prevention of the action of microorganisms can be ensured by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, and the like. It may also be desirable to include isotonic agents, for example sugars, sodium chloride, and the like. Prolonged absorption of the injectable pharmaceutical form can be brought about by the use of agents delaying absorption, for example, aluminum monostearate and gelatin.
[0249] If desired, a pharmaceutical composition of the invention may also contain minor amounts of auxiliary substances such as wetting or emulsifying agents, pH buffering agents, antioxidants, and the like, such as, for example, citric acid, sorbitan monolaurate, triethanolamine oleate, butylalted hydroxytoluene, etc.
[0250] Compositions suitable for parenteral injection may comprise physiologically acceptable sterile aqueous or nonaqueous solutions, dispersions, suspensions or emulsions, and sterile powders for reconstitution into sterile injectable solutions or dispersions. Examples of suitable aqueous and nonaqueous carriers, diluents, solvents or vehicles include water, ethanol, polyols (propyleneglycol, polyethyleneglycol, glycerol, and the like), suitable mixtures thereof, vegetable oils (such as olive oil) and injectable organic esters such as ethyl oleate. Proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersions and by the use of surfactants.
[0251] One preferable route of administration is oral, using a convenient daily dosage regimen that can be adjusted according to the degree of severity of the disease-state to be treated.
[0252] Solid dosage forms for oral administration include capsules, tablets, pills, powders, and granules. In such solid dosage forms, the active compound is admixed with at least one inert customary excipient (or carrier) such as sodium citrate or dicalcium phosphate or (a) fillers or extenders, as for example, starches, lactose, sucrose, glucose, mannitol, and silicic acid, (b) binders, as for example, cellulose derivatives, starch, alignates, gelatin, polyvinylpyrrolidone, sucrose, and gum acacia, (c) humectants, as for example, glycerol, (d) disintegrating agents, as for example, agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, croscarmellose sodium, complex silicates, and sodium carbonate, (e) solution retarders, as for example paraffin, (f) absorption accelerators, as for example, quaternary ammonium compounds, (g) wetting agents, as for example, cetyl alcohol, and glycerol monostearate, magnesium stearate and the like (h) adsorbents, as for example, kaolin and bentonite, and (i) lubricants, as for example, talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate, or mixtures thereof. In the case of capsules, tablets, and pills, the dosage forms may also comprise buffering agents. [0253] Solid dosage forms as described above can be prepared with coatings and shells, such as enteric coatings and others well known in the art. They may contain pacifying agents, and can also be of such composition that they release the active compound or compounds in a certain part of the intestinal tract in a delayed manner. Examples of embedded compositions that can be used are polymeric substances and waxes. The active compounds can also be in microencapsulated form, if appropriate, with one or more of the above-mentioned excipients.
[0254] Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, solutions, suspensions, syrups, and elixirs. Such dosage forms are prepared, for example, by dissolving, dispersing, etc., a compound(s) of the invention, or a pharmaceutically acceptable salt thereof, and optional pharmaceutical adjuvants in a carrier, such as, for example, water, saline, aqueous dextrose, glycerol, ethanol and the like; solubilizmg agents and emulsifiers, as for example, ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propyleneglycol, 1,3- butyleneglycol, dimethylformamide; oils, in particular, cottonseed oil, groundnut oil, corn germ oil, olive oil, castor oil and sesame oil, glycerol, tetrahydrofurfuryl alcohol, polyethyleneglycols and fatty acid esters of sorbitan; or mixtures of these substances, and the like, to thereby form a solution or suspension.
[0255] Suspensions, in addition to the active compounds, may contain suspending agents, as for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar and tragacanth, or mixtures of these substances, and the like.
[0256] Compositions for rectal administrations are, for example, suppositories that can be prepared by mixing the compounds of the present invention with for example suitable non- irritating excipients or carriers such as cocoa butter, polyethyleneglycol or a suppository wax, which are solid at ordinary temperatures but liquid at body temperature and therefore, melt while in a suitable body cavity and release the active component therein.
[0257] Dosage forms for topical administration of a compound of this invention include ointments, powders, sprays, and inhalants. The active component is admixed under sterile conditions with a physiologically acceptable carrier and any preservatives, buffers, or propellants as may be required. Ophthalmic formulations, eye ointments, powders, and solutions are also contemplated as being within the scope of this invention. [0258] Generally, depending on the intended mode of administration, the pharmaceutically acceptable compositions will contain about 1% to about 99% by weight of a compound(s) of the invention, or a pharmaceutically acceptable salt thereof, and 99% to 1% by weight of a suitable pharmaceutical excipient. In one example, the composition will be between about 5% and about 75% by weight of a compound(s) of the invention, or a pharmaceutically acceptable salt thereof, with the rest being suitable pharmaceutical excipients.
[0259] Actual methods of preparing such dosage forms are known, or will be apparent, to those skilled in this art; for example, see Remington's Pharmaceutical Sciences, 18th Ed., (Mack Publishing Company, Easton, Pa., 1990). The composition to be administered will, in any event, contain a therapeutically effective amount of a compound of the invention, or a pharmaceutically acceptable salt thereof, for treatment of a disease-state in accordance with the teachings of this invention.
[0260] The compounds of the invention, or their pharmaceutically acceptable salts, are administered in a therapeutically effective amount which will vary depending upon a variety of factors including the activity of the specific compound employed, the metabolic stability and length of action of the compound, the age, body weight, general health, sex, diet, mode and time of administration, rate of excretion, drug combination, the severity of the particular disease-states, and the host undergoing therapy. The compounds of the present invention can be administered to a patient at dosage levels in the range of about 0.1 to about 1,000 mg per day. For a normal human adult having a body weight of about 70 kilograms, a dosage in the range of about 0.01 to about 100 mg per kilogram of body weight per day is an example. The specific dosage used, however, can vary. For example, the dosage can depend on a number of factors including the requirements of the patient, the severity of the condition being treated, and the pharmacological activity of the compound being used. The determination of optimum dosages for a particular patient is well known to one of ordinary skill in the art.
Utility of compounds of the invention as screening agents
[0261] To employ the compounds of the invention in a method of screening for candidate agents that bind to, for example c-Met, KDR, c-Kit, flt-3, or flt-4, the protein is bound to a support, and a compound of the invention is added to the assay. Alternatively, the compound of the invention is bound to the support and the protein is added. Classes of candidate agents among which novel binding agents may be sought include specific antibodies, non-natural binding agents identified in screens of chemical libraries, peptide analogs, etc. Of particular interest are screening assays for candidate agents that have a low toxicity for human cells. A wide variety of assays may be used for this purpose, including labeled in vitro protein-protein binding assays, electrophoretic mobility shift assays, immunoassays for protein binding, functional assays (phosphorylation assays, etc.) and the like.
[0262] The determination of the binding of the candidate agent to, for example, c-Met, KDR, c-Kit, flt-3, or flt-4 protein may be done in a number of ways. In one example, the candidate agent (the compound of the invention) is labeled, for example, with a fluorescent or radioactive moiety and binding determined directly. For example, thus may be done by attaching all or a portion of the c-Met, KDR, c-Kit, flt-3, or flt-4 protein to a solid support, adding a labeled agent (for example a compound of the invention in which at least one atom has been replaced by a detectable isotope), washing off excess reagent, and determining whether the amount of the label is that present on the solid support. Various blocking and washing steps may be utilized as is known in the art.
[0263] By "labeled" herein is meant that the compound is either directly or indirectly labeled with a label which provides a detectable signal, e.g., radioisotope, fluorescent tag, enzyme, antibodies, particles such as magnetic particles, chemiluminescent tag, or specific binding molecules, etc. Specific binding molecules include pairs, such as biotin and streptavidin, digoxin and antidigoxin etc. For the specific binding members, the complementary member would normally be labeled with a molecule which provides for detection, in accordance with known procedures, as outlined above. The label can directly or indirectly provide a detectable signal.
[0264] In some embodiments, only one of the components is labeled. For example, c-Met, KDR, c-Kit, flt-3, or flt-4 protein may be labeled at tyrosine positions using 125I, or with fluorophores. Alternatively, more than one component may be labeled with different labels; using I for the proteins, for example, and a fluorophor for the candidate agents.
[0265] The compounds of the invention may also be used as competitors to screen for additional drug candidates. "Candidate bioactive agent" or "drug candidate" or grammatical equivalents as used herein describe any molecule, e.g., protein, oligopeptide, small organic molecule, polysaccharide, polynucleotide, etc., to be tested for bioactivity. They may be capable of directly or indirectly altering the cellular proliferation phenotype or the expression of a cellular proliferation sequence, including both nucleic acid sequences and protein sequences. In other cases, alteration of cellular proliferation protein binding and/or activity is screened. In the case where protein binding or activity is screened, some embodiments exclude molecules already known to bind to that particular protein. Exemplary embodiments of assays described herein include candidate agents, which do not bind the target protein in its endogenous native state, termed herein as "exogenous" agents. In one example, exogenous agents further exclude antibodies to c- Met, KDR, c-Kit, flt-3, or flt-4.
[0266] Candidate agents can encompass numerous chemical classes, though typically they are organic molecules having a molecular weight of more than about 100 daltons and less than about 2,500 daltons. Candidate agents comprise functional groups necessary for structural interaction with proteins, particularly hydrogen bonding and lipophilic binding, and typically include at least an amine, carbonyl, hydroxyl, ether, or carboxyl group, for example at least two of the functional chemical groups. The candidate agents often comprise cyclical carbon or heterocyclyl structures and/or aromatic or polyaromatic structures substituted with one or more of the above functional groups. Candidate agents are also found among biomolecules including peptides, saccharides, fatty acids, steroids, purines, pyrimidmes, derivatives, structural analogs, or combinations thereof.
[0267] Candidate agents are obtained from a wide variety of sources including libraries of synthetic or natural compounds. For example, numerous means are available for random and directed synthesis of a wide variety of organic compounds and biomolecules, including expression of randomized oligonucleotides. Alternatively, libraries of natural compounds in the form of bacterial, fungal, plant and animal extracts are available or readily produced. Additionally, natural or synthetically produced libraries and compounds are readily modified through conventional chemical, physical and biochemical means. Known pharmacological agents may be subjected to directed or random chemical modifications, such as acylation, alkylation, esterification, amidification to produce structural analogs.
[0268] In one example, the binding of the candidate agent is determined through the use of competitive binding assays. In this example, the competitor is a binding moiety known to bind to c-Met, KDR, c-Kit, flt-3, or flt-4, such as an antibody, peptide, binding partner, ligand, etc. Under certain circumstances, there may be competitive binding as between the candidate agent and the binding moiety, with the binding moiety displacing the candidate agent.
[0269] In some embodiments, the candidate agent is labeled. Either the candidate agent, or the competitor, or both, is added first to for example c-Met, KDR, c-Kit, flt-3, or flt-4 for a time sufficient to allow binding, if present. Incubations may be performed at any temperature that facilitates optimal activity, typically between 4°C and 40°C
[0270] Incubation periods are selected for optimum activity, but may also be optimized to facilitate rapid high throughput screening. Typically between 0.1 and 1 hour will be sufficient. Excess reagent is generally removed or washed away. The second component is then added, and the presence or absence of the labeled component is followed, to indicate binding.
[0271] In one example, the competitor is added first, followed by the candidate agent. Displacement of the competitor is an indication the candidate agent is binding to c-Met, KDR, c-Kit, flt-3, or flt-4 and thus is capable of binding to, and potentially modulating, the activity of the c-Met, KDR, c-Kit, flt-3, or flt-4. In this embodiment, either component can be labeled. Thus, for example, if the competitor is labeled, the presence of label in the wash solution indicates displacement by the agent. Alternatively, if the candidate agent is labeled, the presence of the label on the support indicates displacement.
[0272] In an alternative embodiment, the candidate agent is added first, with incubation and washing, followed by the competitor. The absence of binding by the competitor may indicate the candidate agent is bound to c-Met, KDR, c-Kit, flt-3, or flt-4 with a higher affinity. Thus, if the candidate agent is labeled, the presence of the label on the support, coupled with a lack of competitor binding, may indicate the candidate agent is capable of binding to c-Met, KDR, c-Kit, flt-3, or flt-4.
[0273] It may be of value to identify the binding site of c-Met, KDR, c-Kit, flt-3, or flt-4. This can be done in a variety of ways. In one embodiment, once c-Met, KDR, c-Kit, flt-3, or flt-4 has been identified as binding to the candidate agent, the c-Met, KDR, c-Kit, flt-3, or flt-4 is fragmented or modified and the assays repeated to identify the necessary components for binding.
[0274] Modulation is tested by screening for candidate agents capable of modulating the activity of c-Met, KDR, c-Kit, flt-3, or flt-4 comprising the steps of combining a candidate agent with c-Met, KDR, c-Kit, flt-3, or flt-4, as above, and determining an alteration in the biological activity of the c-Met, KDR, c-Kit, flt-3, or flt-4. Thus, in this embodiment, the candidate agent should both bind to (although this may not be necessary), and alter its biological or biochemical activity as defined herein. The methods include both in vitro screening methods and in vivo screening of cells for alterations in cell viability, morphology, and the like.
[0275] Alternatively, differential screening may be used to identify drag candidates that bind to native c-Met, KDR, c-Kit, flt-3, or flt-4, but cannot bind to modified c-Met, KDR, c-Kit, flt-3, or flt-4.
[0276] Positive controls and negative controls may be used in the assays. For example, all control and test samples are performed in at least triplicate to obtain statistically significant results. Incubation of samples is for a time sufficient for the binding of the agent to the protein. Following incubation, samples are washed free of non-specifically bound material and the amount of bound, generally labeled agent determined. For example, where a radiolabel is employed, the samples may be counted in a scintillation counter to determine the amount of bound compound.
[0277] A variety of other reagents may be included in the screening assays. These include reagents like salts, neutral proteins, e.g., albumin, detergents, etc which may be used to facilitate optimal protein-protein binding and/or reduce non-specific or background interactions. Also reagents that otherwise improve the efficiency of the assay, such as protease inhibitors, nuclease inhibitors, anti-microbial agents, etc., may be used. The mixture of components may be added in any order that provides for the requisite binding.
Abbreviations and their Definitions
[0278] The following abbreviations and terms have the indicated meanings throughout.
Figure imgf000169_0001
Figure imgf000170_0001
Figure imgf000171_0001
Figure imgf000172_0001
Synthesis of Compounds 79] Schemes 1 and 2 depict general synthetic routes for compounds of the invention and are not intended to be limiting. More specifically, Scheme 1 depicts synthesis of quinazoline compounds, and Scheme 2 depicts synthesis of quinoline compounds. Specific examples are described subsequently to these general synthetic descriptions so as to allow one skilled in the art to make and use either quinazolines or quinolines of the invention.
Scheme 1
Figure imgf000173_0001
4
Figure imgf000173_0002
8
[0280] Referring to Scheme 1, a benzoic ester 1, where R is typically but not necessarily a methyl radical and P is typically but not necessarily an alkyl group, is O-alkylated at the oxygen para to the carboxylate group with an electrophile to afford a substituted derivative 2. P is typically a lower alkyl group, but may be a protecting group that is removed later in a synthesis. When P is a lower alkyl group it may possess functionality initially, or be derivitized to contain such functionality at various stages of the synthesis. The group, E1, may represent either a protecting group, e.g. benzyl, or a group that either has moieties present in compounds of the invention or possesses functionality that serve as a precursors to such groups. Aromatic ring nitration and reduction of the corresponding nitro group are carried out in a regio- and chemoselective manner by methods well known in the art to give anthranilate derivative 3. Formation of quinazolin-4-one 4 is carried out by methods well known in the art, for example by heating 3 in formamide solution in the presence of ammonium formate or for example by heating directly with formamidine hydrochloride. Introduction of 4-position functionality groups is carried out by methods known in the art. For example, quinazolin-4-one 4 is converted to an intermediate quinazoline 5, where "L" represents a leaving group, e.g. chlorine. Quinazoline 5 is then converted to 6 by reaction with a range of nucleophiles, e.g. amines, alcohols, and thiols. After formation of 6, group "Z" is either left "as is" or converted at some subsequent stage to a derivative thereof. For example when Z is -NH-, then the hydrogen on the nitrogen may optionally be replaced with an alkyl group, or when Z is sulfur, then that sulfur atom may be oxidized to, for example, a sulfone. Stracture 6 may represent compounds of the invention or, for example when E1 serves as a protecting group, E1 may be removed to provide phenol 7. Introduction of a group E2 is carried out by methods well established in the art; for example alkylation with an appropriately derivatized alkyl halide (or mesylate or the like) to give 8 which also represents compounds of the invention.
Scheme 2
Figure imgf000174_0001
14 13 12
Figure imgf000174_0002
81] Scheme 2 shows a general route used to make exemplary quinolines of the invention. For example, compound 9 contains an alkyl group, R1, a protecting group, P. The arrangement of the protected and alkylated phenolic oxygens may vary from the pattern depicted in compound 9. Compound 9 is nitrated to provide compound 10. The nitro group of compound 10 is reduced to give aniline 11. Compound 11 is treated, for example, with ethyl formate under basic conditions followed by acidification and isolation to form 4-hydroxy quinoline 12. Quinoline 12 may be converted to compounds of the invention in a number of ways. For example, the 4-oxygen is used as a nucleophile in a nucleophilic aromatic substitution reaction to form quinoline-aryl-ether 13. In another example, compound 13 is further derivatized, via removal of protecting group P, to afford compound 14. The 7-hydroxy of compound 14 is alkylated, for example with electrophile E, to provide a compound of the invention. As discussed in relation to Scheme 1, variations on any of the above steps are possible, and intermediates in these schemes, for example compounds 12, 13, and 14 may also be compounds of the invention according to formula I. Also, for example, the 4-hydroxy quinoline compound 12 are converted to a corresponding 4-nitrogen or 4-sulfur quinoline using chemistry known in the art to make compounds of the invention, or alternatively the corresponding 4-nitrogen or 4-sulfur quinolines are made via routes analogous to that depicted in Schemes 1 and 2.
[0282] Schemes 1 and 2 are illustrative of quinolines and quinazolines having oxygen substitution at their respective 6- and 7-positions; the invention is not so limited, but rather is intended to encompass quinolines and quinazolines not necessarily having substitution, oxygen or otherwise, at their respective 6- or 7-positions.
[0283] Schemes 3 and 4 depict generalized synthetic routes to show the process of the invention to make compounds of formua XXI and is not intended to be limiting. More specifically, Schemes 3 and 4 depict convergent syntheses of quinoline and quinazoline compounds as described herein. Specific examples are described subsequently to this general synthetic description so as to allow one of ordinary skill in the art to practice the invention.
[0284] Referring to Scheme 3, a benzoic ester 16 for example, where R is typically but not necessarily a methyl radical and R1 is typically but not necessarily one or more alkoxy or hydroxy groups. In a typical synthesis, at least one of R1 within Scheme 3 is a hydroxyl which is converted (or protected )via one or more steps to a group important to the activity of the compounds as described as kinase modulators (in the case that -OH itself is desired in the final compound, then deprotection affords the -OH, vide supra). Preferably, but not necessarily, this group is complete once the synthesis of XXII is complete. By building desired complexity into XXII prior to combination with XXIII, convergent syntheses' advantages over serial syntheses are realized more fully. Regioselective aromatic ring nitration, and reduction of the corresponding nitro group, are carried out in a regio- and chemoselective manner by methods well known in the art to give anthranilate derivative 17. Formation of quinazoline or quinoline 4-one 18 is carried out by methods well known in the art. For example by heating 17 in formamide solution in the presence of ammonium formate, or by heating 17 with formamidine hydrochloride, the quinazoline-4-one analog is made. In another example 17 is treated, for example, with ethyl formate under basic
• conditions followed by acidification and isolation to form the 4-hydroxy quinoline analog (a tautomer of the 4-one). In this scheme J' represents either carbon or nitrogen atom with the appropriate number of hydrogens to fill their respective normal valence bonding schemes; J' is a precursor to J. Radicals J and R70 are in accord with formula XXI. Introduction of 4-position functionality is carried out by methods known in the art. For example, 4-one 18 is converted to XXII, where "P1" represents a suitable leaving group (in accord with formula XXI), e.g. chlorine (via dehydration/chlorination of 18 to give XXII). In another example, a 4-hydroxy analog is converted to a sulfonyl ester, e.g. the trifluoromethane sulfonate.
Scheme 3
(R1
Figure imgf000176_0001
16 17 18
Figure imgf000176_0002
XXII 85] Scheme 4 shows a general route used to make compounds of formula XXIII. For example, aromatic compound 19, where "X" is a leaving group, such as fluorine and "E" is an electron withdrawing group such as nitro, is converted to 20 by reaction with a range of nucleophiles, e.g. amines, alcohols, and thiols (where "Z" is oxygen, nitrogen (substituted or not), or sulfur). In this case, "R" represents a removable group, for example benzyl. In a typical synthesis, after formation of 20, group "E" is either left "as is" or converted at some subsequent stage to a derivative thereof. In the example depicted, E is converted to B', a precursor to B in accord with formula XXI, to make 21. For example if E is a nitro, then B' could might be an amino group, made via reduction of the nitro group. Structure 21 may be further derivitized by synthesis of -B-L-T in accord with formula XXI. In scheme 4, this is depicted as a serial process whereby L', a precursor to L, is introduced to give 22, followed by introduction of T' (a precursor to T) to give 23. In some cases, -L-T is preformed and appended to B. One of ordinary skill in the art would appreciate that variations on any of the above steps are possible. Compound 23 is converted to XXIII via conversion of T' to T and introduction of P2 (for example, when R is benzyl, removal of the benzyl after completion of -B-L-T). Scheme 4
Figure imgf000177_0001
22 23 XXiπ
[0286] As discussed above, one aspect of the invention encompasses combination of XXII and XXIII to make compounds of formula XXI. Because of the diversity and complexity of compounds described for kinase modulation (vide supra), methods of the invention provide advantages to serial synthesis.
Figure imgf000177_0002
XXI
Examples
[0287] The following examples serve to more fully describe the manner of using the above-described invention, as well as to set forth the best modes contemplated for carrying out various aspects of the invention. It is understood that these examples in no way serve to limit the true scope of this invention, but rather are presented for illustrative purposes. All references cited herein are incorporated by reference in their entirety. Generally, but not necessarily, each example set out below describes a multi-step synthesis as outlined above. Quinoline and Quinazoline Syntheses
Example 1
Figure imgf000178_0001
[0288] Synthesis of l-(4-Benzyloxy-5-methoxy-2-nitro-phenyl -ethanone. l-(4-
Benzyloxy-3-methoxy-phenyl)-ethanone (200 mmol, 51.3 g) dissolved in DCM (750ml) and the mixture cooled to 0° C. Nitric acid (90%, 300 mmol, 14 ml) was added dropwise to the cooled solution over 20 minutes. Sulfuric acid (96.2%, 300 mmol, 8.75 ml) was then added dropwise over 40 minutes at 0° C.
[0289] Additional nitric acid (200 mmol, 9.4 ml) was added dropwise over 20 minutes. The reaction mixture was diluted with water (300 ml) and wash with water (3 X 200 ml), Sat. NaHCO3 (4 X 200 ml, or until neutral). The organic layer was dried over Na2SO4 and concentrated.
[0290] The crude mixture was recrystallized with DMF to give 22.5 g of the nitro product. The DMF layer was concentrated and recrystallized with ethyl acetate to give additional 8.75g of the product. The ethyl acetate layer was concentrated and purified on silica column using 20% EtOAc/hexanes to gave another 4.75 g of the product. Total yield is 36 g, (-60%). IH NMR (CDC13): 7.647 (IH, s), 7.446-7.333 (5H, m), 6.745 (IH, s), 5.210 (2H, s), 3.968 (3H, s), 2.487 (3H, s).
Example 2
Figure imgf000178_0002
[0291] Synthesis of l-(2-Amino-4-benzyloχy-5-methoxy-phenyl -ethanone. A Mixture of iron powder (477 mmol, 27 g), ammonium acetate (500 mmol, 31.g), l-(4-Benzyloxy-5- methoxy-2-nitro-phenyl)-ethanone (120 mmol, 36 g), toluene (500 ml) and water (500 ml) was refluxed overnight, or until completion. The mixture was filtered through celite and washed with EtOAc. The organic layer was washed with water and Sat. NaCl, dried over Na2SO4, and concentrated to afford the product, 90%. IH NMR (CDC13): 7.408-7.298 (5H, m), 7.130 (IH, s), 6.155 (2H, br), 6.104 (IH, s), 5.134 (2H, s), 3.834 (3H, s), 2.507 (3H, s). LC/MS (M+1 = 272).
Example 3
Figure imgf000179_0001
[0292] Synthesis of 7-Benzyloxy-6-methoxy-quinolin-4-ol. To a solution of l-(2-Amino- 4-benzyloxy-5-methoxy-phenyl)-ethanone (108 mmol, 29.3 g) in DME (700 ml) was added sodium methoxide (432 mmol, 23.35 g). The mixture was stirred for 30 minutes. Ethyl formate (540 mmol, 44 ml) was added and the mixture was stirred overnight. (Additional sodium methoxide may be needed if reaction is not complete as monitored by LC MS.) After the reaction was completion, the mixture was diluted with water (40 ml) and acidified to neutral with 1M HCl. The precipitate was filtered and washed with water, dried in vacuo to afford 22g (72%) of 7-benzyloxy-6-methoxy-quinolin-4-ol. IH NMR (CDC13): 10.7 (IH, br), 7.703 (IH, s), 7.493-7.461 (IH, t), 7.431-7.413 (2H, br d), 7.372- 7.333 (2H, t), 7.296-7.283 (IH, d), 6.839 (IH, s), 6.212-6.193 (IH, d), 5.212 (2H, s), 3.965 (3H, s). LC/MS (M+1 = 282).
Example 4
Figure imgf000179_0002
[0293] 7-Benzyloxy-4-(2-fluoro-4-nitro-phenoxy)-6-methoxy-quinoline. To a round bottom flask equipped with a magnetic stir bar was added 7-Benzyloxy-6-methoxy-lH- quinolin-4-one (12.2 g, 43.3 mmol, 1.0 eq.), acetonitrile (150ml), DMF (150ml) and cesium carbonate (28.2 g, 86.5 mmol, 2.0 eq). The mixture was stirred at room temperature for 30 minutes at which time l,2-difluoro-4-nitro-benzene (7.57 g, 47.6 mmol, 1.1 eq) was added over a 10 minute period. After 2 hours the reaction was complete at which time 75% of the MeCN and DMF was removed and the resulting solution was poured over into ice water. The solid was filtered and dried and further columned with a biotage system. The eluent was 1:3 ethyl acetate/hexane. Removal of the solvent afforded 7-Benzyloxy-4-(2-fluoro-4-nitro-phenoxy)-6-methoxy-quinoline as a pale green solid (7.4 g, 41% yield). 1H NMR (400 MHz, CDC13): 8.53 (d, IH), 8.42 (dd, IH), 8.16 (m, IH), 7.5 (m, 8H), 6.76 (d, IH), 5.31 (s, 2H), 3.92 (s, 3H); MS (El) for C23H27FN2O5: 421 (MH+).
Example 5
Figure imgf000180_0001
94] 4-(2-Fluoro-4-nitro-phenoxy)-6-methoxy-quinolin-7-ol. To a round bottom flask equipped with a magnetic stir bar was added 7-benzyloxy-4-(2-fluoro-4-nitro-phenoxy)-6- methoxy-quinoline (2.9 g, 6.9 mmol, l.Oeq) and 33% HBr in acetic acid (30 ml). The mixture was stirred at room temperature for 3 hours and diluted with ether to give a pale white solid. The solid was filtered, washed with ether and dried to yield 4-(2-Fluoro-4- nitro-phenoxy)-6-methoxy-quinolin-7-ol as a pale white solid (2.74 g, 97.5% yield). 1H NMR (400 MHz, CDC13): 11.89 (bs, IH), 8.87 (d, IH), 8.57 (d, IH), 8.30 (d, IH), 7.89 (m, IH), 7.73 (s, IH), 7.55 (s, IH), 4.03 (s, 3H); MS (El) for Cι6HuFN2O5: 421 (M+H1").
Exam
Figure imgf000180_0002
[0295] 5-[4-(2-Fluoro-4-nitro-phenoxy)-6-methoxy-quinolin-7-yloxymethyl1-hexahydro- cvclopentarclpyrrole-2-carboxylic acid benzyl ester. To a round bottom flask equipped with a magnetic stir bar was added 4-(2-Fluoro-4-nitro-phenoxy)-6-methoxy-quinolin-7-ol (2.74 g, 6.7 mmol, 1.0 eq.), DMA (30ml) and cesium carbonate (6.6 g, 20.2 mmol, 3.0 eq). The mixture was stirred at room temperature for 30 minutes at which time 5- methanesulfonyloxymethyl-hexahydro-cyclopenta[c]pyrrole-2-carboxylic acid benzyl ester (2.6 g, 7.3 mmol, 1.1 eq) was added. The reaction was heated to 75° C and allowed to stir overnight. After allowing the reaction to cool to room temperature the reaction was poured into water. The solid was filtered and was then dissolved in EtOAc and washed 2X water, IX brine and dried over NaSO . The solvent was removed to yield 5-[4-(2- Fluoro-4-nitro-phenoxy)-6-methoxy-quinolin-7-yloxymethyl]-hexahydro- cyclopenta[c]pyrrole-2-carboxylic acid benzyl ester as a cream solid (3.7 g, 94% yield). 1H NMR (400 MHz, CDC13): 8.55 (d, IH), 8.15 (d, IH), 8.09 (d, IH), 7.32 (m, 8H), 6.52 (d, IH), 5.11 (d, 2H), 4.13 (d, 2H), 3.95 (s, 3H), 3.57 (m, 2H), 3.43 (m, 2H), 2.93 (m, 3H), 2.16 (m, 2H), 1.39 (m, 2H); MS (El) for C32H30FN3O7: 588 (M+H1 .
Example 7
Figure imgf000181_0001
[0296] 4-(2-Fluoro-4-nitro-phenoxy -6-methoxy-7-(octahvdro-cvclopenta[c]pyrrol-5- ylmethoxy)-quinoline. To a round bottom flask equipped with a magnetic stir bar was added 5-[4-(2-Fluoro-4-nitro-phenoxy)-6-methoxy-quinolin-7-yloxymethyl]- hexahydrocyclopenta-[c]pyrrole-2-carboxylic acid benzyl ester (2.5 g, 4.1 mmol, l.Oeq), 33% HBr in acetic acid (5 ml) and acetic acid (5 ml). The mixture was stirred at room temperature for 1 hour and diluted with EtOAc to give a pale orange solid. The solid was filtered, washed with EtOAc and dried, giving 4-(2-Fluoro-4-nitro-phenoxy)-6-methoxy-7- (octahydro-cyclopenta[c]pyrrol-5-ylmethoxy)-quinoline (2.1 g, 95% yield). 1H NMR (400 MHz, CDC13): 8.83 (d, IH), 8.32 (m, 2H), 8.02 (s, IH), 7.76 (t, IH), 7.65 (s, IH), 6.89 (d, IH), 5.3 (d, 2H), 4.11 (m, 3H), 3.26 (m, 4H), 2.95 (m, 2H), 2.68 (m, 3H), 2.36 (m, 2H), 1.68 (m, 2H); MS (El) for C24H24FN3O5: 454 (M+H1").
Example 8
Figure imgf000182_0001
[0297] 4-(2-Fluoro-4-nitro-phenoxy)-6-methoxy-7-(2-methyl-octahvdro- cvclopentaFc1pyrrol-5-ylmethoxy)-quinorine. To a round bottom flask equipped with a magnetic stir bar was added 4-(2-Fluoro-4-nitro-phenoxy)-6-methoxy-7-(octahydro- cyclopenta[c]pyrrol-5-ylmethoxy)-quinoline (2.1 g, 3.9 mmol, 1.0 eq.) and acetonitrile/water 1:1 (5ml, 5ml). The reaction mixture was then cooled to 0° C and 37% solution of formaldehyde in water was added (0.2 g, 7.8 mmol, 2.0 eq). While keeping the temperature at 0°C Na(OAc)3BH was added (4.4g, 20.7 mmol, 3.0 eq). After 1 hour the pH was adjusted to 10 and the aqueous was extracted 2 x DCM (100 ml). Removal of the DCM resulted in a white solid. The compound was further purified with a biotage system using an eluent EtOAc and 5% MeOH, affording 4-(2-Fluoro-4-nitro-phenoxy)-6- methoxy-7-(2-methyl-octahydrocyclopenta-[c]pyrrol-5-ylmethoxy)-quinoline (0.9 g, 50% yield). ). 1H NMR (400 MHz, CDC13): 8.57 (d, IH), 8.14 (dd, IH), 8.12 (dd, IH), 7.41 (s, 2H), 7.34 (t, IH), 6.54 (d, IH), 4.19 (d, 2H), 4.01 (s, 3H), 2.61 (m, 4H), 2.43 (m, IH), 2.33 (s, 3H), 2.11 (m, 4H), 1.32 (m, 2H); MS (El) for C25H26FN3O5: 468 (M+H"').
Figure imgf000182_0002
[0298] 3-Fluoro-4-r6-methoxy-7-(2-methyl-octahvdro-cvclopentarclpyrrol-5-ylmethoxy)- quinolin-4-yloxyl-phenylamine. To a par hydrogenation reaction vessel was added 4-(2- fluoro-4-nitro-phenoxy)-6-methoxy-7-(2-methyl-octahydro-cyclopenta[c]pyrrol-5- ylmethoxy)-quinoline (0.800 g, 1.6 mmol, 1.0 eq.), DMF (50 ml), EtoAc (50ml), MeOH (50ml), TEA (5ml) and 10% Pd/C (200 mg). The vessel was placed on the par hydrogenator at 35 psi overnight. The Pd was filtered and the solvent removed to give 3- fluoro-4-[6-methoxy-7-(2-methyl-octahydro-cyclopenta[c]pyrrol-5-ylmethoxy)-quinolin- 4-yloxy]-phenylamine as an off yellow solid (0.78 g, 99% yield). 1H NMR (400 MHz, CDC13): 8.45 (d, IH), 7.57 (s, IH), 7.36 (s, IH), 7.05 (t, IH), 6.54 (m, 2H), 6.39 (d, IH), 4.16 (d, 2H), 4.01 (s, 3H), 3.81 (m, 3H), 2.61 (m, 3H), 2.41 (m, IH), 2.29 (s, 3H), 2.23 (m, 2H), 1.32 (m, 2H); MS (El) for C25H28FN3O3: 438 (M+H4).
Example 10
Figure imgf000183_0001
[0299] l-{3-Fluoro-4-[6-methoxy-7-(2-methyl-octahvdro-cvclopenta[c]pyrrol-5- ylmethoxy)-quinolin-4- ylox v] -phenyl } -3 -phen ylacetyl-thiourea. To a round bottom flask equipped with a magnetic stir bar was added 3-fluoro-4-[6-methoxy-7-(2-methyl- octahydro-cyclopenta[c]pyrrol-5-ylmethoxy)-quinolin-4-yloxy]-phenylamine (0.78 mg, 1.7 mmol, 1.0 eq.), toluene (10ml), ethanol (10ml) and phenyl-acetyl isothiocyanate (1.64 g, 9.2 mmol, 4.5 eq). The reaction mixture was stirred at room temperature overnight. After removal of the solvent the product was purified with a biotage system using an eluent EtOAc and 4% TEA (2L) then EtOAc, 4% TEA, 1% MeOH (IL). The solvent was removed to give l-{3-fluoro-4-[6-methoxy-7-(2-methyl-octahydro-cyclopenta[c]pyrrol-5- ylmethoxy)-quinolin-4-yloxy] -phenyl }-3-phenylacetyl-thiourea (0.5 g, 50% yield). 1H NMR (400 MHz, DMSO): 8.48 (d, IH), 7.92 (dd, IH), 7.53 (s, IH), 7.40 (m, 4H), 7.33 (d, 2H), 7.23 (m, 2H), 6.54 (d, 2H), 6.39 (d, IH), 4.21 (d, 2H), 4.02 (s, 3H), 3.81 (m, 3H), 2.87 (d, 2H), 2.73 (m, 4H), 2.53 (m, IH), 2.27 (m, 2H), 2.01 (s, 3H), 1.36 (m, 2H); MS (El) for C34H35FN4O4S: 615 (M+IT).
Example 11
Figure imgf000184_0001
[0300] 6-(6.7-Dimethoxy-quinolin-4-yloxy)-5-fluoro-benzothiazol-2-ylamine. 4-(6,7- dimethoxy-quinolin-4-yloxy)-3-fluoro-phenylamine (l.OOg, 3.18mmol) was dissolved in AcOH (8.0ml), to which was added NH SCN (486mg, 6.38mmol) and the mixture cooled in an ice bath. Br2 (0.33ml, 6.42mmol) in AcOH (0.33ml) was added dropwise with stirring. After addition was complete, the reaction mixture was stirred at room temperature. After one hour, more NH4SCN (l.Og, 13.1mmol) was added, followed by more Br2 (0.33ml, 6.42mmol) in AcOH (0.33ml), dropwise with stirring. The reaction mixture was then heated to reflux for several minutes. Upon cooling to room temperature, solids were filtered and washed with AcOH, followed by H2O. The volume of the filtrate was reduced in vacuo and the pH adjusted to pH 9-10 with 1.0N NaOH. The resulting solids were filtered, washed with H2O, and dried under high vacuum to give 6-(6,7- dimethoxy-quinolin-4-yloxy)-5-fluoro-benzothiazol-2-ylamine (568mg, 48%). 1H-NMR (400MHz, DMSO): 8.45 (d, IH), 7.82 (d, IH), 7.73 (br s, 2H), 7.53 (s, IH), 7.38 (m, 2H), 6.44 (d, IH), 3.94 (s, 6H). LC/MS Calcd for [M+H]+ 372.1, found 372.2
Example 12
Figure imgf000184_0002
[0301] N-[6-(6,7-Dimethoxy-quinolin-4-yloxy)-5-fluoro-benzothiazol-2-vn-2-phenyl- acetamide. 6-(6,7-dimethoxy-quinolin-4-yloxy)-5-fluoro-benzothiazol-2-ylarnine (95mg, 0.25mmol), Et3N (0.10ml, 0.72mmol), phenylacetyl chloride (0.044ml, 0.33mmol), and THF (1.0ml) were combined and stirred at room temperature for 1 hr. Additional phenylacetyl chloride (0.044ml, 0.33mmol) was added and the mixture heated to reflux for 1-2 hrs. After cooling to room temperature, the reaction mixture was diluted with 1:1 AcCN:H2O (1.0ml) and the resulting solids filtered, washed with 1:1 AcCN:H2O and dried under high vacuum to give N-[6-(6,7-dimethoxy-quinolin-4-yloxy)-5-fluoro-benzothiazol- 2-yl]-2-phenyl-acetamide (72mgs, 59%). 1H-NMR (400MHz, DMSO): 12.80 (s, IH), 8.54 (d, IH), 8.18 (d, IH), 7.91 (d, IH), 7.60 (s, IH), 7.45 (s, IH), 7.34 (m, 4H), 7.28 (m, IH), 6.60 (d, IH), 3.98 (s, 3H), 3.96 (s, 3H), 3.86 (s, 2H). LC/MS Calcd for [M+H]+ 490.1, found 490.0.
Example 13
Figure imgf000185_0001
[0302] 5-[4-(4-Amino-2-fluoro-phenoxy)-6-methoxy-quinazolin-7-yloxymethyl]- hexahydro-cyclopenta[c]pyrrole-2-carboxylic acid benzyl ester. 4-Amino-2-fluoro-phenol (1.53g, 12.0mmol) was dissolved in dry DMF (30ml) to which was added 60% NaH (774mg, 19.3mmol). After the mixture was stirred at room temperature for several minutes, a suspension of 5-(4-chloro-6-methoxy-quinazolin-7-yloxymethyl)-hexahydro- cyclopenta[c]pyrrole-2-carboxylic acid benzyl ester (4.70g, 6.7mmol) in dry DMF (40ml) was added. The reaction mixture was stirred at room temperature for 1-2 hrs, then diluted with EtOAc and washed with sat'd NaHCO3 (3x), H2O (lx), sat'd NaCl (lx), dried (Na2SO ), and concentrated in vacuo to give crude 5-[4-(4-amino-2-fluoro-phenoxy)-6- methoxy-quinazolin-7-yloxymethyl]-hexahydro-cyclopenta[c]pyrrole-2-carboxylic acid benzyl ester (5.6g, -100%) which was used in the next reaction without further purification. 1H-NMR (400MHz, DMSO): 8.50 (s, IH), 7.48 (s, IH), 7.34 (m, 5H), 7.28 (m, IH), 7.02 (t, IH), 6.48 (dd, IH), 6.40 (dd, IH), 5.40 (br s, 2H), 5.05 (s, 2H), 4.16 (d, 2H), 3.92 (s, 3H), 3.48 (m, 2H), 3.30 (m, 2H), 2.65 (m, 2H), 2.52 (m, IH), 2.10 (m, 2H), 1.30 (m, 2H). LC/MS Calcd for [M+H]+ 559.2, found 559.4.
Figure imgf000186_0001
[0303] 5-{4-[2-Fluoro-4-(3-phenylacetyl-thioureido)-phenoxy1-6-methoxy-quinazolin-7- yloxymethyll-hexahvdro-cvclopentarclpyrrole-2-carboxylic acid benzyl ester. Phenylacetyl chloride (2.65ml, 20.0mmol) and AgSCN (4.92g, 29.6mmol) were combined in dry toluene (50ml) and heated to reflux for 2 hrs. The reaction mixture was allowed to cool to room temperature, the solids were filtered through celite and the filtrate concentrated in vacuo. The resulting oil was combined with 5-[4-(4-amino-2-fluoro- phenoxy)-6-methoxy-quinazolin-7-yloxymethyl]-hexahydro-cyclopenta[c]pyrrole-2- carboxylic acid benzyl ester (5.6g, lOmmol) in 1:1 EtOH:toluene (100ml) and the mixture stirred at room temperature for 1-2 hrs. The reaction mixture was diluted with EtOAc and washed with sat'd NaHCO3 (3x), H2O (lx), sat'd NaCl (lx), dried (Na2SO4), and concentrated in vacuo. The resulting oil was purified by flash chromatography (3:1 EtOAc: hexanes) to give 5-{4-[2-fluoro-4-(3-phenylacetyl-thioureido)-phenoxy]-6- methoxy-quinazolin-7-yloxymethyl } -hexahydrocyclopenta[c]pyrrole-2-carboxylic acid benzyl ester (3.61g, 49%) as a dark brown foam. 1H-NMR (400MHz, DMSO): 12.44 (s, IH), 11.80 (s, IH), 8.54 (s, IH), 7.90 (m, IH), 7.53 (s, IH), 7.48 (m, 2H), 7.38 (s, IH), 7.34 (m, 7H), 7.28 (m, 3H), 5.05 (s, 2H), 4.16 (d, 2H), 3.94 (s, 3H), 3.72 (s, 2H), 3.48 (m, 2H), 3.30 (m, 2H), 2.65 (m, 2H), 2.52 (m, IH), 2.10 (m, 2H), 1.30 (m, 2H). LC/MS Calcd for [M+H]+ 736.2, found 736.0.
Figure imgf000186_0002
[0304] l-{3-Fluoro-4-[6-methoxy-7-(octahydro-cyclopenta[c]pyrrol-5-ylmethoxy)- quinazolin-4-yloxy] -phenyl} -3 -phenylacetyl-thiourea, dihydrobromide salt. 5-{4-[2- Fluoro-4-(3-phenylacetyl-thioureido)-phenoxy]-6-methoxy-quinazolin-7-yloxymethyl}- hexahydro-cyclopenta[c]pyrrole-2-carboxylic acid benzyl ester (3.3g, 4.5mmol) was dissolved in AcOH (70ml) to which was added 33%HBr in AcOH (12ml). The reaction mixture was stirred at room temperature for 1 hr, diluted with Et2O (1000ml) and the resulting solids filtered, washed with Et2O, and dried under high vacuum to give the l-{3- fluoro-4-[6-methoxy-7-(octahydro-cyclopenta[c]pyrrol-5-ylmethoxy)-quinazolin-4- yloxy] -phenyl} -3 -phenylacetyl-thiourea, dihydrobromide salt (3.4g, 100%). 1H-NMR (400MHz, DMSO): 12.42 (s, IH), 11.80 (s, IH), 8.84 (br s, 2H), 8.64 (s, IH), 7.92 (m, IH), 7.59 (s, IH), 7.49 (m, 2H), 7.41 (s, IH), 7.33 (m, 4H), 7.27 (m, IH), 4.17 (d, 2H), 3.95 (s, 3H), 3.73 (s, 2H), 3.17 (m, 2H), 3.10 (m, 2H), 2.83 (m, 2H), 2.45 (m, IH), 2.15 (m, 2H), 1.30 (m, 2H). LC/MS Calcd for [M+H]+ 602.2, found 602.1.
Example 16
Figure imgf000187_0001
[0305] l-{3-Fluoro-4-[6-methoxy-7-(2-methyl-octahydro-cyclopenta[c]pyrrol-5- ylmethoxy)-quinazolin-4-yloxy] -phenyl } -3-phenylacetyl-thiourea. 1 - { 3-Fluoro-4-[6- methoxy-7-(octahydro-cyclopenta[c]pyrrol-5-ylmethoxy)-quinazolin-4-yloxy]-phenyl}-3- phenylacetyl-thiourea, dihydrobromide salt (3.4g, 4.5mmol) was dissolved in in a combination of AcCN (100ml), H2O (30ml), and AcOH (2.45ml). Formaldehyde (37% in H2O, 855ml, 10.5mmol) was added and the mixture cooled in an ice bath. Na(OAC)3BH (2.99g, 14.1mmol) was added and the reaction mixture was stirred at 0 C for 1 hr, followed by stirring at room temperature for 2 hrs. The reaction mixture was neutralized with the addition of sat'd NaHCO3 and then concentrated in vacuo. The resulting aqueous mixture was extracted with CH2C12 (3x). The combined extractions were washed with sat'd NaHCO3 (lx), sat'd NaCl (lx), dried (Na2SO4), and concentrated in vacuo. The resulting residue was purified by flash chromatography (100% EtOAc, followed by 4% Et3N in EtOAc) to give the free base of l-{3-fluoro-4-[6-methoxy-7-(2-methyl-octahydro- cyclopenta[c]pyrrol-5-ylmethoxy)-quinazolin-4-yloxy]-phenyl}-3-phenylacetyl-thiourea (1.13g, 40%). The free base was converted to the HCl salt by dissolving the free base in a mixture of 1:1 AcCN:H2O containing 2-3 equivalents of 1 N HCl and lyophilizing to give the HCl salt of l-{3-fluoro-4-[6-methoxy-7-(2-methyl-octahydro-cyclopenta[c]pyrrol-5- ylmethoxy)-quinazolin-4-yloxy] -phenyl} -3 -phenylacetyl-thiourea as a white solid. 1H- NMR (400MHz, DMSO): 12.44 (s, IH), 11.83 (s, IH), 10.24 (br s, IH), 8.59 (s, IH), 7.93 (m, IH), 7.59 (s, IH), 7.50 (m, 2H), 7.42 (s, IH), 7.36 (m, 4H), 7.30 (m, IH), 4.20 (m, 2H), 3.95 (s, 3H), 3.73 (s, 2H), 3.39 (m, 2H), 3.06 (m, 2H), 2.95-2.77 (m, 5H), 2.35 (m, IH), 2.15 (m, 2H), 1.45 (m, 2H). LC/MS Calcd for [M+H]+ 616.2, found 616.2. Alternatively, the free base was converted to the acetate salt by dissolving the free base in a mixture of MeOH and CH2C12 to which was added 3 equivalents of acetic acid. The resulting mixture was concentrated in vacuo and the resulting residue lyophilized from 1:1 AcCN:H2O to give the acetate salt of l-{3-fluoro-4-[6-methoxy-7-(2-methyl-octahydro- cyclopenta[c]pyrrol-5-ylmethoxy)-quinazolin-4-yloxy]-phenyl}-3-phenylacetyl-thiourea as a white solid. 1H-NMR (400MHz, CDC13): d 12.45 (s, IH), 8.65 (s, IH), 7.98 (dd, IH), 7.50 (s, IH), 7.40 (m, 4H), 7.29 (m, 4H), 4.17 (d, 2H), 4.05 (s, 3H), 3.75 (s, 2H), 2.93 (m, 2H), 2.80 (m, 2H), 2.72 (m, 2H), 2.53 (s, 3H), 2.47 (m, IH), 2.25 (m, 2H), 2.02 (s, 3H), 1.35 (m, 2H). LC/MS Calcd for [M+H]+ 616.2, found 616.2.
Example 17
Figure imgf000188_0001
[0306] (6,7-Dimethoxy-quinazolin-4-yl)-(2-fluoro-4-nitro-phenyl)-amine. A mixture of 4- chloro-6,7-dimethoxy-quinazoline (548mg, 2.4mmol), 2-fluoro-4-nitro-phenylamine (392mg, 2.5mmol), AcCN (10ml), and conc'd HCl (0.050ml) was heated to reflux for several hrs. After the reaction mixture was allowed to cool to room temperature, the resulting solids were filtered, washed with AcCN and air-dried to give (6,7-dimethoxy- quinazolin-4-yl)-(2-fluoro-4-nitro-phenyl)-amine (673mgs, 80%). 1H-NMR (400MHz, DMSO): 12.18 (br s, IH), 8.91 (s, IH), 8.45 (s, IH), 8.36 (dd, IH), 8.24 (dd, IH), 7.91 (dd, IH), 7.44 (s, IH), 4.04 (s, 3H), 4.02 (s, 3H). LC/MS Calcd for [M+H]+ 345.1, found 345.4.
Example 18
Figure imgf000189_0001
[0307] N1-(6.7-Dimethoxy-quinazolin-4-yl)-2-fluoro-benzene-l,4-diamine. (6,7-
Dimethoxy-quinazolin-4-yl)-(2-fluoro-4-nitro-phenyl)-amine (673mg, 1.95mmol) was dissolved in a combination of DMF (20ml) and MeOH (20ml), to which was added 10% Pd/C (227mg). The mixture was shaken under an atmosphere of H2 on a Parr hydrogenator at 40psi for 3hrs. The reaction mixture was filtered through celite and the filtrate concentrated in vacuo. The resulting residue was triturated in EtOAc/Et2O. The resulting solids were filtered, washed with Et2O, and dried under vacuum to give N1-(6,7- dimethoxy-quinazolin-4-yl)-2-fluoro-benzene-l,4-cuamine (398mg, 65%) which was used in the next reaction without further purification. 1H-NMR (400MHz, DMSO): 10.80 (br s, IH), 10.30 (br s, IH), 8.63 (s, IH), 8.15 (s, IH), 7.33 (s, IH), 7.15 (m, IH), 6.45 (m, IH), 3.96 (s, 6H). LC/MS Calcd for [M+H]+ 315.1, found 315.4.
Example 19
CU
Figure imgf000189_0002
[0308] 1 - [4-(6 ,7-Dimethoxy-quinazolin-4-ylamino)-3 -fluoro-phenyl] -3 -phenylacetyl- thiourea. Phenylacetyl chloride (0.18ml, 1.4mmol) and AgSCN (338mg, 2.0mmol) were combined in dry toluene (5ml) and heated to reflux for 2 hrs. The reaction mixture was allowed to cool to room temperature, the solids were filtered through celite and the filtrate concentrated in vacuo. The resulting oil was combined with N1-(6,7-Dimethoxy- quinazolin-4-yl)-2-fluoro-benzene-l,4-diamine (398mg, 1.3mmol) in 1:1:2 EtOH:toluene:MeOH (30ml) and the mixture stirred at room temperature overnight. The resulting solids were filtered and washed with toluene, followed by hexanes. The solids were dissolved/suspended in a mixture of EtOAc/MeOH. Insoluble material was filtered and the filtrate concentrated in vacuo. The resulting solids were once again dissolved/suspended in a mixture of EtOAc/MeOH. In soluble material was filtered and the filtrate concentrated in vacuo to give l-[4-(6,7-dimethoxy-quinazolin-4-ylamino)-3- fluoro-phenyl]-3-phenylacetyl-thiourea (105mg, 17%). 1H-NMR (400MHz, DMSO): 12.53 (s, IH), 11.86 (s, IH), 11.44 (br s, IH), 8.81(s, IH), 8.25 (s, IH), 7.94 (dd, IH), 7.54 (m, 2H), 7.16 (m, 5H), 7.10 (m, IH), 4.02 (s, 6H), 3.84 (s, 2H). LC/MS Calcd for [M+H]+ 492.1, found 492.4.
Example 20
Figure imgf000190_0001
[0309] 6,7-Dimethoxy-4-(5-nitro-pyridin-2-yloxy)-quinoline. To a round bottom flask equipped with a magnetic stir bar was added 6,7-dimethoxy-lH-quinolin-4-one (1.8 g, 8.77 mmol, 1.0 eq.), anhydrous acetonitrile (90 mL) and Cs2CO3 (3.13 g, 9.65 mmole, 1.1 eq.). The reaction mixture was stirred at room temperature for 5 minutes. Then, 2-C1-5- nitropyridine (1.53 g, 9.65 mmol, 1.1 eq.) was added. The reaction mixture was stirred at room temperature for 16 hours. The solids were then filtered off and the filtrate was concentrated via rotary evaporation. The resulting material was taken up in EtOAc, and again the solids were filtered off. The EtOAc filtrate was concentrated. Purification was done on Biotage with solvent system EtOAc 100%. The collected pure fractions were concentrated and dried on high vacuum overnight to give 6,7-dimethoxy-4-(5-nitro- pyridin-2-yloxy)-quinoline as a yellow foam solid (0.902 g, 31.4% yield). 1H NMR (400 MHz, CDC13): 9.08 (d, IH), 8.74 (d, IH), 8.60 (dd, IH), 7.49 (s, IH), 7.26 (d, IH), 7.16 (s, IH), 7.07 (d, IH), 4.06 (s, 3H), 3.95 (s, 3H); MS (El) for Cι6H13N3O5: 328 (M+H1).
Example 21
Figure imgf000191_0001
[0310] 6-(6,7-Dimethoxy-quinolin-4-yloxy)-pyridin-3-ylamine. To a round bottom flask equipped with a magnetic stir bar was added 6,7-dimethoxy-4-(5-nitro-pyridin-2-yloxy)- quinoline (0.46 g, 1.41 mmol, 1.0 eq.), and THF (10 mL), MeOH (4 mL), DMF (2 mL), and TEA (2 mL). The 6,7-Dimethoxy-4-(5-nitro-pyridin-2-yloxy)-quinoline was dissolved completely in the above solution mixture, and was flushed with nitrogen for at least 5 minutes. The Pd/C (10% by weight) (0.090 g, 20% by weight) was then added. A balloon filled with H2 was connected to the flask after the nitrogen was vacuumed out. The reaction mixture was stirred at room temperature for 4 hours. The palladium was filtered out through Celite, and the filtrated was collected and concentrated via rotary evaporation. The resulting oil-like product was taken up into 5 mL of water and 1 mL of acetonitrile and lyophilized to yield 6-(6,7-dimethoxy-quinolin-4-yloxy)-pyridin-3-ylamine as a light brown solid (0.411 g, 98.1%). 1H NMR (400 MHz, CDC13): 8.54 (d, IH), 7.85 (d, IH), 7.53 (s, IH), 7.41 (s, IH), 7.18 (dd, IH), 6.96 (d, IH), 6.61 (d, IH), 4.05 (s, 3H), 4.03 (s, 3H), 3.73 (s, 2H); MS (El) for Cι65N3O3: 298 (M+H4).
Example 22
Figure imgf000191_0002
[0311] 1 - r6-(6,7-Dimethoxy-quinolin-4-yloxy)-ρ yridin-3 -yll -3 -phenylacetyl-thiourea. To a round bottom flask equipped with a magnetic stir bar was added 6-(6,7-dimethoxy- quinolin-4-yloxy)-pyridin-3-ylamine (85 mg, 0.0285 mmol, 1.0 eq.), and Phenyl-acetyl isothiocyanate (256 mg, 1.44 mmol, 5.0 eq.) dissolved in EtOAc/MeOH 50:50 (2 mL). The reaction mixture was sittred at room temperature for 12 hours, and the solvent was evaporated via rotary evaporation. Purification was done on Biotage with solvent system 95% EtOAc, 4% TEA and 1% MeOH. The combined pure fractions were concentrated and dried under vacuum overnight to yield l-[6-(6,7-dimethoxy-quinolin-4-yloxy)- pyridin-3-yl]-3-phenylacetyl-thiourea as a light yellow solid (40.4 mg, 29.7%). 1H NMR (400 MHz, CDC13): 8.65 (d, IH), 8.33 (d, IH), 8.27 (dd, IH), 7.35 (m, 7H), 7.15 (d, IH), 6.92 (d, IH), 4.05 (s, 3H), 3.99 (s, 3H), 3.76 (s, 2H); MS (El) for C25H22N4O4S: 475 (M+H4).
Example 23
Figure imgf000192_0001
[0312] N-r4-(6,7-Dimethoxy-quinolin-4-yloxy)-3-fluoro-phenvn-N'-phenethyl-oxalamide. To a solution of 4-(6,7-dimethoxy-quinolin-4-yloxy)-3-fluoro-phenylamine (263 mg, 0.83 mmol) and Et3N (0.223 ml, 1.67 mmol) in CH2C12 (10 mL) was added dropwise a solution of ethyl oxalyl chloride in CH2C12 (1 mL). The stirring was continued for 0.5 h at it. The reaction mixture was then washed with aqueous saturated NaHCO3 and dried over NaSO4. Removal of the solvent gave the crude oxamate, which was treated with neat phenethylamine (1.0 g, 8.3 mmol) at 80 °C for 3 h. Purification by flash column chromatography (hexanes :EtO Ac = 1:3) gave N-[4-(6,7-dimethoxy-quinolin-4-yloxy)-3- fluoro-phenyl]-N'-phenethyl-oxalamide (310 mg, 76%). 1H NMR (400 MHz, CDC13) δ 9.35 (br s, 1 H), 8.70 (d, J = 6.3 Hz, 1 H), 7.83 (dd, / = 11.9, 2.5 Hz, 1 H), 7.60-7.54 (m, 2 H), 7.43 (s, 1 H), 7.38-7.32 (m, 3 H), 7.30-7.20 (m, 4 H), 6.41 (d, J = 5.3 Hz, 1 H), 4.07 (s, 3 H), 4.05 (s, 3 H), 3.67 (dt, / = 7.0, 7.0 Hz, 2 H), 2.92 (t, J = 7.2 Hz, 2 H). LC-MS: 490 [M+H]+ Example 24
Figure imgf000193_0001
[0313] N-{3-Fluoro-4-[6-methoxy-7-(l-methyl-piperidin-4-ylmethoxy)-quinolin-4- yloxy1-phenyl}-N'-phenethyl-oxalamide. To a flask containing 7-benzyloxy-4-(2-fluoro-4- nitro-phenoxy)-6-methoxy-quinoline (850 mg, 2.0 mmol) was added 20 mL of 30% HBr in AcOH. The resulted solution was stirred for 4 h at rt; at this time, a large amount of precipitate formed. The crude product was filtered, washed with Et2O and dried in air, giving 4-(2-fluoro-4-nitro-phenoxy)-6-methoxy-7-hydroxyquinoline (609 mg, 92% yield).
[0314] To a solution of the 4-(2-fluoro-4-nitro-phenoxy)-6-methoxy-7-hydroxyquinoline (609 mg, 1.8 mmol) in DMF (9 mL) was added K2CO3 (1.24 g, 9.0 mmol) and N-Boc-4- piperidinemethanol mesylate (732 mg, 2.5 mmol). The mixture was then stirred at 80 °C for 2.5 h. After it was cooled to rt, the mixture was loaded directly to a Biotage column, and eluted with solvents (hexanes:EtOAc = 1:3). The resulting product, 4-[4-(2-fluoro-4- nitro-phenoxy)-6-methoxy-quinolin-7-yloxymethyl]-piperidine-l-carboxylic acid tert- butyl ester, was obtained as a solid (556 mg, 56%). ,
[0315] To a solution of 4-[4-(2-fluoro-4-nitro-phenoxy)-6-methoxy-quinolin-7- yloxymethyl] -piperidine- 1 -carboxylic acid tert-butyl ester (305 mg, 0.58 mmol) in CH2C12 (1 mL) was added 0.4 mL of TFA. The reaction mixture was stirred for 1.5 h and the solvents were removed under reduced pressure. The crude product was treated with NaBH(OAc)3 (381 mg, 1.80 mmol) and formaldehyde (0.5 mL, 37% in H2O). The stirring was continued for 12 h. The reaction was quenched with sat. aqueous NaHCO3. 15% NaOH was added until PH = 14. The product was extracted with EtOAc. Removal of the solvent in vacuo gave the crude product, 4-(2-fluoro-4-nitro-phenoxy)-6-methoxy-7-(l- methyl-piperidin-4-ylmethoxy)-quinoline, (240 mg, 93%), which was used directly in the next reaction.
[0316] To a solution of 4-(2-Fluoro-4-nitro-phenoxy)-6-methoxy-7-(l-methyl-piperidin-4- ylmethoxy)-quinoline (240 mg, 0.54 mmol) in EtOH (20 mL) was added 10% Pd/C (50 mg). The mixture was then hydrogenated on a Parr hydrogenator (40 psi) for 10 h. AcOH was added to dissolve the intermediate (mostly the hydroxylamine) and the hydrogenation was continued for additional 12 h. LC-MS was used to monitor the reaction progress. The solvents were removed under reduced pressure and the resulting crude product of 3- fluoro-4-[ι6-methoxy-7-(l-methyl-piperidin-4-ylmethoxy)-quinolin-4-yloxy]-phenylamine (about 220 mg) was used directly in the next reaction.
[0317] To a 0 °C solution of 3-fluoro-4-[6-methoxy-7-(l-methyl-piperidin-4-ylmethoxy)- quinolin-4-yloxy]-phenylamine (66 mg, 0.13 mmol) and Et3N (0.34 mL) in CH2C12 (6 mL) was added slowly ethyl oxalyl chloride (98 mg). The reaction mixture was stirred at rt for 30 min, then diluted with CH2C12 and washed with sat. aqueous NaHCO3. After dried over MgSO4 and concentrated, the crude ethyl oxamate was reacted with phenethylamine (80 mg, 0.64 mmol) at 80 °C for 2 h. Purification by HPLC gave product, N-{3-fluoro-4- [6-methoxy-7-(l-methyl-piperidin-4-ylmethoxy)-quinolin-4-yloxy]-phenyl}-N'-phenethyl- oxalamide (52 mg, 68% yield). 1H NMR (400 MHz) δ 9.38 (br s, 1 H), 8.48 (d, J = 5.2 Hz, 1 H), 7.83 (dd, J = 11.7, 2.6 Hz, 1 H), 7.59 (t, J = 6.2 Hz, 1 H), 7.55 (s, 1 H), 7.40-7.20 (8 H), 6.39 (d, J = 5.3 Hz, 1 H), 4.06 (d, J = 6.6 Hz, 2 H), 4.04 (s, 3 H), 3.67 (q, J = 6.8 Hz, 2 H), 2.98 (br d, J = 11.5 Hz, 2 H), 2.92 (t, J = 7.0 Hz, 2 H), 2.34 (s, 3 H), 2.10-1.80 (m, 5 H), 1.60-1.54 (m, 2 H).
Example 25
Figure imgf000194_0001
[0318] 1 -(4-Fluoro-phenylcarbamoyl)-cyclopropanecarboxyιic acid. The title compound was prepared based on a modified procedure of Shih and Rankin [Synthetic Communications, 1996, 26(4), 833-836]: To a mixture of cyclopropane- 1,1 -dicarboxylic acid (21.2 g, 0.163 mol, 1.0 eq.) in anhydrous THF (200 mL) under nitrogen was added dropwise triethylamine (16.49 g, 0.163 mol, 1.0 eq.) with stirring for 30 minutes at 0°C, followed by the addition of thionyl chloride (19.39 g, 0.163 mol, 1.0 eq.) with stirring for another 30 minutes at 0°C. To the resulting mixture under nitrogen was added dropwise a solution of 4-fluoroaniline (19.92 g, 0.179 mol, 1.1 eq.) in anhydrous THF (100 mL) with stirring for 1.5 hours at 0°C. The reaction mixture was diluted with ethyl acetate and washed with IN NaOH. The layers were separated, and the ethyl acetate layer was concentrated in vacuo to give a brownish solid. The brownish solid was washed with small amount of cold ethyl acetate, filtered and dried under vacuum to yield l-(4-fluoro- phenylcarbamoyl)-cyclopropanecarboxylic acid as a white solid (23.71 g, 65.18%). 1H NMR (400 MHz, CD3OD): 7.57-7.53 (m, 2H), 7.05-7.00 (m, 2H) 1.46-1.43 (m, 2H), 1.40- 1.37 (m, 2H).
Example 26
Figure imgf000195_0001
[0319] l-(4-Fluoro-phenylcarbamoyl)-cyclobutanecarboxylic acid. To a mixture of cyclobutane-l,l-dicarboxylic acid (10.0 g, 69.4 mmol, 1.0 eq.) in anhydrous THF (100 mL) under nitrogen was added dropwise triethylamine (7.02 g, 69.4 mmol, 1.0 eq.) with stirring for 30 minutes at 0°C, followed by the addition of thionyl chloride (8.25 g, 69.4 mmol, 1.0 eq.) with stirring for another 30 minutes at 0°C. To the resulting mixture under nitrogen was added dropwise a solution of 4-fluoroaniline (8.48 g, 76.3 mmol, 1.1 eq.) in anhydrous THF (50 mL) with stirring for 1.5 hours at 0°C. The reaction mixture was diluted with ethyl acetate and extracted with 2N NaOH. The aqueous phase was titrated with 2N HCl to pH 1-2 and then extracted with ethyl acetate. The organic phase was dried with sodium sulfate and concentrated in vacuo to give l-(4-fluoro-phenylcarbamoyl)- cyclobutanecarboxylic acid as a light pink solid (5.75 g, 34.9%). 1H NMR (400 MHz, CDC13 w/ldrop CD3OD): 7.53-7.48 (m, 2H), 7.06-7.00 (m, 2H), 2.81-2.63 (m, 4H), 2.14- 2.02 (m, 2H).
Example 27
Figure imgf000195_0002
[0320] 1 -Benzylcarbamoyl-cyclopropanecarboxylic acid. The title compound was prepared based on a modified procedure of Shih and Rankin [Synthetic Communications, 1996, 26(4), 833-836]: To a mixture of cyclopropane- 1,1-dicarboxylic acid (5.0 g, 38.4 mmol, 1.0 eq.) in anhydrous THF (50 mL) under nitrogen was added dropwise triethylamine (3.89 g, 38.4 mmol, 1.0 eq.) with stirring for 30 minutes at 0°C, followed by the addition of thionyl chloride (4.57 g, 38.4 mmol, 1.0 eq.) with stirring for another 30 minutes at 0°C. To the resulting mixture under nitrogen was added dropwise a solution of benzylamine 5 (4.53 g, 42.3 mmol, 1.1 eq.) in anhydrous THF (25 mL) with stirring for 1.5 hours at 0°C. The reaction mixture was diluted with ethyl acetate and extracted with 2N NaOH (to pH 10). The aqueous phase was titrated with 2N HCl to pH 1-2 and then extracted with ethyl acetate. The organic phase was dried with sodium sulfate and concentrated in vacuo to give 1-Benzylcarbamoyl-cyclopropanecarboxylic acid as a white solid (4.39 g, 52.15%). 1H NMR (400 MHz, CDC13): 8.44 (br s, IH), 7.37-7.33 (m, 2H), 7.32-7.26 (m, 3H), 1.82-1.70 (m, 4H).
Example 28
H0A o o κγ^Y
[0321] l-Phenylcarbamoyl-cyclopropanecarboxylic acid. To a mixture of cyclopropane- 1,1-dicarboxylic acid (5.29 g, 40.7 mmol, 1.0 eq.) in anhydrous THF (50 mL) under nitrogen was added dropwise triethylamine (4.12 g, 40.7 mmol, 1.0 eq.) with stirring for 30 minutes at 0°C, followed by the addition of thionyl chloride (4.84 g, 40.7 mmol, 1.0 eq.) with stirring for another 30 minutes at 0°C. To the resulting mixture under nitrogen was added dropwise a solution of phenylamine 9 (4.17 g, 44.8 mmol, 1.1 eq.) in anhydrous THF (25 mL) with stirring for 1.5 hours at 0°C. The reaction mixture was diluted with ethyl acetate and extracted with 2N NaOH (to pH >10). The aqueous phase was titrated with 2N HCl to pH 1-2 and then extracted with ethyl acetate. The organic phase was dried with sodium . sulfate and concentrated in vacuo to give 1-phenylcarbamoyl- cyclopropanecarboxylic acid as a white solid (5.08 g, 60.8%). 1H NMR (400 MHz, CDC13): 10.50 (br s, IH), 7.56-7.54 (m, 2H), 7.35-7.31 (m, 2H), 7.15-7.10 (m, IH), 1.94- 1.91 (m, 2H), 1.82-1.79 (m, 2H). Example 29
Figure imgf000197_0001
[0322] 7-Benzyloxy-4-chloro-6-methoxy-quinoline. Dry DMF (8.0ml, 103mmol) was dissolved in dry CHC13 (40ml) and cooled in an ice bath. Oxalyl chloride (9.0ml, 105mmol) in CH2C12 (10ml) was added dropwise with stirring at 0C. When the bubbling had ceased, this solution was added slowly to an ice-cold solution of 7-benzyloxy-6- methoxy-3H-quinazolin-4-one (lO.Og, 35.4mmol) in dry CHC13 (60ml) and the mixture was then heated to reflux for 2-3hrs. After cooling to room temperature, H O (100ml) was added and the phases were separated. The aqueous phase was further extracted with CHC13 (2x). The combined CHC13 extractions were washed with sat'd NaCl (lx), dried (Na2SO4) and concentrated in vacuo. The resulting residue was purified by flash chromatography (1:1 hexanes:EtOAc, followed by 100%EtOAc) to give 7-benzyloxy-4- chloro-6-methoxy-quinoline (5.1 lg, 48%). LC/MS Calcd for [M+H]+ 301.1, found 301.1.
Example 30
Figure imgf000197_0002
[0323] Cyclopropane- 1.1-dicarboxylic acid r3-fluoro-4-(7-hvdroxy-6-methoxy-quinolin- 4-yloxy)-phenyl1-amide(4-fluoro-phenyl)-amide. To a solution of cyclopropane- 1,1- dicarboxylic acid [4-(7-benzyloxy-6-methoxy-quinolin-4-yloxy)-3-fluoro-phenyl]-amide (4-fluoro-phenyl)-amide (1.18g, 2.0 mmol) in EtOH (20 mL) was added 1,4- cyclohexadiene (2.0 mL, 20 mmol) and 10% Pd/C (300 mg). The reaction mixture was then heated to reflux and the stirring was continued for 2 h. It was cooled to room temperature, filtered through celite and washed with MeOH. The MeOH solution was then concentrated under reduced pressure. The residue was taken into EtOAc (200 mL). The EtOAc solution was washed with water, and dried over Na2SO4. Removal of the solvent under reduced pressure gave 900 mg (89%) of the crude product (90% purity by analytical HPLC), which was used in the next reaction without further purification.
Example 31
Figure imgf000198_0001
[0324] N-(4-ir7-([2-(Diethylamino)ethylloxy}-6-(methyloxy)quinolin-4-yl1oχy}-3- fluorophenyl)-N'-(4-fluorophenyl)cvclopropane-l, 1-dicarboxamide. To a mixture of cyclopropane-l,l-dicarboxylic acid [3-fluoro-4-(7-hydroxy-6-methoxy-quinolin-4-yloxy)- phenyl]-amide(4-fluoro-phenyl)-amide (186 mg, 0.36 mmol) in CH2C12 (10 mL) was added 2-(diethylamino)ethanol (63 mg, 0.54 mmol), and PPh3 (141 mg, 0.54 mmol). DIAD (109 mg, 0.54 mmol) was then added as a CH2C12 (1 mL) solution. The resulted solution was stirred at room temperature for 2 h and the solvent was removed under reduced pressure. To the residue was added 1 N HCl (50 mL), and it was washed with EtOAc (50 mL x 2). The aqueous phase was basified by adding 15% NaOH aqueous solution until pH =11-13, and then extracted with ether (50 mL x 2). The combined organic layer was dried (MgSO4), and concentrated in vacuo. The residue was purified on preparative HPLC to give N-(4-{[7-{[2-(diethylamino)ethyl]oxy}-6-(methyloxy)quinolin- 4-yl]oxy}-3-fluorophenyl)-N'-(4-fluoro-phenyl)cyclopropane-l, 1-dicarboxamide (74 mg, 34%) as a pale yellow solid. 1H NMR (400 MHz, DMSO-- ) δ 10.40 (br s, 1 H), 10.02 (br s, 1 H), 8.47 (d, / = 5.2 Hz, 1 H), 7.91 (br d, J = 13.9 Hz, 1 H), 7.54-7.52 (m, 2 H), 7.55- 7.50 (m, 1 H), 7.52 (s, 1 H), 7.50-7.40 (m, 1 H), 7.41 (s, 1 H), 7.16 (br t, / = 8.7 Hz, 2 H), 6.41 (br d, J = 4.7 Hz, 1 H), 4.18 (t, J = 6.0 Hz, 2 H), 3.94 (s, 3 H), 2.87 (br t, J = 6.3 Hz, 2 H), 2.59 (q, J= 7.1 Hz, 4 H), 1.47 (br s, 4 H), 1.00 (t, /= 7.0 Hz, 6 H). Example 32
Figure imgf000199_0001
Fe, HC02NH4, Δ
Figure imgf000199_0002
[0325] l-(4-Benzyloxy-3-methoxyphenyl)ethanone. A solution of 4-hydroxy-3- methoxyacetophenone (40 g, 240 mmol), benzyl bromide (31.4 mL, 260 mmol) and potassium carbonate (99.6 g, 360 mmol) in DMF (800 mL) was heated to 40 °C overnight. The solution was cooled to room temperature, poured over ice and the resultant solid was filtered. This material was washed with water and dried to give l-(4-benzyloxy-3- methoxyphenyl)ethanone (61 g, 99 %).
[0326] l-(4-Benzyloxy-5-methoxy-2-nitrophenyl)ethanone. A stirred solution of l-(4- benzyloxy-3-methoxyphenyl)ethanone (51.3 g, 200 mmol) in dichloromethane (750 mL) was cooled to 0 °C. Nitric acid (90 %, 14 mL, 300 mmol) was added dropwise to the cooled solution over 20 min. Sulfuric acid (96.2 %, 16.3 mL, 300 mmol) was then added dropwise over 40 min at 0 °C. Additional nitric acid (9.4 mL, 200 mmol) was added dropwise over 20 min. The reaction mixture was washed with water (3 x 200 mL), and saturated sodium bicarbonate (4 x 200 mL, or until neutral). The organic layer was dried over Na2SO4 and concentrated. The crude mixture was recrystallized from DMF to give l-(4-benzyloxy-5-methoxy-2-nitroρhenyl)ethanone (36 g, 60 %). 1H NMR (400 MHz, CDC13): δ 7.65 (s, IH), 7.45-7.33 (m, 5H), 6.74 (s, IH), 5.21 (s, 2H), 3.97 (s, 3H), 2.49 (s, 3H).
[0327] l-(2-Amino-4-benzyloxy-5-methoxyphenyl)ethanone. A mixture of iron powder (27 g, 0.48 g atoms), ammonium formate (31 g, 500 mmol), l-(4-benzyloxy-5-methoxy-2- nitrophenyl)ethanone (36 g, 120 mmol), toluene (500 mL) and water (500 mL) was heated to reflux overnight. The mixture was filtered through celite and washed with ethyl acetate. The combined organic layers were washed with water and brine. The organic layer was dried over Na2SO and concentrated to afford l-(2-amino-4-benzyloxy-5- methoxyphenyl)ethanone (29.3 g, 90 %). 1H NMR (CDC13): δ 7.41-7.30 (m, 5H), 7.13 (s, IH), 6.16 (br s, 2H), 6.10 (s, IH), 5.13 (s, 2H), 3.83 (s, 3H), 2.51 (s, 3H). LC/MS (M+H =
272).
[0328] 7-Benzyloxy-6-methoxyquinorin-4-ol. Sodium ethoxide (74.8 g, 1.1 mol) was added to a solution of l-(2-amino-4-benzyloxy-5-methoxyphenyl)ethanone (29.3 g, 108 mmol) in DME (700 mL) and stirred for 30 min. Ethyl formate (44 mL, 540 mmol) was added and the mixture was stirred overnight (in case of incomplete reaction, additional sodium ethoxide can be added and the reaction monitored by LC/MS). After the reaction was complete, the mixture was diluted with water (40 mL) and acidified to neutral pH with 1M HCl. The solid was filtered, washed with water and dried to afford 7-benzyloxy- 6-methoxyquinolin-4-ol (22 g, 72%). 1H NMR (400 MHz, CDC13): δ 10.7 (br s, IH), 7.70 (s, IH), 7.49-7.46 (t, IH), 7.43-7.41 (br d, 2H), 7.37-7.33 (t, 2H), 7.30-7.28 (d, IH), 6.84 (s, IH), 6.21-6.19 (d, IH), 5.21 (s, 2H), 3.96 (s, 3H). LC/MS (M+H = 282).
[0329] 7-Benzyloxy-4-chloro-6-methoxyquinoline. Phosphoras oxychloride (300 mL) was added to 7-benzyloxy-6-methoxyquinolin-4-ol (40 g, 140 mmol) and the mixture heated to reflux for 2 h. The mixture was carefully poured into a mixture of ice and sodium carbonate. The solution was adjusted to pH 8 with the addition of solid sodium bicarbonate and stirred at room temperature overnight. The solid was filtered and washed with water and dried to give 7-benzyloxy-4-chloro-6-methoxyquinoline as a pale brown solid (40.2 g, 95%). 1H NMR (400 MHz, d6-OMSO): δ 8.61 (s, IH), 7.57-7.37 (m, 8H), 5.32 (s, 2H), 3.98 (s, 3H); 13C NMR (100 MHz, 6-DMSO): δ 152.4, 151.5, 148.5, 146.2, 139.6, 137.0, 129.2, 128.8, 121.7, 120.4, 110.1, 101.9, 70.8, 56.5; IR (cm-1): 2359, 2341, 1506, 1456, 1435, 1252, 1227, 1146, 999, 845, 752, 698, 667; LC/MS (M+H = 300).
Example 33
Figure imgf000200_0001
[0330] Trifluoromethanesulfonic acid 7-benzyloxy-6-methoxy-quinolin-4-yl ester. To a dry 2L RBF containing 7-benzyloxy-6-methoxyquinolin-4-ol (75.3 g, 267 mmol) was added DCM (1 L), 4-dimethylaminopyridine (3.28 g, 26.8 mmol) and 2,6-lutidine (62 mL, 534 mmol). The mixture was cooled to -20°C by controlled addition of dry ice to an acetone bath. Trifluoromethanesulfonyl chloride (37 mL, 350 mmol) was added dropwise to the cooled solution with magnetic stirring over 25 minutes. After addition was complete, the mixture was stirred in bath for 20 minutes, then at room temperature for 3 hours. LCMS indicated reaction completion. The reaction mixture was concentrated in vacuo and placed under high vacuum to remove residual 2,6-lutidine. To the resulting brown solids was added methanol (3.5 L). The resulting slurry was stirred with mechanical stirrer for 30 min before adding water (1.5 L). The solids were isolated by filtration, followed by a water wash. The resulting solid was dried under high vacuum overnight yielding trifluoromethanesulfonic acid 7-benzyloxy-6-methoxy-quinolin-4-yl ester as a light brown solid (92.2 g, 83.8%). 1H NMR (400MHz, DMSO, d6): δ 8.82 (d, IH), 7.67 (s, IH), 7.59 (d, IH), 7.54-7.52 (m, 2H), 7.46-7.42 (m, 2H), 7.39-7.36 (m, IH), 7.23 (s, IH), 5.35 (s, 2H), 3.97 (s, 3H). LC/MS: M+H = 414.
Example 34
Figure imgf000201_0001
[0331] Trifluoromethanesulfonic acid 6,7-dimethoxyquinolin-4-yl ester from 6,7- Dimethoxy-quinolin-4-ol . To a dry IL RBF containing 6,7-dimethoxy-quinolin-4-ol (20.9 g, 102 mmol), which can be prepared according to the procedure of Riegel, B. (7. Amer. Chem. Soc. 1946, 68, 1264), was added DCM (500 mL), 4-dimethylaminopyridine (1.24 g, 10 mmol) and 2,6-lutidine (24 mL, 204 mmol). The mixture was vigorously stirred at RT. Trifluoromethanesulfonyl chloride (14 mL, 132 mmol) was added dropwise to the solution. After addition was complete, the mixture was stirred ice bath for 2 to 3 hrs. On LC/MS indicating the reaction completion, the reaction mixture was concentrated in vacuo and placed under high vacuum to remove residual 2,6-lutidine. To the resulting brown solids was added methanol (250 mL). The resulting slurry was stirred for 30 min before adding water (1 L). The solids were isolated by filtration, followed by a water wash. The resulting solid was dried under high vacuum overnight yielding trifluoromethanesulfonic acid 6, 7-dimethoxy-quinolin-4-yl ester as a light brown solid (27 g, 80%). 1H NMR (400MHz, DMSO, d6): δ 8.82 (d, IH), 7.59 (m, 2H), 7.20 (s, IH), 3.97 (d, 6H). LC/MS: M+H = 338. Example 35
Figure imgf000202_0001
[0332] l-Benzyloxy-2-fluoro-4-nitrobenzene. A solution of 2-fluoro-4-nitrophenol (50.0 g, 318 mmol), benzyl bromide (42 mL, 350 mmol) and potassium carbonate (66.0 g, 478 mmol) in DMF (200 mL) was heated to 40 °C overnight. The solution was cooled to room temperature, poured over ice and the resultant solid was filtered. This material was washed with water and dried to give l-benzyloxy-2-fluoro-4-nitrobenzene (75.0 g, 95 %). 1H NMR (400 MHz, -DMSO): δ 8.19-8.11 (m, 2H), 7.53-7.37 (m, 6H), 5.36 (s, 2H); 13C NMR (100 MHz, -i6-DMSO): δ 152.8, 152.4, 149.9, 140.9, 136.1, 129.3, 129.1, 128.7, 122.0, 115.2, 112.8, 112.6, 71.6; IR (cm"1): 1499, 1346, 1279, 1211, 1142, 1072, 986, 885, 812, 789, 754, 742, 700, 648, 577.
[0333] 4-Benzyloxy-3-fluoroaniline. A mixture of iron powder (45.2 g, 0.809 g atoms), ammonium formate (53.6 g, 0.850 mol), l-benzyloxy-2-fluoro-4-nitrobenzene (50.0 g, 0.200 mol), toluene (400 mL) and water (400 mL) was heated to reflux overnight. The mixture was filtered through Celite and washed with hot ethyl acetate. The combined organic layers were washed with water and brine, then dried over sodium sulfate and concentrated to afford 4-benzyloxy-3-fluoroaniline (44 g, 100 %). 1H NMR (400 MHz, 6-DMSO): δ 7.43-7.26 (m, 5H), 6.90 (dd, IH), 6.49 (dd, IH), 6.34 (m, IH), 4.99 (br s, 2H), 4.98 (s, 2H); 13C NMR (100 MHz, 6-DMSO): δ 171.1, 155.1, 152.7, 144.9, 138.0, 137.2, 129.6, 129.0, 128.5, 118.9, 110.0, 102.9, 72.5; IR (cm-1): 1510, 1454, 1277, 1215, 1126, 1007, 957, 843, 800, 789, 739, 694, 604; LC/MS (M+H = 218).
[0334] Ethyl [(4-benzyloxy-3-fluorophenyl)amino1(oxo)acetate. Ethyl oxalyl chloride (44 mL, 390 mmol) was added to a solution of 4-benzyloxy-3-fluoroaniline (44 g, 180 mmol) in diisopropylethylamine (69 mL, 400 mmol) and stirred at room temperature for 15 min. The mixture was extracted with dichloromethane and washed with water and brine. The organic layer was dried over sodium sulfate and concentrated to afford ethyl [(4- benzyloxy-3-fluorophenyl)amino](oxo)acetate (58.4 g, 100 %). 1H NMR (400 MHz, d6- DMSO): δ 10.87 (s, IH), 7.73 (d, IH), 7.69 (d, IH), 7.53 (d, IH), 7.46-7.40 (m, 4H), 5.17 (s, 2H), 4.31 (q, 2H), 1.31(t, 3H); IR (cm-1): 1732, 1705, 1558, 1541, 1508, 1456, 1273, 1186, 1167, 1101, 999, 858, 741, 694; LC/MS (M+H = 318).
[0335] N-(4-Benzyloxy-3-flourophenyl)-N'-(2-phenylethyl)ethanedi amide. Phenethylamine (33 mL, 520 mmol) was added to ethyl [(4-benzyloxy-3- fluorophenyl)amino](oxo)acetate (81 g, 260 mmol) and the mixture was sonicated at room temperature for 30 min. The resulting solid was filtered, washed with water and dried to give N-(4-benzyloxy-3-flourophenyl)-N'-(2-phenylethyl)ethanediamide (100 g, 99 %). 1H ΝMR (400 MHz, d6-DMSO): δ 10.72 (br s, IH), 9.05 (m, IH), 8.78 (m, IH), 7.77 (m, IH), 7.59 (m, IH), 7.46-7.19 (m, 8H), 5.16 (m, 2H), 3.45 (m, 2H), 2.83 (m, 2H); IR (cm" x): 2980, 2883, 1653, 1522, 1506, 1441, 1385, 1221, 1122, 951, 808, 746, 696, 584; LC/MS (M+H = 393).
[0336] N-(3-Fluoro-4-hyckoxyphenyl)-N'-(2-phenylethyl)ethanediamide. A mixture of N- (4-benzyloxy-3-flourophenyl)-N,-(2-phenylethyl)ethanediamide (40 g, 100 mmol) and 38% hydrobromie acid in acetic acid (250 mL) was stirred at room temperature overnight. The resulting solid was filtered, washed with water and dried to give N-(3-fluoro-4- hydroxyphenyl)-N'-(2-phenylethyl)ethanediamide as a slightly yellow solid (30.6 g, 99 %yield). 1H ΝMR (400 MHz, 6-DMSO): δ 10.60 (s, IH), 9.02 (t, IH), 7.70 (d, IH), 7.47 (d, IH), 7.32-7.20 (m, 3H), 6.91 (t, IH), 3.43 (m, 2H), 2.81 (m, 2H); 13C ΝMR (100 MHz, -DMSO): δ 160.5, 158.8, 152.0, 149.6, 142.2, 139.8, 130.3, 129.3, 129.0, 126.8, 118.1, 117.4, 109.6, 109.3 IR (cm-1): 3279, 1653, 1518, 1456, 1279, 1190, 742, 696, 584; LC/MS (M+H = 303).
Example 36
Figure imgf000203_0001
[0337] l-Benzyloxy-2-fluoro-4-nitro-benzene. To a slurry of sodium hydride (60% dispersion is oil, 693 mmol, 27.7 g) and dimethylacetamide (600 ml) was added benzyl alcohol (462 mmol, 48 ml) dropwise with stirring under N2. The mixture was stirred for 1 hour at RT and then cooled to 0°C. 3,4-difluoronitrobenzene (508 mmol, 56.2 ml) was added to the cooled solution and stirred for 1 hour. Reaction mixture poured onto saturated ammonium chloride solution (800 ml) and stirred for 30 minutes, filtered and washed with water. The solid was stirred in ethyl acetate (500 mL), and filtered to give 54g of product. The ethyl acetate filtrate, after concentrated in vacuo, was triturated with diethyl ether (500 mL), sonicated for 2 hours, and filtered to give another 30g of product. The ether layer was concentrated and column purified using 5% EtOAc/hexanes as eluent to gave additional 15g of product. The total yield of l-benzyloxy-2-fluoro-4-nitro- benzene was 95g (83%). (Note: the product contains ca. 5% of 3,4-Bis-benzyloxy- nitrobenzene, which is carried into the next step without further purification.) 1H NMR (400MHz, CDC13): δ 8.04 -8.00 (m, 2H), 7.43-7.37 (m, 5H), 7.08 (t, IH), 5.26 (s, 2H).
[0338] 4-Benzyloxy-3-fluoro-phenylamine. A mixture of l-benzyloxy-2-fluoro-4-nitro- benzene (44g, 178 mmol), toluene (400 ml), ammonium formate (35 g), iron (30 g), and water (400 ml) was heated to reflux with stirring overnight. The reaction mixture was filtered through celite and washed with ethyl acetate (400ml). The organic layer was separated and washed with brine (300 ml), dried over sodium sulfate and concentrated to give 4-benzyloxy-3-fluoro-phenylamine as an oil (33.7 g, 87%). 1H NMR (400MHz, CDC13): δ 7.41-7.29 (m, 5H), 6.79 (t, IH), 6.45 (dd, IH), 6.14 (dd, IH), 5.02 (s, 2H), 3.50 (s, 2H). LC/MS: (M+1) 218.
[0339] Cyclopropane- 1,1-dicarboxylic acid (4-benzyloxy-3-fluoro-phenyl)-amide (4- fluoro-phenvD-amide. To a stirred mixture of 4-benzyloxy-3-fluoro-phenylamine (155.3 mmol, 33.7 g), l-(4-fluoro-phenylcarbamoyl)-cyclopropanecarboxylic acid (170.8 mmol, 38.13 g) and anhydrous dichloromethane (600 ml) was added EDCI (233.9 mmol, 44.7 g) in portions. After stirring at RT for 1 hr, the reaction mixture was diluted with saturated sodium bicarbonate (400 ml) and stirred for 30 minutes. The precipitate was filtered and air dried to give the 1st crop of product. The biphasic filtrate was separated, and the organic phase was washed with brine (300 ml), dried over sodium sulfate, and concentrated. The residue was taken up in DCM (100 ml), stirred for 15 minutes, and filtered to give a 2nd crop of product. The combined yield of cyclopropane- 1,1- dicarboxylic acid (4-benzyloxy-3-fluoro-phenyl)-amide (4-fluoro-phenyl)-amide was 64.5 g (98%). 1H NMR (400MHz, CDC13): δ 8.92 (br s, IH), 8.88 (br s, IH), 7.50-7.32 (m, 8H), 7.06-7.02 (m, 3H), 6.97-6.92 (t, IH), 5.13 (s, 2H), 1.65 (s, 4H). LC/MS: ( M+1) 423.
[0340] Cyclopropane- l.l-dicarboxylic acid (3-fluoro-4-hvdroxy-phenyl)-amide (4-fluoro- phenvD-amide. A mixture of cyclopropane- 1,1-dicarboxylic acid (4-benzyloxy-3-fluoro- ρhenyl)-amide (4-fluoro-phenyl)-amide (152.8 mmol, 64.5), ethanol (800 ml), cyclohexadiene (764 mmol, 71 ml), and 10% Pd/C (2 g) was refluxed for 2 hours. Reaction mixture cooled and filtered through celite and washed with methanol. The combined filtrate was concentrated and stirred in 10% EtOAc/ether (350 ml). The resulting precipitate was filtered and washed with ether to give a 1st crop of product. The filtrate was concentrated and stirred in DCM (150 ml) to give another precipitate, which was then filtered to give a 2nd crop of product. The combined yield of cyclopropane- 1,1- dicarboxylic acid (3-fluoro-4-hydroxy-phenyl)-amide (4-fluoro-phenyl)-amide was 43 g (85%) in 95% purity by HPLC (UV @ 254 nm). 1H NMR (400MHz, DMSO-D6): δ 10.07 (br s, IH), 9.92 (br s, IH), 9.64 (br s, IH), 7.64-7.60 (m, 2H), 7.55-7.51 (m, IH), 7.17-7.12 (m, 3H), 6.89-6.84 (t, IH), 1.43 (s, 4H). LC/MS: (M+1) 333.
Example 37
Figure imgf000205_0001
Figure imgf000205_0002
EtOH, reflux, 93%
Figure imgf000205_0003
[0341] Cyclopropane- l.l-dicarboxylic acid (4-benzyloxy-phenyl)-amide (4-fluoro- phenvD-amide. To a 0 °C suspension of 4-benzyloxyaniline hydrochloride (47.0 g, 200 mmol) and l-(4-fluoro-phenylcarbamoyl)-cyclopropanecarboxyιic acid (49.1 g, 220 mmol) in CH2C12 (400 mL) was added EDCI (38.2 g, 200 mmol). Stirring was continued at rt for 2-4 h until the reaction was complete. CH2C12 was removed under reduced pressure. H2O (300 mL) and MeOH (200 mL) were added, and the resulting mixture was stirred at rt for 30 min. After filtration and wash with H2O, the solid was transferred to another flask containing 300 mL of sat. aqueous NaHCO3 solution. The mixture was stirred for another 30 min. The solid was filtered, washed with water, and dried over night on a lyophilizer, affording cyclopropane- 1,1-dicarboxylic acid (4-benzyloxy-phenyl)- amide (4-fluoro-phenyl)-amide (75.8g, 95% yield) as an off-white solid.
[0342] Cyclopropane- 1,1 -dicarboxylic acid (4-fluoro-phenyl -amide (4-hvdroxy-phenyl)- amide. To a refluxing mixture of cyclopropane- 1,1 -dicarboxylic acid (4-benzyloxy- phenyl)-amide (4-fluoro-phenyl)-amide (46 g, 113 mmol), 10% Pd/C (2 g) in EtOH (400 mL) was added dropwise 1,4-cyclohexadiene (62.7 mL, 678 mmol). Stirring was continued for 2-5 h until the reaction was complete. The mixture was cooled to rt, filtered through celite, and washed with EtOH. The solution was then concentrated under reduced pressure. To the flask containing the crude product was added CHC13 (200 mL). The resulting suspension was stirred for 15 min at rt. The solid was filtered, and dried in the air to give cyclopropane- 1,1 -dicarboxylic acid (4-fluoro-phenyl)-amide (4-hydroxy- phenyl)-amide (34.4 g, 95%, yield).
Example 38
Figure imgf000206_0001
[0343] Alternate Synthesis of Cvclopropane-l,l-dicarboxylic acid (4-fluoro-phenyl)- amide (4-hvdroxy-phenyl)-amide. To a solution of 4-aminophenol (2.93 g, 26.9 mmol) and l-(4-fluoro-ρhenylcarbamoyl)-cyclopropanecarboxylic acid (5.00 g, 22.4 mmol) in DMA (30 mL) was added EDCI (5.15 g, 26.9 mmol). The mixture was stirred vigorously until the reaction was complete (~ 3 h). With vigorous stirring, the reaction mixture was then poured into a flask containing sat. aqueous NaHCO3 solution (200 mL). The stirring was continued for 1 h. The resulting suspension was then filtered. The solid was washed with water (50 mL), chloroform (50 mL) and dried under vacuum, affording l-(4-fluoro- phenylcarbamoyl)-cyclopropanecarboxylic acid (6.22g, 88% yield) as a powder (>95% purity by HPLC and 1H NMR). Example 39
Figure imgf000207_0001
[0344] N-{4-r(7-Benzyloxy-6-methoxyquinolin-4-yl)oxy1-3-fluorophenyl}-N'-(2- phenylethvDethanediamide. A mixture of 7-benzyloxy-4-chloro-6-methoxyquinoline (30 g, 100 mmol), N-(3-fluoro-4-hydroxyphenyl)-N'-(2-phenylethyl)ethanediamide (32 g, 106 mmol), DMAP (125 g, 1.02 mol) and bromobenzene (500 mL) was heated to reflux for 6 h. The mixture was cooled to room temperature and the bromobenzene was removed under reduced pressure. Methanol (500 mL) was added to the residue and the mixture was stirred at room temperature for 2 h. The resulting solid was filtered, washed with methanol and dried to give N-{4-[(7-benzyloxy-6-methoxyquinolin-4-yl)oxy]-3-fluorophenyl}-N'- (2-phenylethyl) ethanediamide (34 g, 61 %). 1H ΝMR (400 MHz, 6-DMSO): δ 11.05 (s, IH), 9.15 (s, IH), 8.47 (d, IH), 8.05 (d, IH), 7.84 (d, IH), 7.56-6.36 (m, 13H), 6.46 (d, IH), 5.32 (s, 2H), 3.97 (s, 3H), 3.47 (q, 2H), 2.86 (t, 2H); 13C ΝMR (100 MHz, d6- DMSO): δ 160.5, 160.2, 159.9, 159.5, 155.2, 152.7, 152.2, 150.3, 149.6, 146.9, 139.7, 137.4, 137.3, 137.2, 137.1, 129.3, 129.2, 129.1, 129.0, 128.9, 128.7, 128.6, 126.9, 124.8, 117.9, 115.3, 109.9, 102.8, 99.8, 70.6, 56.5, 41.3, 35.2; IR (cm"1): 1657, 1510, 1481, 1433, 1416, 1352, 1310, 1252, 1215, 1609, 986, 891, 868, 850, 742, 696; LC/MS (M+H = 566).
Example 40
Figure imgf000207_0002
[0345] N-l3-Fluoro-4-[(7-hvdroxy-6-methoxyquinolin-4-yl)oxy1phenyl|-N,-(2- phenylethvDethanedi amide. To a solution of Λ/-{4-[(7-benzyloxy-6-methoxyquinolin-4- yl)oxy]-3-fluorophenyl}-N'-(2-phenylethyl)ethanediamide (32 g, 56 mmol) in methanol (200 mL), DMF (100 mL), dichloromethane (100 mL), ethyl acetate (100 mL) and acetic acid (5 mL) was added palladium hydroxide (4.2 g) and the mixture was shaken on a Parr hydrogenator under a hydrogen pressure of 45 psi for 4 h. The resulting suspension was filtered through celite and the solid residue was washed with boiling dichloromethane (2 L) and acetone (2 L). The combined filtrates were evaporated to yield N-{3-fluoro-4-[(7- hydroxy-6-methoxyquinolin-4-yl)oxy]phenyl}-N'-(2-phenylethyl)ethanediamide as an off- white solid (25.6 g, 95 %). 1H ΝMR (400 MHz, dβ-DMSO): δ 11.06 (s, IH), 10.25 (br s, IH), 9.12 (t, IH), 8.40 (d, IH), 8.01 (dd, IH), 7.50-7.44 (m, 2H), 7.31-7.23 (m, 6H), 6.39 (d, IH), 3.95 (s, 3H), 2.85 (t, 2H), 2.50 (m, 2H); IR (cm-1): 1666, 1624, 1585, 1520, 1481, 1427, 1377, 1256, 1211, 1194, 1022, 880, 850, 839, 802, 750, 700; LC/MS (M+H = 476).
Figure imgf000208_0001
[0346] N-(3-Fluoro-4-{[6-methoxy-7-(3-morpholin-4-ylpropoxy)quinolin-4- yl] oxy }phen yl)-N '- (2-phenylethyl)ethanediamide. A solution of N-{3-fluoro-4-[(7- hydroxy-6-methoxyquinolin-4-yl)oxy]phenyl } -N'-(2-phenylethyl)ethanediamide (25.6 g, 54 mmol), N-(3-chloropropyl)morpholine hydrochloride (11.7 g, 592 mmol) and potassium carbonate (16.6 g, 120 mmol) in DMF (300 mL) was heated to 80 °C overnight. Upon cooling, a majority of the DMF (250 mL) was removed on a rotary evaporator, 5% aqueous LiCI (300 mL) was added and the mixture was sonicated at room temperature. The solid was filtered, suspended in IN HCl and washed with ethyl acetate (2 x 300 mL). The solution was adjusted to pH 14 using 2Ν sodium hydroxide and subsequently extracted with dichloromethane (3 x 200 mL). The organic layer was dried over sodium sulfate, filtered and evaporated to give N-(3-fluoro-4-{[6-methoxy-7-(3-morpholin-4- ylpropoxy)quinolin-4-yl]oxy}phenyl)-N'-(2-phenylethyl)ethanediamide as a yellow solid (24 g, 74 %). 1H ΝMR (400 MHz, CDC13): δ 9.37 (s, IH), 8.46 (d, IH), 7.81 (dd, IH), 7.57 (t, IH), 7.53 (s, IH), 7.42 (s, 2H), 7.34-7.20 (m, 6H), 6.39 (d, IH), 4.27 (t, 2H), 4.03 (s, 3H), 3.71 (m, 4H), 3.65 (q, 2H), 2.91 (t, 2H), 2.56 (br s, 4H), 2.13 (m, 2H); 13C ΝMR (100 MHz, - -DMSO): δ 160.1, 160.0, 159.5, 155.2, 152.7, 152.6, 150.2, 149.5, 147.1, 139.7, 137.3, 137.1, 129.3, 129.1, 126.9, 124.8, 117.9, 115.1, 109.2, 102.7, 99.6, 67.4, 66.9, 56.5, 55.5, 54.1, 41.3, 35.2, 26.4; IR (cm"1): 1655, 1506, 1483, 1431, 1350, 1302, 1248, 1221, 1176, 1119, 864, 843, 804, 741, 700; LC/MS (M+H = 603).
Example 42
Figure imgf000209_0001
2,6-lutidine, reflux
Figure imgf000209_0002
Figure imgf000209_0003
[0347] Cyclopropane- 1 , 1 -dicarboxylic acid [4-(7-benzyloxy-6-methoxy-quinolin-4- yloxy)-3 -fluoro-phenyl] -amide (4-fluoro-phenyl)-amide. To a flask containing cyclopropane-l,l-dicarboxylic acid (3-fluoro-4-hydroxy-phenyl)-amide (4-fluoro-phenyl)- amide (2.25 g, 6.7 mmol) and trifluoromethanesulfonic acid 7-benzyloxy-6-methoxy- quinolin-4-yl ester (1.87 g, 4.5 mmol) was added dry 2,6-lutidine (9 mL). The reaction mixture was heated to reflux (143°C) with vigorous stirring. The reaction progress was monitored by LC-MS. 2,6-Lutidine was removed under reduced ( pressure when the reaction was complete (about 6 h). The residue was treated with charcoal (1.5 g) in refluxing EtOAc (50 mL) for 15 min, and filtered through celite. The volume of the filtrate was reduced to about 20 mL and was added 20 mL of 1 N HCl. The crude product precipitated as the HCl salt, which was filtered and washed with EtOAc and H2O (88% purity by analytical HPLC). The HCl salt was free-based with saturated aqueous NaHCO3 solution. Further purification by column chromatography (hexans:EtOAc = 1:4) gave cyclopropane- 1 , 1 -dicarboxylic acid [4-(7-benzyloxy-6-methoxy-quinolin-4-yloxy)-3- fluoro-phenyl] -amide (4-fluoro-phenyl)-amide as an off-white solid (1.3 g, 48% yield. 1H NMR (400 MHz, DMSO, d6): 10.41 (s, IH), 10.02 (s, IH), 8.48 (d, IH), 7.92 (dd, IH), 7.65 (m, 2H), 7.54 (m, 5H), 7.41 (m, 4H), 7.17 (m, 2H), 6.43 (d, IH), 5.32 (s, 2H), 3.97 (s, 3H), 1.48 (m, 4H). LC/MS Calcd for [M+H]+ 596.2, found 596.3. Example 43
Figure imgf000210_0001
[0348] Cyclopropane- 1,1 -dicarboxylic acid r3-fluoro-4-(7-hvdroxy-6-methoxy-quinolin- 4-yloxy)-phenyl1 -amide (4-fluoro-phenyl)-amide. To a solution of the cyclopropane- 1,1- dicarboxylic acid [4-(7-benzyloxy-6-methoxy-quinolin-4-yloxy)-3-fluoro-phenyl]-amide (4-fluoro-ρhenyl)-amide (22.4 g, 37.6 mmol) in EtOH (340 mL) was added 1,4- cyclohexadiene (35 mL, 376 mmol) and 10% Pd/C (2.08 g). The reaction mixture was then heated at 65°C with stirring for 3 h (Caution: H2 gas is released from the reaction). It was then allowed to cool to room temperature, and filtered through celite follwed by a MeOH wash. The solution was then concentrated under reduced pressure. The yellow residue was taken into EtOAc (1 L). The EtOAc solution was washed with water (IX), brine (2X), dried over MgSO4 and concentrated in vacuo. Cyclopropane- 1,1-dicarboxylic acid [3-fluoro-4-(7-hydroxy-6-methoxy-quinolin-4-yloxy)-phenyl]-amide (4-fluoro- phenyl)-amide was obtained as a yellow solid (17.3 g, 91.1% yield), which were carried on to the next reaction without further purification. 1H NMR (400 MHz, DMSO, d6): 10.39 (s, IH), 10.15 (s, IH), 10.00 (s, IH), 8.38 (d, IH), 7.88 (dd, IH), 7.63 (m, 2H), 7.50 (m, 2H), 7.40 (t, IH), 7.27 (s, IH), 7.14 (m, 2H), 6.33 (d, IH), 3.95 (s, 3H), 1.47 (m, 4H). LC/MS Calcd for [M+H]+ 506.2, found 506.3. Anal. HPLC: 99.4% pure.
Figure imgf000210_0002
[0349] N-[3-fluoro-4-({6-(methyloxy)-7-[(3-morpholin-4-ylpropyl oxylquinolin-4- yl }oxy)phenvn-N-(4-fluorophenyl)cvclopropane- 1 , 1-dicarboxamide. To a mechanically stirred slurry of cyclopropane- 1,1 -dicarboxylic acid [3-fluoro-4-(7-hydroxy-6-methoxy- quinolin-4-yloxy)-phenyl]-amide (4-fluoro-phenyl)-amide (16.6 g, 32.8 mmol) and potassium carbonate (13.6 g, 98.6 mmol) in DMF (250 mL) was added 4-(3- chloropropyl)-morpholine hydrochloride (13, 7.92 g, 39.6 mmol). The resulting mixture was heated at 90°C for 5 hours (until phenol completely consumed). The reaction mixture was allowed to cool to room temperature, then dumped into water (900 mL), followed by extraction with EtOAc (3X). The combined extracts were washed with 5% LiCI (aq.) (3X) and brine (IX) followed by drying over MgSO4 and concentration in vacuo. The crude (18.8g) obtained as brown solid was further purified by flash chromatography [silica gel, 4-stage gradient system: 1) EtOAc; 2) EtOAc:MeOH:7N NH3/MeOH (95:5:0.5); 3) DCM:MeOH:7N NH3/MeOH (95:5:0.5); 4) DC MeOH: 7N NH3/MeOH (93:8:1)], affording N- [3 -fluoro-4-( { 6- (methyloxy)-7- [(3 -morpholin-4-ylpropyl)oxy] quinolin-4- yl}oxy)phenyl]-N'-(4-fluorophenyl)cyclopropane- 1,1 -dicarboxamide was obtained as an off white solid (15.0 g, 72% yield). IH NMR (400 MHz, DMSO-d6): 10.41 (s, IH), 10.02 (s, IH), 8.47 (d, IH), 7.91 (dd, IH), 7.65 (m, 2H), 7.53 (m, 2H), 7.42 (t, IH), 7.40 (s, IH), 7.16 (m, 2H), 6.42 (d, IH), 4.20 (t, 2H), 3.96 (s, 3H), 3.59 (t, 4H), 2.47 (t, 2H), 2.39 (br, s, 4H), 1.98 (m, 2H), 1.48 (m, 4H). LC/MS Calcd for [M+H]+ 633.3, found 633.0.
Example 45
Figure imgf000211_0001
[0350] Cyclopropane- 1 , 1 -dicarboxylic acid [4-(7-benzyloxy-6-methoxy-quinazolin-4- yloxy)-3 -fluoro-phenyl] -amide (4-fluoro-phenyl)-amide: A mixture of 7-benzyloxy-4- chloro-6-methoxy-quinazoline (5 g, 16.67 mmol), cyclopropane- 1,1 -dicarboxylic acid (3- fluoro-4-hydroxy-phenyl)-amide (4-fluoro-phenyl)-amide (8.3 g, 25 mmol), potassium carbonate (125 mmol, 17.25 g), and dimethylacetamide (125 ml) was heated 50° C with stirring for 16h. Reaction mixture was poured onto ice/water (600 ml) and stirred for 30 minutes, and filtered. The soϊid was dissolved in ethyl acetate and washed with water (lx), brine, and concentrated. The crude was purified on silica get column eluting with 30% acetone in hexanes to yield cyclopropane-l,l-dicarboxylic acid [4-(7-benzyloxy-6- methoxy-quinazolin-4-yloxy)-3-fluoro-phenyl]-amide (4-fluoro-phenyl)-amide (7.5 g, 76%). 1H NMR (CDC13): 8.64 (IH, br. s), 8.55 (IH, s), 8.33 (IH, br. s), 7.74-7.71 (IH, dd), 7.54 (IH, s), 7.48-7.33 (8H, m), 7.31-7.24 (2H, m), 7.06-7.02 (2H, m), 5.32 (2H, s), 4.06 (3H, s), 1.77-1.74 (2H, m), 1.63-1.61 (2H, m).
Example 46
Figure imgf000212_0001
[0351] Cyclopropane- 1 , 1 -dicarboxylic acid r3-fluoro-4-(7-hydroxy-6-methoxy- quinazolin-4-yloxy)-phenvn-amide (4-fluoro-phenyl)-amide. To a mixture of cyclopropane-l,l-dicarboxylic acid [4-(7-benzyloxy-6-methoxy-quinazolin-4-yloxy)-3- fluoro-phenyl] -amide (4-fluoro-phenyl)-amide (7.5 g, 12.6 mmol), acetic acid (few drops), dichloromethane (50 ml) and methanol (100 ml) was added 10% Pd/C (700 mg). The mixture was agitated in hydrogen gas (40 psi) until the reaction was complete (ca. 4 hr). The solution was filtered through celite and concentrated to give a crade product as a solid. The crade product was triturated with ether, and filtered. The filter cake was dried in vacuo to yield cyclopropane- 1,1 -dicarboxylic acid [3-fluoro-4-(7-hydroxy-6-methoxy- quinazolin-4-yloxy)-phenyl]-amide (4-fluoro-phenyl)-amide (6.1 g, 95% yield). 1H NMR (dmso-d6): 10.86 (IH, br. s), 10.34 (IH, br. s), 10.04 (IH, br. s), 8.46 (IH, s), 7.84-7.80 (IH, dd), 7.66-7.62 (2H, m), 7.55 (IH, s), 7.47-7.45 (IH, m), 7.41-7.37 (IH, m), 7.24 (IH, s), 7.18-7.13 (2H, t), 3.98 (3H, s), 1.46 (4H, s).
Figure imgf000212_0002
[0352] N-[3-Fluoro-4-(|6-(methyloxy)-7-[(3-morpholin-4-ylpropyl)oxy]quinazolin-4- yl}oxy phenyl1-N'-(4-fluorophenyl)cyclopropane-l, 1-dicarboxamide. To a mixture of cyclopropane- 1 , 1 -dicarboxylic acid [3-fluoro-4-(7-hydroxy-6-methoxy-quinazolin-4- yloxy)-phenyl] -amide (4-fluoro-phenyl)-amide (1.5 g, 2.96 mmol), 4-(3- hydroxypropyl)morpholine (0.623 mL, 4.5 mmol), triphenylphosphine (1.18 g, 4.5 mmol), and dichloromethane (50 mL) was added diisopropyl azodicarboxylate (0.886 mL, 4.5 mmol). The mixture was stirred at room temperature for 16 h, monitored by LCMS. After removal of solvent, the crude mixture was separated by flash column chromatography (silica), eluting with 5% methanol in dichloromethane to give N-[3-fluoro-4-({6- (methyloxy)-7-[(3-morpholin-4-ylpropyl)oxy]quinazolin-4-yl}oxy)phenyl]-N'-(4- fluorophenyl)cyclopropane-l,l-dicarbox-amide (890 mg, 47% yield). 1HNMR (400MHz, DMSO-d6): d 10.36 (br s, IH), 10.05 (br s, IH), 8.55 (s, IH), 7.83 (m, IH), 7.64 (m, 2H), 7.57 (s, IH), 7.44 (m, 3H), 7.18 (t, 2H), 4.27 (m, 2H), 3.99 (s, 3H), 3.61 (m, 6H), 2.40 (m, 4H), 2.01 (m, 2H), 1.47 (m, 4H). LC/MS Calcd for [M+H]+ 634.2, found 634.3.
Example 48
Figure imgf000213_0001
[0353] N-(4-{[6,7-bis(methyloxy)quinolin-4-yl]oxy}phenyl)-N'-(4-fluorophenyl)cyclo- propane- 1 , 1 -dicarboxamide. To a solution of cyclopropane- 1,1 -dicarboxylic acid (4- fluoro-phenyl)-amide (4-hydroxy-phenyl)-amide (6.98 g, 22.2 mmol) in anhydrous 2,6- lutidine (50 mL) was added trifluoromethanesulfonic acid 6, 7-dimethoxy-quinolin-4-yl ester (5 g, 14.8 mmol). The reaction mixture was heated at 165°C in a sealed pressure tube with stirring for 18 h. The reaction mixture was concentrated on high vacuum to completely remove lutidine. The resulting solid material was dissolved in DCM (250 mL), and washed several times with 1 N sodium hydroxide to remove the excess phenol. The crude mixture was loaded on a silica gel flash column and eluted with 75% EtOAc- hexanes, affording N-(4-{ [6,7-bis(methyloxy)quinolin-4-yl]oxy }phenyl)-N'-(4- fluorophenyl)cyclopropane-l, 1-dicarboxamide (3.2 g, 44%). 1H NMR (400 MHz, dg- DMSO): δ 10.2 (s, IH), 10.05 (s, IH), 8.4 (s, IH), 7.8 (m, 2H), 7.65 (m, 2H), 7.5 (s, IH), 7.35 (s, IH), 7.25 (m, 2H), 7.15(m, 2H), 6.4 (s, IH), 4.0 (d, 6H), 1.5 (s, 4H). LC/MS: M+H= 502.
Figure imgf000214_0001
[0354] 4,7-Dichloroquinoline. Phosphorus oxychloride (4mL, 429 mmol) was added to 7-chloro-4-hydroxyquinoline 2.86g, 15.9mmol) in a round bottom flask equipped with a reflux condenser. The mixture was heated to reflux for 2h, then allowed to cool to room temperature. The solution was concentrated in vacuo to a thick oil, then dumped over cracked ice. The resulting solution was neutralized with saturated NaHCO3 (aq). The slurry was filtered and washed with water. The solids were dried under vacuum, afforded 4,7-dichloroquinoline as a white solid (2.79g, 88.5% yield).
Example 50
Figure imgf000214_0002
[0355] 4-[4-(2-Fluoro-4- { [ 1 -(4-fluoro-phenylcarbamoyl)-cvclopropanecarbonyl1 -amino } - phenoxy)-6-methoxy-quinazolin-7-yloxymethyl]-piperidine-l-carboχylic acid tert-butyl ester. Cyclopropane- 1,1 -dicarboxylic acid [3-fluoro-4-(7-hydroxy-6-methoxy-quinazolin- 4-yloxy)-phenyl]-amide (4-fluoro-phenyl)-amide (325 mg, 0.64 mmol), 4- methanesulfonyloxymethyl-piperidine-1 -carboxylic acid tert-butyl ester (193 mg, 0.66 mmol), K2CO3 (181 mg, 1.31 mmol) were combined in DMF (5 ml) and heated to 80°C overnight. The reaction was not complete and more 4-methanesulfonyloxymethyl- piperidine-1 -carboxylic acid tert-butyl ester (90 mg, 0.31 mmol) and K2CO3 (90 mg, 0.65 mmol) were added and heating at 80°C continued for another night. The reaction mixture was allowed to cool to room temperature, then diluted with EtOAc and washed with H2O (3x), sat'd NaCl (lx), dried (Na2SO ) and concentrated in vacuo. The resulting crude material was purified by flash chromatography (1:1 hexanes :EtO Ac, followed by 1:3 hexanes:EtOAc) to give 4-[4-(2-fluoro-4-{[l-(4-fluoro-phenylcarbamoyl)- cyclopropanecarbonyl]-amino}-phenoxy)-6-methoxy-quinazolm-7-yloxymethyl]- piperidine-1 -carboxylic acid tert-butyl ester (273 mg, 60%). LC/MS Calcd for [M+H]+ 704.3, found 704.4.
Example 51
Figure imgf000215_0001
[0356] Cyclopropane- 1,1 -dicarboxylic acid {3-fluoro-4-[6-methoxy-7-(piperidin-4- ylmethoxy)-quinazolin-4-yloxy1 -phenyl] -amide (4-fluoro-phenyl)-amide, TFA salt. 4- [4- (2-Fluoro-4- { [ 1 -(4-fluoro-phenylcarbamoyl)-cyclopropanecarbonyl]-amino } -phenoxy)-6- methoxy-quinazolin-7-yloxymethyl] -piperidine- 1 -carboxylic acid tert-butyl ester (273 mg, 0.39 mmol) was dissolved in CH2C12 (8 ml) to which was added TFA (8 ml) and the mixture stirred at room temperature for lhr. The reaction mixture was concentrated in vacuo and the resulting oil triturated with Et2O. The resulting solids were filtered, washed with Et2O and dried under high vacuum to give cyclopropane- 1,1 -dicarboxylic acid {3- fluoro-4-[6-methoxy-7-(piperidin-4-ylmethoxy)-quinazolin-4-yloxy]-phenyl}-amide (4- fluoro-phenyl)-amide, TFA salt (222 mg, 80%). LC/MS Calcd for [M+H]+ 604.2, found 604.3.
Example 52
Figure imgf000215_0002
[0357] N-{3-Fluoro-4-r(6-(methyloxy)-7-{ [(l-methylpiperidin-4-yl)methyl1oχy}quin- azolin-4-yl)oxy1phenyl } -N'-(4-fluorophenyl)cvclopropane- 1 , 1 -dicarboxamide. Cyclopropane- 1,1 -dicarboxylic acid {3-fluoro-4-[6-methoxy-7-(piperidin-4-ylmethoxy)- quinazolin-4-yloxy]-phenyl}-amide (4-fluoro-phenyl)-amide, TFA salt (222 mg, 0.31 mmol), H2O (3 ml), 37% formaldehyde in H2O (0.18 ml) and acetic acid (27 drops) were combined in acetonitrile (9 ml) to which was slowly added triacetoxyborohydride (561 mg, 2.65 mmol). The mixture was stirred at room temperature for 1-2 hr, then diluted with IN NaOH and H2O and extracted with CH2C12 (3x). The combined CH2C12 extractions were washed with sat'd NaCl (lx), dried (Na2SO4) and concentrated in vacuo. The resulting residue was dissolved in a minimum of 1:1 dioxane:EtOAc to which was added 4M HCl in dioxane (1-2 ml). The resulting solids were filtered, washed with EtOAc and dried under high vacuum to give N-{3-fluoro-4-[(6-(methyloxy)-7-{[(l- methylpiperidin-4-yl)methyl]oxy}quinazolin-4-yl)oxy] phenyl } -N'-(4- fluorophenyl)cyclopropane- 1,1 -dicarboxamide, HCl salt (167 mg, 83%). lHNMR (400MHz, DMSO-d6): δ 10.40 (s, IH), 10.17 (br s, IH) 10.07 (s, IH), 8.61 (s, IH), 7.85 (m, IH), 7.65 (m, 2H), 7.48 (m, 2H), 7.42 (t, IH), 7.16 (t, 2H), 4.12 (2, 2H), 4.00 (s, 3H), 3.46 (m, 2H), 2.99 (m, 2H), 2.73 (d, 3H), 2.13 (m, IH), 2.01 (m, 2H), 1.63 (m, 2H), 1.47 (m, 4H). LC/MS Calcd for [M+H]+ 618.2, found 618.3.
Synthesis of Bridged Bicyclics
[0358] The following describes synthesis of bridged bicyclics with appended leaving groups for use as, for example, alkylating agents. In the context of this invention, these alkylating agents are used, for example, to alkylate the quinazoline or quinolines on the 6- or 7-oxygens to make compounds of the invention. The invention is not limited to alkylation chemistry to append such bridged bicyclics, but rather the aforementioned description is meant only to be illustrative of an aspect of the invention.
Example 53
[0359] l,4:3.6-dianhvdro-2-( -methyl-5-O-(methylsulfonyl)-D-glucitol: To a solution of l,4:3,6-dianhydro-2-0-methyl-D-glucitol (1.19g, 7.4 mmol) in dichloromethane was added pyridine (lmL, 12.36 mmol) followed by methanesulfonyl chloride (0.69mL, 8.92 mmol) and the mixture was allowed to stir at room temperature over 12 hours. The solvent was removed and the amorphous residue was partitioned with ethyl acetate and 0.1M aqueous hydrochloric acid. The aqueous phase was extracted once with additional ethyl acetate and the combined organic layers were washed with saturated aqueous sodium chloride then dried over anhydrous magnesium sulfate. Filtration and concentration followed by drying in vacuo afforded l,4:3,6-dianhydro-2-(9-methyl-5-( - (methylsulfonyl)-D-glucitol (1.67g, 94% yield) as a colorless oil. GC/MS calculated for C8H14SO6: 238 (M+).
Example 54
[0360] l,4:3,6-dianhvdro-5-Q-(phenylcarbonyl)-D-fructose ethylene glycol acetal: A solution of l,4:3,6-dianhydro-5-0-(phenylcarbonyl)-D-fructose (2.00g, 8.06 mmol), ethylene glycol (5.00g, 80.6 mmol), and p-toluenesulfonic acid (1.53g, 8.06 mmol) in benzene (lOOmL) was refluxed for 90 min using a Dean-Stark Trap apparatus. The reaction mixture was diluted with ethyl acetate (lOOmL), washed with saturated aqueous sodium bicarbonate (2 x 50mL) then brine (50mL), and dried over anhydrous sodium sulfate. Filtration, concentration and column chromatography on silica (1:1 hexane/ethyl acetate) provided 1.44g (61% yield) of l,4:3,6-dianhydro-5-0-(phenylcarbonyl)-D- fructose ethylene glycol acetal as a colorless solid. 1H NMR (400 MHz; CDC13): 8.08 (m, 2H), 7.58 (m, IH), 7.54 (m, 2H), 5.38 (dd, IH), 4.97 (t, IH), 4.21-4.02 (m, 7H), 3.86 (d, IH), 3.75 (d, IH).
Example 55
[0361] l,4:3,6-dianhvdro-D-fructose ethylene glycol acetal: To a solution of 1,4:3,6- dianhydro-5-0-(phenylcarbonyl)-D-fructose ethylene glycol acetal (1.44g, 4.93 mmol) in methanol (40mL) was added 50% aqueous sodium hydroxide (0.38 g, 4.75 mmol) and the mixture was stirred at room temperature for 30 minutes. Neutralization with 1M HCl, followed by concentration and column chromatography on silica (1:2 hexane/ethyl acetate) provided 0.74g (80% yield) of l,4:3,6-dianhydro-D-fractose ethylene glycol acetal as a colorless solid. 1H NMR (400 MHz; CDC13): 4.60 (t, IH), 4.32 (m, IH), 4.14 (d, IH), 4.05-3.98 (m, 5H), 3.82 (s, 2H), 3.62 (dd, IH), 2.65 (d, IH).
[0362] l,4:3,6-dianhvdro-5-0-(methylsulfonyl)-D-fructose ethylene glycol acetal: To a solution of l,4:3,6-dianhydro-D-fractose ethylene glycol acetal (0.74g, 3.93 mmol) and triethylamine (1.20g, 11.86 mmol) in dichloromethane (40mL) was added methanesulfonyl chloride (0.90g, 7.88 mmol) at 0°C under nitrogen. The solution was warmed to room temperature and stirred for 13 h. Dichloromethane (50mL) was added, and the organic layer was washed with saturated aqueous sodium bicarbonate (30mL), water (30mL), and brine (30mL) then dried over anhydrous sodium sulfate. Filtration and concentration provided 1.02g (97%) of l,4:3,6-dianhydro-5-0-(methylsulfonyl)-D- fractose ethylene glycol acetal as a yellow oil. 1H NMR (400 MHz; CDC13): 5.08 (m, IH), 4.82 (t, IH), 4.13 (dd, IH), 4.04 (m, 4H), 3.93 (dd, IH), 3.87 (d, IH), 3.81 (d, IH), 3.13 (s, 3H).
Example 56
[0363] l,4:3,6-dianhydro-2-deoxy-2-methylidene-D- rαbmo-hexitol: To a solution of l,4:3,6-dianhydro-2-deoxy-2-methylidene-5-0-(phenylcarbonyl)-D-αrflbϊ>ιo-hexitol (329mg, 1.34 mmol) in methanol (lOmL) was added 50% aqueous sodium hydroxide (95mg, 1.19 mmol) and the mixture was stirred at room temperature for 30 minutes. Neutralization with 4M hydrogen chloride in 1,4-dioxane followed by concentration and column chromatography on silica (1:1 hexane/ethyl acetate) provided 141mg (74%) of l,4:3,6-dianhydro-2-deoxy-2-methylidene-D- rΩ& o-hexitol as a colorless solid. 1H NMR (400 MHz; CDC13): 5.37 (m, IH), 5.20 (m, IH), 4.80 (m, IH), 4.54 (m, 2H), 4.43 (m, IH), 4.26 (m, IH), 3.95 (dd, IH), 3.54 (dd, IH), 2.70 (d, IH).
[0364] l,4:3,6-dianhvdro-2-deoxy-2-methylidene-5-0-(methylsulfonyl -D-flr b o- hexitol: To a solution of l,4:3,6-dianhydro-2-deoxy-2-methylidene-D- r b o-hexitol (135mg, 0.95 mmol) and triethylamine (288mg, 2.85 mmol) in dichloromethane (lOmL) was added methanesulfonyl chloride (222mg, 1.94 mmol) at 0°C under nitrogen. The solution was warmed to room temperature and stirred for 18 h. Dichloromethane (50mL) was added and the organic layer was washed with saturated aqueous sodium bicarbonate (2 x 25mL), water (25mL) and brine (25mL) then dried over anhydrous sodium sulfate. Filtration and concentration provided 213mg (72%) of l,4:3,6-dianhydro-2-deoxy-2- methylidene-5-0-(methylsulfonyl)-D-αra-j o-hexitol as a yellow oil. 1H NMR (400 MHz; CDC13): 5.40 (m, IH), 5.23 (m, IH), 5.04 (m, IH), 4.85 (m, IH), 4.73 (t, IH), 4.58 (m, IH), 4.41 (m, IH), 4.08 (dd, IH), 3.86 (dd, IH), 3.14 (s, 3H). Example 57
[0365] l,4:3,6-dianhvdro-2-deoxy-5-0-(phenylcarbonyl)-L-arøb o-hex-l-enitol: To a mixture of l,4:3,6-dianhydro-5-O-(phenylcarbonyl)-(D)-glycitol (4.32g, 17.3 mmol), triethylamine (4.91 mL, 35.3 mmol) and 4-dimethylaminopyridine (0.63g, 5.2 mmol) in dichloromethane (50 mL) at -10 ° to -15° was added trifluromethanesulfonic anhydride (3.48mL, 20.7 mmol) dropwise over ten minutes and the resulting mixture was stirred at this temperature for 3 hours. The mixture was poured into 100 mL of ice- water and extracted with dichloromethane (3 x 50 mL). The combined organic layers were washed with brine, dried over anhydrous sodium sulfate, filtered then concentrated. The crade triflate was suspended in toluene (50 mL) followed by addition of 1,8- diazabicyclo[4,5,0]undec-7-ene (5.25 mL, 34.6 mmol) and the mixture was stirred at room temperature for 18 hours. The reaction mixture was poured into ice-water and partitioned then the aqueous portion was extracted with dichloromethane (3 x 50 mL). The combined organic portion was washed with brine, dried over anhydrous sodium sulfate, filtered and concentrated. The residue was purified by flashed chromatography (silica gel, 5-20% ethyl acetate-hexane) to give l,4:3,6-dianhydro-2-deoxy-5-( -(phenylcarbonyl)-L- αrab o-hex-1-enitol, as a white solid, 3.10g, 77% yield. 1H NMR (400MHz; CDC13): 8.08-8.06 (m, 2H), 7.61-7.57 (m, IH), 7.56-7.43 (m, 2H), 6.62-6.61 (d, IH), 5.48-5.46 (m,lH), 5.32-5.26 (m,lH), 5.13-5.10 (m, 2H), 4.18-4.14 (tr,lH), 3.61-3.56 (tr, IH).
Example 58
[0366] Methyl 3,6-anhvdro-5-Q-(phenylcarbonyl)-β-L-glucofuranoside: To a solution of l,4:3,6-dianhydro-2-deoxy-5-0-(phenylcarbonyl)-L-arabino-hex-l-enitol (l.OOg, 4.3 mmol) in methanol (17 mL) at -4°C was added 3-chloroperoxybenzoic acid (85%, 1.35g, 8.6 mmol), and the resulting mixture was slowly warmed to room temperature and stirred for 18 hours. The reaction mixture was concentrated, diluted with dichloromethane (50 mL), washed with saturated aqueous sodium bicarbonate solution, dried over sodium sulfate, filtered and concentrated. The residue was purified by flash chromatography (silica gel, 25-60% ethyl acetate-hexane) to give methyl 3,6-anhydro-5-(9- (phenylcarbonyl)-β-L-glucofuranoside as a white solid, 1.03g, 83% yield. 1H NMR (400MHz; CDC13): 8.11-8.08 (d, 2H), 7.61-7.56 (tr, IH), 7.48-7.44 (m, 2H), 5.24-5.17 (m, 2H), 4.96 (s, IH), 4.57-4.56 (d, IH), 4.27 (s, IH), 4.22-4.18 (dd, IH), 4.08-4.04 (dd, IH) 3.36 (s, 3H).
[0367] Methyl 3,6-anhvdro-2-0-methyl-5-0-(phenylcarbonyl)-β-L-glucofuranoside: A mixture of methyl 3,6-anhydro-5-0-(phenylcarbonyl)-β-L-glucofuranoside (1.03g, 3.7 mmol), silver (I) oxide (0.85g, 3.7 mmol) and methyl iodide (0.34 mL, 5.5 mmol) in DMF (2 mL) was heated at 60°C for 1 hour. After cooling to room temperature the reaction mixture was diluted with ethyl acetate (50 mL), filtered over celite, adsorbed on silica gel (lOg) and purified by flash chromatography (silica gel, 5-30% ethyl acetate-hexane) to give methyl 3,6-anhydro-2-0-methyl-5-0-(phenylcarbonyl)-β-L-glucofuranoside as a colorless oil, 0.82g, 76% yield. 1H NMR (400MHz; CDC13): 8.11-8.09 (d, 2H), 7.60-7.56 (m, IH), 7.46-7.44 (m, 2H), 5.24-5.20 (m, IH), 5.18-5.09 (tr, IH), 4.99 (s, IH), 4.61-4.60 (d, IH), 4.21-4.17 (tr, IH), 4.08-4.03 (tr, IH), 3.81 (s, IH), 3.40 (s, 3H), 3.57 (s, 3H).
[0368] Methyl 3 ,6-anhvdro-2-Q-methyl- -D-idofuranoside: A solution of methyl 3,6- anhydro-2-0-methyl-5-0-(phenylcarbonyl)-β-L-glucofuranoside (820mg, 3.1mmol) and 50% sodium hydroxide (248 mg, 3.1 mmol) in methanol (lOmL) was stirred at room temperature for 30 minutes. The material was adsorbed on silica gel (5g) and passed through a short column (15% ethyl acetate in hexanes to 5% methanol in ethyl acetate) to give methyl 3,6-anhydro-2-0-methyl- -D-idofuranoside as a colorless oil, 420 mg, 85% yield. 1H NMR (400MHz; CDC13): 5.04 (s, IH), 5.84-5.81 (tr, IH), 4.44-4.42 (tr, IH), 4.25-4.19 (m, IH), 3.85-3.75 (m, IH), 3.49 (s, 3H), 3.43 (s, 3H), 2.75-2.72 (d, IH).
[0369] Methyl 3,6-anhydro-2-Q-methyl-5-( -(methylsulfonyl)-β-L-glucofuranoside:
Methyl 3,6-anhydro-2-(9-methyl-α-D-idofuranoside (420 mg, 2.6 mmol) was dissolved in dichloromethane (10 mL) and pyridine (0.36 mL, 3.7 mmol) at 0°C. Methanesulfonyl chloride (0.14 mL, 3.1 mmol) was added and the resulting mixture was stirred at 0°C for 1 hour then at room temperature for 2 hours. The reaction mixture was washed with water and saturated aqueous sodium bicarbonate solution, dried over anhydrous sodium sulfate, filtered and concentrated to give methyl 3,6-anhydro-2-O-methyl-5-( -(methylsulfonyl)-β- L-glucofuranoside as a colorless oil, 669mg, 95% yield, which was used without further purification. Example 59
[0370] 3,6-anhvdro-5-0-(phenylcarbonyl)-g-L-glucofuranose: A mixture of osmium tetroxide (4% in water, 0.25 mL, 0.03 mmol) and N-methylmorpholine (505 mg, 4.3 mmol) in 3 mL of 50% acetone in water was warmed to 60°C. A solution of 1,4:3,6- dianhydro-2-deoxy-5-O-(phenylcarbonyl)-L-αrab ø-hex-l-enitol (2.00g, 8.6 mmol) in 6 mL of 50% acetone in water was added over 3 hours. During this time an additional amount of N-methylmorpholine (l.Olg, 8.6 mmol) was added in small portions periodically. Upon completion of the addition process the reaction was stirred for another hour and cooled to room temperature. The crade mixture was applied to a column of silica gel and flashed (0-6% methanol in 1:1 ethyl acetate:hexane) to give 3,6-anhydro-5-0- (phenylcarbonyl)- -L-glucofuranose as a white solid, 1.5g, 65% yield. 1H NMR (400MHz; DMSO-dβ): 8.01-7.95, (m, 2H), 7.68-7.66 (m, IH), 7.57-7.53 (m, 2H), 5.18- 5.11 (m, 2H), 4.85-4.81 (m, IH, m), 4.37-4.35 (m, IH), 4.05-3.96 (m, 2H), 3.85-3.83 (m, IH).
[0371] 3,6-anhydro-2-0-methyl-5-0-(phenylcarbonyl)-α-L-glucofuranoside: 3,6-
Anhydro-5-0-(phenylcarbonyl)-α-L-glucofuranose (576 mg, 2.2 mmol) was added to a mixture of sodium hydride (60% oil dispersion, 346 mg, 8.7 mmol) and methyl iodide (0.54mL, 8.7 mmol) in 5 mL of DMF at 0°C and the resulting mixture was stirred for 1 hour. The reaction mixture was diluted with ethyl acetate and quenched with water (5 mL). The aqueous portion was extracted with ethyl acetate (3 x 5 mL). The combined organic portion was washed with brine, dried over anhydrous sodium sulfate, filtered, and concentrated. The residue was purified by flashed chromatography (silica gel, 5-20% ethyl acetate in hexane) to give 3,6-anhydro-2- -methyl-5-0-(phenylcarbonyl)-α-L- glucofuranoside as a white solid, 270 mg, 42% yield. 1H NMR (400MHz; CDC13): 8.09- 8.07 (m, 2H), 7.6,1-7.57 (m, IH), 7.48-7.27 (m, 2H), 5.25-5.22 (m, IH), 5.07-5.06 (d, IH), 4.94-4.91 (m, IH), 4.73-4.71 (m, IH), 4.20-4.16 (m, IH), 3.96-3.94 (m, IH), 3.85-3.83 (tr, IH), 3.50 (s, 3H), 3.42 (s, 3H).
[0372] Methyl 3,6-anhvdro-2- -methyl-5-( -(methylsulfonyl)- -L-glucofuranoside: A solution of methyl 3,6-anhydro-2-( -methyl-5-( -(phenylcarbonyl)- -L-glucofuranoside (230mg, 0.92 mmol) and 50% sodium hydroxide (74 mg, 0.92 mmol) in methanol (5 mL) was stirred at room temperature for 30 minutes. The mixture was adsorbed on silica gel (2g) and passed through a short column (15% ethyl acetate in hexanes to 5% methanol in ethyl acetate) to afford a colorless oil which was employed directly in the next step, 140 mg, 0.72 mmol, 95% yield. The alcohol was dissolved in dichloromethane (5 mL) and pyridine (121 μL, 1.03 mmol) was added at 0°C. Methanesulfonyl chloride (27μL, 0.88 mmol) was added and the resulting mixture was stirred at 0°C for 1 hour then at room temperature for 2 hours. The reaction mixture was washed with water and saturated aqueous sodium bicarbonate solution, dried over sodium sulfate, filtered and concentrated to give methyl 3,6-anhydro-2-0-methyl-5-(9-(methylsulfonyl)-α-L-glucofuranoside as a colorless oil, 190 mg, 96% yield.
Example 60
[0373] 3,6-Anhydro-l,2- -(l-methylethylidene)-5-0-(phenylcarbonyl)-α-L-gluco- furanose: A mixture of 3,6-anhydro-5-0-(phenylcarbonyl)-α-L-glucofuranose (l.OOg), 2,2-dimethoxy propane (0.63 mL), p-toluenesulfonic acid (20 mg) and benzene (10 mL) was heated at reflux for 3 hours. The reaction mixture was cooled then adsorbed on silica gel (lOg) and purified by flash chromatography (silica gel, 5-35% ethyl acetate in hexanes) to give 3,6-anhydro-l,2-( -(l-methylethylidene)-5-0-(phenylcarbonyl)- -L- glucofuranose as colorless oil, 0.85g, 74% yield. 1H NMR (400MHz; CDC13): 8.08-8.06 (d, 2H), 7.59-7.56 (tr, IH), 7.46-7.42 (m, 2H), 5.99-5.98 (d, IH), 5.35-5.31 (tr, IH), 5.10- 5.08 (d, IH), 4.66-4.65 (d, IH), 4.61-4.60 (d, IH), 4.20-4.16 (dd, IH), 3.91-3.74 (tr, IH,), 1.50 (s, 3H), 1.34 (s, 3H).
[0374] 3,6-Anhvdro-l,2-0-(l-methylethylidene)-5-O-(methylsulfonyl)- -L-gluco- furanose: A solution of 3,6-anhydro-l,2-0-(l-methylethylidene)-5-0-(phenylcarbonyl)-α- L-glucofuranose (850mg) and 50% sodium hydroxide (111 mg) in methanol (lOmL) was stirred at room temperature for 30 minutes. The mixture was then adsorbed on silica gel (5g) and passed through a short column (15% ethyl acetate in hexanes to 5% methanol in ethyl acetate) and the alcohol intermediate, 390 mg, 70% yield, was used immediately in the next step. The alcohol was dissolved in dichloromethane (10 mL) and pyridine (0.32 mL) at 0°C. Methanesulfonyl chloride (0.12 mL) was added and the resulting mixture was stirred at 0°C for 1 hour then at room temperature for 2 hours. The reaction mixture was washed with water and saturated aqueous sodium bicarbonate solution, dried over anhydrous sodium sulfate, filtered and concentrated to give 3,6-anhydro-l,2-0-(l- methylethylidene)-5-0-(methylsulfonyl)-α-L-glucofuranose as a colorless oil, 485 mg, 90% yield, which was immediately employed in the next step. Example 61
[0375] (3S.8aSV3-(Chloromethvnhexahvdro-lH-pyrrolor2,l-ciri.41oxazine: (S)-(+)-
Prolinol (6.00 g, 59.3 mmol) was added to epichlorohydrin (47 mL, 600 mmol) at 0°C. The solution was stirred at 40°C for 0.5 h and then concentrated in vacuo. The residual oil was cooled in an ice bath and concentrated sulfuric acid (18 mL) was added dropwise with stirring. The mixture was heated at 170-180°C for 1.5 h, poured into ice (300 mL) and then basified with sodium carbonate to pΗ~8. The mixture was partitioned with ethyl acetate/hexanes and filtered. The filtrate was separated and the aqueous portion was extracted twice with ethyl acetate. The combined organic portion was dried over sodium sulfate, filtered and concentrated in vacuo to afford oil that was purified by column chromatography (ethyl acetate for less polar product and then 30% methanol in ethyl acetate). (3S,8aS)-3-(Chloromethyl)hexahydro-lH-pyrrolo[2,l-c][l,4]oxazine (less polar product) (1.87 g, 10.7 mmol, 18% yield): 1H NMR (400 MHz, CDC13): 4.06 (dd, IH), 3.79-3.71 (m, IH), 3.60-3.48 (m, 2H), 3.36 (dd, IH), 3.15 (dd, IH), 3.13-3.06 (m, IH), 2.21-2.01 (m, 3H), 1.90-1.68 (m, 3H), 1.39-1.24 (m, IH); MS (El) for C8H14NOCl: 176 (MH4). (3R,8aS)-3-(Chloromethyl)hexahydro-lH-pyrrolo[2,l-c][l,4]oxazine (1.54 g, 8.77 mmol, 15% yield): 1H NMR (400 MHz, CDC13): 3.94-3.77 (m, 4H), 3.55 (dd, IH), 3.02- 2.93 (m, 2H), 2.45 (dd, IH), 2.29-2.15 (m, 2H), 1.88-1.64 (m, 3H), 1.49-1.38 (m, IH); MS (El) for C8H14NOCl: 176 (MH4").
[0376] Using the same or analogous synthetic techniques and/or substituting with alternative starting materials, the following were prepared:
[0377] (3R,8aR)-3-(Chloromethyl)hexahvdro-lH-pyrrolor2.1-ciri,41oxazine: 1H NMR (400 MHz, CDC13): 4.05 (dd, IH), 3.79-3.70 (m, IH), 3.61-3.48 (m, 2H), 3.35 (dd, IH), 3.15 (dd, IH), 3.13-3.07 (m, IH), 2.21-2.01 (m, 3H), 1.89-1.67 (m, 3H), 1.39-1.25 (m, IH); MS (El) for C8H14NOCl: 176 (MH4").
[0378] (3S,8aR)-3-(Chloromethyl)hexahvdro-lH-pyrrolor2,l-cl[l,41oxazine: 1H NMR (400 MHz, CDC13): 3.93-3.77 (m, 4H), 3.55 (dd, IH), 3.02-2.93 (m, 2H), 2.45 (dd, IH), 2.30-2.15 (m, 2H), 1.88-1.64 (m, 3H), 1.49-1.37 (m, IH); MS (El) for C8H14NOCl: 176 (MH4). Example 62
[0379] r3S.8aS)-Hexahvdro-lH-pyrrolo[2,l-cin,41oxazin-3-ylmethyl acetate: (3S,8aS)-3- (Chloromethyl)hexahydro-lH-pyrrolo[2,l-c][l,4]oxazine (2.30 g, 13.1 mmol) and potassium acetate (12.8 g, 131 mmol) were stirred in dimethylformamide (25 mL) at 140°C for 20 h. The mixture was partitioned between ethyl acetate and water. The organic portion was washed twice with water, then with brine, dried over sodium sulfate, filtered and concentrated in vacuo to afford (3S,8aS)-hexahydro-lH-pyrrolo[2,l- c][l,4]oxazin-3-ylmethyl acetate as a brown oil (2.53 g, 12.7 mmol, 97% yield). 1H NMR (400 MHz, CDC13): 4.14-4.02 (m, 3H), 3.81-3.72 (m, IH), 3.37-3.31 (m, IH), 3.09 (dt, IH), 3.00 (dd, IH), 2.21-2.00 (m, 3H), 2.10 (s, 3H), 1.90-1.67 (m, 3H), 1.39-1.24 (m, IH); MS (El) for Ci0Hi7NO3: 200 (MH4).
[0380] (3S,8aS)-Hexahvdro-lH-pyrrolo[2,l-cl[1.41oxazin-3-ylmethanol: (3S,8aS)-
Ηexahydro-lH-pyrrolo[2,l-c][l,4]oxazin-3-ylmethyl acetate (2.36 g, 11.9 mmol) was treated with sodium methoxide (25 wt% solution in methanol; 2.7 mL) for 0.5 h. The mixture was cooled in an ice bath and a solution of 4M ΗC1 in 1,4-dioxane (3 mL, 12.0 mmol) was added slowly. The mixture was stirred at room temperature for 5 minutes and then was concentrated in vacuo to afford a suspension which was diluted with dichloromethane, filtered and the filtrate was concentrated in vacuo to afford (3S,8aS)- hexahydro-lH-pyrrolo[2,l-c][l,4]oxazin-3-ylmethanol as a brown oil (1.93 g, >100% yield). 1H NMR (400 MHz, CDC13): 4.05 (dd, IH), 3.73-3.65 (m, 2H), 3.62-3.56 (m, IH), 3.39-3.34 (m, IH), 3.10 (dt, IH), 3.00-2.95 (m, IH), 2.24-1.98 (m, 4H), 1.97-1.70 (m, 3H), 1.44-1.28 (m, IH); MS (El) for C8H15NO2: 158 (MH4).
[0381] (3S,8aS)-hexahvdro-lH-pyrrolo[2, 1-c] [1 ,4]oxazin-3-ylmethyl methanesulfonate: (3S,8aS)-Ηexahydro-lH-pyrrolo[2,l-c][l,4]oxazin-3-ylmethanol (1.00 g, 6.37 mmol) was dissolved in dichloromethane (10 mL) and triethylamine (2.4 mL, 17.3 mmol) was added at 0°C followed by dropwise addition of methanesulfonyl chloride (0.93 mL, 12.0 mmol). The solution was warmed to room temperature and stirred for 1.25 h and then was concentrated in vacuo. The residue was partitioned between ethyl acetate and saturated sodium bicarbonate solution. The organic portion was washed with saturated sodium bicarbonate solution. The combined aqueous portion was extracted with ethyl acetate. The combined organic portion was washed with brine, dried over sodium sulfate, filtered and concentrated in vacuo to afford (3S,8aS)-hexahydro-lH-pyrrolo[2,l-c][l,4]oxazin-3- ylmethyl methanesulfonate as an orange-brown oil (1.20 g, 5.1 mmol, 80% yield). MS (El) for C9H17NO4S: 236 (MH4).
Example 63
[0382] Octah ydro-2H-quinolizin-3 - ylmethanol : Ethyl octahydro-2H-quinolizine-3- carboxylate (2.35 g, 11.1 mmol) was added dropwise to a stirred suspension of lithium aluminum hydride (1 M solution in tetrahydrofuran, 33 mL, 33 mmol) in tetrahydrofuran (50 mL) at 0°C. The reaction was stirred at room temperature for 3 h. The mixture was cooled in an ice bath and ethyl acetate (6 mL) was added slowly, followed by water (1.25 mL), 15% aqueous sodium hydroxide solution (5 mL) and water (1.25 mL). The mixture was filtered through a pad of celite and washed with ether. The filtrate was concentrated in vacuo and dried rigorously to afford octahydro-2H-quinolizin-3-ylmethanol as a yellow oil (1.66 g, 9.82 mmol, 88% yield). MS (El) for Cι0Η19NO: 170 (MH4).
[0383] Octahvdro-2H-quinolizin-3-ylmethyl methanesulfonate: Octahydro-2H-quinolizin- 3-ylmethanol (600 mg, 3.55 mmol) was dissolved in dichloromethane (8 mL) and triethylamine (1.5 mL, 10.8 mmol) was added at 0°C followed by dropwise addition of methanesulfonyl chloride (0.56 mL, 7.16 mmol). The solution was warmed to room temperature and stirred for 1.25 h and then was concentrated in vacuo. The residue was partitioned between ethyl acetate and saturated sodium bicarbonate solution. The aqueous portion was extracted with ethyl acetate. The combined organic portion was washed with brine, dried over sodium sulfate, filtered and concentrated in vacuo to afford octahydro- 2H-quinolizin-3-ylmethyl methanesulfonate as an orange oil (796 mg, 3.22 mmol, 91% yield). MS (El) for CπΗ21NO3S: 248 (MH4).
Example 64
[0384] (3S,8aS)-3-(Hvdroxymethyl)hexahvdropyrrolo[l,2-fllpyrazin-l(2H)-one: A solution of methyl l-[(2S)-3-hydroxy-2-({[(phenylmethyl)oxy]carbonyl}amino)propyl]-L- prolinate (3.50 g, 10.4 mmol) in methanol was added to 5% palladium on carbon (50 wt.% in water) in methanol and treated with hydrogen at 40 psi for 1 h. The mixture was filtered and the filtrate was brought to reflux briefly and then cooled and concentrated in vacuo to afford (3S,8aS)-3-(hydroxymethyl)hexahydropyrrolo[l,2-α]pyrazin-l(2H)-one as a colorless solid (1.50 g, 8.83 mmol, 85% yield). 1H NMR (400 MHz, CDC13): 7.28-7.22 (m, IH), 3.83-3.75 (m, IH), 3.69 (dd, IH), 3.56 (dd, IH), 3.31 (t, IH), 3.08 (dd, IH), 2.92 (dt, IH), 2.76-2.70 (m, IH), 2.66 (dd, IH), 2.28-2.16 (m, IH), 2.02-1.73 (m, 3H); MS (El) for C8H14N2O2: 171 (MH4).
[0385] (3S,8aS)-3-({r(l,l-Dimethylethyl)(dimethvnsilvnoxy}methvnhexahvdro- pyrrolo[ 1 ,2-αlpyrazin- 1 (2H)-one: To a solution of (3S,8aS)-3-(hydroxymethyl) hexahydropyrrolo[l,2-α]pyrazin-l(2H)-one (1.49 g, 8.82 mmol) in dimethylformamide (20 mL) was added triethylamine (2.45 mL, 17.6 mmol) and 4-dimethylaminopyridine (90 mg, 0.882 mmol). The solution was cooled in an ice bath and tert-butyldimethylsilyl chloride (2.66 g, 17.6 mmol) was added. The mixture was warmed to room temperature and stirred for 14 h. The mixture was concentrated in vacuo and the residue was partitioned between ethyl acetate and water. The aqueous portion was extracted twice with ethyl acetate. The combined organic portion was dried over sodium sulfate, filtered and concentrated in vacuo to afford a pale brown solid which was triturated with ethyl acetate to afford (3S,8aS)-3-({[(l,l-dimethylethyl)(dimethyl)silyl]oxy}methyl) hexahydropyrrolo[l,2-α]pyrazin-l(2H)-one as an off-white solid (1.74 g, 5.84 mmol, 66% yield). 1H NMR (400 MHz, CDC13): 6.09-5.90 (m, IH), 3.86-3.76 (m, IH), 3.63 (dd, IH), 3.44 (dd, IH), 3.25 (t, IH), 3.10 (ddd, IH), 2.98-2.90 (m, IH), 2.68-2.60 (m, IH), 2.52 (dd, IH), 2.28-2.18 (m, IH), 2.06-1.95 (m, IH), 1.93-1.74 (m, 2H), 0.90 (s, 9H), 0.07 (s, 6H); MS (El) for C14H28N2O2Si: 285 (MH4).
[0386] (3S.8aS)-3-({[(l,l-Dimethylethyl)(dimethyl)silyl1oxy)methyl)-2-methylhexahvdro pyrrolof 1.2- lpyrazin- 1 (2H)-one: (3S,8aS)-3-({ [(l,l-Dimethylethyl)(dimethyl)silyl]oxy} methyl)hexahydropyrrolo[l,2- ]pyrazin-l(2H)-one (1.51g, 5.32mmol) in dimethylformamide (8 mL) was added to an ice-cooled suspension of sodium hydride (60 wt.% dispersion in oil; 213 mg, 5.32 mmol) in dimethylformamide (8 mL). The mixture was stirred at 0°C for 0.25 h and then iodomethane (0.332 mL, 5.32 mmol) was added dropwise. The mixture was stirred at room temperature for 0.5 h and then was stirred at 70°C for 2 h. The mixture was concentrated in vacuo and the residue was partitioned between ethyl acetate and water. The aqueous portion was extracted with ethyl acetate. The combined organic portion was dried over sodium sulfate, filtered and concentrated in vacuo to afford (3S,8aS)-3-({[(l,l-dimethylethyl)(dimethyl)silyl]oxy}methyl)-2- methylhexahydropyrrolo[l,2-α]pyrazin-l(2H)-one as a yellow oil (1.552 g, 5.21 mmol) which was dissolved in tetrahydrofuran (20 mL) and treated with tetrabutylammonium fluoride (l.OM solution in tetrahydrofuran; 10.4 mL, 10.4 mmol) for 2 h at room temperature. The mixture was concentrated in vacuo and purified by column chromatography (10% methanol in dichloromethane) to afford (3S,8aS)-3- (hydroxymethyl)-2-methylhexahydropyrrolo[l,2-α]pyrazin-l(2H)-one as a yellow oil (496mg, 2.70mmol, 51% yield from (3S,8aS)-3-({[(l,l- dimethylethyl)(dimethyl)silyl]oxy} methyl)hexahydropyrrolo[l,2- ]pyrazin-l(2H)-one). 1H NMR (400 MHz, CDC13): 3.98-3.93 (m, IH), 3.86 (dd, IH), 3.61-3.55 (m, IH), 3.29- 3.25 (m, IH), 3.09-3.03 (m, IH), 3.03-2.97 (m, IH), 3.02 (s, 3H), 2.93 (dd, IH), 2.87-2.79 (m, IH), 2.32-2.21 (m, IH), 2.00-1.86 (m, 2H), 1.83-1.64 (m, IH); MS (El) for C9H16N2O2: 185 (MH4).
Example 65
[0387] 1.2-Dideoxy- 1 -r(2S)-2-(methoxycarbonyl)- 1 -pyrrolidinyl] -2- [ [(phenylmethoxy) carbonyl] amino] -D-g/ycero-hexitol : To a solution of 2-deoxy-2-{[(phenylmethyloxy) carbonyl] amino }-D-glycero-hexopyranose (5.0 g, 0.016 mol) in methanol (500 mL) was added L-proline methyl ester hydrochloride (2.8 g, 0.022 mol) and sodium cyanoborohydride (3.4 g, 0.054 mol). The solution was heated to 64 °C for 14 h. After cooling to room temperature, the reaction mixture was concentrated in vacuo to afford 1,2- dideoxy-l-[(2S)-2-(methoxycarbonyl)-l-pyrrolidinyl]-2-[[(phenylmethoxy)carbonyl] amino] -D-g/ycero-hexitol (6.81 g, 100%) as a clear and colorless oil. MS (El) for C20H31N2O8: 427 (MH4).
Example 66
[0388] Methyl l-r(2S)-3-hvdroxy-2-((r(phenylmethyl)oxy1carbonyl}amino)propyl1-L- prolinate: 1 ,2-dideoxy- 1 - [(2S)-2-(methoxycarbonyl)- 1 -pyrrolidinyl] -2- [ [(phenylmethoxy) carbonyl]amino]-D-g/ycero-hexitol (6.81 g, 0.016 mol) was taken into water (100 mL) and the resulting solution was cooled to 0°C. Sodium periodiate (14.8 g, 0.069 mol) dissolved in water was added dropwise and the resulting mixture was stirred at 0°C for 2 h. The reaction mixture was partitioned with dichloromethane (3x100 mL), dried over anhydrous magnesium sulfate, filtered and concentrated in vacuo. The residue was taken up in methanol (200 mL) and the resulting solution was cooled to 0°C. Sodium borohydride (1.98 g, 0.052 mol) was added and the reaction mixture was stirred for 1 h at 0°C. The reaction mixture was concentrated in vacuo and partitioned with dichloromethane and saturated aqueous ammonium chloride. The organic layer was dried over anhydrous magnesium sulfate, filtered and concentrated in vacuo. The resulting crude product was purified by column chromatography (5% methanol in dichloromethane) to yield methyl 1- [(2S)-3-hydroxy-2-({ [(phenylmethyl)oxy] carbonyl }amino)propyl]-L-prolinate (4.9 g, 92%) as a white solid. MS (El) for d7H25N2O5: 337 (MH4).
[0389] Methyl l-[(2S)-3-[(methylsulfonyl)oxyl-2-({ [(phenylmethyl)oxylcarbonyl } amino propyl] -L-prolinate: Methyl l-[(2S)-3-hydroxy-2-({[(phenylmethyl)oxy]carbonyl}amino) propyl] -L-prolinate (200 mg, 0.594 mmol) was dissolved in dichloromethane (5 mL) followed by the addition of 4-(dimethylamino)pyridine (3.6 mg, 0.039 mmol) and triethylamine (0.125 mL, 0.891 mmol) and the resulting mixture was cooled to' 0 °C. Methanesulfonyl chloride (0.060 mL, 0.773 mmol) was added dropwise and the reaction mixture was stirred for 1 h at 0°C. The mixture was partitioned between dichloromethane and saturated aqueous sodium bicarbonate. The organic layer was dried over anhydrous magnesium sulfate, filtered and concentrated in vacuo to afford methyl l-[(2S)-3- [(methylsulfonyl)oxy]-2-({ [(phenylmethyl)oxy]carbonyl } amino)propyl] -L-prolinate (246 mg, 100%) as a clear and colorless oil. MS (El) for C18H27N2O7S: 415 (MH4).
Example 67
[0390] 1 , l-Dimethylethyl (3aR,6aS)-5-(hydroxymethyl)hexahvdrocvclopenta[c]pyrrole- 2(lH)-carboxylate: Under a nitrogen atmosphere, borane tetrahydrofuran complex (1M in TΗF, 42 mL, 41.9 mmol) was diluted with tetrahydrofuran (42 mL) and cooled with an ice bath. Neat 2,3-dimethylbut-2-ene (5.0 mL, 41.9 mmol) was added in portions over 0.25 h and the solution was stirred at 0°C for 3 h. A solution of 1,1-dimethylethyl (3aR,6aS)-5- methylidenehexahydrocyclopenta[c]pyrrole-2(lH)-carboxylate (1.98 g, 8.88 mmol) in tetrahydrofuran (10 mL) was added slowly,, and the solution was warmed to room temperature and stirred 12 h. After cooling to 0°C, 10% aqueous sodium hydroxide (17 mL, 41.7 mmol) was added slowly, followed by 30% aqueous hydrogen peroxide (13 mL, 128 mmol) and the solution was warmed to room temperature. The solvent was removed in vacuo and the solution was partitioned between water and diethyl ether. The layers were separated and the aqueous layer was further extracted (3 x 50 mL diethyl ether). The combined organic layers were dried over anhydrous sodium sulfate, filtered and concentrated in vacuo to provide 2.04 (95%) of 1,1-dimethylethyl (3aR,6aS)-5- (hydroxymethyl)hexahydrocyclopenta[c]pyrrole-2(lH)-carboxylate, which was used without purification. 1H NMR (400 MHz, CDC13): 8.50 (broad s, IH), 3.66-3.46 (m, 3H), 3.20-3.00 (m, 2H), 2.70-2.59 (m, 2H), 2.37-2.18 (m, IH), 2.04 (m, IH), 1.84 (broad s, IH), 1.70-1.55 (m, IH), 1.46 (s, 9H), 1.17 (m, IH), 0.93 (m, IH).
[0391] 1 , 1 -Dimethylethyl (3 aR,6aS)-5- { [(methylsulf on vDoxylmethyl }hexahvdrocyclo- penta [c1pyrrole-2(lH)-carboxylate: Methanesulfonyl chloride (0.2mL, 2.48mmol), was added dropwise to a solution of 1,1-dimethylethyl (3aR,6aS)-5-(hydroxymethyl)hexahydro cyclopenta[c] pyrrole-2(lH)-carboxylate (0.40 g, 1.65 mmol) and triethylamine (0.69 mL, 4.95 mmol) in 20 mL dichloromethane at 0°C and the reaction mixture was stirred for 1 h at room temperature. The solvent was evaporated, the resulting crude mixture was diluted with 100 mL ethyl acetate and washed with water (30 mL), 1M aqueous sodium hydroxide, brine, 1M aqueous hydrochloric acid and brine again. The organic layer was dried over anhydrous sodium sulfate, filtered and concentrated in vacuo. The resulting 1,1-dimethylethyl (3aR,6aS)-5-{[(methylsulfonyl)oxy]methyl}hexahydrocyclo- penta[c]pyrrole-2(lH)-carboxylate was used without further purification. MS (El) for C14Η25NO5S: 320 (MH4), 264 (M-fBu).
Example 68
[0392] 1,1-Dimethylethyl (3aR,6aS)-5-(hvdroxy)-hexahvdrocvclopentarc1 pyrrole-2QH)- carboxylate: Sodium borohydride (0.15 g, 4.00 mmol), was added to a solution of 1,1- dimethylethyl (3aR,6aS)-5-oxo-hexahydrocyclopenta[c] pyrrole-2(lH)-carboxylate (0.45 g, 2.00 mmol) in 10 mL methanol at 0°C and the reaction mixture was stirred for 1 h at this temperature. The solvent was evaporated, the crade mixture was diluted with 100 mL ethyl acetate and washed with water (30 mL), 1M aqueous hydrochloric acid and brine. The organic layer was dried over anhydrous sodium sulfate, filtered and concentrated to give 1,1-dimethylethyl (3aR,6aS)-5-(hydroxy)-hexahydrocyclopenta[c] pyrrole-2(lH)- carboxylate (0.44g, 98%). 1H NMR (400 MHz, dg-DMSO): 4.08 (m, IH), 3.40 (m, 2H), 3.30 (m, 2H), 2.50 (m, 2H), 1.98 (m, 2H), 1.40 (s, 9H), 1.30 (m, 2H). MS (El) for C12H2ιNO3: 228 (MH4). [0393] 1,1 -Dimethylethyl (3aR,6aS)-5-( [(methylsulfonyl)oxyl Ihexahydrocyclo- penta[c]pyrrole-2(lH)-carboxylate: Methanesulfonyl chloride (0.18 mL, 2.33 mmol), was added dropwise to a solution of 1,1-dimethylethyl (3aR,6aS)-5-(hydroxy)- hexahydrocyclopenta[c] pyrrole-2(lH)-carboxylate (0.44 g, 1.94 mmol) and triethylamine (0.81 mL, 5.81 mmol) in 10 mL dichloromethane at 0°C and the reaction mixture was stirred for 1 h at room temperature. The solvent was evaporated, the resulting crade mixture was diluted with 100 mL ethyl acetate and washed with water (30 mL), brine, 1M aqueous hydrochloric acid and brine again. The organic layer was dried over anhydrous sodium sulfate, filtered and concentrated. The resulting crade 1,1-dimethylethyl (3aR,6aS)-5-{[(methylsulfonyl)oxy]}hexahydrocyclopenta[c]pyrrole-2(lH)-carboxylate was used without further purification. MS (El) for C13Η23NO5S: 306 (MH4).
Example 69
[0394] 3-(Chloromethyl)hexahydro- IH- [ 1 ,4]oxazino[3 ,4-cl [ 1 ,4] oxazine: A solution of (3R)-morpholin-3-ylmethanol (4.21 g, 36.0 mmol) in 2-(chloromethyl)oxirane (28.2 mL, 0.360 mol) was heated to 40°C for 3 h and then the solution was concentrated in vacuo. The intermediate was cooled in an ice bath and treated with 30.0 mL of concentrated sulfuric acid. The mixture was heated to 170°C for 2 h and then allowed to cool to room temperature. The mixture was poured into ice-water and solid sodium bicarbonate was carefully added until the solution was basic. 10% methanol in ethyl acetate was added and the biphasic mixture was filtered. The layers were separated and the aqueous layer was extracted (3 x 100 mL 10% methanol in ethyl acetate). The combined organic layers were dried over anhydrous sodium sulfate, filtered and concentrated in vacuo. Column chromatography (SiO2, 2:5 hexanes:ethyl acetate) provided 3-(chloromethyl)hexahydro- lH-[l,4]oxazino[3,4-c][l,4]oxazine 2.44g (35%) as two separated diastereomers. (3R,9aS)-3-(chloromethyl)hexahydro-lH-[l,4]oxazino[3,4-c][l,4]oxazine: (0.886 g, 13% yield): 1H NMR (400 MHz, CDC13): 3.91 (m, 3H), 3.82 (m, IH), 3.68 (dt, IH), 3.61 (dd, IH), 3.47 (dd, IH), 3.35 (t, IH), 3.19 (t, IH), 2.80 (d, IH), 2.54 (m, 2H), 2.40 (m, 2H); MS (El) for C8H14NO2Cl: 192 (MH4). (3S,9aS)-3-(chloromethyl)hexahydro-lH- [l,4]oxazino[3,4-c][l,4]oxazine: (1.55 g, 22% yield): 1H NMR (400 MHz, CDC13): 3.85 (m, 2H), 3.73 (m, 3H), 3.50 (m, 2H), 3.29 (t, IH), 3.18 (t, IH), 2.85 (dd, IH), 2.64 (dd, IH), 2.40 (m, 2H), 2.17 (t, IH); MS (El) for C8Hi4NO2Cl: 192 (MH4). [0395] Hexahydro- IH- [ 1 ,41oxazino [3 ,4-cl F 1 ,41 oxazin-3-ylmethyl acetate: A suspension of (3R,9aS)-3-(chloromethyl)hexahydro-lH-[l,4]oxazino[3,4-c][l,4]oxazine (1.97 g, 10.3 mmol) and potassium acetate (10.1 g, 102 mmol) in DMF (20.0 mL) was stirred at 140°C for 16 h, and then at 150°C for another 12 h. The reaction mixture was partitioned between water (250 mL) and ethyl acetate (250 mL), the organic layer was washed with 5% lithium chloride (2 x 100 mL) and brine (100 mL) then dried over anhydrous sodium sulfate and concentrated in vacuo. Column chromatography (SiO2, 1:1 hexane:ethyl acetate, then 100% ethyl acetate) afforded 0.92 g (42%) of hexahydro- 1H- [l,4]oxazino[3,4-c][l,4]oxazin-3-ylmethyl acetate as a yellow oil. Distinct diastereomers as described above were converted in this step to give: (3R,9aS)-hexahydro-lH- [l,4]oxazino[3,4-c][l,4]oxazin-3-ylmethyl acetate: 1H NMR (400 MHz, CDC13): 4.18 (dd, IH), 4.00 (m, IH), 3.80 (dd, IH), 3.68 (dt, IH), 3.60 (dd, IH), 3.46 (m, 2H), 3.22 (t, IH), 2.64 (dd, IH), 2.53 (m, 2H), 2.43-2.35 (m, 2H), 2.10 (s, 3H), and (3S,9aS)- hexahydro-lH-[l,4]oxazino[3,4-c][l,4]oxazin-3-ylmethyl acetate: 1H NMR (400 MHz, CDC13): 4.09 (d, 2H), 3.90-3.82 (m, 2H), 3.75-3.64 (m, 3H), 3.27 (t, IH), 3.18 (t, IH), 2.69 (dd, IH), 2.63 (m, IH), 2.46-2.33 (m, 2H), 2.16 (t, IH), 2.10 (s, 3H).
[0396] (3R,9aS)-Hexahvdro- IH- r 1 ,4] oxazinoβ ,4-cl [ 1 ,41 oxazin-3- ylmethyl methanesulfonate: To a solution of (3R,9aS)-hexahydro-lH-[l,4]oxazino[3,4-c][l,4]oxazin-3- ylmethyl acetate (0.922 g, 4.28 mmol) in methanol (14.0 mL) was added 1.03 mL (4.50 mmol) of sodium methoxide (25% wt. in methanol) dropwise at room temperature. After 5 min., 1.6 mL (6.43 mmol) of 4.0M hydrogen chloride in dioxane was added and a pink precipitate formed. The solution was concentrated in vacuo and the pink solid was taken up in 30.0 mL dichloromethane. This slurry was cooled in an ice bath and triethylamine (3.0 mL, 21.5 mmol) was added, followed by methanesulfonyl chloride (0.37 mL, 4.71 mmol). The resultant yellow solution was stirred for 30 minutes at room temperature. The mixture was then partitioned between dichloromethane and saturated aqueous sodium bicarbonate then the aqueous layer was extracted (3 x 50 mL dichloromethane). The combined organic layers were dried over anhydrous sodium sulfate, filtered and concentrated in vacuo to provide crade (3R,9aS)-hexahydro-lH-[l,4]oxazino[3,4- c][l>4] oxazin-3 -ylmethyl methanesulfonate which was taken on to the following reaction without purification. Example 70
[0397] (8aR)-6-(Chloromethyl)tetrahvdro-lH-[l,31thiazolo[4,3-ciri,41oxazine: A solution of (4R)-l,3-thiazolidin-4-ylmethanol (0.300 g, 2.52 mmol) in 2- (chloromethyl)oxirane (2.0 mL, 25.5 mmol) was heated under nitrogen to 40°C for 12 h. The solution was then cooled to room temperature and 2-(chloromethyl)oxirane was removed in vacuo. The crude intermediate was cooled in ice, and was taken up in 2.0 mL of concentrated sulfuric acid. The resulting mixture was heated to 200°C for 0.5 h then poured carefully onto wet ice, which was allowed to melt. The aqueous solution was carefully made basic using solid sodium bicarbonate and the resulting mixture was filtered using water and 10% methanol in ethyl acetate as eluent. The layers were separated and the aqueous layer was extracted with 10% methanol in ethyl acetate. The combined organic layers were dried over anhydrous sodium sulfate, filtered, and concentrated in vacuo to give 11.6 mg (2.4% yield) of crade (8aR)-6-(chloromethyl)tetrahydro-lH- [l,3]thiazolo[4,3-c][l,4]oxazine as a mixture of diastereomers which was directly taken on to the next step.
Example 71
[0398] 1 , 1-Dimethylethyl (3-endo)-3- { 2- [(methylsulfonyl)oxyl ethyl } -8-azabicvclo \3.2.11 octane-8-carboxylate: To a solution of 1,1-dimethylethyl (3-e7idø)-3-(2-hydroxyethyl)-8- azabicyclo[3.2.1]octane-8-carboxylate (30.3 mg, 1.19 mmol) in dichloromethane (4.0 mL), was added triethylamine (0.5 mL, 3.56 mmol) and the solution was cooled to 0°C under nitrogen. Methanesulfonyl chloride (0.11 mL, 1.42 mmol) was added slowly and mixture was allowed to warm to room temperature and stirred for lh. The reaction mixture was partitioned between dichloromethane and water. The aqueous phase was extracted with dichloromethane (2 x 100 mL). The combined organic layers were dried over anhydrous sodium sulfate, filtered and concentrated in vacuo to provide 35.1 mg (89%) of 1,1- dimethylethyl (3-e/z σ)-3-{2-[(methylsulfonyl)oxy]ethyl}-8-azabicyclo[3.2.1]octane-8- carboxylate, which was carried forward for alkylation without purification. Example 72
Figure imgf000233_0001
[0399] N-[3-Fluoro-4-({7-(methyloxy)-6-r(3-morpholin-4-ylpropyl)oxy1quinazolin-4- yl }oxy)phenyl1 -N'-(4-fluorophenyl)cvclopropane- 1 , 1 -dicarboxamide. Crude cyclopropane- 1 , 1 -dicarboxylic acid [3-fluoro-4-(6-hydroxy-7-methoxy-quinazolin-4- yloxy)-phenyl] -amide (4-fluoro-phenyl)-amide (333mg, 0.66mmol), PS-PPh3 resin, (loading = 2.33mmol/g, 797mg, 1.86mmol), 3-morpholin-4-yl-propan-l-ol (0.26ml, 1.88mmol), and DEAD (0.31ml, 1.91mmol) were combined in CH2C12 (10ml) and stirred at room temperature for l-2hrs. The reaction mixture was filtered and the resin thoroughly washed with CH2C12. The filtrate was concentrated in vacuo and the resulting residue was dissolved in EtOAc and washed with H2O (4x) and sat'd NaCl (lx) and then extracted with IN HCl (3x). The combined IN HCl extractions were washed with EtOAc (2x). The acidic aqueous phase was then basified with IN NaOH and extracted with EtOAc (3x). The combined EtOAc extractions were washed with H2O (lx), sat'd NaCl (lx), dried (Na2SO4), and concentrated in vacuo. The resulting residue was purified by preparative reverse phase HPLC (25mM NHjOAc/acetonitrile) and the pure fractions were lyophilized to give cyclopropane- 1,1 -dicarboxylic acid {3-fluoro-4-[7-methoxy-6-(3-morpholin-4-yl- propoxy)-quinazolin-4-yloxy] -phenyl} -amide (4-fluoro-phenyl)-amide (42.6mg, 10%). 1HNMR (400MHz, DMSO-d6): d 10.37 (br s, IH), 10.05 (br s, IH), 8.55 (s, IH), 7.84 (m, IH), 7.65 (m, 2H), 7.58 (s, IH), 7.43 (m, 3H), 7.16 (t, 2H), 4.27 (m, 2H), 4.00 (s, 3H), 3.60 (m, 6H), 2.39 (m, 4H), 1.99 (m, 2H), 1.47 (m, 4H). LC/MS Calcd for [M+H]+ 634.2, found 634.1.
[0400] Using the same or analogous synthetic techniques and/or substituting with alternative starting materials, the following were prepared:
[0401] N-{3-fluoro-4-[(7-(methyloxy)-6-{[(l-methylpiperidin-4-yl methvnoxy}quin- azolin-4-yl)oxy1phenyl } -N'-(4-fluorophenyl)cyclopropane- 1 , 1 -dicarboxamide: 1H NMR (400MHz, CDC13): δ 9.67 (s, IH), 8.59 (s, IH), 8.43 (s, IH), 7.75 (d, IH), 7.52 (s, IH), 7.46 (m, 2H), 7.31 (s, IH), 7.20 (m, 2H), 7.06 (t, 2H), 4.04 (d, 2H), 4.03 (s, 3H), 2.98 (d, 2H), 2.34 (s, 3H), 2.12-2.1.95 (m, 5H), 1.76 (m, 2H), 1.64 (m, 2H), 1.57 (m, 2H).
Example 73
Figure imgf000234_0001
[0402] Preparation of 1 - [4-(6 ,7-dimethoxy-quinolin-4- yloxy)-phenylcarbamoyl] - cyclopropanecarboxylic acid. To the cyclopropyl di-carboxylic acid (449 mg, 3.45 mmol) in THF (3.5 mL) was added TEA (485 μL, 3.45 mmol). The resulting solution was stirred at room temperature under a nitrogen atmosphere for 40 minutes before adding thionyl chloride (250 μL, 3.44 mmol). The reaction was monitored by LCMS for the formation of mono acid chloride (quenched the sample with MeOH and looked for corresponding mono methyl ester). After 3 hours stirring at room temperature, 4-(6,7-dimethoxy-quinolin-4- yloxy)-phenylamine (1.02 g, 3.44 mmol) was added as a solid, followed by more THF (1.5 mL). Continued to stir at room temperature for 16 hours. The resulting thick slurry was diluted with EtOAc (10 mL) and extracted with IN NaOH. The biphasic slurry was filtered and the aqueous phase was acidified with cone. HCl to pH = 6 and filtered. Both solids were combined and washed with EtOAc, then dried under vacuum. The desired product, 1 - [4-(6 ,7-dimethoxy-quinolin-4-yloxy)-phenylcarbamoyl] - cyclopropanecarboxylic acid, was obtained (962 mg, 68.7% yield, 97% pure) as a white solid. 1H NMR (D2O/NaOH): 7.97 (d, IH), 7.18 (d, 2H), 6.76 (m, 4H), 6.08 (d, IH), 3.73 (s, 3H), 3.56 (s, 3H), 1.15 (d, 4H).
Example 74
Figure imgf000234_0002
[0403] 'N-(4-{ r6,7-Bis(methyloxy ouinolin-4-yl1oxy}phenyl)-N'-[r4-fluoroρhenyl)methyll cyclopropane- 1 , 1 -dicarboxamide. To a solution of l-[4-(6,7-dimethoxy-quinolin-4-yloxy)- phenylcarbamoyl]-cyclopropanecarboxylic acid (74.3 mg, 0.182 mmol), 4-Fluoro benzylamine (25 μL, 0.219 mmol), DIEA (90.0 μL, 0.544 mmol) in DMA (1.0 mL) was added HATU (203 mg, 0.534 mmol). The resulting solution was stirred at room temperature for 1 hour before adding dropwise to water (10 mL) with stirring. The slurry was sonicated, filtered and the solids were washed with 1 N NaOH followed by water. After air drying, the solids were further purified by prep HPLC, affording N-(4-{[6,7- bis(methyloxy)quinolin-4-yl]oxy}phenyl)-N'-[(4-fluorophenyl)methyl]cyclopropane-l,l- dicarboxamide (33 mg, 35% yield, 98% pure) as a white solid. 1H NMR (DMSO, d6): 10.82 (s, IH), 8.80 (d, IH), 8.50 (t, IH), 7.83 (d, 2H), 7.74 (s, IH), 7.56 (s, IH), 7.30-7.38 (m, 4H), 7.15 (t, 2H), 6.80 (d, IH), 4.32 (d, 2H), 4.04 (s, 3H), 4.03 (s, 3H), 1.42 (s, 4H).
[0404] The following compounds were prepared, in a similar manner as above, from the coupling of l-[4-(6,7-dimethoxy-quinolin-4-yloxy)-phenylcarbamoyl]-cyclopropane carboxylic acid with a corresponding alkylamine or arylamine.
[0405] N-(4-{[6,7-Bis(methyloxy)quinolin-4-ylloxy}phenyl)-N'-[2-(piperidin-l- ylmethvDphenyllc vclopropane- 1 , 1 -dicarboxamide. IH NMR (DMSO-d6): 10.62 (s, IH), 8.79 (d, IH), 8.24 (t, IH), 7.83 (d, 2H), 7.72 (s, IH), 7.58 (s, IH), 7.37 (d, 2H), 6.76 (d, IH), 4.04 (s, 3H), 4.03 (s, 3H), 3.98 (m, 2H), 3.66 (m, 2H), 3.49 (m, 4H), 3.25 (t, 2H), 3.13 (br., 2H), 1.42 (d, 4H).
[0406] N-(4-{[6,7-Bis(methyloxy)quinolin-4-yl1oxy}phenyl)-N'-[2-(piperidin-l- ylmethyl)phenyllcvcloproρane-l, 1-dicarboxamide. IH NMR (DMSO-d6): 10.78 (s, IH), 10.53 (s, IH), 8.43 (d, IH), 8.12 (d, IH), 7.82 (d, 2H), 7.49 (s, IH), 7.37 (s, IH), 7.20-7.28 (m, 3H), 7.15 (dd, IH), 7.01 (td, IH), 6.35 (d, IH), 3.93 (s, 3H), 3.92 (s, 3H), 3.47 (s, 2H), 2.17 (br., 4H), 1.49 (m, 4H), 1.41 (m, 4H), 1.32 (br., 2H).
[0407] 'N-(4- { [6,7-Bis(methyloxy)quinolin-4-yl1oχy}phenyl)-N'-r2-(pyrrolidin-l - ylmethvDphenyllc vclopropane- 1 , 1 -dicarboxamide. IH NMR (DMSO-d6): 10.98 (s, IH), 10.56 (s, IH), 8.42 (d, IH), 8.10 (dd, IH), 7.81 (m, 2H), 7.49 (s, IH), 7.37 (s, IH), 7.17- 7.27 (m, 4H), 7.01 (td, IH), 6.35 (d, IH), 3.93 (s, 3H), 3.92 (s, 3H), 3.61 (s, 2H), 2.30 (br., 4H), 1.47 (br., 4H), 1.43 (m, 4H).
[0408] N-(4-{[6,7-Bis(methyloxy)quinolin-4-ylloxy1phenyl)-N'-r3-(morρholin-4- ylmeth vDphenyll cyclopropane- 1 , 1 -dicarboxamide. IH NMR (DMSO-d6): 10.12 (s, IH), 10.03 (s, IH), 8.44 (d, IH), 7.74 (d, 2H), 7.57 (s, IH), 7.53 (d, IH), 7.48 (s, IH), 7.37 (s, IH), 7.21 (m, 3H), 6.98 (d, IH), 6.40 (d, IH), 3.93 (s, 3H), 3.92 (s, 3H), 3.56 (t, 4H), 3.41 (s, 2H), 2.34 (br., 4H), 1.48 (s, 4H).
[0409] 'N-(4-{[6,7-Bis(methyloxy)quinolin-4-vnoxy1phenyl)-N'-[2-(moroholin-4- ylmethyl)phenyl1cvclopropane-l.1-dicarboxamide. IH NMR (DMSO-d6): 10.54 (s, IH), 10.47 (s, IH), 8.43 (d, IH), 8.08 (d, IH), 7.78 (d, 2H), 7.49 (s, IH), 7.37 (d, IH), 7.18- 7.30 (m, 4H), 7.03 (t, IH), 6.37 (d, IH), 3.94 (s, 3H), 3.93 (s, 3H), 3.50 (s, 2H), 3.44 (br., 4H), 2.20 (br., 4H), 1.48 (d, 4H).
[0410] 'N-(4-{ [6,7-Bis(methyloxy)quinolin-4-yl1oχy}phenyl)-N'-[3-(piperidin-l- ylmethvDphen vile vclopropane- 1 , 1 -dicarboxamide. IH NMR (DMSO-d6): 10.0-10.2 (br., 2H), 8.46 (d, IH), 7.76 (d, 2H), 7.53 (m, 3H), 7.39 (s, IH), 7.24 (m, 3H), 6.98 (d, IH), 6.43 (d, IH), 3.95 (s, 3H), 3.93 (s, 3H), 3.37 (s, 2H), 2.31 (br., 4H), 1.48 (m, 8H), 1.39 (br., 2H).
[0411] 'N-(4-{[6,7-Bis(methyloxy)quinolin-4-yl1oχy}phenyl)-N'-r3-(ρyrrolidin-l- ylmeth vDphenyll cyclopropane- 1 , 1 -dicarboxamide. IH NMR (DMSO-d6): 10.0-10.2 (br., 2H), 8.46 (d, IH), 7.77 (d, 2H), 7.59 (s, IH), 7.53 (d, IH), 7.51 (s, IH), 7.39 (s, IH), 7.23 (m, 3H), 6.99 (d, IH), 6.43 (d, IH), 3.95 (s, 3H), 3.93 (s, 3H), 3.52 (s, 2H), 2.42 (br., 4H), 1.69 (br, 4H), 1.48 (s, 4H).
Example 75
Figure imgf000236_0001
Figure imgf000237_0001
[0412] Synthesis of N-(4-f r6,7-bis(methyloxy)-2-(methylthio)quinolin-4-ylloxy}-3- fluorophenyl)-N'-(4-fluorophenyl)c vclopropane- 1 , 1 -dicarboxamide Commercially available 5-(bis-methylsulfanyl-methylene)-2,2-dimethyl-[l,3]dioxane-4,6-dione (3.5 g, 14 mmol) and 3,4-dimethoxyoaniline (2.2 g, 14 mmol) were reflux in EtOH (20 mL) for 2 hours. The EtOH was removed under reduced pressure and EtOAc was added to the residue. The product was filtered and washed with cold EtOAc (3X). 5-[(3,4-dimethoxy- phenylamino)-methylsulfanyl-methylene]-2,2-dimethyl-[l,3]dioxane-4,6-dione was obtained as a white solid (1.7 g, 47% yield) and used without further purification. LCMS: m/z 352 (M-H)".
[0413] A mixture of 5-[(3,4-dimethoxy-phenylamino)-methylsulfanyl-methylene]-2,2- dimethyl-[l,3]dioxane-4,6-dione (1.7 g, 6.6 mmol) and diphenylether (3.5 g, 21 mmol) were heated at 260 °C for 10 minutes. The mixture was cooled to room temperature and heptane was added. 6,7-Dimethoxy-2-methylsulfanyl-quinolin-4-ol was filtered and isolated as an orange solid and used without further purification (1.4 g, 83% yield). LCMS: m/z 352 (M+H)+.
[0414] A mixture of 6,7-dimethoxy-2-methylsulfanyl-quinolin-4-ol (1.0 g, 4.0 mmol), 3,4- difluoronitrobenzene (0.48 mL, 4.3 mmol), cesium carbonate (2.6 g, 8.0 mmol), and DMF (15 mL) was stirred at room temperature for 12 hours, after which time, the mixture was filtered. The filtrate was extracted with DCM, washed with 10% LiCl(aq.), water, (IX) and brine (IX), followed by drying over Na2SO and concentration in vacuo. The crude solids were purified by flash chromatography (silica gel, 5% MeOH in DCM), affording the nitroquinoline (1.3 g, 85.8% yield) as an orange solid. LCMS: m/z 391 (M+H)+. A mixture of nitroquinoline (0.33 g, 0.85 mmol), 5% Pt/S on carbon (0.050 g), ammonium formate (0.40 g, 6.3 mmol) in EtOH (5 mL) was heated at 80 °C for 1 hour The mixture was cooled to room temperature and the solvent removed under reduced pressure. The residue was dissolved in DCM, the mixture filtered, and the precipitate discarded. Removal of the organic solvent afforded 4-(6,7-dimethoxy-2-methylsulfanyl-quinolin-4- yloxy)-3-fluoro-phenylamine as an orange oil (220 mg, 73% yield). LCMS: m/z 361 (M+H)+. [0415] To a mixture of 4-(6,7-dimethoxy-2-methylsulfanyl-quinolin-4-yloxy)-3-fluoro- phenylamine (0.22 g, 0.61 mmol) and l-(4-Fluoro-phenylcarbamoyl)- cyclopropanecarboxylic acid (0.16 g, 0.73 mmol) in DMF (5 mL) was added TEA (0.25 mL, 1.8 mmol) followed by HATU (0.57 g, 1.5 mmol). The resulting solution was stirred overnight at room temperature. The reaction mixture was dumped into water and extracted with DCM (2X). The combined extracts were washed with 5% LiCl(aq.) (3X), water, (IX) and brine (IX), followed by drying over Na2SO4 and concentration in vacuo. The crade solids were purified by preparatory HPLC with ammonium acetate, affording N-(4-{[6,7-bis(methyloxy)-2-(methylthio)quinolin-4-yl]oxy}-3-fluorophenyl)-N'-(4- fluorophenyl)cyclopropane-l, 1-dicarboxamide (0.39 g, 11% yield) as a white solid. 1H NMR DMSO-d6) δ 10.34 (s, IH), 9.94 (s, IH), 7.83 (d, IH), 7.59 (m, 2H), 7.56 (m, IH), 7.40 (m, 2H), 7.23 (s, IH), 7.09 (t, 2H), 6.12 (s, IH), 3.88 (s, 3H), 3.85 (s, 3H), 2.48 (s, 3H), 1.40 (m, 4H).
Example 76
Figure imgf000238_0001
Figure imgf000238_0002
2. Ammonium formate, Fe, Tol/H20 reflux
Figure imgf000238_0003
[0416] Synthesis Of N-(4- { [2-amino-6,7-bis(methyloxy)quinolin-4-yl1 oxy } -3- fluorophenyl)-N'-(4-fluorophenyl)cvclopropane-l, 1-dicarboxamide. A mixture of 5- [(3 ,4- dimethoxy-phenylamino)-methylsulfanyl-methylene]-2,2-dimethyl-[l,3]dioxane-4,6-dione (1.0 g, 2.8 mmol), 30% ammonium hydroxide (8.5 mL), HgCl2 (0.76 g, 2.8 mmol) in THF (5 mL) was stirred at room temperature for 30 minutes. The mixture was extracted with DCM and water (3X) and dried with Na2SO . Concentration in vacuo afforded 5-[amino- (3,4-dimethoxy-phenylamino)-methylene]-2,2-dimethyl-[l,3]dioxane-4,6-dione as a white solid (0.90 g, 97% yield) and this compound was used without further purification. LCMS: m/z 321 (M-H)". [0417] A mixture of 5-[amino-(3,4-dimethoxy-phenylamino)-methylene]-2,2-dimethyl- [l,3]dioxane-4,6-dione (0.90 g, 2.8 mmol) and diphenylether (3.0 g, 18 mmol) was heated at 260 °C for 30 minutes. The mixture was cooled to room temperature and heptane was added. Product 2-amino-6,7-dimethoxy-quinolin-4-ol was filtered and isolated as an orange solid and used without further purification (0.31 g, 33% yield). LCMS: m/z 221 (M+H)+.
[0418] 4-(4-Amino-2-fluoro-phenoxy)-6,7-dimethoxy-quinolin-2-ylamine was synthesized from 2-amino-6,7-dimethoxy-quinolin-4-ol in a similar manner as 4-(6,7- dimethoxy-2-methylsulfanyl-quinolin-4-yloxy)-3-fluoro-phenylamine, and obtained as a white solid (4.0% yield). LCMS: m/z 330 (M+H)+.
[0419] N-(4-{[2-amino-6,7-bis(methyloxy)quinolin-4-yl]oxy}-3-fluorophenyl)-N'-(4- fluorophenyl)cyclopropane- 1,1 -dicarboxamide was synthesized from 4-(4-amino-2-fluoro- phenoxy)-6,7-dimethoxy-quinolin-2-ylamine in a similar manner as N-(4-[[6,7- bis(methyloxy)-2-(methylthio)quinolin-4-yl]oxy}-3-fluorophenyl)-N'-(4- fluorophenyl)cyclopropane-l, 1-dicarboxamide. It was purified by preparatory HPLC using ammonium acetate and isolated as a white solid (4.0% yield). 1H NMR (DMSO-dβ) δ 10.34 (s, IH), 9.95 (s, IH), 7.82 (d, IH), 7.58 (m, 2H), 7.44 (d, IH), 7.33 (t, IH), 7.25 (s, IH), 7.09 (t, 2H), 7.07 (s, IH), 6.17 (br s, 2H), 5.66 (s, IH), 3.79 (s, 3H), 3.77 (s, 3H), 1.40 (d, 4H). LCMS: m/z 535 (M+H)+.
Example 77
Figure imgf000239_0001
Figure imgf000239_0002
[0420] Synthesis of 'N-(3-fluoro-4-{ [2-(methylamino)-6,7-bis(methyloxy)quinolin-4- ylloxy}phenyl)-N'-(4-fluorophenyl)cyclopropane-l, 1-dicarboxamide. A mixture of 5- [(3,4-dimethoxy-phenylamino)-methylsulfanyl-methylene]-2,2-dimethyl-[l,3]dioxane-4,6- dione (0.50 g, 1.4 mmol), methylamine (2 M in THF, 0.75 mL 1.5 mmol), HgCl2 (0.38 g, 1.4 mmol) in THF (5 mL) was stirred at room temperature for 30 minutes. The mixture was extracted with DCM and water (3X) and dried with Na2SO4. Concentration in vacuo afforded 5-[(3,4-dimethoxy-phenylamino)-methylamino-methylene]-2,2-dimethyl-
[l,3]dioxane-4,6-dione as a yellow solid (0.48 g, 99% yield) and this compound was used without further purification. LCMS: m/z 335 (M-H)".
[0421] A mixture of 5-[(3,4-dimethoxy-phenylamino)-methylamino-methylene]-2,2- dimethyl-[l,3]dioxane-4,6-dione (0.40 g, 2.8 mmol) and diphenylether (3.0 g, 18 mmol) was heated at 260 °C for 15 minutes. The mixture was cooled to room temperature and heptane was added. Product 6,7-dimethoxy-2-methylamino-quinolin-4-ol was filtered and isolated as a tan solid and used without further purification (0.30 g, quantitative yield). LCMS: m/z 235 (M+H)+.
[0422] [4-(4-Amino-2-fluoro-phenoxy)-6,7-dimethoxy-quinolin-2-yl]-methyl-amine was synthesized from 6,7-dimethoxy-2-methylamino-quinolin-4-ol in a similar manner as 4-(4- Amino-2-fluoro-phenoxy)-6,7-dimethoxy-quinolin-2-ylamine, and isolated as a yellow oil (58% yield). LCMS: m/z 330 (M+H)"4.
[0423] N-(3-fluoro-4-{[2-(methylamino)-6,7-bis(methyloxy)quinolin-4-yl]oxy}phenyl)- N'-(4-fluorophenyl)cyclopropane- 1,1 -dicarboxamide was synthesized from [4-(4-amino-2- fluoro-phenoxy)-6,7-dimethoxy-quinolin-2-yl]-methyl-amine in a similar manner as N-(4- {[6,7-bis(methyloxy)-2-(methylthio)quinolin-4-yl]oxy}-3-fluorophenyl)-N'-(4-fluoro- phenyl)cyclopropane- 1,1 -dicarboxamide. It was purified by preparatory HPLC using ammonium acetate and isolated as a white solid (6.0 mg, 4.0% yield). 1H NMR (DMSO- d6) δ 10.42 (s, IH), 9.91 (s, IH), 7.88 (dd, IH), 7.56 (m, 2H), 7.44 (m, 4H), 7.09 (t, 2H), 5.90 (s, IH), 3.88 (s, 3H), 3.85 (s, 3H), 3.39 (br s, IH), 2.92 (s, 3H), 1.41 (dt, 4H). LCMS: m/z 535 (M+H)+.
Example 78
Figure imgf000240_0001
[0424] 'N-(4- ( [6- 1 r3-(diethylamino)propylloxy 1 -7-(methylox ylqui nolin-4- yll oxy } -3 - fluorophenyl)-N'-(4-fluorophenyl)cvclopropane- 1 , 1 -dicarboxamide. To a slurry of cyclopropane-l,l-dicarboxylic acid [3-fluoro-4-(6-hydroxy-7-methoxy-quinolin-4-yloxy)- phenyl]-amide (4-fluoro-phenyl)-amide (0.12 g, 0.23 mmol), hydroxypropyldiethylamine (0.090 mL, 0.61 mmol), triphenylphosphine (0.20 g, 0.76 mmol) in DCM (10 mL) was added DIAD (0.17 mL, 0.86 mmol). The resulting mixture was stirred at room temperature for 12 hours, after which time, the solvent was removed under reduced pressure. The residue was extracted with EtOAc and IN HCl (6X) and brine (IX) followed by drying with Na2SO4. Concentration of the organic fraction in vacuo afforded 'N-(4-{[6-{[3-(diethylamino)propyl]oxy}-7-(methyloxy)quinolin-4-yl]oxy}-3- fluorophenyl)-N'-(4-fluorophenyl)cyclopropane-l, 1-dicarboxamide as a yellow oil (0.18 g, wet, ca 95% purity by analytical HPLC). Further purification by preparatory HPLC using ammonium acetate afforded the product in 99% purity by analytical HPLC. LCMS: m/z 619 (M+H)+. 1H NMR (DMSO-d6) δ 10.37 (br s, IH), 10.00 (s, IH), 8.44 (d, IH), 7.87 (d, IH), 7.62 (m, 2H), 7.49 (m, 2H), 7.41 (m, 2H), 7.13 (t, 2H), 6.40 (d, IH), 4.17 (t, 2H), 3.93 (s, 3H), 2.59 (t, 2H), 2.49 (m, 6H), 1.91 (m, 4H), 0.94 (t, 6H).
Example 79
Figure imgf000241_0001
[0425] 'N-(4-fluorophenylVN'-(4-(r2-methyl-6,7-bis(methyloxy')quinazolin-4- yll ox y IphenyDc vclopropane- 1 , 1 -dicarboxamide. Commercially available 2-amino-4,5- dimethoxy-benzoic acid methyl ester (3g, 0.014 mol) and acetic anhydride (4.03 mL, 0.0426 mol) were heated in heptane at 100°C for 3 hours. After removal of heptane in vaccuo, the crade product of 2-acetylamino-4,5-dimethoxy-benzoic acid methyl ester was obtained and used without further purification. LC/MS: m/z 254 (M+H).
[0426] To the crade 2-acetylamino-4,5-dimethoxy-benzoic acid methyl ester obtained above was added ammonium acetate (7.98g, 0.104 mol) and acetic acid (10 mL). The mixture was heated at reflux in a pressure tube until the formation of the desired cyclization product, as indicated by LC/MS: m/z 221 (M+H). After cooling to RT, the reaction mixture was diluted with water, and extracted with EtOAc 3 times. The combined organic phase was basified with aq. NaOH solution, and washed 3 times with EtOAc. The aqueous layer was then acidified with aq. HCl and extracted three times with EtOAc. The combined organic extract was dired over Na2SO4 and concentrated in vacuo, affording 6,7-dimethoxy-2-methyl-quinazolin-4-ol (0.15g), which was used without further purification. LC/MS: m/z 221 (M+H).
[0427] A mixture of 6,7-dimethoxy-2-methyl-quinazolin-4-ol obtained from previous step (0.15g, 0.68 mmol) and POC13 (1.59 mL, 17.04 mmol) was heated at reflux for 48 hours. The reaction mixture was poured into ice water, neutralized with NaHCO3; and adjusted to basic with K2CO3. The mixture was cooled to 0°C with stirring. The resulting precipitate was filtered, giving 4-chloro-6,7-dimethoxy-2-methyl-quinazoline (0.094g), which was used without further purification.
[0428] A mixture of the chloro quinazoline (0.094g, 0.397 mmol) obtained above, 4- nitrophenol (0.1 lg, 0.795 mmol) and bromobenzene (3mL) was heated at 160°C for 48 hours. The solvent was then removed and the reaction was taken up in MeOH. Et2O was added and the reaction stirred 30 min and the precipitate was filtered, affording 6,7- dimethoxy-2-methyl-4-(4-nitro-phenoxy)-quinazoline (0.08 lg) as a very light yellow solid. LC/MS: m/z 342 (M+H).
[0429] A mixture of 6,7-dimethoxy-2-methyl-4-(4-nitro-phenoxy)-quinazoline (0.08 lg, 0.236 mmole), Pt/S (0.008g, 15 mol%), ammounium formate (0.098g, 1.56 mmol) and EtOH (3mL) was heated with stirring at 70°C for 3 hours. The reaction mixture was then filtered while hot and washed with hot EtOH. The crude product of 4-(6,7-dimethoxy-2- methyl-quinazolin-4-yloxy)-phenylamine (0.924g) was obtained as a yellow solid, which was used in the next reaction without further purification. LC/MS: m/z 312 (M+H).
[0430] To a mixture of 4-(6,7-dimethoxy-2-methyl-quinazolin-4-yloxy)-phenylamine (O.lOOg, 0.321 mmol) and l-(4-fluoro-phenylcarbamoyl)-cyclopropanecarboxylic acid (0.056g, 0.386 mmol) in DMF was added DIEA (0.168 mL, 0.963 mmol), followed by HATU (0.183g, 0.482 mmol). The reaction mixture was stirred at RT for 15 hours. The mixture was diluted with EtOAc, washed with 5% LiCI aq solution three times, dried over Na2SO4, and concentrated in vacuo. The crude product was purified on preparative HPLC to give 'N-(4-fluorophenyl)-N'-(4-{[2-methyl-6,7-bis(methyloxy)quinazolin-4- yl]oxy}phenyl)cyclopropane-l, 1-dicarboxamide (3.2 mg) as a white solid. IH NMR (DMSO-d6) 10.15 (bs, IH), 10.01 (bs, IH), 7.69-7.75 (m, 2H), 7.61-7.68 (m, 2H), 7.52 (s, IH), 7.32 (s, IH), 7.23-7.29 (m, 2H), 7.12-7.19 (m, 2H), 3.93 (d, 6H), 2.43 (s, 3H), 1.53 (s, 4H).
Example 80
Figure imgf000243_0001
Figure imgf000243_0002
[0431] Preparation of 1 -(4-Fluoro-phenylcarbamoyl)-2-methyl-cyclopropanecarboxylic acid. 2-Methylcyclopropane- 1,1 -dicarboxylic acid methyl ester was prepared by following the literature procedure (Baldwin, J. E.; Adlington, R. M.; Rawlings, B. J. Tetrahedron Lett. 1985, 481.) The carboxylic acid (700 mg, 4.4 mmol) was dissolved in CH2C12 (10 mL). To the resulting solution was added 4-fluoroaniline (590 mg, 5.3 mmol), HOBt (890 mg, 6.6 mmol) and EDCI (2.5 g, 13.2 mmol). The stirring was continued for 3 h at rt. CH2C12 (30 mL) was added to the reaction mixture, and the resulting solution was washed with brine, and dried over Na2SO4. CH2C12 was removed under reduced pressure. Further purification by column chromatography gave 635 mg (57%) of the desired amide. [0432] The methyl ester obtained above was then treated with LiOH»H2O (116 mg, 2.78 mmol, 1.1 eqiv.) in THF (2 mL) and H2O (1 mL) for 3h at rt. THF was removed under reduced pressure. The aqueous solution was diluted with 20 mL of H2O, washed with ether (10 mL), and acidified with 1 N HCl. The solid was filtered, dissolved in EtOAc, and dried over Na2SO4. Removal of EtOAc gave the crade product of l-(4-fluoro- ρhenylcarbamoyl)-2-methyl-cyclopropane-carboxylic acid, which was used in the next reaction. 1H NMR (400 MHz, DMSO-d6) δ 12.99 (br s, 1 H), 10.33 (br s, 1 H), 7.59 (dd, J = 9.0, 5.0 Hz, 2 H), 7.11 (dd, J = 9.0, 9.0 Hz, 2 H), 1.86-1.78 (m, 1 H), 1.43 (dd, J = 9.0, 4.2 Hz, 1 H), 1.30 (dd, J = 7.8, 4.3 Hz, 1 H), 1.19 (d, J = 6.3 Hz, 3 H).
Example 81
Figure imgf000244_0001
Figure imgf000244_0002
[0433] Synthesis of (lS,2R)-N-[3-fluoro-4-({6-(methyloxy)-7-[(3-morpholin-4-ylpropyl) oxy]quinolin-4-yl}oxy)phenyl]-N'-(4-fluorophenyl)-2-methylcyclopropane-l,l- dicarboxamide. To a solution of 4-(7-benzyloxy-6-methoxy-quinolin-4-yloxy)-3-fluoro- phenylamine (150 mg, 0.38 mmol) in CH2C12 (3 mL) was added DIEA (341 mg, 2.64 mmol), l-(4-fluoro-phenylcarbamoyl)-2-methyl-cyclopropanecarboxylic acid (120 mg, 0.49 mmol) and PyBOP (686 mg, 1.32 mmol). The reaction mixture was stirred at rt for 6 h. After standard workup, the crade product was purified by column chromatography.
[0434] The coupling product (130 mg, 0.21 mmol) obtained above was dissolved in EtOH (2 mL). 1,4-cyclohexadiene (170 mg, 2.1 mmol) and 10% Pd/C (10 mg) were added. The mixture was stirred for 2 h under reflux. After cooling, the mixture was filtered through Celite, and washed with MeOH. Removal of the solvents gave the crade product (136 mg), which was used in the next reaction.
[0435] To a solution of the 7-hydroxyquinoline (136 mg, 0.26 mmol) in DMF (2 mL) was added 4-(3-chloropropyl)morpholine hydrochloride (70 mg, 0.35 mmol) and K2CO3 (69 mg, 0.50 mmol). The reaction mixture was then stirred at 80 °C for 5 h. After cooling, EtOAc (20 mL) was added. The EtOAc solution was washed twice with brine, and dried over Na2SO4. Removal of EtOAc and purification by column chromatography (CH2C12 : MeOH = 10:1) gave '(lS,2R)-N-[3-fluoro-4-({6-(methyloxy)-7-[(3-morpholin-4- ylpropyl)oxy] quinolin-4-yl } oxy)phenyl] -N'-(4-fluorophenyl)-2-methylcyclopropane- 1,1- dicarboxamide. The product was then dissolved in ethyl ether, and treated with 1.5 equiv. of 1 N HCl/ether. Filtration and lyophilization gave the HCl salt of '(lS,2R)-N-[3-fluoro- 4-({6-(methyloxy)-7-[(3-morpholin-4-ylpropyl)oxy]quinolin-4-yl}oxy)phenyl]-N'-(4- fluorophenyl)-2-methylcyclopropane-l, 1-dicarboxamide: 1H NMR (400 MHz, DMSO-d6) δ 10.49 (br s, 1 H), 10.26 (br s, 1 H), 10.15 (br s, 1 H), 8.74 (br s, 1 H), 7.95 (br d, J = 13.2 Hz, 1 H), 7.8-7.5 (m, 6 H), 7.16 (t, J = 8.9 Hz, 2 H), 6.82 (br s, 1 H), 4.34 (t, J = 5.9 Hz, 2 H), 4.02 (s, 3 H), 3.99 (br s, 2 H), 3.77 (br t, J = 12.0 Hz, 2 H), 3.56-3.30 (m, 4 H), 3.17- 3.07 (m, 2 H), 2.40-2.30 (m, 2 H), 2.04-1.95 (m, 1 H), 1.45 (dd, J = 7.2, 4.7 Hz, 1 H), 1.36 (dd, J = 8.5, 4.5 Hz, 1 H), 1.09 (d, J = 6.2 Hz, 3 H). /
Example 82
Figure imgf000245_0001
Figure imgf000245_0002
Figure imgf000245_0003
[0436] Synthesis of (lR,2R)-N-[3-fluoro-4-((6-(methyloxy)-7-[(3-morpholin-4- ylprop vPoxyl -quinolin-4- yl } oxy)phenyl1 -N'-(4-fluorophenyl)-2-methylc vclopropane- 1,1- dicarboxamide. To a solution of 4-(7-benzyloxy-6-methoxy-quinolin-4-yloxy)-3-fluoro- phenylamine (322 mg, 0.82 mmol) and 2-Methyl-cyclopropane- 1,1 -dicarboxylic acid methyl ester (195 mg, 1.23 mmol) in CH2C12 (4 mL) was added HOBt (61 mg, 0.32 mmol) and EDCI (211 mg, 1.64 mmol). The stirring was continued for 12 h at rt. The reaction mixture was then diluted with EtOAc and washed with brine. Removal of organic solvents in vacuo and further purification by column chromatography gave the desired coupling product (153 mg).
[0437] The product (153 mg, 0.29 mmol) obtained above was treated with LiOH«H2O (15 mg, 0.35 mmol) in THF (1 mL) and H2O (1 mL) for 2 h. THF was removed. 10 mL of H2O was added to the mixture. The aqueous solution was washed with ether, and acidified with 1 N HCl. The solid was then filtered and dried under vacuum.
[0438] The crude carboxylic acid (118 mg, 0.23 mmol) and 4-fluoroaniline (111 mg, 0.27 mmol) were dissolved in DMF (2 mL). To this solution was added DIEA (178 mg, 1.38 mmol) and PyBOP (358 mg, 0.69 mmol). The mixture was stirred overnight at rt. It was then diluted with EtOAc, washed twice with brine. Removal of EtOAc and column chromatography gave the desired product.
[0439] The product (66 mg, 0.11 mmol) obtained above was dissolved in EtOH (2 mL). 1,4-cyclohexadiene (80 mg, 1.1 mmol) and 10% Pd/C (10 mg) were added. The mixture was stirred for 2 h under reflux. After cooling, the mixture was filtered through Celite, and washed with MeOH. Removal of the solvents gave the crade product (70 mg), which was used in the next reaction.
[0440] To a solution of the 7-hydroxyquinoline (80 mg, 0.15 mmol) in DMF (2 mL) was added 4-(3-chloropropyl)morpholine hydrochloride (62 mg, 0.31 mmol) and K2CO3 (64 mg, 0.46 mmol). The reaction mixture was then stirred at 80 °C for 5 h. After cooling, EtOAc (20 mL) was added. The EtOAc solution was washed twice with brine, and dried over Na2SO4. Removal of EtOAc and purification by column chromatography (CH2C12 : MeOH = 10:1) gave '(lR,2R)-N-[3-fluoro-4-({6-(methyloxy)-7-[(3-morpholin-4- ylpropyl)oxy]quinolin-4-yl}oxy)phenyl]-N'-(4-fluorophenyl)-2-methylcyclopropane-l,l- dicarboxamide. The product was then dissolved in ethyl ether, and treated with 1.5 equiv. of 1 N HCl/ether. Filtration and lyophilization gave the HCl salt of '(lR,2R)-N-[3-fluoro- 4-({6-(methyloxy)-7-[(3-morpholin-4-ylpropyl)oxy]quinolin-4-yl}oxy)phenyl]-N'-(4- fluorophenyl)-2-methylcyclopropane-l, 1-dicarboxamide: 1H NMR (400 MHz, DMSO-dβ) δ 10.65 (br s, 1 H), 10.54 (br s, 1 H), 9.74 (s, 1 H), 8.75 (br s, 1 H), 8.01 (br d, J = 12.9 Hz, 1 H), 7.80-7.50 (m, 6 H), 7.20-7.10 (m, 2 H), 6.84 (br s, 1 H), 4.34 (br t, J = 5 Hz, 2 H), 4.04 (s, 3 H), 4.05-3.95 (m, 2 H), 3.77 (br t, J = 11 Hz, 2 H), 3.52 (br d, J = 12.7 Hz, 4 H), 3.12 (br q, J = 9.0 Hz, 2 H), 2.40-2.30 (m, 2 H), 2.10-1.95 (m, 1 H), 1.40-1.30 (m, 2 H), 1.10 (d, J = 6.2 Hz, 3 H).
Example 83
Figure imgf000247_0001
Figure imgf000247_0002
[0441] Synthesis of '(2R,3R)-N-[3-fluoro-4-({6-(methyloxy)-7-r(3-morpholin-4- ylpropyl)oxyl-quinolin-4-yl}oxy)phenyn-N'-(4-fluorophenyl)-2,3-dimethylcvclopropane- 1 , 1-dicarboxamide. 2,3-trans-Dimethyl-cyclopropane-l,l-dicarboxylic acid diethyl ester was prepared by following the literature procedure. (Ohishi, J. Synthesis, 1980, 690.) To a solution of 2,3-trans-dimethyl-cyclopropane-l,l-dicarboxylic acid diethyl ester (6.75 g, 31.5 mmol) in MeOH (30 mL) was added 33 mL of 1 N NaOH aqueous solution. The mixture was stirred at 85 °C for 5 h. MeOH was removed under reduced pressure; the residue was diluted with 40 mL of H2O. The aqueous solution was washed with 20 mL of ether, and acidified with 1 N HCl. Filtration and drying under vacuum gave 4.72 g 80 %) of the desired carboxylic acid.
[0442] The aniline (1.08 g, 2.78 mmol) and the carboxylic acid (518 mg, 2.78 mmol) prepared above were dissolved in CH2C12 (15 mL). HATU (2.11 g, 5.56 mmol) and DIEA (1.8 mL, 11.1 mmol) were added. The reaction mixture was stirred at rt overnight. It was then concentrated and diluted with EtOAc. The EtOAc solution was then washed with 5% NaOH and brine. Removal of EtOAc gave the crade coupling product, which was hydrolyzed to the corresponding carboxylic acid by treatment with LiOH»H2O (175 mg, 4.17 mmol) in THF (100 mL) -H2O (50 mL) at 60 °C for 10 h.
[0443] The carboxylic acid (850 mg, 1.60 mmol) and 4-fluoroaniline (355 mg, 3.20 mmol) were dissolved in DMF (8 mL). HATU (3.89 g, 3.2 mmol) and DIEA (1.1 ml, 6.4 mmol) were added. The reaction mixture was stirred at rt overnight. H2O (10 mL) was added to the reaction, and a precipitate formed. The solid was filtered, washed with aqueous sat. Na2CO3 and ether. Further purification by column chromatography gave 596 mg (60%) of the desired product. Debenzylation was done by following the standard procedure.
[0444] To a solution of the 7-hydroxyquinoline (261 mg, 0.49 mmol) in DMF (5 mL) was added 4-(3-chloropropyl)morpholine hydrochloride (195 mg, 0.98 mmol) and K2CO3 (202 mg, 1.46 mmol). The reaction mixture was then stirred at 80 °C for 4 h. After cooling, EtOAc (20 mL) was added. The EtOAc solution was washed twice with brine, and dried over Na2SO4. Removal of EtOAc and purification by column chromatography (CH2C12 : MeOH = 10:1) gave 122 mg (37%) of '(2R,3R)-N-[3-fluoro-4-({6-(methyloxy)-7-[(3- morpholin-4-ylpropyl)oxy]quinolin-4-yl}oxy)phenyl]-N'-(4-fluorophenyl)-2,3-dimethyl- cyclopropane-1, 1-dicarboxamide. 1H NMR (400 MHz, CDC13) δ 8.44 (d, J = 5.1 Hz, 1 H), 8.11 (br s, 1 H), 7.77-7.70 (m, 2 H), 7.53 (s, 1 H), 7.50-7.44 (m, 2 H), 7.40 (s, 1 H), 7.22- 7.16 (m, 2 H), 7.06-6.98 (m, 2 H), 6.36 (br d, J = 5.1 Hz, 1 H), 4.26 (t, J = 7.0 Hz, 2 H), 4.02 (s, 3 H), 3.72 (t, J = 4.4 Hz, 4 H), 2.57 (t, J = 7.3 Hz, 2 H), 2.50-2.42 (m, 4 H), 2.18- 2.10 (m, 2 H), 1.80-1.66 (m, 2 H), 1.30-1.24 (m, 6 H). Example 84
Figure imgf000249_0001
[0445] Synthesis of 'N-(4-{ [6,7-bis(methyloxy)quinolin-4-yl1oχy}phenyl)-N'-(4- fluorophenyl)- 1 -(phenylmethyl)azetidine-3 ,3-dicarboxamide. l-Benzyl-azetidine-3,3- dicarboxylic acid was prepared by following the literature procedure (Miller, R. A.; et al. Syn. Comm. 2003, 33, 3347). To a solution of 4-(6,7-dimethoxy-quinolin-4-yloxy)- phenylamine (4.2 mmol, 1 equiv.) and 4-fluoroaniline (4.2 mmol, 1 equiv.) in DMF (20 mL) was charged with DIEA (12.6 mmol, 3 equiv.) and a solution of 1-benzyl-azetidine- 3,3-dicarboxylic acid ( 4.2 mmol, 1 equiv.) in DMF (10 mL). The reaction mixture was allowed to stir at RT and monitored by LCMS. The reaction was complete in 6 h. The reaction mixture was diluted with ethyl acetate and washed with 10% LiCI (3x), brine (3x), dried with sodium sulfate, filtered and the solvent was reduced in vacuo. The crade product was purified by silica gel chromatography eluting with 2% of MeOH in EtOAc. The fractions containing the desired product were further purified using preparative HPLC to give 'N-(4-{ [6,7-bis(methyloxy)quinolin-4-yl]oxy}phenyl)-N'-(4-fluorophenyl)-l- (phenylmethyl)azetidine-3,3-dicarboxamide (300 mg, 12% yield) as a white solid. 1HNMR (DMSO-d6): 10.0 (s, IH), 9.90 (s, IH), 8.45 (d, IH), 7.80 (d, 2H), 7.70 (m, 2H), 7.50 (s, IH), 7.40 (s, IH), 7.48-7.15 (m, 9H), 3.95 (s, 6H), 3.70 (s, 4H), 3.60 (s, 2H). LCMS (POS): 607.2 (M+H).
[0446] N-(4-{[6,7-bis(methyloxy)quinolin-4-yl]oxy}phenyl)-N'-(4-fluorophenyl) azetidine-3,3-dicarboxamide. To a solution of 'N-(4-{[6,7-bis(methyloxy)quinolin-4- yl]oxy}phenyl)-N'-(4-fluorophenyl)-l-(phenylmethyl)azetidine-3,3-dicarboxamide (300 mg, 0.5 mmol) in MeOH (50 mL) was charged with Pd/C (50% wet, 10% mmol, 265 mg) and acetic acid (2 mL). The reaction mixture was subjected to hydrogenolysis condition under H2 (50 psi) on a Parr Hydrogenator for 16 hr. The reaction mixture was filtered through celite and washed with MeOH. After removal of solvent in vacuo, the crude product was purified using preparative HPLC (solvent system: MeCN/H2O/NH4OAc), affording N-(4-{[6,7-bis(methyloxy)quinolin-4-yl]oxy}phenyl)-N'-(4-fluorophenyl) azetidine-3,3-dicarboxamide (82 mg, 32% yield) as a white solid. XHNMR (DMSO-d6): 8.46 (d, IH), 7.84 (d, 2H), 7.70 (m, 2H), 7.50 (s, IH), 7.40 (s, IH), 7.24 (d, 2H), 7.20 (t, 2H), 6.44 (d, IH), 4.03 (s, 4H), 3.95 (s, 6H), 1.90 (s, 3H, acetate salt). LCMS (POS): 517.3 (M+H).
Example 85
Figure imgf000250_0001
[0447] N- { 3 -fluoro-4- [(6-(methyloxy -7- { \( 1 -methylpiperidin-4- vDmethyl] oxy } quinolin- 4-yl)oxy1phenyl 1 -N'-(4-fluorophenyl)cyclopropane- 1 , 1 -dicarboxamide. To a solution of cyclopropane- 1,1 -dicarboxylic acid {3-fluoro-4-[6-methoxy-7-(piperidin-4-ylmethoxy)- quinolin-4-yloxy] -phenyl} -amide (4-fluoro-phenyl)-amide, TFA salt (~500 mg, 0.71 mmol) in C1CH2H2C1 (8 mL) were added 30% formaldehyde (4 mL) and NaBH(OAc)3 (752 mg, 3.55 mmol). The reaction mixture was stirred overnight. It was then quenched with aqueous sat. NaHCO3, extracted with EtOAc. The organic phase was washed with brine and dried over Na2SO4. Drying salts were filtered, washed with EtOAc and the filtrate concentrated in vacuo to give 210mgs of crade product. The resulting residue was redissolved in EtOAc and any insoluble material filtered. To the filtrate was added 4M HCl in dioxane (200μl) and the mixture was stirred at room temperature for 1 hour. Solids were filtered, washed with EtOAc, dried under high vacuum, dissolved in 50% aqueous AcCN and lyophilized to give 'N-{3-fluoro-4-[(6-(methyloxy)-7-{[(l-methylpiperidin-4- yl)methyl]oxy}quinolin-4-yl)oxy]phenyl}-N'-(4-fluorophenyl)cyclopropane-l, 1-dicarboxamide, HCl salt (113 mg, -25% yield). 1HNMR (400MHz, DMSO-d6): δ 10.51 (s, IH), 10.30 (br. s, IH), 10.04 (s, IH), 8.80 (d, IH), 7.99 (dd, IH), 7.55 (m, 2H), 7.67-7.53 (m, 4H), 7.16 (t, 2H), 6.89 (d, IH), 4.13 (d, 2H), 4.05 (s, 3H), 3.47 (m, 2H), 3.00 (m, 2H), 2.74 (d, 3H), 2.17 (m, IH), 2.03 (m, 2H), 1.68 (m, 2H), 1.49 (m, 4H). LC/MS Calcd for [M+H]+ 617.3, found 617.4. Anal. HPLC (8 min gradient): 98% pure, 3.11 min. Example 86
Figure imgf000251_0001
■ HCl
[0448] (lR,2R,3S)-N-(4-(r7-{r2-rdiethylamino)ethylloxy}-6-(methyloxy)ouinolin-4- yl1oxy}-3-fluorophenyl)-N'-(4-fluorophenyl)-2,3-dimethylcvclopropane-l,l- dicarboxamide. 2,3-Dimethyl-cyclopropane-l,l-dicarboxylic acid [3-fluoro-4-(7-hydroxy- 6-methoxy-quinolin-4-yloxy)-phenyl]-amide (4-fluoro-phenyl)-amide (210 mg, 0.39mmol), DMA (2mls), (2-chloro-ethyl)-diethyl-amine, HCl salt (73 mg, 0.42mmol) and K2CO3 (136 mg, 0.98mmol) were combined and heated at 80C overnight. The reaction mixture was then diluted with H2O and sonicated. The resulting solids were filtered, washed with H O and dried under high vacuum. The crade product was then purified by preparative HPLC using an ammonium acetate buffer system and lyophilized to give '( lR,2R,3S)-N-(4- { [7- { [2-(diethylamino)ethyl] oxy } -6-(methyloxy)quinolin-4-yl]oxy } -3- fluorophenyl)-N'-(4-fluorophenyl)-2,3-dimethylcyclopropane-l, 1-dicarboxamide (39 mg, 16% yield). 1HNMR (400MHz, DMSO-d6): δ 10.14 (s, IH), 9.61 (s, IH), 8.46 (d, IH), 7.87 (dd, IH), 7.67 (m, 2H), 7.57 (m, IH), 7.51 (s, IH), 7.42 (s, IH), 7.39 (m, IH), 7.15 (t, 2H), 6.41 (d, IH), 4.20 (m, 2H), 3.94 (s, 3H), 2.87 (m, 2H), 2.60 (m, 4H), 1.80 (m, 2H), 1.18 (s, 3H), 1.17 (s, 3H), 1.01 (m, 6H). Note: 0.5eq of AcOH is present by NMR. LC/MS Calcd for [M+H]"4 633.3, found 633.4. Anal. HPLC (25 min gradient): 96% pure, 18.52 min.
Example 87
Figure imgf000251_0002
■HCl [0449] N-(4- { [7- 1 [2-(diethylamino)ethyll oxy } -6-(methylox v ouinazolin-4- yll oxy I -3 - fluorophenyl)-N'-(4-fluorophenyl)-2,2-dimethylcvclopropane-l, 1-dicarboxamide. 2,2- Dimethyl-cyclopropane- 1 , 1 -dicarboxylic acid [3 -fluoro-4-(7-hydroxy-6-methoxy- quinazolin-4-yloxy)-phenyl]-amide (4-fluoro-phenyl)-amide (203 mg, 0.38mmol), DMA (2mls), (2-chloro-ethyl)-diethyl-amine, HCl salt (73 mg, 0.42mmol) and K2CO3 (146 mg, 1.05mmol) were combined and heated at 80C overnight. The reaction mixture was then diluted with H2O and extracted with CH2C12 (3x). The combined CH2C12 extractions were washed with sat'd NaHCO3 (lx), sat'd NaCl (lx), dried (Na2SO ), and concentrated in vacuo. The resulting crade product was purified by flash chromatograghy (Silica Gel 60, 100% EtOAc, followed by 10% MeOH, 1% triethylamine in EtOAc), then dissolved in 50% aqueous AcCN and lyophilized to give 'N-(4-{[7-{[2-(diethylamino)ethyl]oxy}-6- (methyloxy)quinazolin-4-yl]oxy}-3-fluorophenyl)-N'-(4-fluorophenyl)-2,2-dimethyl- cyclopropane-1, 1-dicarboxamide (70 mg, 29% yield). 1HNMR (400MHz, DMSO): δ 10.24 (s, IH), 10.00 (s, IH), 8.54 (s, IH), 7.84 (dd, IH), 7.66 (m, 2H), 7.56 (s, IH), 7.51 (m, IH), 7.43 (m, 2H), 7.18 (t, 2H), 4.26 (m, 2H), 3.98 (s, 3H), 2.88 (m, 2H), 2.59 (m, 4H), 1.58 (m, 2H), 1.18 (s, 6H), 1.00 (t, 6H). LC/MS Calcd for [M+H]+ 634.3, found 634.4. Anal. HPLC (25 min gradient): 94% pure, 24.08 min.
Example 88
Figure imgf000252_0001
Figure imgf000252_0002
[0450] Synthesis of 'N-r3-(aminomethyl)ρhenyl1-N'-(4-{ [6,7-bis(methyloxy)quinolin-4- ylloxylphenyDcyclopropane- 1 , 1-dicarboxamide. (4-Nitro-benzyl)-carbamic acid tert-butyl ester. 4-Nitro-benzylamine, HCl salt (5.19g, 27.5mmol) was dissolved in dioxane (lOOmls). NaOH (3.4g, 85.0mmol) in H2O (20mls) was added, followed by Boc anhydride (7.6g, 34.8mmol). The mixture was stirred at room temperature. After 3hrs, the reaction mixture was diluted with EtOAc and washed with H2O (3x), sat'd NaCl (lx), dried (Na2SO ), and concentrated in vacuo. The resulting residue was triturated with hexanes, the resulting solids filtered, washed with hexanes and dried under vacuum to give (4-nitro-benzyl)-carbamic acid tert-butyl ester (6.34g, 91% yield). LC/MS Calcd for [M+H]4" 253.1, found 197.0 (minus t-butyl).
[0451] (4-Amino-benzyl)-carbamic acid tert-butyl ester. (4-Nitro-benzyl)-carbamic acid tert-butyl ester (6.34g, 25.1mmol), iron powder (6.5g, 116mmol), ammonium formate (13.0g, 206mmol), H2O (75mls), and toluene (75mls) were combined and heated to reflux. After 3hrs the reaction mixture was allowed to cool and filtered through Celite with thorough washing with EtOAc. The filtrate was transferred to a separatory funnel and the phases separated. The organic phase was further washed with H2O (lx), sat'd NaCl (lx), dried (Na2SO4), and concentrated in vacuo to give (4-amino-benzyl)-carbamic acid tert- butyl ester (5.02g, 90% yield). LC/MS Calcd for [M+H]+ 223.1, found 167.1 (minus t- butyl).
[0452] [3-({ l-[4-(6,7-Dimethoxy-quinolin-4-yloxy)-phenylcarbamoyl]-cyclopropane- carbonyl}-amino)-benzyl] -carbamic acid tert-butyl ester. l-[4-(6,7-Dimethoxy-quinolin- 4-yloxy)-phenylcarbamoyl]-cyclopropanecarboxylic acid (254 mg, 0.62mmol), (4-amino- benzyl)-carbamic acid tert-butyl ester (164 mg, 0.74mmol), dry DMA (lOmls), HATU (714 mg, 1.88mmol), and DIEA (325ml, 1.86mmol) were combined and stirred at room temperature. After 2hrs, the reaction mixture is diluted with H2O and the resulting solids are filtered, washed with H2O, followed by sat'd NaHCO3, and dried under high vacuum to give crade [3-({ l-[4-(6,7-dimethoxy-quinolin-4-yloxy)-phenylcarbamoyl]- cyclopropanecarbonyl}-amino)-benzyl] -carbamic acid tert-butyl ester (301 mg, 79% yield) which was used in the next reaction without further purification. LC/MS Calcd for [M+H]+ 613.3, found 613.1.
[0453] N-[3-(Aminomethyl)phenyl]-N'-(4-{[6,7-bis(methyloxy)quinolin-4-yl]oxy}phenyl) cyclopropane- 1,1 -dicarboxamide, TFA salt. [3-({ l-[4-(6,7-Dimethoxy-quinolin-4- yloxy)-phenylcarbamoyl]-cyclopropanecarbonyl}-amino)-benzyl]-carbamic acid tert-butyl ester (50 mg, 0.081mmol) was dissolved in 50% TFA in CH2C12 (lOmls) and stirred at room temperature. After 2hrs, the reaction mixture was concentrated in vacuo and the resulting residue was triturated with Et2O. The resulting solids were filtered, washed with Et2O and dried under high vacuum to give 'N-[3-(aminomethyl)phenyl]-N'-(4-{[6,7- bis(methyloxy)quinolin-4-yl]oxy}phenyl)cyclopropane-l, 1-dicarboxamide as the TFA salt (54 mg, 100%). 1HNMR (400MHz, DMSO-d6): δ 10.28 (s, IH), 10.19 (s, IH), 8.77 (m, IH), 8.21 (m, 3H), 7.84 (m, 2H), 7.76 (m, IH), 7.71 (m, IH), 7.58 (m, 2H), 7.38 (m, 3H), 7.19 (m, IH), 6.76 (m, IH), 4.03 (s, 6H), 3.39 (m, 2H), 1.53 (m, 4H). Note: all peaks are very broad and unresolved. LC/MS Calcd for [M+H]"4 513.2, found 513.4. Anal. HPLC (25 min gradient): 88% pure, 12.39 min.
[0454] Table 3 contains 1H-NMR data for selected compounds of the invention.
Table 3
Figure imgf000254_0001
Table 3
Figure imgf000255_0001
Table 3
Figure imgf000256_0001
Table 3
Figure imgf000257_0001
Table 3
Figure imgf000258_0001
Table 3
Figure imgf000259_0001
Table 3
Figure imgf000260_0001
Table 3
Figure imgf000261_0001
Table 3
Figure imgf000262_0001
Table 3
Figure imgf000263_0001
Table 3
Figure imgf000264_0001
Assays
[0455] Kinase assays were performed by measurement of incorporation of γ- P ATP into immobilized myelin basic protein (MBP). High binding white 384 well plates (Greiner) were coated with MBP (Sigma #M-1891) by incubation of 60ul/well of 20μg/ml MBP in Tris-buffered saline (TBS; 50mM Tris pH 8.0, 138mM NaCl, 2.7mM KCl) for 24 hours at 4° C. Plates were washed 3X with lOOμl TBS. Kinase reactions were carried out in a total volume of 34μl in kinase buffer (5mM Hepes pH 7.6, 15mM NaCl, 0.01% bovine gamma globulin (Sigma #1-5506), lOmM MgCl2, ImM DTT, 0.02% TritonX-100). Compound dilutions were performed in DMSO and added to assay wells to a final DMSO concentration of 1%. Each data point was measured in duplicate, and at least two duplicate assays were performed for each individual compound determination. Enzyme was added to final concenfrations of lOnM or 20nM, for example. A mixture of unlabeled ATP and γ- 33 p Aτp wag added t0 start the reacuon (2χlθ6 cpm of γ-33P ATP per well
(3000Ci/mmole) and either lOμM or 30μM unlabeled ATP, typically. The reactions were carried out for 1 hour at room temperature with shaking. Plates were washed 7x with TBS, followed by the addition of 50μl/well scintillation fluid (Wallac). Plates were read using a Wallac Trilux counter. This is only one format of such assays, various other formats are possible, as known to one skilled in the art.
[0456] The above assay procedure can be used to determine the IC50 for inhibition and/or the inhibition constant, Kj. The IC50 is defined as the concentration of compound required to reduce the enzyme activity by 50% under the conditions of the assay. Exemplary compositions have ICso's of, for example, less than about 100 μM, less than about 10 μM, less than about 1 μM, and further for example having ICso's of less than about 100 nM, and still further, for example, less than about 10 nM. The Ki for a compound may be determined from the IC50 based on three assumptions. First, only one compound molecule binds to the enzyme and there is no cooperativity. Second, the concentrations of active enzyme and the compound tested are known (i.e., there are no significant amounts of impurities or inactive forms in the preparations). Third, the enzymatic rate of the enzyme-inhibitor complex is zero. The rate (i.e., compound concenttation) data are fitted to the equation:
Figure imgf000265_0001
where V is the observed rate, Vmax, is the rate of the free enzyme, I0 is the inhibitor concentration, E0 is the enzyme concentration, and Ka is the dissociation constant of the enzyme-inhibitor complex.
Kinase specificity assays:
[0457] Kinase activity and compound inhibition are investigated using one or more of the three assay formats described below. The ATP concentrations for each assay are selected to be close to the Michaelis-Menten constant (KM) for each individual kinase. Dose- response experiments are performed at 10 different inhibitor concentrations in a 384- well plate format. The data are fitted to the following four-parameter equation:
Y = Min + (Max - Min) / (1 + (X/IC50)ΛH) where Y is the observed signal, X is the inhibitor concentration, Min is the background signal in the absence of enzyme (0% enzyme activity), Max is the signal in the absence of inhibitor (100% enzyme activity), IC50 is the inhibitor concenttation at 50% enzyme inhibition and H represents the empirical Hill's slope to measure the cooperativity. Typically H is close to unity.
c-Met Assay
[0458] c-Met biochemical activity was assessed using a Luciferase-Coupled Chemiluminescent Kinase assay (LCCA) format as described above. Again, kinase activity was measured as the percent ATP remaining following the kinase reaction. Remaining ATP was detected by luciferase-luciferin-coupled chemiluminescence. Specifically, the reaction was initiated by mixing test compounds, lμM ATP, lμM poly- EY and lOnM c-Met (baculoviras expressed human c-Met kinase domain P948-S1343) in a 20uL assay buffer (20mM Tris-HCL pH7.5, lOmM MgCl2, 0.02% Triton X-100, lOOmM DTT, 2mM MnCl2). The mixture is incubated at ambient temperature for 2hours after which 20uL luciferase-luciferin mix is added and the chemiluminescent signal read using a Wallac Victor2 reader. The luciferase-luciferin mix consists of 50 mM HEPES, pH 7.8, 8.5ug/mL oxalic acid (pH 7.8), 5 (or 50) mM DTT, 0.4% Triton X-100, 0.25 mg/mL coenzyme A, 63 uM AMP, 28 ug/mL luciferin and 40,000 units of light/mL luciferase.
KDR Assay
[0459] KDR biochemical activity was assessed using a Luciferase-Coupled Chemiluminescent Kinase assay (LCCA) format. Kinase activity was measured as the percent ATP remaining following the kinase reaction. Remaining ATP was detected by luciferase-luciferin-coupled chemiluminescence. Specifically, the reaction was initiated by mixing test compounds, 3 μM ATP, 1.6 μM poly-EY and 5 nM KDR (baculoviras expressed human KDR kinase domain D807-V1356) in a 20uL assay buffer (20mM Tris- HCL pH7.5, lOmM MgCl2, 0.01% Triton X-100, ImM DTT, 3mM MnCl2). The mixture is incubated at ambient temperature for 4 hours after which 20uL luciferase-luciferin mix is added and the chemiluminescent signal read using a Wallac Victor2 reader. The luciferase-luciferin mix consists of 50 mM HEPES, pH 7.8, 8.5ug/mL oxalic acid (pH 7.8), 5 (or 50) mM DTT, 0.4% Triton X-100, 0.25 mg/mL coenzyme A, 63 uM AMP, 28 ug/mL luciferin and 40,000 units of light/mL luciferase.
flt-4 Assay
[0460] Biochemical activity for flt-4 was assessed using an Alphascreen Tyrosine Kinase protocol. AlphaScreen™ (Perkin Elmer) technology is a proximity assay employing microparticles. Singlet oxygen derived from a donor bead following laser excitation results in chemiluminescence when in proximity (100 A) to an acceptor bead due to biomolecular interactions. For the Flt-4 assay, donor beads coated with stteptavidin and acceptor beads coated with PY100 anti-phosphotyrosine antibody were used (Perkin Elmer). Biotinylated poly(Glu,Tyr) 4:1 (Perkin Elmer) was used as the substrate. Substrate phosphorylation was measured by addition of donor/acceptor beads by chemiluminescence following donor-acceptor bead complex formation. Test compounds, 5 μM ATP, 3 nM biotinylated poly(Glu, Tyr) and 1 nM Flt-4 (baculoviras expressed human Flt-4 kinase domain D725-R1298) were combined in a volume of 20 μL in a 384- well white, medium binding microtiter plate (Greiner). Reaction mixtures were incubated for 1 hr at ambient temperature. Reactions were quenched by addition of 10 uL of 15-30 mg/mL AlphaScreen bead suspension containing 75 mM Hepes, pH 7.4, 300 mM NaCl, 120 mM EDTA, 0.3% BSA and 0.03% Tween-20. After 2-16 hr incubation at ambient temperature plates were read using an AlphaQuest reader (Perkin Elmer). IC50 values correlate well with those determined by radiometric assays.
flt-3 Assay
[0461] Biochemical activity for flt-3 was assessed using a Luciferase-Coupled Chemiluminescent Kinase assay (LCCA) format. Kinase activity was measured as the percent ATP remaining following the kinase reaction. Remaining ATP was detected by luciferase-luciferin-coupled chemiluminescence. Specifically, the reaction was initiated by mixing test compounds, 5 μM ATP, 3 μM poly-EY and 5 nM Flt-3 (baculoviras expressed human Flt-3 kinase domain R571-S993) in a 20uL assay buffer (20mM Tris- HCL pH7.5, lOmM MgCl2, 0.01% Triton X-100, ImM DTT, 2mM MnCl2). The mixture is incubated at ambient temperature for 3 hours after which 20uL luciferase-luciferin mix is added and the chemiluminescent signal read using a Wallac Victor2 reader. The luciferase-luciferin mix consists of 50 mM HEPES, pH 7.8, 8.5ug/mL oxalic acid (pH 7.8), 5 (or 50) mM DTT, 0.4% Triton X-100, 0.25 mg/mL coenzyme A, 63 uM AMP, 28 ug/mL luciferin and 40,000 units of light/mL luciferase.
c-Kit Assay
[0462] c-Kit biochemical activity was assessed using AlphaScreen TM (Perkin Elmer) technology, described above. Test compounds, ATP, biotinylated poly(Glu, Tyr) and c- Kit kinase were combined in a volume of 20 μL in a 384-well white, medium binding microtiter plate (Greiner). Reaction mixtures were incubated for 1 hr at ambient temperature. Reactions were quenched by addition of 10 uL of 15-30 mg/mL AlphaScreen bead suspension containing 75 mM Hepes, pH 7.4, 300 mM NaCl, 120 mM EDTA, 0.3% BSA and 0.03% Tween-20. After 16 hr incubation at ambient temperature plates were read using an AlphaQuest reader (Perkin Elmer).
Structure Activity Relationships 63] Table 4 shows structure activity relationship data for selected compounds of the invention. Inhibition is indicated as IC50 with the following key: A = IC50 less than 50 nM, B = IC50 greater than 50 nM, but less than 500 nM, C = IC50 greater than 500 nM, but less than 5000 nM, and D = IC50 greater than 5,000 nM. Depending upon the functionality about the quinazoline or quinoline, exemplary compounds of the invention exhibit selectivity for any of c-Met, KDR, c-Kit, flt-3, and flt-4. Abbreviations for enzymes listed in Tables 2-3 are defined as follows: c-Met refers to hepatocyte growth factor receptor kinase; KDR refers to kinase insert domain receptor tyrosine kinase; flt-4, fms-like tyrosine kinase-4, representative of the FLK family of receptor tyrosine kinases; c-Kit, also called stem cell factor receptor or steel factor receptor; and flt-3, fms-like tyrosine kinase-3. Empty cells in the tables indicate lack of data only.
Table 4
Figure imgf000268_0001
Figure imgf000269_0001
Figure imgf000270_0001
Figure imgf000271_0001
Figure imgf000272_0001
Figure imgf000273_0001
Figure imgf000274_0001
Figure imgf000275_0001
Figure imgf000276_0001
Figure imgf000277_0001
Figure imgf000278_0001
Figure imgf000279_0001
Figure imgf000280_0001
Figure imgf000281_0001
Figure imgf000282_0001
Figure imgf000283_0001
Figure imgf000284_0001
Figure imgf000285_0001
Figure imgf000286_0001
Figure imgf000287_0001
Figure imgf000288_0001
Figure imgf000289_0001
Figure imgf000290_0001
Figure imgf000291_0001
Figure imgf000292_0001
Figure imgf000293_0001
Figure imgf000294_0001

Claims

Wliat is claimed is:
1. A compound for modulating kinase activity according to formula I,
Figure imgf000295_0001
I or a pharmaceutically acceptable salt, hydrate, or prodrug thereof, wherein,
R1 is selected from -H, halogen, -OR3, -NO2, -NH2, -NR3R4, and optionally substituted lower alkyl;
A1 is selected from =N-, =C(H)-, and =C(CN)-;
Z is selected from -S(O)0-2-, -O-, and -NR5-;
Ar is either a group of formula II, or of formula III,
Figure imgf000295_0002
II III wherein,
R is selected from -H, halogen, trihalomethyl, -CN, -NO2, -NH2, -ORJ, -NR 3TrT»4, -S(O)o-2R3, -SO2NR3R3, -CO2R3, -C(O)NR3R3, -N(R3)SO2R3, -N(R3)C(O)R3, -N(R3)CO2R3, -C(O)R3, and optionally substituted lower alkyl; q is 0 to 4;
G is a group -B-L-T, wherein
B is selected from absent, -N(R13)-, -N(SO2R13)-, -O-, -S(O)0-2-, and -C(=O)-;
L is selected from absent, -C(=S)N(R13)-, -C(=NR14)N(R13)-, -SO2N(R13)-, -SO2-, -C(=O)N(R13)-, -N(R13)-, -C(=O)C1-2alkylN(R13)-, -N(R13)C1-2alkylC(=O)-, -C(=O)C0-ialkylC(=O)N(R13)-, -C0-4alkylene-, -C(=O)C0-1alkylC(=O)OR3-, -C(=NR14)Co-1alkylC(=O)-, -C(=O)-, -C(=O)C0-1alkylC(=O)-, and an optionally substituted four to six-membered heterocyclyl containing between one and three annular heteroatoms including at least one nitrogen; and
T is selected from -H, -R13, -C0-4alkyl, -C0-4alkylQ, -OC0-4alkylQ, -C0- alkylOQ, -N(R13)C0-4alkylQ, -SO2C0- alkylQ, -C(=O)C0-4alkylQ, -Co- alkylN(R13)Q, and -C(=O)N(R13)Co-4alkylQ, wherein each of the aforementioned Co-4alkyl is optionally substituted;
J is selected from -S(O)0-2-, -O-, and -NR15-;
R3 is -H or R4;
R4 is selected from optionally substituted lower alkyl, optionally substituted aryl, optionally substituted lower arylalkyl, optionally substituted heterocyclyl, and optionally substituted lower heterocyclylalkyl; or
R3 and R4, when taken together with a common nitrogen to which they are attached, form an optionally substituted five- to seven-membered heterocyclyl, said optionally substituted five- to seven-membered heterocyclyl optionally containing at least one additional annular heteroatom selected from N, O, S, and P;
A2 and A3 are each independently selected from =N-, =C(R2)-;
R5 is -H or optionally substituted lower alkyl;
D is selected from -O-, -S(O)0-2-, and -NR15-;
R50 is either R3, or according to formula IV;
Figure imgf000296_0001
IV wherein X1, X2, and optionally X3, represent the atoms of a saturated bridged ring system, said saturated bridged ring system comprising up to four annular heteroatoms represented by any of X1, X2, and X3; wherein, each X1 is independently selected from -C(R6)R7-, -O-, -S(O)0-2-, and -NR8-; each X2 is independently an optionally substituted bridgehead methine or a bridgehead nitrogen; each X3 is independently selected from -C(R6)R7-, -O-, -S(O)0-2-, and -NR8-; Y is either: an optionally substituted lower alkylene linker, between D and either 1) any annular atom of the saturated bridged ring system, except X2 when X2 is a bridgehead nitrogen, or 2) any heteroatom, represented by any of R or R ; provided there are at least two carbon atoms between D and any annular heteroatom of the saturated bridged ring system or any heteroatom represented by any of R6 or R7; or Y is absent, when Y is absent, said saturated bridged ring system, is directly attached to D via an annular carbon of said saturated bridged ring system, unless D is -SO2-, in which case said saturated bridged ring system, is directly attached to D via an any annular atom of said saturated bridged ring system; m and p are each independently 1-4; n is 0-2, when n = 0, then there is a single bond between the two bridgehead X 's;
R6 and R7 are each independently selected from -H, halogen, trihalomethyl, -CN, -NH2, -NO2, -OR3, -NR3R4, -S(O)0-2R4, -SO2NR3R4, -CO2R3, -C(O)NR3R4, -N(R3)SO2R4, -N(R3)C(O)R3, -NCO2R3, -C(O)R3, optionally substituted lower alkyl, optionally substituted aryl, optionally substituted lower arylalkyl, optionally substituted heterocyclyl, optionally substituted lower heterocyclylalkyl, and a bond to either Y or D; or
R and R , when taken together are oxo; or
R6 and R7, when taken together with a common carbon to which they are attached, form a optionally substituted three- to seven-membered spirocyclyl, said optionally substituted three- to seven-membered spirocyclyl optionally containing at least one additional annular heteroatom selected from N, O, S, and P;
R8 is selected from -R3, Y, -SO2NR3R4, -CO2R4, -C(O)NR3R3, -SO2R4, and -C(O)R3;
R13 is selected from -H, -C(=O)R3, -C(=O)OR3, -C(=O)SR3, -SO2R4, -C(=O)N(R3)R3, and optionally substituted lower alkyl,
1 " two R , together with the atom or atoms to which they are attached, can combine to form a heteroalicyclic optionally substituted with between one and four of R60, said heteroalicyclic can have up to four annular heteroatoms, and said heteroalicyclic can have an aryl or heteroaryl fused thereto, in which case said aryl or heteroaryl is optionally substituted with an additional one to four of R60;
R14 is selected from -H, -NO2, -NH2, -N(R3)R4, -CN, -OR3, optionally substituted lower alkyl, optionally substituted heteroalicyclylalkyl, optionally substituted aryl, optionally substituted arylalkyl and optionally substituted heteroalicyclic;
R15 is a group -M^M2, wherein M1 is selected from absent, -C(=S)N(R13)-, -C(=NR14)N(R13)-, ' -SO2N(R13)-, -SO2-, -C(=O)N(R13)-, -C(=O)C(=O)N(R13)-, -C0- 4alkylene-, -C(=O)-, and an optionally substituted four to six-membered hetercyclyl annular containing between one and three heteratoms including at least one nitrogen; and M2 is selected from -H, -C0-6alkyl, alkoxy, -C(=O)C0- alkylQ, -C0- alkylQ, -OC0- alkylQ-, -N(R13)C0-4alkylQ-, and -C(=O)N(R13)C0- alkylQ; and
Q is a five- to ten-membered ring system, optionally substituted with between zero and four of R20;
R20 is selected from -H, halogen, trihalomethyl, -CN, -NO2, -NH2, -OR3, -NR3R4, -S(O)0-2R3, -SO2NR3R3, -CO2R3, -C(O)NR3R3, -N(R3)SO2R3, -N(R3)C(O)R3, -N(R3)CO2R3, -C(O)R3, and optionally substituted lower alkyl;
R60 is selected from -H, halogen, trihalomethyl, -CN, -NO2, -NH2, -OR3, -NR3R4, -S(O)0-2R3, -SO2NR3R3, -CO2R3, -C(O)NR3R3, -N(R3)SO2R3, -N(R3)C(O)R3, -N(R3)CO2R3, -C(O)R3, optionally substituted lower alkyl, optionally substituted aryl, optionally substituted heteroarylalkyl, and optionally substituted arylalkyl; two of R60, when attached to a non-aromatic carbon, can be oxo; with the proviso, only when Ar is according to formula II, if Y is a C1-6 alkylene; Z is -NH- or -N(CH3)-; R1 is a C1-6alkyl optionally substituted in the 2-position by -OH or a . 4alkoxy group; R2 is -H or halogen; n = 0; and the atoms, X1, of one bridge of the saturated bridged ring system, when combined with both bridgehead atoms, X , of the saturated bridged ring system, represent:
1) either a pyrrolidine or a piperidine, and any atom, X1 or X2, of either of said pyrrolidine or said piperidine is attached to Y, then the other bridge of said saturated bridged ring system cannot be any one of -OC(O)CH2-, -CH2OC(O)-, -OC(O)CH2CH2-, -CH2OC(O)CH2-, -CH2CH2OC(O)-, -OC(O)CH2NH-, -OC(O)CH2N(C1-4alkyl)-, and -OC(O)CH2O-; or 2) either a piperazine or a 4-(Cι- alkyl)-piperazine, and any atom, X1 or X2, of either of said piperazine or said 4-(C1-4alkyl)-piperazine is attached to Y, then the other bridge of said saturated bridged ring system, only when attached via the 2- and the 3-position of either of said piperazine or said 4-(C1- alkyl)-piperazine, cannot be one of -CH2OC(O)CH2-, -CH2CH2OC(O)-, and either of the two aforementioned bridges optionally substituted by one or two C1-2alkyl groups; or
3) a piperazine, and any atom, X1 or X2, of said piperazine is attached to Y, then the other bridge of said saturated bridged ring system, only when attached via the 3- and the 4-position of said piperazine, cannot be one of -C(O)OCH2CH2-, -CH2OC(O)CH2-, and either of the two aforementioned bridges optionally substituted by one or two C1-2alkyl groups, and only when either of the two aforementioned bridges are attached to the 3-position of said piperazine via their left-hand end as depicted above; or
4) a 2-oxomorpholine, said 2-oxomorpholine attached to Y via its 4-position, then the other bridge of said saturated bridged ring system, only when attached via the 5- and the 6-position of said 2-oxomorpholine, cannot be one of -(CH2)g-, -CH2WCH2-, -CH2WCH2CH2-, and -CH2CH2WCH2-, wherein W is -O-, -S(O)o-2-, -NH-, or -N(C1-4alkyl)- wherein g is 2, 3, or 4; and with the proviso that when Z is-O-, Ar is according to formula II, and the portion of G directly attached to Ar is selected from:
Figure imgf000299_0001
then R ,5Q must be of formula IV; and with the proviso that when Ar is phenylene or substituted phenylene, Z is -S(O)o-2- or
-O-, then the portion of G directly attached to Ar cannot contain
Figure imgf000300_0001
when R70 is selected from -H, C1-4alkyl, and Cι-4alkoxyl.
2. The compound according to claim 1, wherein In one example, the compound is according to paragraph [0033], wherein Z is either -O- or -NR5-.
The compound according to claim 2, wherein G is selected from the following:
Figure imgf000300_0002
Figure imgf000301_0001
Figure imgf000302_0001
wherein wherein Q, R20, and R13 are as defined above; each E is selected from -O-, -N(R13)-, -CH2-, and -S(O)0-2-; M is selected from -O-, -N(R13)-, -CH2-, and -C(=O)N(R13)-; each V is independently either =N- or =C(H)-; each methylene in any of the above formulae is independently optionally substituted with R25; and R25 is selected from halogen, trihalomethyl, -CN, -NO2, -NH2, -ORd, -NR 3TrT>4, -S(O)0-2R , -SO2NR >3"rR>3J, -CO2R3, -C(O)NR3R3, -N(R3)SO2R3, -N(R3)C(O)R3, -N(R3)CO2R3, -C(O)R3, optionally substituted aryl, optionally substituted arylalkyl, heteroarylalkyl, and optionally substituted lower alkyl; two of R , together with the carbon or carbons to which they are attached, can combine to form a three- to seven-membered alicyclic or heteroalicyclic, two of R25 on a single carbon can be oxo.
4. The compound according to claim 3, wherein Ar is according to one of formula IIa, IIb, and IIIa.
Figure imgf000302_0002
Ila lib Ilia
5. The compound according to claim 4, wherein D is -O- and R is -OR .
6. The compound according to claim 5, wherein -O-R50 and R1 are interchangeably located at the 6-position and 7-position of the quinazoline or quinoline according to formula I.
7. The compound according to claim 6, wherein R1 is -OH or -OC1-6alkyl.
8. The compound according to claim 7, wherein A1 is =N- or =C(H)-.
9. The compound according to claim 8, wherein G is selected from:
Figure imgf000303_0001
wherein Q, R , R , E, and R are as defined above; each methylene in any of the above formulae, other than those in a depicted ring, is independently optionally substituted with R25; and R25 is selected from halogen, trihalomethyl, oxo, -CN, -NO2, -NH2, -OR3, -NR3R4, -S(O)0-2R3, -SO2NR3R3, -CO2R3, -C(O)NR3R3, -N(R3)SO2R3, -N(R3)C(O)R3, -N(R3)CO2R3, -C(O)R3, optionally substituted aryl, optionally substituted arylalkyl, heteroarylalkyl, and optionally substituted lower alkyl; two of R , together with the carbon or carbons to which they are attached, can combine to form a three- to seven- membered alicyclic or heteroalicyclic.
10. The compound according to claim 9, wherein Q is selected from:
Figure imgf000303_0002
wherein R ,20 is defined as above, and P is a five- to seven-membered ring, including the two shared carbons of the aromatic ring to which P is fused, P optionally containing between one and three heteroatoms.
11. The compound according to claim 10, wherein Ar is according to formula Ila, and G is selected from:
Figure imgf000304_0001
wherein Q, R , R , E, and R are as defined above, and each methylene in any of the above formulae, other than those in a depicted ring, is independently optionally substituted with R25; and R25 is selected from halogen, trihalomethyl, oxo, -CN, -NO2, -NH2, -OR3, -NR3R4, -S(O)o-2R3, -SO2NR3R3, -CO2R3, -C(O)NR3R3, -N(R3)SO2R3, -N(R3)C(O)R3, -N(R3)CO2R3, -C(O)R3, optionally substituted aryl, optionally substituted arylalkyl, heteroarylalkyl, and optionally substituted lower alkyl; two of R , together with the carbon or carbons to which they are attached, can combine to form a three- to seven- membered alicyclic or heteroalicyclic.
12. The compound according to claim 10, wherein Ar is according to formula lib, and G is selected from: *
Figure imgf000304_0002
wherein Q, R20, R13, E, and R60 are as defined above, and each methylene in any of the above formulae, other than those depicted in a ring, is independently optionally substituted with R25; and R25 is selected from halogen, trihalomethyl, oxo, -CN, -NO2, -NH2, -OR3, -NR3R4, -S(O)0-2R3, -SO2NR3R3, -CO2R3, -C(O)NR3R3, -N(R3)SO2R3, -N(R3)C(O)R3, -N(R3)CO2R3, -C(O)R3, optionally substituted aryl, optionally substituted arylalkyl, heteroarylalkyl, and optionally substituted lower alkyl; two of R25, together with the carbon or carbons to which they are attached, can combine to form a three- to seven- membered alicyclic or heteroalicyclic.
13. The compound according to claim 12, wherein the methylene between the two carbonyls of the depicted formulae is di-substituted with either optionally substituted lower alkyl, or an optionally substituted spirocycle.
14. The compound according to claim 11 or claim 12, wherein R50 is a heteroalicylic or a C1-6alkyl-heteroalicylic.
15. The compound according to claim 14, wherein at least one of R2 is halogen.
16. The compound according to claim 14, wherein R50 is according to formula IV.
17. The compound according to claim 16, wherein the saturated bridged ring system according to formula IV has a geometry selected from the group consisting of [4.4.0], [4.3.0], [4.2.0], [4.1.0], [3.3.0], [3.2.0], [3.1.0], [3.3.3], [3.3.2], [3.3.1], [3.2.2], [3.2.1], [2.2.2], and [2.2.1].
18. The compound according to claim 17, wherein Y is selected from -CH2CH2CH2CH2-, -CH2CH2CH2-, -CH2CH2-, -CH2-, and absent.
19. The compound according to claim 18, wherein n is 0 and the saturated bridged ring system according to formula IV has a geometry selected from the group consisting of [4.4.0], [4.3.0], [4.2.0], [4.1.0], [3.3.0], [3.2.0], and [3.1.0].
20. The compound according to claim 19, wherein said saturated bridged ring system contains at least one annular nitrogen or at least one annular oxygen.
21. The compound according to claim 20, wherein said saturated bridged ring system contains -NR8-, wherein R8 is selected from -H, optionally substituted lower alkyl, -CO2R3, -C(O)NR3R3, -SO2R3, and -C(O)R3.
22. The compound according to claim 20, wherein said saturated bridged ring system is of formula V,
Figure imgf000306_0001
v wherein U1 is selected from -O-, -S(O)0-2-, -NR8-, -CR6R7-, and absent; and e is 0 or 1.
23. The compound according to claim 22, wherein Y is -CH2-.
24. The compound according to claim 23, wherein U1 is -NR8-, wherein R8 is selected from -H, optionally substituted lower alkyl, -CO2R3, -C(O)NR3R3, -SO2R3, and -C(O)R3.
25. The compound according to claim 23, wherein TJ1 is -O-.
26. The compound according to claim 23, wherein U1 is absent.
27. The compound according to claim 20, wherein Y is selected from -CH2CH2-, -CH2-, and absent.
28. The compound according to claim 27, wherein said saturated bridged ring system is of formula VI,
Figure imgf000306_0002
VI wherein R9, R10, and R11 are each independently selected from -H, and -OR12; or
R9 is selected from -H, and -OR12, and R10 and R11, when taken together, are either an optionally substituted alkylidene or an oxo;
R12 is selected from -H, -C(O)R3, optionally substituted lower alkylidyne, optionally substituted lower arylalkylidyne, optionally substituted lower heterocyclylalkylidyne, optionally substituted lower alkylidene, optionally substituted lower alkylidenearyl, optionally substituted lower alkylideneheterocyclyl, optionally substituted lower alkyl, optionally substituted lower alkylaryl, optionally substituted aryl, optionally substituted lower heterocyclylalkyl, and optionally substituted heterocyclyl; or two R 's, when taken together, form 1) a corresponding spirocyclic ketal when said two R12's stem from R10 and R11, or 2) a corresponding cyclic ketal when said two R12's stem from R9 and one of R10 and R11.
29. The compound according to claim 28, wherein one of R10 and R11 is -OR12, wherein R12 is selected from -H, -C(O)R3, and optionally substituted lower alkyl; and R9 and the other of R10 and R11 are both -H.
30. The compound according to claim 29, wherein Y is either -CH2- or absent.
31. The compound according to claim 30, wherein R9 is an alkyl group containing at least one fluorine substitution thereon.
32. The compound according to claim 21, wherein said saturated bridged ring system is of formula VII.
Figure imgf000307_0001
VII
33. The compound according to claim 32, wherein Y is either -CH2- or absent.
34. The compound according to claim 33, wherein R8 is methyl or ethyl.
35. The compound according to claim 21, wherein said saturated bridged ring system is of formula VIII.
Figure imgf000307_0002
VIII
36. The compound according to claim 35, wherein Y is -CH2-.
37. The compound according to claim 36, wherein R8 is methyl or ethyl.
38. The compound according to claim 20, wherein said saturated bridged ring system is of formula IX
Figure imgf000308_0001
IX wherein U2 is selected from -O-, -S(O)0-2-, -NR8-, -CR6R7-, and absent.
39. The compound according to claim 38, wherein R of formula IX is selected from -H and optionally substituted alkyl.
40. The compound according to claim 39, wherein U2 is either -CR6R7- or absent.
41. The compound according to claim 40, wherein U2 is either -CH2- or absent.
42. The compound according to claim 41, wherein Y is -CH2-.
43. The compound according to claim 21, wherein said saturated bridged ring system is according to formula X.
Figure imgf000308_0002
X
44. The compound according to claim 43, wherein R is methyl or ethyl.
45. The compound according to claim 1, selected from Table 1.
Table 1
Figure imgf000308_0003
Table 1
Figure imgf000309_0001
Table 1
Figure imgf000310_0001
Table 1
Figure imgf000311_0001
Table 1
Figure imgf000312_0001
Table 1
Figure imgf000313_0001
Table 1
Figure imgf000314_0001
Table 1
Figure imgf000315_0001
Table 1
Figure imgf000316_0001
Table 1
Figure imgf000317_0001
Table 1
Figure imgf000318_0001
Table 1
Figure imgf000319_0001
Table 1
Figure imgf000320_0001
Table 1
Figure imgf000321_0001
Table 1
Figure imgf000322_0001
Table 1
Figure imgf000323_0001
Table 1
Figure imgf000324_0001
Table 1
Figure imgf000325_0001
Table 1
Figure imgf000326_0001
Table 1
Figure imgf000327_0001
Table 1
Figure imgf000328_0001
Table 1
Figure imgf000329_0001
Table 1
Figure imgf000330_0001
Table 1
Figure imgf000331_0001
Table 1
Figure imgf000332_0001
Table 1
Figure imgf000333_0001
Table 1
Entry Name Structure
N-{ 3-Fluoro-4-[6-methoxy-7-
(piperidin-4-ylmethoxy)-
143 quinolin-4-yloxy] -phenyl } -2- oxo-2-(2-phenyl-morpholin-4- yl)-acetamide
N-(2-Dimethylamino-2- phenyl-ethyl)-N'-{ 3-fluoro-4-
144 [6-methoxy-7-(piperidin-4- ylmethoxy)-quinolin-4- yloxy] -phenyl } -oxalamide
N-{ 3-Fluoro-4-[6-methoxy-7- (piperidin-4-ylmethoxy)-
145 quinolin-4-yloxy] -phenyl } -N'-
(2-oxo-2-phenyl-ethyl)- oxalamide
N-[5-Chloro-6-(6,7-
146 dimethoxy-quinolin-4-yloxy)- pyridin-3-yl]-2,2-difluoro-N'- (4-fluoro-phenyl)-malonamide
Figure imgf000334_0001
Table 1
Entry Name Structure
N-Benzyl-N'-{3-fluoro-4-[6- methoxy-7-( 1 -methyl-
147 piperidin-4-ylmethoxy)- quinolin-4-yloxy] -phenyl } - oxalamide
N-{3-Fluoro-4-[6-methoxy-7-
(piperidin-4-ylmethoxy)-
148 quinolin-4-yloxy] -phenyl } -N'-
[2-(2-fluoro-phenyl)-ethyl]- oxalamide
N-[2-(3-Chloro-phenyl)- ethyl]-N'-{3-fluoro-4-[6-
149 methoxy-7-(piperidin-4- ylmethoxy)-quinolin-4- yloxy] -phenyl } -oxalamide
N-{3-Fluoro-4-[6-methoxy-7-
(piperidin-4-ylmethoxy)-
150 quinolin-4-yloxy] -phenyl } -N'-
[2-(2-methoxy-phenyl)-ethyl] - oxalamide
Figure imgf000335_0001
Table 1
Figure imgf000336_0001
Table 1
Entry Name Structure
N-[2-(2-Ethoxy-phenyl)- ethyl]-N'-{3-fluoro-4-[6-
155 methoxy-7-(piperidin-4- ylmethoxy)-quinolin-4- yloxy] -phenyl } -oxalamide
N- [2-(2,4-Dimethyl-phenyl)- ethyl]-N'-{3-fluoro-4-[6-
156 methoxy-7-(piperidin-4- ylmethoxy)-quinolin-4- yloxy] -phenyl } -oxalamide
N- { 3-Fluoro-4- [6-methoxy-7-
(piperidin-4-ylmethoxy)-
157 quinolin-4-yloxy] -phenyl }-N'-
(15 -phenyl-2-ρ-tolyl-ethyl)- oxal amide
N-[2-(4-Chloro-ρhenyl)- ethyl]-N'-{3-fluoro-4-[6-
158 methoxy-7-(piperidin-4- ylmethoxy)-quinolin-4- yloxy] -phenyl }-oxal amide
Figure imgf000337_0001
Table 1
Figure imgf000338_0001
Table 1
Entry Name Structure
N-(l ,2-Diphenyl-ethyl)-N'- { 3- fluoro-4- [6-methoxy-7-
163 (piperidin-4-ylmethoxy)- quinolin-4-yloxy] -phenyl } - oxalamide
N- [2-(2,4-Dichloro-phenyl)- ethyl]-N'-{3-fluoro-4-[6-
164 methoxy-7-(piperidin-4- ylmethoxy)-quinolin-4- yloxy] -phenyl } -oxalamide
N-[2-(3,4-Dimethoxy- ρhenyl)-ethyl]-N'-{ 3-fluoro-4-
165 [6-methoxy-7-(piperidin-4- ylmethoxy)-quinolin-4- yloxy] -phenyl } -oxalamide
N-[2-(4-Ethyl-phenyl)-ethyl]- N'-{3-fluoro-4-[6-methoxy-7-
166 (piperidin-4-ylmethoxy)- quinolin-4-yloxy] -phenyl } - oxalamide
Figure imgf000339_0001
Table 1
Entry Name Structure
N-[2-(4-Ethoxy-phenyl)- ethyl]-N'-{3-fluoro-4-[6-
167 methoxy-7-(piperidin-4- ylmethoxy)-quinolin-4- yloxy] -phenyl } -oxalamide
N-[2-(4-Ethoxy-3-methoxy- phenyl)-ethyl]-N'-{ 3-fluoro-4-
168 [6-methoxy-7-(piperidin-4- ylmethoxy)-quinolin-4- yloxy] -phenyl } -oxalamide
N-{ 3-Fluoro-4-[6-methoxy-7-
(piperidin-4-ylmethoxy)-
169 quinolin-4-yloxy] -phenyl } -N'-
[2-(4-phenoxy-phenyl)-ethyl]- oxalamide
Figure imgf000340_0001
Table 1
Entry Name Structure
N-[2-(3-Ethoxy-4-methoxy- phenyl)-ethyl]-N'-{ 3-fluoro-4-
170 [6-methoxy-7-(piperidin-4- ylmethoxy)-quinolin-4- yloxy] -phenyl } -oxalamide
N-{3-Fluoro-4-[6-methoxy-7-
(piperidin-4-ylmethoxy)-
171 quinolin-4-yloxy] -phenyl } -N'-
(2-ρyridin-2-yl-ethyl)- oxalamide
N-{ 3-Fluoro-4-[6-methoxy-7-
(piperidin-4-ylmethoxy)-
172 quinolin-4-yloxy] -phenyl }-N'-
(2-pyridin-4-yl-ethyl)- oxalamide
N-{3-Fluoro-4-[6-methoxy-7-
(piperidin-4-ylmethoxy)-
173 quinolin-4-yloxy]-phenyl } -N'-
[2-(4-fluoro-phenyl)-ethyl]- oxalamide
Figure imgf000341_0001
Table 1
Figure imgf000342_0001
Table 1
Entry Name Structure
N-{ 3-Fluoro-4-[6-methoxy-7-
( 1 -methyl-piperidin-4-
178 ylmethoxy)-quinolin-4- yloxy] -phenyl } -N'-isobutyl- oxalamide
N- { 3-Fluoro-4- [6-methoxy-7- ( 1 -methyl-piperidin-4-
179 ylmethoxy)-quinolin-4- yloxy]-phenyl}-N'-(3-methyl- butyl)-oxalamide
N-{3-Fluoro-4-[6-methoxy-7-
( 1 -methyl-piperidin-4-
180 ylmethoxy)-quinolin-4- yloxy]-phenyl}-N'-(2R- phenyl-propyl)-oxalarnide
N-{3-Fluoro-4-[6-methoxy-7- ( 1 -methyl-piperidin-4-
181 ylmethoxy)-quinolin-4- yloxy]-phenyl}-N'-(2-phenyl- propyl)-oxalamide
Figure imgf000343_0001
Table 1
Figure imgf000344_0001
Table 1
Entry Name Structure
N-[2-(2,6-Dichloro-phenyl)- ethyl]-N'-{3-fluoro-4-[6-
186 methoxy-7-(piperidin-4- ylmethoxy)-quinolin-4- yloxy] -phenyl } -oxalamide
N-[2-(2,4-Dichloro-phenyl)- ethyl]-N*-{3-fluoro-4-[6-
187 methoxy-7-(piperidin-4- ylmethoxy)-quinolin-4- yloxy] -phenyl } -oxalamide
N-(2-Benzo[l,3]dioxol-5-yl- ethyl)-N*-{3-fluoro-4-[6-
188 methoxy-7-(piperidin-4- ylmethoxy)-quinolin-4- yloxy] -phenyl } -oxalamide
N-[2-(3-Bromo-4-methoxy- phenyl)-ethyl]-N'-{ 3-fluoro-4-
189 [6-methoxy-7-(piperidin-4- ylmethoxy)-quinolin-4- yloxy] -phenyl } -oxalamide
Figure imgf000345_0001
Table 1
Figure imgf000346_0001
Table 1
Entry Name Structure
N-[2-(3-Ethoxy-ρhenyl)- ethyl]-N'-{3-fluoro-4-[6-
193 methoxy-7-(piperidin-4- ylmethoxy)-quinolin-4- yloxy] -phenyl } -oxalamide
N-[2-(3,4-Dimethyl-phenyl)- ethyl]-N'-{3-fluoro-4-[6-
194 methoxy-7-(piperidin-4- ylmethoxy)-quinolin-4- yloxy] -phenyl } -oxalamide
N-[2-(2,5-Dimethyl-phenyl)- ethyl]-N'-{3-fluoro-4-[6-
195 methoxy-7-(piperidin-4- ylmethoxy)-quinolin-4- yloxy] -phenyl } -oxalamide
N-[2-(3-Chloro-4-propoxy- phenyl)-ethyl] -N'- { 3-fluoro-4-
196 [6-methoxy-7-(piperidin-4- ylmethoxy)-quinolin-4- yloxy] -phenyl } -oxalamide
Figure imgf000347_0001
Table 1
Entry Name Structure
N-[2-(4-Butoxy-3-chloro- phenyl)-ethyl]-N'-{3-fluoro-4-
197 [6-methoxy-7-(piperidin-4- ylmethoxy)-quinolin-4- yloxy] -phenyl } -oxalamide
N-[2-(4-tert-Butyl-phenyl ethyl]-N'-{3-fluoro-4-[6-
198 methoxy-7-(piperidin-4- ylmethoxy)-quinolin-4- yloxy] -phenyl } -oxalamide
N-{3-Fluoro-4-[6-methoxy-7- (piperidin-4-ylmethoxy)-
199 quinolin-4-yloxy] -phenyl } -N'-
[2-(4-sulfamoyl-phenyl)- ethyl] -oxalamide
Figure imgf000348_0001
Table 1
Entry Name Structure
N-{ 3-Fluoro-4-[6-methoxy-7- (piperidin-4-ylmethoxy)-
200 quinolin-4-yloxy] -phenyl }-N'- [2-(4-hydroxy-3 -methoxy- phenyl)-ethyl] -oxalamide
N- { 3-Fluoro-4- [6-methoxy-7- (piperidin-4-ylmethoxy)-
201 quinolin-4-yloxy] -phenyl } -N'- [2-(3-hydroxy-4-methoxy- phenyl)-ethyl]-oxalamide
N-(2,4-Dichloro-benzyl)-N'-
{ 3-fluoro-4-[6-methoxy-7-
202 (piperidin-4-ylmethoxy)- quinolin-4-yloxy] -phenyl } - oxalamide
Table 1
Entry Name Structure
N-{3-Fluoro-4-[6-methoxy-7-
(piperidin-4-ylmethoxy)-
203 quinolin-4-yloxy] -phenyl }-N'-
(4-fluoro-2-trifluoromethyl- benzyl)-oxalamide
N-{3-Fluoro-4-[6-methoxy-7-
204 (piperidin-4-ylmethoxy)- quinolin-4-yloxy] -phenyl }-N'-
( 1 -p-tolyl-ethyl)-oxalamide
N-{3-Fluoro-4-[6-methoxy-7-
(piperidin-4-ylmethoxy)-
205 quinolin-4-yloxy] -phenyl }-N'-
(3-fluoro-4-trifluoromethyl- benzyl)-oxalamide
Figure imgf000350_0001
Table 1
Entry Name Structure
N-(3-Chloro-4-fluoro-benzyl)-
N'-{3-fluoro-4-[6-methoxy-7-
206 (piρeridin-4-ylmethoxy)- quinolin-4-yloxy] -phenyl } - oxalamide
N- { 3-Fluoro-4- [6-methoxy-7-
(piperidin-4-ylmethoxy)-
207 quinolin-4-yloxy] -phenyl } -N'-
[ 1 -(3-methoxy-phenyl)-ethyl] - oxalamide
N-{3-Fluoro-4-[6-methoxy-7- (piperidin-4-ylmethoxy)-
208 quinolin-4-yloxy] -phenyl }-N'-
( 1 -naphthalen-2-yl-ethyl)- oxalamide
Figure imgf000351_0001
Table 1
Figure imgf000352_0001
Table 1
Figure imgf000353_0001
Table 1
Figure imgf000354_0001
Table 1
Entry Name Structure
N-(2-Fluoro-benzyl)-N'-{3- fluoro-4- [6-methoxy-7-
220 (piperidin-4-ylmethoxy)- quinolin-4-yloxy] -phenyl } - oxalamide
N-[2-(3,4-Dichloro-ρhenyl)- ethyl]-N'-{ 3 -fluoro-4- [6-
221 methoxy-7-(piperidin-4- ylmethoxy)-quinolin-4- yloxy] -phenyl } -oxalamide
N-(4-Fluoro-benzyl)-N'-{3- fluoro-4- [6-methoxy-7-
222 (piperidin-4-ylmethoxy)- quinolin-4-yloxy] -phenyl } - oxalamide
N-(2,3-Difluoro-benzyl)-N'-
{ 3-fluoro-4-[6-methoxy-7-
223 (piperidin-4-ylmethoxy)- quinolin-4-yloxy] -phenyl } - oxalamide
Figure imgf000355_0001
Table 1
Entry Name Structure
N-{ 3-Fluoro-4-[6-methoxy-7-
224 (piperidin-4-ylmethoxy)- quinolin-4-yloxy] -phenyl } -N1-
(2-phenoxy-ethyl)-oxalamide
N-(2,2-Diphenyl-ethyl)-N {3- fluoro-4- [6-methoxy-7-
225 (piperidin-4-ylmethoxy)- quinolin-4-yloxy] -phenyl } - oxalamide
N-{ 3-Fluoro-4-[6-methoxy-7-
(piperidin-4-ylmethoxy)-
226 quinolin-4-yloxy] -phenyl }-N'-
[2-(4-methoxy-phenyl)-ethyl]- oxalamide
N-{3-Fluoro-4-[6-methoxy-7-
(piperidin-4-ylmethoxy)-
227 quinolin-4-yloxy] -phenyl } -N'-
(2-phenyl-propyl)-oxalamide
Figure imgf000356_0001
Table 1
Entry Name Structure
N-[2-(4-Bromo-phenyl)- ethyl]-N'-{ 3-fluoro-4-[6-
228 methoxy-7-(piperidin-4- ylmethoxy)-quinolin-4- yloxy] -phenyl } -oxalamide
N-{4-[7-(l-Ethyl-piperidin-4- ylmethoxy)-6-methoxy-
229 quinolin-4-yloxy] -3 -fluoro- phenyl } -2-oxo-2-(2-phenyl- morpholin-4-yl)-acetamide
N-{3-Fluoro-4-[6-methoxy-7-
(piperidin-4-ylmethoxy)-
230 quinolin-4-yloxy] -phenyl } -N'-
(3-fluoro-5-trifluoromethyl- benzyl)-oxalamide
N-(3 ,5-Difluoro-benzyl)-N'-
{ 3 -fluoro-4- [6-methoxy-7-
231 (piperidin-4-ylmethoxy)- quinolin-4-yloxy] -phenyl } - oxalamide
Figure imgf000357_0001
Table 1
Entry Name Structure
N-(2-Chloro-5- trifluoromethyl-benzyl)-N'-
{ 3-fluoro-4-[6-methoxy-7-
232 (piperidin-4-ylmethoxy)- quinolin-4-yloxy] -phenyl } - oxalamide
N-[4-(6,7-Dimethoxy- quinolin-4-yloxy)-3 -fluoro-
233 phenyl]-N'-(2-dimethylamino-
2-phenyl-ethyl)-oxalamide
N-{ 3-Fluoro-4-[6-methoxy-7-
(piperidin-4-ylmethoxy)-
234 quinolin-4-yloxy] -phenyl } -N'-
(4-methoxy-benzyl)- oxalamide
Figure imgf000358_0001
Table 1
Entry Name Structure
N-{ 3-Fluoro-4-[6-methoxy-7-
(piperidin-4-ylmethoxy)-
235 quinolin-4-yloxy] -phenyl } -N'-
(4-trifluoromethyl-benzyl)- oxalamide
N-{ 3-Fluoro-4-[6-methoxy-7-
(piperidin-4-ylmethoxy)-
236 quinolin-4-yloxy] -phenyl }-N'-
(3-methoxy-benzyl)- oxalamide
N- { 3-Fluoro-4-[6-methoxy-7-
(piperidin-4-ylmethoxy)-
237 quinolin-4-yloxy] -phenyl } -N'-
(3 -trifluoromethyl-benzyl)- oxalamide
N-{ 3-Fluoro-4-[6-methoxy-7-
(piperidin-4-ylmethoxy)-
238 quinolin-4-yloxy] -phenyl }-N'-
(3-trifluoromethoxy-benzyl)- oxalamide
Figure imgf000359_0001
Table 1
Entry Name Structure
N- { 3 -Fluoro-4- [6-methoxy-7-
(piperidin-4-ylmethoxy)-
239 quinolin-4-yloxy] -phenyl }-N'-
(2-methoxy-benzyl)- oxalamide
N- { 3-Fluoro-4- [6-methoxy-7-
(piperidin-4-ylmethoxy)-
240 quinolin-4-yloxy] -phenyl } -N'-
(2-trifluoromethyl-benzyl)- oxalamide
N-(3-Chloro-benzyl)-N'- { 3- fluoro-4- [6-methoxy-7-
241 (piperidin-4-ylmethoxy)- quinolin-4-yloxy] -phenyl } - oxalamide
N-{ 3-Fluoro-4-[6-methoxy-7-
(piperidin-4-ylmethoxy)-
242 quinolin-4-yloxy] -phenyl } -N'-
(2-trifluoromethoxy-benzyl)- oxalamide
Figure imgf000360_0001
Table 1
Entry Name Structure
N-(2-Chloro-benzyl)-N'-{3- fluoro-4-[6-methoxy-7-
243 (piperidin-4-ylmethoxy)- quinolin-4-yloxy] -phenyl } - oxalamide
N-{ 3-Fluoro-4-[6-methoxy-7-
(piperidin-4-ylmethoxy)-
244 quinolin-4-yloxy] -phenyl } -N'-
(4-trifluoromethoxy-benzyl)- oxalamide
N- { 3 -Fluoro-4- [6-methoxy-7-
( 1 -methyl-piperidin-4-
245 ylmethoxy)-quinolin-4- yloxy]-phenyl }-N'-(4- methoxy-benzyl)-oxalamide
Figure imgf000361_0001
Table 1
Figure imgf000362_0001
Table 1
Entry Name Structure
N-[5-Chloro-6-(6,7-
250 dimethoxy-quinolin-4-yloxy)- pyridin-3-yl]-N'-(2,4-difluoro- phenyl)-malonamide
N-[5-Chloro-6-(6,7- dimethoxy-quinolin-4-yloxy)-
251 pyridin-3-yι]-N'-(4-fluoro- phenyl)-N'-methyl- malonamide
N- { 3 -Fluoro-4- [6-methoxy-7-
(piperidin-4-ylmethoxy)-
252 quinolin-4-yloxy] -phenyl }-N'-
(lR-phenyl-propyl)- oxalamide
Figure imgf000363_0001
Table 1
Figure imgf000364_0001
Table 1
Figure imgf000365_0001
Table 1
Figure imgf000366_0001
Table 1
Entry Name Structure
N-{5-Chloro-6-[7-(3- diethylamino-propoxy)-6-
265 methoxy-quinolin-4-yloxy] - pyridin-3-yl}-N'-(4-fluoro- phenyl)-malonamide
N-(4-Chloro-benzyl)-N'- { 3- fluoro-4-[6-methoxy-7-
266 (piperidin-4-ylmethoxy)- quinolin-4-yloxy] -phenyl } - oxalamide
N-(3 ,5-Dimethoxy-benzyl)-
N'- { 3-fluoro-4-[6-methoxy-7-
267 (piperidin-4-ylmethoxy)- quinolin-4-yloxy] -phenyl }- oxalamide
Figure imgf000367_0001
Table 1
Figure imgf000368_0001
Table 1
Figure imgf000369_0001
Table 1
Entry Name Structure
N-{ 3-Fluoro-4-[6-methoxy-7-
(piperidin-4-ylmethoxy)-
275 quinolin-4-yloxy] -phenyl } -N'-
(2-fluoro-3 -trifluoromethyl- benzyl)-oxalamide
N- [2-(2-Bromo-6-methoxy- phenyl)-ethyl]-N'-{ 3-fluoro-4-
276 [6-methoxy-7-(piperidin-4- ylmethoxy)-quinolin-4- yloxy] -phenyl } -oxalamide
N-[2-(3,4-Dimethoxy- phenyl)-ethyl]-N'-{3-fluoro-4-
[6-methoxy-7-(piperidin-4-
277 ylmethoxy)-quinolin-4- yloxy] -phenyl } -N-methyl- oxalamide
N-[2-(5-Bromo-2-methoxy- ρhenyl)-ethyl]-N'-{3-fluoro-4-
278 [6-methoxy-7-(piperidin-4- ylmethoxy)-quinolin-4- yloxy] -phenyl } -oxalamide
Figure imgf000370_0001
Table 1
Entry Name Structure
N- { 3 -Fluoro-4- [6-methoxy-7-
(piperidin-4-ylmethoxy)-
279 quinolin-4-yloxy] -phenyl }-N'-
(2-fluoro-5 -trifluoromethyl- benzyl)-oxalamide
N-{3-Fluoro-4-[6-methoxy-7-
(piperidin-4-ylmethoxy)-
280 quinolin-4-yloxy] -phenyl } -N'-
[ 1 -(4-fluoro-phenyl)-ethyl] - oxalamide
N-(lS-Benzyl-2-oxo-2- pyrrolidin- 1 -yl-ethyl)-N'- { 3 -
281 fluoro-4-[6-methoxy-7-
(piperidin-4-ylmethoxy)- quinolin-4-yloxy] -phenyl } - oxalamide
N-{3-Fluoro-4-[6-methoxy-7- (octahydro-
282 cyclopenta[c]pyrrol-5- ylmethoxy)-quinazolin-4- yloxy] -phenyl } -N'-phenethyl- oxalamide
Figure imgf000371_0001
Table 1
Entry Name Structure
N-[2-(4-Amino-phenyl)- ethyl]-N'-{3-fluoro-4-[6-
283 methoxy-7-(piperidin-4- ylmethoxy)-quinolin-4- yloxy] -phenyl } -oxalamide
2-(4-Benzyl-piperidin- 1 -yl)- N-{ 3-fluoro-4-[6-methoxy-7-
284 (piperidin-4-ylmethoxy)- quinolin-4-yloxy] -phenyl } -2- oxo-acetamide
N-[4-(6,7-Dimethoxy-
285 quinolin-4-yloxy)-phenyl] -N'- (4-fluoro-phenyl)-malonamide
Figure imgf000372_0001
Table 1
Figure imgf000373_0001
Table 1
Figure imgf000374_0001
Table 1
Entry Name Structure
N-{3-Fluoro-4-[6-methoxy-7-
(piperidin-4-ylmethoxy)-
295 quinolin-4-yloxy] -phenyl } -2- oxo-2-pyrrolidin- 1 -yl- acetamide
N-Ethyl-N'-{ 3-fluoro-4-[6- methoxy-7-(piperidin-4-
296 ylmethoxy)-quinolin-4- yloxy] -phenyl } -N-methyl- oxalamide
Figure imgf000375_0001
46. A compound for modulating kinase activity of formula A-B-C, or a pharmaceutically acceptable salt, hydrate, or prodrug thereof, wherein, A is selected from:
Figure imgf000375_0002
Figure imgf000376_0001
B is selected from:
Figure imgf000376_0002
and, C is selected from:
Figure imgf000376_0003
Figure imgf000377_0001
wherein R2 is selected from -H, halogen, trihalomethyl, -CN, -NH2, -NO2, -OR3, -NR3R3,
-S(O)0-2R", -SO2NR >3Trl3J, -CO2RJ, -C(O)NR 3^r>3, -N(R SO2R", -N(RJ)C(O)RJ, -N(R3)CO2R3, -C(O)R3, and optionally substituted lower alkyl; q is 0 to 2; each R3 is independently selected from -H, optionally substituted lower alkyl, optionally substituted aryl, optionally substituted arylalkyl, and optionally substituted heteroarylalkyl; two R3, together with the nitrogen to which they are attached, form a four- to seven- membered heteroalicyclic, said four- to seven-membered heteroalicyclic optionally containing one additional heteroatom; when one said additional heteroatom is a nitrogen, then said nitrogen is optionally substituted with a group selected from -H, trihalomethyl, -SO2R5, -SO2NR5R5, -CO2R5, -C(O)NR5R5, -C(O)R5, and optionally substituted lower alkyl; each R35 is independently selected from -H, -C(=O)R3, -C(=O)OR3, -C(=O)SR3, -SO2R3, -C(=O)N(R3)R3, and optionally substituted lower alkyl; two R , together with the nitrogen to which they are attached, can combine to form a heteroalicyclic optionally substituted with between one and four of R60, said heteroalicyclic may have an additional annular heteroatom, and said heteroalicyclic may have an aryl fused thereto, said aryl optionally substituted with an additional one to four of R60;
A1 is selected from =N-, =C(H)-, and =C(CN)-;
A2 is either =N- or =C(H)-;
R5 is -H or optionally substituted lower alkyl;
R8 is selected from R3, -SO2NR3R3, -CO2R3, -C(O)NR3R3, -SO2R3, and -C(O)R3;
R9, R10, and R11 are each independently selected from -H, and -OR12; or
R9 is selected from -H, and -OR12, and R10 and R11, when taken together, are either an optionally substituted alkylidene or an oxo; and
R12 is selected from -H, -C(O)R3, optionally substituted lower alkylidyne, optionally substituted lower arylalkylidyne, optionally substituted lower heterocyclylalkylidyne, optionally substituted lower alkylidene, optionally substituted lower alkylidenearyl, optionally substituted lower alkylideneheterocyclyl, optionally substituted lower alkyl, optionally substituted lower alkylaryl, optionally substituted aryl, optionally substituted lower heterocyclylalkyl, and optionally substituted heterocyclyl; or two R12's, when taken together, form 1) a corresponding spirocyclic ketal when said two R12's stem from R10 and R11, or 2) a corresponding cyclic ketal when said two R12's stem from R9 and one of R10 and R11;
E1 is selected from -O-, -CH2-, -N(R5)-, and -S(O)0-2-;
Q is a five- to ten-membered ring system, optionally substituted with between zero and four of R20;
R20 is selected from -H, halogen, trihalomethyl, -CN, -NO2, -NH2, -OR3, -NR3R3, -S(O)o-2R3, -SO2NR3R3, -CO2R3, -C(O)NR3R3, -N(R3)SO2R3, -N(R3)C(O)R3, -N(R3)CO2R3, -C(O)R3, and optionally substituted lower alkyl;
R60 is selected from -H, halogen, trihalomethyl, -CN, -NO2, -NH2, -OR3, -NR3R3, -S(O)0-2R3, -SO2NR3R3, -CO2R3, -C(O)NR3R3, -N(R3)SO2R3, -N(R3)C(O)R3, -N(R3)CO2R3, -C(O)R3, optionally substituted lower alkyl, optionally substituted aryl, optionally substituted heteroarylalkyl, and optionally substituted arylalkyl; two of R60, when attached to a non-aromatic carbon, can be oxo; each methylene in any of the above formulae is independently optionally substituted with R25; each R is independently selected from halogen, trihalomethyl, -CN, -NO2, -NH2, -OR , -NR3R3, -S(O)o-2R3, -SO2NR3R3, -CO2R3, -C(O)NR3R3, -N(R3)SO2R3, -N(R3)C(O)R3, -N(R3)CO2R3, -C(O)R3, optionally substituted aryl, optionally substituted arylalkyl, heteroarylalkyl, and optionally substituted lower alkyl; two of R25, together with the carbon or carbons to which they are attached, can combine to form a three- to seven- membered alicyclic or heteroalicyclic, two of R25 on a single carbon can be oxo; with the proviso that when B is selected from:
Figure imgf000379_0001
and C contains
Figure imgf000379_0002
and the remaining portion of C contains one of:
Figure imgf000379_0003
directly attached to
Figure imgf000380_0001
then A must be one of:
Figure imgf000380_0004
and with the proviso that when C contains
Figure imgf000380_0002
and B is selected from
Figure imgf000380_0003
then the portion of C directly attached to
Figure imgf000381_0001
cannot contain
Figure imgf000381_0002
when R 70 is selected rom _Hj-4alkyl, and C1-4alkoxyl.
47. The compound according to claim 46, wherein Q is selected from phenyl, napthyl, 1,2,3,4-tetrahydronaphthyl, indanyl, benzodioxanyl, benzofuranyl, phenazinyl, phenothiazinyl, phenoxazinyl, tetrahydroisoquinolyl, pyrrolyl, pyrazolyl, pyrazolidinyl, imidazolyl, imidazolinyl, imidazolidinyl, tettahydropyridinyl, pyridinyl, pyrazinyl, pyrimidinyl, pyridazinyl, oxazolyl, oxazolinyl, oxazolidinyl, triazolyl, isoxazolyl, isoxazolidinyl, thiazolyl, thiazolinyl, thiazolidinyl, isothiazolyl, isothiazolidinyl, indolyl, isoindolyl, indolinyl, isoindolinyl, octahydroindolyl, octahydroisoindolyl, quinolyl, isoquinolyl, benzimidazolyl, thiadiazolyl, benzopyranyl, benzothiazolyl, benzoxazolyl, furyl, thienyl, benzothieliyl, and oxadiazolyl; each optionally substituted with between one and four of R20; wherein each R20 is independently selected from -H, halogen, trihalomethyl, -CN, -NO2, -NH2, -OR3, -NR3R3, -CO2R3, -C(O)NR3R3, -N(R3)SO2R3, -N(R3)C(O)R3, -N(R3)CO2R3, -C(O)R3, and optionally substituted lower alkyl.
48. The compound according to claim 47, wherein B is either of the following:
Figure imgf000381_0003
wherein A1 is either =N- or =C(H)-.
49. The compound according to claim 48, wherein B is
Figure imgf000381_0004
50. The compound according to claim 49, wherein C is selected from:
Figure imgf000382_0001
Figure imgf000383_0001
wherein R2, R3, R5, R20, R25 and R60 are as defined above.
51. The compound according to claim 50, R2 is selected from halogen, trihalomethyl, -CN, -NO2, -OR3, -NR3R3, -CO2R3, -C(O)NR3R3, -N(R3)C(O)R3, -N(R3)CO2R3, -C(O)R3, and optionally substituted lower alkyl
52. The compound according to claim 51 , wherein R2 is halogen.
53. The compound according to claim 52, wherein R2 is either fluorine or chlorine.
54. A compound for modulating kinase activity according to Formula XI,
Figure imgf000383_0002
or a pharmaceutically acceptable salt, hydrate, or prodrug thereof, wherein, each R1 is independently selected from halogen, -OR3, -NO2, -NH2, -NR3R4, -D-R50 and optionally substituted C1-6alkyl;
R70 is selected from -H, halogen, -OR3, -S(O)0-2R3, -NO2, -NH2, -NR3R4, and optionally substituted C1-6alkyl;
Q is selected from =N-, =C(H)-, and =C(CN)-; Z is selected from -S(O)0-2-, -O-, and -NR5-;
Ar is either a five- or six-membered arylene or a five- or six-membered heteroarylene containing between one and three heteroatoms;
G is either an optionally substituted cycloalkyl or an optionally substituted heteroalicyclic; each R2 is independently selected from halogen, trihalomethyl, -CN, -NO2, -NH2, -OR3, -NR3R4, -S(O)0-2R3, -SO2NR3R3, -CO2R3, -C(O)NR3R3, -N(R3)SO2R3, -N(R3)C(O)R3, -N(R3)CO2R3, -C(O)R3, and optionally substituted C1-6alkyl; each R3 is independently -H or R4; each R4 is independently selected from optionally substituted C1-6alkyl, optionally substituted aryl, optionally substituted aryl C1-6alkyl, optionally substituted heterocyclyl, and optionally substituted heterocyclyl C1-6alkyl; or
R3 and R4, when taken together with a common nitrogen to which they are attached, form an optionally substituted five- to seven-membered heterocyclyl, said optionally substituted five- to seven-membered heterocyclyl optionally containing at least one additional annular heteroatom selected from N, O, S, and P; ,
R5 is -H or optionally substituted C1-6alkyl; each D is independently selected from -O-, -S(O)0-2-, and -NR5-; each R50 is independently either R3, or according to formula XII;
Figure imgf000384_0001
xπ wherein X1, X2, and optionally X3, represent the atoms of a saturated bridged ring system, said saturated bridged ring system comprising up to four annular heteroatoms represented by any of X1, X2, and X3; wherein, each X1 is independently selected from -C(R6)R7-, -O-, -S(O)0-2-, and -NR8-; each X2 is independently an optionally substituted bridgehead methine or a bridgehead nitrogen; each X3 is independently selected from -C(R6)R7-, -O-, -S(O)0-2-, and -NR8-;
Y is either: an optionally substituted lower alkylene linker, between D and either 1) any annular atom of the saturated bridged ring system, except X2 when X2 is a bridgehead nitrogen, or 2) any heteroatom, represented by any of R6 or R7; provided there are at least two carbon atoms between D and any annular heteroatom of the saturated bridged ring system or any heteroatom represented by any of R6 or R7; or Y is absent, when Y is absent, said saturated bridged ring system, is directly attached to D via an annular carbon of said saturated bridged ring system, unless D is -SO2-, in which case said saturated bridged ring system, is directly attached to D via an any annular atom of said saturated bridged ring system; m and p are each independently one to four; n is zero to two, when n equals zero, then there is a single bond between the two bridgehead X2 's;
R6 and R7 are each independently selected from -H, halogen, trihalomethyl, -CN, -NH , -NO2, -OR3, -NR3R4, -S(O)0-2R4, -SO2NR3R4, -CO2R3, -C(O)NR3R4, -N(R3)SO2R4, -N(R3)C(O)R3, -NCO2R3, -C(O)R3, optionally substituted C1-6alkyl, optionally substituted aryl, optionally substituted aryl C1-6alkyl, optionally substituted heterocyclyl, optionally substituted heterocyclyl C1-6alkyl, and a bond to either Y or D; or
R and R , when taken together are oxo; or
R6 and R7, when taken together with a common carbon to which they are attached, form a optionally substituted three- to seven-membered spirocyclyl, said optionally substituted three- to seven-membered spirocyclyl optionally containing at least one additional annular heteroatom selected from N, O, S, and P;
R8 is selected from -R3, Y, -SO2NR3R4, -CO2R4, -C(O)NR3R3, -SO2R4, and -C(O)R3; and each R30 is independently selected from halogen, trihalomethyl, -CN, -NO2, -NH2, -OR3, -NR3R4, -S(O)0-2R3, -SO2NR3R3, -CO2R3, -C(O)NR3R3, -N(R3)SO2R3, -N(R3)C(O)R3, -N(R3)CO2R3, -C(O)R3, and optionally substituted C1-6alkyl.
55. The compound according to claim 54, wherein Z is either -O- or -NR5-.
56. The compound according to claim 55, wherein at least one of R1 is -D-R50.
57. The compound according to claim 56, wherein D is -O- and at least one other R1 is -OR3.
58. The compound according to claim 57, of formula XHIa or XHIb:
Figure imgf000386_0001
XHIa Xlllb wherein Q1 is either =N- or =C(H)-
59. The compound according to claim 58, wherein R 150 is selected from C1-6alkyl optionally substituted with at least one of optionally substituted amino, optionally substituted C1-6alkyl amino, optionally substituted C1-6dialkyl amino, optionally substituted heteroalicylic, and a group of formula XII.
60. The compound according to claim 59, wherein R3a is C1-6alkyl.
61. The compound according to claim 60, wherein Z is -O-.
62. The compound according to claim 61, wherein G is selected from cyclopropyl, aziradine, cyclobutyl, and azetidine, each optionally substituted with between zero and four of R30.
63. The compound according to claim 62, wherein Q is either =N- or =C(H)-.
64. The compound according to claim 63, wherein R is selected from -H, halogen, C . β alkyl and perfluoro Cι-6 alkyl.
65. The compound according to claim 64, wherein -N(R3b)R4 is selected from the following:
Figure imgf000387_0001
wherein J, is a five- to ten-membered ring, optionally substituted with between zero and five of R20; each R is independently selected from -H, halogen, trihalomethyl, -CN, -NO2, -NH2, -OR3, -NR3R4, -S(O)o-2R3, -SO2NR3R3, -CO2R3, -C(O)NR3R3, -N(R3)SO2R3, -N(R3)C(O)R3, -N(R3)CO2R3, -C(O)R3, optionally substituted Cι-6alkyl, optionally substituted aryl, optionally substituted aryl C1-6alkyl, optionally substituted heterocyclyl, and optionally substituted heterocyclyl Cι-6alkyl; two of R20, together with the atom or atoms to which they are attached, combine to form an optionally substituted three- to seven-membered heteroalicyclic, said optionally substituted three- to seven-membered heteroalicyclic either spiro- to J or fused to J;
E is selected from -O-, -N(R5)-, -CH2-, and -S(O)0-2-; each R60 is independently selected from halogen, trihalomethyl, -CN, -NO2, -NH2, -OR3, -NR3R4, -S(O)0-2R3, -SO2NR3R3, -CO2R3, -C(O)NR3R3, -N(R3)SO2R3, -N(R3)C(O)R3, -N(R3)CO2R3, -C(O)R3, optionally substituted C1-6alkyl, optionally substituted aryl, optionally substituted heteroaryl C1-6alkyl, and optionally substituted aryl C1-6alkyl; each methylene in any of the above formulae, other than those in a depicted ring, is independently optionally substituted with R25; and
R25 is selected from halogen, trihalomethyl, oxo, -CN, -NO2, -NH2, -OR3, -NR3R4, -S(O)0-2R3, -SO2NR3R3, -CO2R3, -C(O)NR3R3, -N(R3)SO2R3, -N(R3)C(O)R3, -N(R3)CO2R3, -C(O)R3, optionally substituted aryl, optionally substituted aryl Cι-6alkyl, heteroaryl C1-6alkyl, and optionally substituted C1-6alkyl; or two of R , together with the carbon or carbons to which they are attached, can combine to form a three- to seven-membered alicyclic or heteroalicyclic;
R is equivalent to R as defined above; and
R4 and R5 are as defined above.
66. . The compound according to claim 65, of formula XlVa or XlVb:
Figure imgf000388_0001
XlVa XlVb
67. The compound according to claim 66, wherein R50 is C1-6alkyl optionally substituted with a group selected from optionally substituted amino, an optionally substituted alkylamino, optionally substituted dialkylamino, and optionally substituted heteroalicylic.
68. The compound according to claim 67, wherein the heteroalicyclic portion of said optionally substituted heteroalicyclic of R50 is selected from the group consisting of piperidine, piperazine, morpholine, thiomorpholine, thiomorpholine 1 -oxide, thiomorpholine 1,1 -dioxide, 2-oxo-morpholine, pyrrolidine, and azepine.
69. The compound according to claim 67, wherein R50 is according to formula XII.
70. The compound according to claim 69, wherein the saturated bridged ring system according to formula XII has a geometry selected from the group consisting of [4.4.0], [4.3.0], [4.2.0], [4.1.0], [3.3.0], [3.2.0], [3.1.0], [3.3.3], [3.3.2], [3.3.1], [3.2.2], [3.2.1], [2.2.2], and [2.2.1].
71. The compound according to claim 70, wherein Y is selected from -CH2CH2CH2CH2-, -CH2CH2CH2-, -CH2CH2-, -CH2-, and absent.
72. The compound according to claim 71, wherein n is 0 and the saturated bridged ring system according to formula XII has a geometry selected from the group consisting of [4.4.0], [4.3.0], [4.2.0], [4.1.0], [3.3.0], [3.2.0], and [3.1.0].
73. The compound according to claim 72, wherein said saturated bridged ring system contains at least one annular nitrogen or at least one annular oxygen.
74. The compound according to claim 73, wherein said saturated bridged ring system contains --NNRR88--,, wwhheerreeiinn RR88 iiss sseelleeccted from -H, optionally substituted Chalky!, -CO2R3, -C(O)NR3R3, -SO2R3, and -C(O)R3.
75. The compound according to claim 73, wherein said saturated bridged ring system is of formula XV,
Figure imgf000389_0001
xv wherein U1 is selected from -O-, -S(O)0-2-, -NR8-, -CR6R7-, and absent; and e is 0 or 1.
76. The compound according to claim 75, wherein Y is -CH2-.
77. The compound according to claim 76, wherein U1 is -NR8-, wherein R8 is selected from -H, optionally substituted lower alkyl, -CO2R3, -C(O)NR3R3, -SO2R3, and -C(O)R3.
78. The compound according to claim 76, wherein U1 is -O-.
79. The compound according to claim 76, wherein U1 is absent.
80. The compound according to claim 71, wherein Y is selected from -CH2CH2-, -CH2-, and absent.
81. The compound according to claim 80, wherein said saturated bridged ring system is of formula XVI,
Figure imgf000390_0001
XVI wherein R9, R10, and R11 are each independently selected from -H, and -OR12; or
R9 is selected from -H, and -OR12, and R10 and R11, when taken together, are either an optionally substituted alkylidene or an oxo;
R is selected from -H, -C(O)R , optionally substituted lower alkylidyne, optionally substituted lower arylalkylidyne, optionally substituted lower heterocyclylalkylidyne, optionally substituted lower alkylidene, optionally substituted lower alkylidenearyl, optionally substituted lower alkylideneheterocyclyl, optionally substituted lower alkyl, optionally substituted lower alkylaryl, optionally substituted aryl, optionally substituted lower heterocyclylalkyl, and optionally substituted heterocyclyl;
19 or two R 's, when taken together, form 1) a corresponding spirocyclic ketal when said two R12's stem from R10 and R11, or 2) a corresponding cyclic ketal when said two R12's stem from R9 and one of R10 and R11.
82. The compound according to claim 81, wherein one of R10 and R11 is -OR12, wherein R12 is selected from -H, -C(O)R3, and optionally substituted lower alkyl; and R9 and the other of R10 and R11 are both -H.
83. The compound according to claim 82, wherein Y is either -CH2- or absent.
84. The compound according to claim 81, wherein R9 is an alkyl group containing at least one fluorine substitution thereon.
85. The compound according to claim 74, wherein said saturated bridged ring system is of formula XVII.
R8-CD-?
XVII
86. The compound according to claim 85, wherein Y is either -CH2- or absent.
87. The compound according to claim 86, wherein R8 is methyl or ethyl.
88. The compound according to claim 87, wherein at least one of R2 is halogen.
89. The compound according to claim 74, wherein said saturated bridged ring system is of formula XVIII.
Figure imgf000391_0001
XVIII
90. The compound according to claim 89, wherein Y is -CH2-.
91. The compound according to claim 90, wherein R is methyl or ethyl.
92. The compound according to claim 73, wherein said saturated bridged ring system is of formula XIX
Figure imgf000391_0002
XIX wherein U2 is selected from -O-, -S(O)0-2-, -NR8-, -CR6R7-, and absent.
93. The compound according to claim 92, wherein R3 of formula XIX is selected from -H and optionally substituted alkyl.
94. The compound according to claim 93, wherein U is either -CR R - or absent.
95. The compound according to claim 94, wherein U2 is either -CH2- or absent.
96. The compound according to claim 95, wherein Y is -CH2-.
97. The compound according to claim 74, wherein said saturated bridged ring system is according to formula XX.
Figure imgf000391_0003
XX
98. The compound according to claim 97, wherein R8 is methyl or ethyl.
99. The compound according to any of claims 67 through 98, wherein R2 is selected from C1-6 alkyl, perfluoro Cι-6 alkyl, and halogen.
100. The compound according to claim 99, wherein R2 is selected from perfluoro C1-3 alkyl and halogen.
101. The compound according to any of claims 67 through 98, wherein R20 is selected from halogen, -CN, -NO2, -NH2, -OR3, -NR3R4, -N(R3)SO2R3, -N(R3)C(O)R3, -N(R3)CO2R3, optionally substituted heterocyclyl, and optionally substituted heterocyclyl C1-6alkyl, and (two of R20) together with the atom or atoms to which they are attached, an optionally substituted three- to six-membered heteroalicyclic, said optionally substituted three- to six-membered heteroalicyclic fused to the phenyl as in XlVa or XlVb.
102. The compound according to claim 101, wherein R is selected from halogen, -NR3R4, optionally substituted heterocyclyl, and optionally substituted heterocyclyl Q. 6alkyl, and (two of R20) together with the atom or atoms to which they are attached, an optionally substituted five- to six-membered heteroalicyclic, said optionally substituted five- to six-membered heteroalicyclic fused to the phenyl as in XlVa or XlVb.
103. The compound according to claim 102, wherein R is selected from C1-6 alkyl, perfluoro C1-6 alkyl, and halogen.
104. The compound according to claim 103, wherein R is selected from perfluoro C1-3 alkyl and halogen.
105. The compound according to claim 54, selected from Table 2.
Table 2
Figure imgf000393_0001
Table 2
Figure imgf000394_0001
Table 2
Figure imgf000395_0001
Table 2
Figure imgf000396_0001
Table 2
Figure imgf000397_0001
Table 2
Figure imgf000398_0001
Table 2
Figure imgf000399_0001
Table 2
Figure imgf000400_0001
Table 2
Figure imgf000401_0001
Table 2
Figure imgf000402_0001
Table 2
Figure imgf000403_0001
Table 2
Figure imgf000404_0001
Table 2
Figure imgf000405_0001
Table 2
Figure imgf000406_0001
Table 2
Figure imgf000407_0001
Table 2
Figure imgf000408_0001
Table 2
Figure imgf000409_0001
Table 2
Figure imgf000410_0001
Table 2
Figure imgf000411_0001
Table 2
Figure imgf000412_0001
Table 2
Figure imgf000413_0001
Table 2
Figure imgf000414_0001
Table 2
Figure imgf000415_0001
106. A pharmaceutical composition comprising a compound according to any one of claims 1 - 105 and a pharmaceutically acceptable carrier.
107. A metabolite of the compound or the pharmaceutical composition according to any one of claims 1 - 106.
108. A method of modulating the in vivo activity of a kinase, the method comprising administering to a subject an effective amount of the compound or the pharmaceutical composition according to any of claims 1 - 105.
109. The method according to claim 108, wherein modulating the in vivo activity of the kinase comprises inhibition of said kinase.
110. The method according to claim 108, wherein the kinase is at least one of c-Met, KDR, c-Kit, flt-3, and flt-4.
111. The method according to claim 110, wherein the kinase is c-Met.
112. A method of treating diseases or disorders associated with uncontrolled, abnormal, and/or unwanted cellular activities, the method comprising administering, to a mammal in need thereof, a therapeutically effective amount of the compound or the pharmaceutical composition as described in any one of claims 1 - 106.
113. A method of screening for a modulator of a kinase, said kinase selected from c- Met, KDR, c-Kit, flt-3, and flt-4, the method comprising combining a compound according to any one of claims 1 - 105, and at least one candidate agent and determining the effect of the candidate agent on the activity of said kinase.
114. A method of inhibiting proliferative activity in a cell, the method comprising administering an effective amount of a composition comprising a compound according any one of claims 1 - 105 to a cell or a plurality of cells.
115. A process for preparing a compound of Formula XXI,
Figure imgf000416_0001
XXI comprising reaction of a compound of Formula XXII, with a compound of Formula XXIII
Figure imgf000416_0002
Figure imgf000416_0003
XXIII wherein, each R1 is independently selected from halogen, -OR3, -NO2, -NH2, -NR3R3, -D-R50 and optionally substituted Cι-6alkyl;
R70 is selected from -H, halogen, -OR3, -S(O)0-2R3, -NO2, -NH2, -NR3R3, and optionally substituted C1-6alkyl;
J is selected from =N-, =C(H)-, =C(halogen)-, and =C(CN)-;
Z is selected from -S(O)0-2-, -O-, and -NR5-; each R5 is independently selected from -H, optionally substituted Chalky!, optionally substituted aryl, and optionally substituted aryl C1-6alkyl;
Ar is either a five- to ten-membered arylene or a five- to ten-membered heteroarylene containing between one and three heteroatoms;
R2 is selected from -H, halogen, trihalomethyl, -CN, -NO2, -NH2, -OR3, -NR3R3, -S(O)0-2R3, -SO2NR3R3, -CO2R3, -C(O)NR3R3, -N(R3)SO2R3, -N(R3)C(O)R3, -N(R3)CO2R3, -C(O)R3, and optionally substituted Cι-6alkyl; each R3 is independently selected from -H, -Si(R5)(R5)R5, optionally substituted lower alkyl, optionally substituted aryl, optionally substituted arylalkyl, and optionally substituted heteroarylalkyl; two R3, together with the nitrogen to which they are attached, form a four- to seven- membered heteroalicyclic, said four- to seven-membered heteroalicyclic optionally containing one additional heteroatom; when one said additional heteroatom is a nitrogen, then said nitrogen is optionally substituted with a group selected from -H, trihalomethyl, -SO2R5, -SO2NR5R5, -CO2R5, -C(O)NR5R5, -C(O)R5, and optionally substituted lower alkyl;
B is selected from absent, -N(R13)-, -N(SO2R13)-, -O-, -S(O)0-2-, and -C(=O)-;
L is selected from absent, -C(=S)N(R13)-, -C(=NR14)N(R13)-, -SO2N(R13)-, -SO2-, -C(=O)N(R13)-, -N(R13)-, -C(=O)C1-2alkylN(R13)-, -N(R13)C1-2alkylC(=O)-,
-C(=O)C0-ιalkylC(=O)N(R13)-, -C(=O)-, -C0-4alkylene-, -C(=O)C0-1alkylC(=O)OR3-, -C(=NR14)C0-1alkylC(=O)-, -C(=O)C0-1alkylC(=O)-, and an optionally substituted four- to six-membered heterocyclyl containing between one and three annular heteroatoms and comprising at least one nitrogen;
T is selected from -H, -R13, -C0-4alkyl, -C0-4alkylQ, -OC0-4alkylQ, -C0- alkylOQ, -N(R13)C0-4alkylQ, -SO2C0-4alkylQ, -C(=O)C0-4alkylQ, -C0-4alkylN(R13)Q, and -C(=O)N(R13)Co-4alkylQ, wherein each of the aforementioned Co-4alkyl is optionally substituted;
Q is a five- to ten-membered ring system, optionally substituted with between zero and four of R20; each R20 is independently selected from -H, halogen, trihalomethyl, -CN, -NO2, -NH2, -OR3, -NR3R3, -S(O)o-2R3, -SO2NR3R3, -CO2R3, -C(O)NR3R3, -N(R3)SO2R3, -N(R3)C(O)R3, -N(R3)CO2R3, -C(O)R3, optionally substituted C1-6alkyl, optionally substituted aryl, optionally substituted aryl C1-6alkyl, optionally substituted heterocyclyl, and optionally substituted heterocyclyl Cι-6alkyl; two of R20, together with the atom or atoms to which they are attached, combine to form an optionally substituted three- to seven-membered heteroalicyclic, said optionally substituted three- to seven-membered heteroalicyclic either spiro- to Q or fused to Q;
D is selected from -O-, -S(O)0-2-, and -NR15-; R50 is either R3, or according to formula XXIV;
Figure imgf000418_0001
XXIV wherein X1, X2, and optionally X3, represent the atoms of a saturated bridged ring system, said saturated bridged ring system comprising up to four annular heteroatoms represented
1 ^ by any of X , X , and X ; wherein, each X1 is independently selected from -C(R6)R7-, -O-, -S(O)0-2-, and -NR8-; each X2 is independently an optionally substituted bridgehead methine or a bridgehead nitrogen; each X3 is independently selected from -C(R6)R7-, -O-, -S(O)0-2-, and -NR8-;
Y is either: an optionally substituted C1-6alkylene linker, between D and either 1) any annular atom of the saturated bridged ring system, except X when X 9 is a bridgehead nitrogen, or 2) any heteroatom, represented by any of R or R ; provided there are at least two carbon atoms between D and any annular heteroatom of the saturated bridged ring system or any heteroatom represented by any of R6 or R7; or Y is absent, when Y is absent, said saturated bridged ring system, is directly attached to D via an annular carbon of said saturated bridged ring system, unless D is -SO2-, in which case said saturated bridged ring system, is directly attached to D via an any annular atom of said saturated bridged ring system; m and p are each independently one to four; n is zero to two, when n is zero, then there is a single bond between the two bridgehead
X2's;
R6 and R7 are each independently selected from -H, halogen, trihalomethyl, -CN, -NH2, -NO2, -OR3, -NR3R3, -S(O)0-2R3, -SO2NR3R3, -CO2R3, -C(O)NR3R3, -N(R3)SO2R3, -N(R3)C(O)R3, -NCO2R3, -C(O)R3, optionally substituted C1-6alkyl, optionally substituted aryl, optionally substituted aryl C1-6alkyl, optionally substituted heterocyclyl, optionally substituted heterocyclyl a C1-6lkyl, and a bond to either Y or D; or
R and R , when taken together are oxo; or
R and R , when taken together with a common carbon to which they are attached, form a optionally substituted three- to seven-membered spirocyclyl, said optionally substituted three- to seven-membered spirocyclyl optionally containing at least one additional annular heteroatom selected from N, O, S, and P;
R8 is selected from -R3, Y, -SO2NR3R3, -CO2R3, -C(O)NR3R3, -SO2R3, and -C(O)R3;
R13 is selected from -H, -C(=O)R3, -C(=O)OR3, -C(=O)SR3, -SO2R3, -C(=O)N(R3)R3, and optionally substituted C1-6alkyl; two R13, together with the atom or atoms to which they are attached, can combine to form a heteroalicyclic optionally substituted with between one and four of R60, said heteroalicyclic comprising up to four annular heteroatoms, and said heteroalicyclic optionally comprising an aryl or heteroaryl fused thereto, in which case said aryl or heteroaryl is optionally substituted with an additional one to four of R60;
R14 is selected from -H, -NO2, -NH2, -N(R3)R3, -CN, -OR3, optionally substituted C1-6alkyl, optionally substituted heteroalicyclyl Cι-6alkyl, optionally substituted aryl, optionally substituted aryl Cι-6alkyl and optionally substituted heteroalicyclic;
R15 is a group -M^M2, wherein M1 is selected from absent, -C(=S)N(R13)-, -C(=NR14)N(R13)-, -SO2N(R13)-, -SO2-, -C(=O)N(R13)-, -C(=O)C(=O)N(R13)-, -Co-4alkylene-, -C(=O)-, and an optionally substituted four to six-membered heterocyclyl containing between one and three heteroatoms but comprising at least one nitrogen; and M2 is selected from -H, -C0-6alkyl, alkoxy, -C(=O)C0. alkylQ, -C0-4alkylQ, -OC0- alkylQ-, -N(R13)C0-4alkylQ-, and -C(=O)N(R13)C0- alkylQ;
R60 is selected from -H, halogen, trihalomethyl, -CN, -NO2, -NH2, -OR3, -NR3R3, -S(O)o-2R3, -SO2NR3R3, -CO2R3, -C(O)NR3R3, -N(R3)SO2R3, -N(R3)C(O)R3, -N(R3)CO2R3, -C(O)R3, optionally substituted C1-6alkyl, optionally substituted aryl, optionally substituted heteroaryl C1-6alkyl, and optionally substituted aryl C1-6alkyl; two of R60, when attached to a non-aromatic carbon, can be oxo;
P is a suitable leaving group; and P2 is selected from -H, a metal, and a group removed in-situ when combining XXII and XXIII to make XXI.
116. The process according to claim 115, wherein Ar is pαr -phenylene as defined by the substitution pattern of -Z- and -B-L-T about said phenylene.
117. The process according to claim 116, wherein Z is either -O- or -NR5-.
118. The process according to claim 117, wherein -B-L-T is selected from the following:
Figure imgf000420_0001
Figure imgf000421_0001
Figure imgf000422_0001
wherein Q, R20, and R13 are as defined above; each E is selected from -O-, -N(R13)-, -CH2, and -S(O)o-2-; M is selected from -O-, -N(R13)-, -CH2-, and -C(=O)N(R13)-; each V is independently either =N- or =C(H)-; each methylene in any of the above formulae is independently optionally substituted with R25; and R25 is selected from halogen, trihalomethyl, -CN, -NO2, -NH2, -OR3, -NR3R3, -S(O)0-2R3, -SO2NR3R3, -CO2R3, -C(O)NR3R3, -N(R3)SO2R3, -N(R3)C(O)R3, -N(R3)CO2R3, -C(O)R3, optionally substituted aryl, optionally substituted aryl C1-6alkyl, heteroaryl Cι-6alkyl, and optionally substituted -βalkyl; two of R25, together with the carbon or carbons to which they are attached, can combine to form an optionally substituted three- to seven-membered alicyclic or heteroalicyclic; two of R ,25 on a single carbon can be oxo.
119. The process according to claim 118, wherein there is one of R1 that is -D-R50 and another of R1 that is -OR3a.
120. The process according to claim 119, wherein D is -O-.
121. The process according to claim 120, wherein -O-R50 and -OR3a are interchangeably located at the 6-position and 7-position of the quinazoline or quinoline according to Formula XXI.
122. The process according to claim 121, wherein -OR3a is selected from -OH, -OSi(R5)(R5)R5, and optionally substituted -OC1-6alkyl.
123. The process according to claim 122, wherein J is =N- or =C(H)-.
124. The process according to claim 123, wherein -B-L-T is selected from:
Figure imgf000422_0002
Figure imgf000423_0001
wherein Q, R , R , E, and R are as defined above; each methylene in any of the above formulae, other than those in a depicted ring, is independently optionally substituted with
R .25 D.; and R ,2/5D is selected from halogen, trihalomethyl, oxo, -CN, -NO2, -NH2, -OR% -NR3R3, -S(O)0-2R3, -SO2NR3R3, -CO2R3, -C(O)NR3R3, -N(R3)SO2R3, -N(R3)C(O)R3, -N(R3)CO2R3, -C(O)R3, optionally substituted aryl, optionally substituted aryl C1-6alkyl, heteroaryl C1-6alkyl, and optionally substituted C1-6alkyl; two of R , together with the carbon or carbons to which they are attached, can combine to form a three- to seven- membered optionally substituted alicyclic or heteroalicyclic.
125. The process according to claim 124, wherein Q is selected from the following three formula:
Figure imgf000423_0002
wherein R20 is defined as above, and P is a five- to seven-membered ring, including the two shared carbons of the aromatic ring to which P is fused, P optionally containing between one and three heteroatoms.
126. The process according to claim 125, wherein -B-L-T is either of formula XXV or formula XXVI, ?
Figure imgf000424_0001
XXV XXVI wherein R ,2υ0 is defined as above; G is either an optionally substituted cycloalkyl or an optionally substituted heteroalicyclic; each R30 is independently selected from halogen, trihalomethyl, -CN, -NO2, -NH2, -OR3, -NR 3JτR33J, -S(O)0-2RΛ, -SO2NR >3"rR»3Λ, -CO2RJ,
-C(O)NR ,33τR,3J, -N(RJ)SO2RJ, -N(RJ)C(O)R°, -N(RJ)CO2R3, -C(O)R3, and optionally s suubbssttiittuutteedd CC11--66aallkkyyll;; aanndd RR33aa and R3b are each independently selected from -H and optionally substituted C1-6alkyl.
127. The process according to claim 126, wherein a compound of formula XXIIa is combined with a compound of formula XXIIIa to make a compound of formula XXIa,
Figure imgf000424_0002
XXIa wherein -B-L-T, Z, J, R50, and R2 are as defined above; R70 is selected from -H, -NO2, -NH2, and -NR3R3; provided when Z is -N(R5)- that R5 is selected from -H, C1-3alkyl, and aryl Cι-3alkyl; P1 is selected from halogen, optionally substituted alkyl-S(O)o-2-, optionally substituted arylsulfonate, optionally substituted alkylsulfonate, a group containing boron, an azide, a group containing phosphorus, and a metal; and P2 is selected from -H and a metal.
128. The process according to claim 127, wherein P2 is selected from -H, lithium, sodium, potassium, cesium, copper, palladium, and titanium.
129. The process according to claim 128, wherein Z is -O-.
130. The process according to claim 129, wherein P1 is selected from chlorine, bromine, a toluene sulfonate, and trifluoromethansulfonate.
131. The process according to claim 130, wherein R is -H.
132. The process according to claim 131, wherein J is =C(H)-.
133. The process according to claim 132, wherein R2 is selected from C1-6 alkyl, perfluoro C1-6 alkyl, and halogen.
134. The process according to claim 133, wherein XXIIa and XXIIIa are heated together, optionally with a base, optionally with microwave radiation, to form XXIa.
135. The process according to claim 134, wherein the base is selected from an organic base, an inorganic base, and a combination of an organic base and an inorganic base.
136. The process according to claim 135, wherein the base is selected from 2,6-lutidine, 4-N,N-dimethylaminopyridine, and a metal carbonate.
137. The process according to claim 136, wherein XXIIa and XXIIIa are heated together in a solvent with said base, at between about 40°C and 200°C for between about one hour and twenty-four hours to form XXIa.
138. The process according to claim 137, wherein the solvent is an organic solvent.
139. The process according to claim 138, wherein one molar equivalent of XXIIa is combined with between about one quarter and four molar equivalents of XXIIIa.
140. The process according to claim 139, wherein one molar equivalent of XXIIa is combined with more than one but less than two molar equivalents of XXIIIa.
141. The process according to claim 140, wherein XXIIa is combined with XXIIIa and said base in an aromatic solvent to form a mixture, and said mixture is heated to between about 100°C and 200°C for between about one and ten hours to form la.
142. The process according to claim 141, wherein the aromatic solvent is an optionally substituted benzene.
143. The process according to claim 142, wherein the aromatic solvent is bromobenzene.
144. The process according to claim 143, wherein the base is 4-N,N- dimethylaminopyridine.
145. The process according to claim 144, wherein said mixture is heated to reflux for between about three and seven hours.
146. The process according to claim 145, wherein said mixture is heated to reflux for between about four and six hours.
147. The process according to claim 140, wherein XXIIa is combined with XXIIIa and said base in a non-aromatic solvent to form a mixture, and said mixture is heated to between about 40°C and 100°C for between about one and twenty hours to form XXIa.
148. The process according to claim 147, wherein the non-aromatic solvent comprises a functional group selected from an amide, and ether, a nitrile, a halide, an ester, an amine, and a ketone.
149. The process according to claim 148, wherein the non-aromatic solvent is N,N- dimethylacetamide.
150. The process according to claim 149, wherein the base is potassium carbonate.
151. The process according to claim 150, wherein said mixture is heated to about 50°C between about ten and twenty hours.
152. The process according to claim 151, wherein the aromatic solvent is an optionally substituted pyridine.
153. The process according to claim 152, wherein the aromatic solvent is 2,6-lutidine.
154. The process according to claim 153, wherein the base is 2,6-lutidine.
155. The process according to claim 154, wherein said mixture is heated to reflux for between about three and seven hours.
156. The process according to claim 155, wherein said mixture is heated to reflux for between about four and six hours.
157. The process according to claim 139, wherein one molar equivalent of XXIIIa is combined with more than one but less than two molar equivalents of XXIIa.
158. The process according to claim 157, wherein XXIIa is combined with XXIIIa and said base in an aromatic solvent to form a mixture, and said mixture is heated to between about 100°C and 200°C for between about ten and twenty hours to form XXIa.
159. The process according to claim 158, wherein the aromatic solvent is an optionally substituted pyridine.
160. The process according to claim 159, wherein the aromatic solvent is 2,6-lutidine.
161. The process according to claim 160, wherein the base is 2,6-lutidine.
162. The process according to claim 161, wherein said mixture is heated to between about 150°C and 200°C for between about fifteen and twenty hours.
163. The process according to any of claims 134 - 162, wherein a compound of formula XXIIb is substituted for the compound of formula XXIIa, and either a compound of formula XXIIIb or a compound of formula XXIIIc is substituted for the compound of formula XXIIIa, in order to make a compound of formula XXIb or a compound of formula XXIc, respectively,
Figure imgf000427_0001
XXIIb
Figure imgf000427_0002
XXIIIb XXIIIc
Figure imgf000428_0001
Figure imgf000428_0002
wherein J, R50, R20 and R2 are as defined above.
164. The process according to claim 163, wherein R , if present, is halogen.
165. The process according to claim 164, wherein R2, if present, is fluorine.
166. The process according to claim 165, wherein R2, if present, is up to two fluorines ortho to the oxygen of the phenylene to which R2 is attached.
167. The process according to claim 115, used to make a compound listed in either Table l or Table 2.
168. The process according to any of claims 115 - 167, further comprising converting said compound to a pharmaceutically acceptable salt, hydrate, or prodrug thereof.
PCT/US2004/031523 2003-09-26 2004-09-24 C-met modulators and methods of use WO2005030140A2 (en)

Priority Applications (22)

Application Number Priority Date Filing Date Title
AT04789057T ATE532782T1 (en) 2003-09-26 2004-09-24 C-MET MODULATORS AND APPLICATION METHODS
JP2006528265A JP4638436B2 (en) 2003-09-26 2004-09-24 c-Met modulators and uses thereof
AU2004275842A AU2004275842B2 (en) 2003-09-26 2004-09-24 c-MET modulators and methods of use
EP17190013.7A EP3329918B1 (en) 2003-09-26 2004-09-24 C-met modulators and methods of use
CA2537812A CA2537812C (en) 2003-09-26 2004-09-24 C-met modulators and method of use
EP04789057A EP1673085B1 (en) 2003-09-26 2004-09-24 C-met modulators and methods of use
US11/586,751 US9174947B2 (en) 2003-09-26 2006-10-26 c-Met modulators and methods of use
US11/753,503 US8476298B2 (en) 2003-09-26 2007-05-24 c-Met modulators and method of use
US11/753,462 US8178532B2 (en) 2003-09-26 2007-05-24 c-Met modulators and method of use
US11/753,514 US8067436B2 (en) 2003-09-26 2007-05-24 c-Met modulators and methods of use
US12/393,806 US7579473B2 (en) 2003-09-26 2009-02-26 c-Met modulators and methods of use
AU2010204461A AU2010204461B2 (en) 2003-09-26 2010-07-27 c-MET modulators and methods of use
AU2010204459A AU2010204459B2 (en) 2003-09-26 2010-07-27 c-MET modulators and methods of use
US12/911,442 US20110077233A1 (en) 2003-09-26 2010-10-25 C-Met Modulators and Method of Use
US13/249,815 US8497284B2 (en) 2003-09-26 2011-09-30 C-met modulators and method of use
US13/427,093 US20120184523A1 (en) 2003-09-26 2012-03-22 C-Met Modulators and Method of Use
US13/888,592 US20140155378A9 (en) 2003-09-26 2013-05-07 c-Met Modulators and Method of Use
US13/928,977 US20140155396A1 (en) 2003-09-26 2013-06-27 C-Met Modulators and Method of Use
US14/847,801 US20150376133A1 (en) 2003-09-26 2015-09-08 C-Met Modulators and Methods of Use
US15/066,872 US20160185725A1 (en) 2003-09-26 2016-03-10 c-Met Modulators and Method of Use
US15/688,310 US11124482B2 (en) 2003-09-26 2017-08-28 C-met modulators and methods of use
US16/921,542 US20200331860A1 (en) 2003-09-26 2020-07-06 C-Met Modulators and Methods of Use

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US50618103P 2003-09-26 2003-09-26
US60/506,181 2003-09-26
US53537704P 2004-01-09 2004-01-09
US60/535,377 2004-01-09
US57738404P 2004-06-04 2004-06-04
US60/577,384 2004-06-04

Related Child Applications (3)

Application Number Title Priority Date Filing Date
US10573336 A-371-Of-International 2006-09-18
US11/586,751 Continuation US9174947B2 (en) 2003-09-26 2006-10-26 c-Met modulators and methods of use
US14/847,801 Continuation US20150376133A1 (en) 2003-09-26 2015-09-08 C-Met Modulators and Methods of Use

Publications (2)

Publication Number Publication Date
WO2005030140A2 true WO2005030140A2 (en) 2005-04-07
WO2005030140A3 WO2005030140A3 (en) 2005-05-19

Family

ID=34397024

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2004/031523 WO2005030140A2 (en) 2003-09-26 2004-09-24 C-met modulators and methods of use

Country Status (19)

Country Link
US (14) US9174947B2 (en)
EP (9) EP2210607B1 (en)
JP (14) JP4638436B2 (en)
AT (3) ATE517091T1 (en)
AU (3) AU2004275842B2 (en)
BE (1) BE2014C055I2 (en)
CA (2) CA2744997A1 (en)
CY (3) CY1111956T1 (en)
DK (4) DK2392564T3 (en)
ES (6) ES2925655T3 (en)
HK (5) HK1141799A1 (en)
HR (4) HRP20110707T1 (en)
HU (1) HUS1400052I1 (en)
LU (1) LU92508I2 (en)
NL (1) NL300678I2 (en)
PL (3) PL2392565T3 (en)
PT (2) PT2210607E (en)
SI (4) SI2392565T1 (en)
WO (1) WO2005030140A2 (en)

Cited By (194)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005082855A1 (en) * 2004-02-27 2005-09-09 Eisai Co., Ltd. Novel pyridine derivative and pyrimidine derivative (2)
EP1674466A1 (en) * 2004-12-27 2006-06-28 4Sc Ag 2,5- and 2,6-disubstituted benzazole analogues useful as protein kinase inhibitors
WO2006060318A3 (en) * 2004-11-30 2006-07-20 Amgen Inc Quinolines and quinazoline analogs and their use as medicaments for treating cancer
WO2007103308A2 (en) 2006-03-07 2007-09-13 Array Biopharma Inc. Heterobicyclic pyrazole compounds and methods of use
WO2008023698A1 (en) * 2006-08-23 2008-02-28 Eisai R & D Management Co., Ltd. Salt of phenoxypyridine derivative or crystal thereof and process for producing the same
JP2008510839A (en) * 2004-08-25 2008-04-10 ターゲジェン インコーポレーティッド Heterocyclic compounds and methods of use
WO2008049855A2 (en) * 2006-10-27 2008-05-02 Glaxo Group Limited 7-azaindole derivatives as c-met kinase inhibitors
EP1964837A1 (en) * 2005-11-22 2008-09-03 Eisai R&D Management Co., Ltd. Anti-tumor agent for multiple myeloma
WO2008079291A3 (en) * 2006-12-20 2008-11-06 Amgen Inc Substituted heterocycles and methods of use
DE102007026341A1 (en) 2007-06-06 2008-12-11 Merck Patent Gmbh Benzoxazolonderivate
JP2009504804A (en) * 2005-05-20 2009-02-05 メチルジーン インコーポレイテッド Inhibitors of VEGF receptor and HGF receptor signaling
US7576090B2 (en) * 2004-12-27 2009-08-18 4Sc Ag Benzazole analogues and uses thereof
JP2009534410A (en) * 2006-04-19 2009-09-24 ノバルティス アクチエンゲゼルシャフト 6-O-substituted benzoxazole and benzothiazole compounds and methods for inhibiting CSF-1R signaling
WO2009125597A1 (en) 2008-04-10 2009-10-15 大鵬薬品工業株式会社 Acylthiourea compound or salt thereof, and use of the compound or the salt
WO2009127417A1 (en) 2008-04-16 2009-10-22 MAX-PLANCK-Gesellschaft zur Förderung der Wissenschaften e.V. Quinoline derivatives as axl kinase inhibitors
JP2009539878A (en) * 2006-06-08 2009-11-19 アレイ バイオファーマ、インコーポレイテッド Quinoline compounds and methods of use
US7683060B2 (en) 2006-08-07 2010-03-23 Incyte Corporation Triazolotriazines as kinase inhibitors
US20100081805A1 (en) * 2008-09-26 2010-04-01 Nicole Marie Deschamps Preparation of a Quinolinyloxydiphenylcyclopropanedicarboxamide
US20100093727A1 (en) * 2008-10-14 2010-04-15 Ning Xi Compounds and methods of use
JP2010516680A (en) * 2007-01-19 2010-05-20 エックスカバリー,インコーポレイテッド Kinase inhibitor compounds
WO2010056960A1 (en) * 2008-11-13 2010-05-20 Exelixis Inc. Methods of preparing quinoline derivatives
US7732465B2 (en) 2005-08-30 2010-06-08 Novartis Vaccines And Diagnostics, Inc. Substituted benzimidazoles and methods of their use
US7732613B2 (en) 2005-09-14 2010-06-08 Bristol-Myers Squibb Company Met kinase inhibitors
WO2010065838A1 (en) 2008-12-04 2010-06-10 Exelixis, Inc. Methods of preparing quinoline derivatives
DE102009003975A1 (en) 2009-01-07 2010-07-08 Merck Patent Gmbh Benzothiazolonderivate
WO2010083414A1 (en) 2009-01-16 2010-07-22 Exelixis, Inc. Malate salt of n- (4- { [ 6, 7-bis (methyloxy) quin0lin-4-yl] oxy}phenyl-n' - (4 -fluorophenyl) cyclopropane-1-dicarboxamide, and crystalline forms therof for the treatment of cancer
US7767670B2 (en) 2003-11-13 2010-08-03 Ambit Biosciences Corporation Substituted 3-carboxamido isoxazoles as kinase modulators
US7767677B2 (en) 2004-09-20 2010-08-03 Xenon Pharmaceuticals Inc. Heterocyclic derivatives and their use as stearoyl-CoA desaturase inhibitors
US7767675B2 (en) 2006-11-22 2010-08-03 Incyte Corporation Imidazotriazines and imidazopyrimidines as kinase inhibitors
US7777036B2 (en) 2004-09-20 2010-08-17 Xenon Pharmaceuticals Inc. Heterocyclic derivatives and their use as therapeutic agents
US7790885B2 (en) 2006-08-31 2010-09-07 Eisai R&D Management Co., Ltd. Process for preparing phenoxypyridine derivatives
WO2010111063A1 (en) 2009-03-21 2010-09-30 Ning Xi Amino ester derivatives, salts thereof and methods of use
US7829712B2 (en) 2004-09-20 2010-11-09 Xenon Pharmaceuticals Inc. Pyridazine derivatives for inhibiting human stearoyl-CoA-desaturase
WO2010140092A1 (en) 2009-06-05 2010-12-09 Pfizer Inc. L- ( piperidin-4-yl) -pyrazole derivatives as gpr 119 modulators
US7855290B2 (en) 2005-08-24 2010-12-21 Eisai R&D Management Co., Ltd. Pyridine derivatives and pyrimidine derivatives (3)
US7858623B2 (en) 2005-04-27 2010-12-28 Amgen Inc. Substituted amide derivatives and methods of use
US7872017B2 (en) 2006-05-19 2011-01-18 Abbott Laboratories Fused bicycloheterocycle substituted azabicyclic alkane derivatives
WO2011009095A1 (en) 2009-07-17 2011-01-20 Exelixis, Inc. Crystalline forms of n-[3-fluoro-4-({6-(methyloxy)-7-[(3-morpholin-4-ylpropyl)oxy]-quin0lin-4-yl}oxy)phenyl]-n'-(4-fluorophenyl)cyclopropane-1, 1-dicarboxamide
US7884114B2 (en) 2007-08-15 2011-02-08 Glaxo Group Limited Compounds
WO2011017639A1 (en) 2009-08-07 2011-02-10 Exelixis,Inc Methods of using c-met modulators
US7919496B2 (en) 2004-09-20 2011-04-05 Xenon Pharmaceuticals Inc. Heterocyclic derivatives for the treatment of diseases mediated by stearoyl-CoA desaturase enzymes
EP2311809A1 (en) * 2009-10-16 2011-04-20 Max-Planck-Gesellschaft zur Förderung der Wissenschaften e.V. Quinolinyloxyphenylsulfonamides
US7951805B2 (en) 2004-09-20 2011-05-31 Xenon Pharmaceuticals Inc. Heterocyclic derivatives and their use as mediators of stearoyl-CoA desaturase
CN102083824A (en) * 2008-05-05 2011-06-01 葛兰素史密斯克莱有限责任公司 Method of treating cancer using a cMET and AXL inhibitor and an ErbB inhibitor
CN102086211A (en) * 2009-12-08 2011-06-08 深圳市东阳光实业发展有限公司 Aromatic heterocyclic compounds serving as protein kinase inhibitor
DE102009056886A1 (en) 2009-12-03 2011-06-09 Bayer Schering Pharma Aktiengesellschaft cMet inhibitors for the treatment of endometriosis
CN102093421A (en) * 2011-01-28 2011-06-15 广州盈升生物科技有限公司 Phosphorus substituent group-containing quinoline compound and preparation method of quinoline compound as well as pharmaceutical composition containing quinoline compound and application of pharmaceutical composition
US7973164B2 (en) 2006-03-02 2011-07-05 Astrazeneca Ab Quinoline derivatives
US7973160B2 (en) 2000-10-20 2011-07-05 Eisai R&D Management Co., Ltd. Nitrogen-containing aromatic derivatives
US7998948B2 (en) 2007-11-30 2011-08-16 Eisai R&D Management Co., Ltd. Pharmaceutical composition for treating esophageal cancer
WO2011112896A1 (en) 2010-03-12 2011-09-15 Exelixis, Inc Hydrated crystalline forms of n-[3-fluoro-4-({6-(methyloxy)-7-[(3-morpholin-4-ylpropyl)oxy]-quinolin-4-yl}oxy)phenyl]-n'-(4-fluorophenyl)cyclopropane-1,1-dicarboxamide
US8026360B2 (en) 2004-09-20 2011-09-27 Xenon Pharmaceuticals Inc. Substituted pyridazines as stearoyl-CoA desaturase inhibitors
CN102212062A (en) * 2010-04-02 2011-10-12 东莞市长安东阳光新药研发有限公司 Derivative of amino ester, salt thereof and using method
US8058474B2 (en) 2003-11-11 2011-11-15 Eisai R&D Management Co., Ltd. Urea derivative and process for preparing the same
WO2011145035A1 (en) 2010-05-17 2011-11-24 Indian Incozen Therapeutics Pvt. Ltd. Novel 3,5-disubstitued-3h-imidazo[4,5-b]pyridine and 3,5- disubstitued -3h-[1,2,3]triazolo[4,5-b] pyridine compounds as modulators of protein kinases
US8071603B2 (en) 2004-09-20 2011-12-06 Xenon Pharmaceuticals Inc. Heterocyclic derivatives and their use as stearoyl-CoA desaturase inhibitors
WO2011162835A1 (en) 2010-02-03 2011-12-29 Incyte Corporation Imidazo[1,2-b][1,2,4]triazines as c-met inhibitors
US8088771B2 (en) 2008-07-28 2012-01-03 Gilead Sciences, Inc. Cycloalkylidene and heterocycloalkylidene inhibitor compounds
WO2012003338A1 (en) 2010-07-01 2012-01-05 Takeda Pharmaceutical Company Limited COMBINATION OF A cMET INHIBITOR AND AN ANTIBODY TO HGF AND/OR cMET
WO2012009723A1 (en) 2010-07-16 2012-01-19 Exelixis, Inc. C-met modulator pharmaceutical compositions
WO2012009722A1 (en) 2010-07-16 2012-01-19 Exelixis, Inc. C-met modulator pharmaceutical compositions
US8124764B2 (en) 2008-07-14 2012-02-28 Gilead Sciences, Inc. Fused heterocyclyc inhibitor compounds
US8134000B2 (en) 2008-07-14 2012-03-13 Gilead Sciences, Inc. Imidazolyl pyrimidine inhibitor compounds
WO2012044574A1 (en) 2010-09-27 2012-04-05 Exelixis, Inc. Dual inhibitors of met and vegf for the treatment of castration resistant prostate cancer and osteoblastic bone metastases
WO2012042421A1 (en) 2010-09-29 2012-04-05 Pfizer Inc. Method of treating abnormal cell growth
WO2012044572A1 (en) 2010-09-27 2012-04-05 Exelixis, Inc. Dual inhibitors of met and vegf for the treatment of castration- resistant prostate cancer and osteoblastic bone metastases
WO2012044577A1 (en) 2010-09-27 2012-04-05 Exelixis, Inc. Dual inhibitors of met and vegf for the treatment of castration resistant prostate cancer and osteoblastic bone metastases
US8153643B2 (en) 2004-10-12 2012-04-10 Astrazeneca Ab Quinazoline derivatives
WO2012109510A1 (en) 2011-02-10 2012-08-16 Exelixis, Inc. Processes for preparing quinoline compounds and pharmaceutical compositions containing such compounds
EP2125780B1 (en) * 2006-12-20 2012-08-29 Amgen Inc. Substituted heterocycles and methods of use
US8258316B2 (en) 2009-06-08 2012-09-04 Gilead Sciences, Inc. Alkanoylamino benzamide aniline HDAC inhibitor compounds
US8283357B2 (en) 2009-06-08 2012-10-09 Gilead Sciences, Inc. Cycloalkylcarbamate benzamide aniline HDAC inhibitor compounds
WO2012151326A1 (en) * 2011-05-02 2012-11-08 Exelixis, Inc. Method of treating cancer and bone cancer pain
US8318752B2 (en) 2003-09-19 2012-11-27 Astrazeneca Ab 4-(3-chloro-2-fluoroanilino)-7-methoxy-6-{[1-(N-methylcarbamoyl-methyl)piperidin-4-yl]oxy}quinazoline, its pharmaceutically acceptable salts, and pharmaceutical compositions comprising the same
CN102030705B (en) * 2009-09-30 2012-12-19 上海睿智化学研究有限公司 Synthesis method of 7- benzyloxy-6-methoxyl-4-hydroxyquinoline
US8344018B2 (en) 2008-07-14 2013-01-01 Gilead Sciences, Inc. Oxindolyl inhibitor compounds
US8389541B2 (en) 2007-08-29 2013-03-05 Methylgene Inc. Inhibitors of protein tyrosine kinase activity
CN102964308A (en) * 2012-11-30 2013-03-13 中国药科大学 Novel pyrimidine compound, preparation method thereof, pharmaceutical composition containing novel pyrimidine compound and application of novel pyrimidine compound
WO2013036676A1 (en) 2011-09-06 2013-03-14 New York Blood Center, Inc. Hiv inhibitors
WO2013040801A1 (en) 2011-09-19 2013-03-28 广州盈升生物科技有限公司 Hydroxamic acid compound containing quinolyl and preparation method thereof, and pharmaceutical composition containing this compound and use thereof
WO2013043840A1 (en) 2011-09-22 2013-03-28 Exelixis, Inc. Method for treating osteoporosis
US8420645B2 (en) 2008-05-21 2013-04-16 Incyte Corporation Salts of 2-fluoro-N-methyl-4-[7-(quinolin-6-yl-methyl)-imidazo[1,2-b][1,2,4]triazin-2-yl]benzamide and processes related to preparing the same
WO2013059788A1 (en) 2011-10-20 2013-04-25 Exelixis, Inc. Process for preparing quinoline derivatives
WO2013070903A1 (en) 2011-11-08 2013-05-16 Exelixis, Inc. Method of quantifying cancer treatment
WO2013070890A1 (en) 2011-11-08 2013-05-16 Exelixis, Inc. Dual inhibitor of met and vegf for treating cancer
CN103172641A (en) * 2011-12-20 2013-06-26 钱卫 Heterocyclic amino and alkoxy-replaced quinazoline derivative and application thereof
US8481536B2 (en) 2004-04-08 2013-07-09 Targegen, Inc. Benzotriazine inhibitors of kinases
EP2621904A2 (en) * 2010-09-12 2013-08-07 Advenchen Laboratories, LLC Compounds as c-met kinase inhibitors
US8541457B2 (en) 2005-06-03 2013-09-24 Xenon Pharmaceuticals Inc. Aminothiazole derivatives as human stearoyl-CoA desaturase inhibitors
WO2013144737A2 (en) 2012-03-30 2013-10-03 Rhizen Pharmaceuticals Sa Novel 3,5-disubstitued-3h-imidazo[4,5-b]pyridine and 3,5- disubstitued -3h-[1,2,3]triazolo[4,5-b] pyridine compounds as modulators of c-met protein kinases
WO2013152252A1 (en) 2012-04-06 2013-10-10 OSI Pharmaceuticals, LLC Combination anti-cancer therapy
US8569319B2 (en) 2010-04-29 2013-10-29 Deciphera Pharmaceuticals, LLS Pyridone amides and analogs exhibiting anti-cancer and anti-proliferative activities
WO2013166296A1 (en) 2012-05-02 2013-11-07 Exelixis, Inc. A dual met - vegf modulator for treating osteolytic bone metastases
US8618289B2 (en) 2008-03-17 2013-12-31 Ambit Biosciences Corporation RAF kinase modulator compounds and methods of use thereof
WO2014000686A1 (en) * 2012-06-28 2014-01-03 辰欣药业股份有限公司 Pyrimidobenzoazepine compound and use thereof as antitumour drug
EP2684874A1 (en) 2006-10-23 2014-01-15 Cephalon, Inc. Fused bicyclic derivatives of 2,4-diaminopyrimidine as alk and C-met inhibitors
WO2014029251A1 (en) * 2012-08-24 2014-02-27 辰欣药业股份有限公司 Benazepine compound and application thereof
WO2014029250A1 (en) * 2012-08-24 2014-02-27 辰欣药业股份有限公司 Benazepine compound and application thereof
CN103664776A (en) * 2012-09-26 2014-03-26 正大天晴药业集团股份有限公司 Preparation method for tyrosine kinase inhibitor and midbody thereof
CN103705521A (en) * 2012-09-28 2014-04-09 韩冰 Compound for treating cerebral infarction and application thereof
CN103800328A (en) * 2012-11-07 2014-05-21 韩冰 Compound for treating neurodegenerative diseases and application thereof
CN103804291A (en) * 2012-11-07 2014-05-21 韩冰 Compound for treating neurodegenerative diseases and application thereof
CN103804381A (en) * 2012-11-06 2014-05-21 韩冰 Compound for treatment of ischemic brain damage and application thereof
CN103804361A (en) * 2012-11-07 2014-05-21 韩冰 Compound for treating neurodegenerative diseases and application thereof
CN103800340A (en) * 2012-11-09 2014-05-21 韩冰 Compounds for treating glaucoma and application thereof
CN103804298A (en) * 2012-11-09 2014-05-21 韩冰 Compounds for treating glaucoma and application thereof
CN103804290A (en) * 2012-11-07 2014-05-21 韩冰 Compound for treating neurodegenerative diseases and application thereof
CN103804363A (en) * 2012-11-14 2014-05-21 韩冰 Compound with neuroprotective effect and use thereof
CN103804362A (en) * 2012-11-12 2014-05-21 韩文毅 Compounds for treating diabetes and application thereof
CN103804382A (en) * 2012-11-05 2014-05-21 韩文毅 Compound for treating eczema and application thereof
CN103965104A (en) * 2013-01-29 2014-08-06 正大天晴药业集团股份有限公司 Preparation methods of tyrosine kinase inhibitor and intermediates thereof
WO2014145715A1 (en) 2013-03-15 2014-09-18 Exelixis, Inc. Metabolites of n-(4-{[6,7-bis(methyloxy)quinolin-4-yl]oxy}phenyl)-n'-(4-fluorophenyl) cyclopropane-1,1-dicarboxamide
WO2014145693A1 (en) * 2013-03-15 2014-09-18 Exelixis, Inc. Metabolites of n-[3-fluoro-4-({ 6-(methyloxy)-7-[(3-morpholin-4-ylpropyl)oxy]quinolin-4-yl}oxy)phenyl]-n'-(4-fluorophenyl)cyclopropane-1,1-dicarboxamide
WO2014165779A1 (en) 2013-04-04 2014-10-09 Exelixis, Inc. Drug combinations to treat cancer
WO2014165786A1 (en) 2013-04-04 2014-10-09 Exelixis, Inc. Cabozantinib dosage form and use in the treatment of cancer
CN104109124A (en) * 2013-04-19 2014-10-22 正大天晴药业集团股份有限公司 Crystal of cabozantinib*0.5 malate
CN104109128A (en) * 2013-04-19 2014-10-22 连云港润众制药有限公司 Cabozantinib malate and preparation method thereof
US20140364431A1 (en) * 2011-12-30 2014-12-11 Shenyang Pharmaceutical University Quinoline and cinnoline derivatives and their applications
CN104370811A (en) * 2013-08-15 2015-02-25 广东东阳光药业有限公司 New quinoline compound crystal form and preparation method thereof
US8969379B2 (en) 2004-09-17 2015-03-03 Eisai R&D Management Co., Ltd. Pharmaceutical compositions of 4-(3-chloro-4-(cyclopropylaminocarbonyl)aminophenoxy)-7=methoxy-6-quinolinecarboxide
US8999982B2 (en) 2010-08-28 2015-04-07 Lead Discovery Center Gmbh Pharmaceutically active compounds as Axl inhibitors
CN104628657A (en) * 2013-11-06 2015-05-20 韩冰 Class of compounds for treating ischemic brain damage and purpose thereof
CN104788372A (en) * 2014-07-25 2015-07-22 上海圣考医药科技有限公司 Deuterated cabozantinib derivative, preparation method and application thereof, and intermediate of deuterated cabozantinib derivative
WO2015123639A1 (en) 2014-02-14 2015-08-20 Exelixis, Inc. Crystalline solid forms of n-{4-[(6,7-dimethoxyquinolin-4-yl)oxy]phenyl}-n'-(4-fluorophenyl) cyclopropane-1, 1-dicarboxamide, processes for making, and methods of use
KR20150095652A (en) * 2012-11-05 2015-08-21 션양 파마슈티컬 유니버시티 Novel quinoline derivatives and their applications
US9133123B2 (en) 2010-04-23 2015-09-15 Cytokinetics, Inc. Certain amino-pyridines and amino-triazines, compositions thereof, and methods for their use
WO2015142928A1 (en) 2014-03-17 2015-09-24 Exelixis, Inc. Dosing of cabozantinib formulations
WO2015164869A1 (en) 2014-04-25 2015-10-29 Exelixis, Inc. Method of treating lung adenocarcinoma
CN105218445A (en) * 2015-08-25 2016-01-06 江苏中邦制药有限公司 The preparation method of a kind of TYR enzyme inhibitors Foretinib
WO2016019285A1 (en) 2014-07-31 2016-02-04 Exelixis, Inc. Method of preparing fluorine-18 labeled cabozantinib and its analogs
WO2016022697A1 (en) 2014-08-05 2016-02-11 Exelixis, Inc. Drug combinations to treat multiple myeloma
CN105541798A (en) * 2016-02-03 2016-05-04 中国人民解放军第二军医大学 Quinoline multi-target kinase inhibitor with antitumor activity and preparation method thereof
WO2016091891A1 (en) 2014-12-09 2016-06-16 INSERM (Institut National de la Santé et de la Recherche Médicale) Human monoclonal antibodies against axl
CN105906568A (en) * 2016-04-28 2016-08-31 西安交通大学 Cyclopropane diamide compound with antitumor activity and preparation method and application thereof
WO2016135066A1 (en) 2015-02-26 2016-09-01 INSERM (Institut National de la Santé et de la Recherche Médicale) Fusion proteins and antibodies comprising thereof for promoting apoptosis
WO2016150966A1 (en) 2015-03-25 2016-09-29 Sandoz Ag Crystalline forms of cabozantinib phosphate and cabozantinib hydrochloride
CN106008371A (en) * 2016-06-24 2016-10-12 谢阳 1-aryl ureido naphthenic base-1-formamide compound and pharmaceutical composition and application thereof
CN106083715A (en) * 2016-06-01 2016-11-09 谢阳 A kind of quinoline, quinazoline compounds and pharmaceutical composition thereof and application
WO2017029362A1 (en) 2015-08-19 2017-02-23 Sandoz Ag Asymmetric bisamidation of malonic ester derivatives
US9604965B2 (en) 2010-04-23 2017-03-28 Cytokinetics, Inc. Substituted pyridazines as skeletal muscle modulators
CN106543143A (en) * 2015-09-22 2017-03-29 合肥中科普瑞昇生物医药科技有限公司 New FLT3 kinase inhibitors of one class and application thereof
CN106632028A (en) * 2016-12-22 2017-05-10 上海再启生物技术有限公司 Cabozantinib preparation method
US9730886B2 (en) 2010-04-23 2017-08-15 Cytokinetics, Inc. Amino-pyrimidine skeletal muscle modulators
CN107129465A (en) * 2016-02-26 2017-09-05 中国科学院上海药物研究所 Aryl guanidine compound and its production and use
WO2017173049A1 (en) * 2016-03-31 2017-10-05 Merck Patent Gmbh Compounds for the inhibition of cyclophilins and uses thereof
CN107235896A (en) * 2016-09-13 2017-10-10 上海翔锦生物科技有限公司 Tyrosine kinase inhibitor and its application
CN107709320A (en) * 2015-05-21 2018-02-16 中国科学院上海药物研究所 A kind of pyrido nitrogen heterocyclic and its production and use
WO2018064191A1 (en) 2016-09-27 2018-04-05 The Usa Of America, As Represented By The Secretary, Department Of Health & Human Services Method of treating urothelial carcinoma and other genitourinary malignancies using n-(4-(6,7-dimethoxyquinolin-4-yloxy)-phenyl)-n'(4-fluorophenyl)cyclopropane-1,1-dicarboxamide
US9945862B2 (en) 2011-06-03 2018-04-17 Eisai R&D Management Co., Ltd. Biomarkers for predicting and assessing responsiveness of thyroid and kidney cancer subjects to lenvatinib compounds
WO2018072614A1 (en) * 2016-10-18 2018-04-26 北京康辰药业股份有限公司 Quinolinyl-substituted carboxylic acid compound or pharmaceutically acceptable salt thereof, pharmaceutical composition thereof, and use thereof
CN108264482A (en) * 2018-02-05 2018-07-10 南京法恩化学有限公司 It is a kind of to block the rich preparation method for Buddhist nun
WO2018136796A1 (en) 2017-01-20 2018-07-26 Exelixis, Inc. Combinations of cabozantinib and atezolizumab to treat cancer
WO2018133159A1 (en) * 2017-01-17 2018-07-26 南昌弘益药业有限公司 Method for preparing novel quinoline compound
WO2018218233A1 (en) 2017-05-26 2018-11-29 Exelixis, Inc. Crystalline solid forms of salts of n-{4-[(6,7-dimethoxyquinolin-4-yl) oxy]phenyl}-n'-(4-fluorphenyl) cyclopropane-1,1-dicarboxamide, processes for making, and methods of use
WO2018227119A1 (en) 2017-06-09 2018-12-13 Exelixis, Inc. Liquid dosage forms to treat cancer
WO2019014402A1 (en) * 2017-07-14 2019-01-17 Innate Tumor Immunity, Inc. Nlrp3 modulators
US10240207B2 (en) 2014-03-24 2019-03-26 Genentech, Inc. Cancer treatment with c-met antagonists and correlation of the latter with HGF expression
US10259791B2 (en) 2014-08-28 2019-04-16 Eisai R&D Management Co., Ltd. High-purity quinoline derivative and method for manufacturing same
WO2019079783A1 (en) 2017-10-20 2019-04-25 Vanderbilt University Antagonists of the muscarinic acetylcholine receptor m4
CN109824587A (en) * 2017-11-23 2019-05-31 上海翔锦生物科技有限公司 The preparation method of tyrosine kinase inhibitor XJF007 and its intermediate
CN109896997A (en) * 2017-12-08 2019-06-18 中国药科大学 The preparation method and its usage of N- anilid class c-Met kinase inhibitor
WO2019148043A1 (en) * 2018-01-26 2019-08-01 Exelixis, Inc. Compounds for the treatment of kinase-dependent disorders
CN110117254A (en) * 2019-06-20 2019-08-13 江苏君若医药有限公司 Preparation method of cabozantinib
CN110156803A (en) * 2017-02-27 2019-08-23 北京赛特明强医药科技有限公司 Dioxanes and quinolines and the preparation method and application thereof
WO2019180141A1 (en) 2018-03-23 2019-09-26 Bayer Aktiengesellschaft Combinations of rogaratinib
US10517861B2 (en) 2013-05-14 2019-12-31 Eisai R&D Management Co., Ltd. Biomarkers for predicting and assessing responsiveness of endometrial cancer subjects to lenvatinib compounds
CN110862398A (en) * 2018-08-27 2020-03-06 北京赛特明强医药科技有限公司 Urea substituted aromatic ring-linked dioxane quinazoline or quinoline compound, composition and application thereof
WO2020118753A1 (en) * 2018-12-12 2020-06-18 安徽中科拓苒药物科学研究有限公司 Pan-kit kinase inhibitor having quinoline structure and application thereof
WO2020123800A1 (en) * 2018-12-13 2020-06-18 Exelixis, Inc. Crystalline forms and salt forms of a kinase inhibitor
US10799503B2 (en) 2016-12-01 2020-10-13 Ignyta, Inc. Methods for the treatment of cancer
WO2020216188A1 (en) * 2019-04-22 2020-10-29 北京康辰药业股份有限公司 Crystal forms of compound, preparation method therefor, pharmaceutical composition and application thereof
WO2020247019A1 (en) * 2019-06-03 2020-12-10 Exelixis, Inc. Crystalline salt forms of a kinase inhibitor
CN112312909A (en) * 2018-01-26 2021-02-02 埃克塞里艾克西斯公司 Compounds for the treatment of kinase-dependent disorders
US11059808B2 (en) 2014-09-19 2021-07-13 New York Blood Center, Inc. Substituted phenylpyrrolecarboxamides with therapeutic activity in HIV
US11090386B2 (en) 2015-02-25 2021-08-17 Eisai R&D Management Co., Ltd. Method for suppressing bitterness of quinoline derivative
US11110062B2 (en) 2017-02-15 2021-09-07 Taiho Pharmaceutical Co., Ltd. Pharmaceutical composition
US11124482B2 (en) 2003-09-26 2021-09-21 Exelixis, Inc. C-met modulators and methods of use
US20210309668A1 (en) * 2018-08-01 2021-10-07 Agency For Science, Technology And Research Bicyclic compounds as kinase modulators, methods and uses thereof
US11141413B2 (en) 2016-04-15 2021-10-12 Exelixis, Inc. Method of treating renal cell carcinoma using N-(4-(6,7-dimethoxyquinolin-4-yloxy)phenyl)-N′-(4-fluorophenyl)cyclopropane-1,1-dicarboxamide, (2S)-hydroxybutanedioate
CN113577066A (en) * 2020-04-30 2021-11-02 中国科学院上海药物研究所 Use of arylguanidine compounds or pharmaceutically acceptable salts thereof
IT202000027678A1 (en) 2020-11-18 2022-05-18 Indena Spa CABOZANTINIB-(S)-MALATO AMORPHOUS SOLID DISPERSIONS AND PROCESSES FOR THEIR PREPARATION
US11352344B2 (en) 2017-10-31 2022-06-07 Vanderbilt University Antagonists of the muscarinic acetylcholine receptor M4
US11358949B2 (en) 2017-12-20 2022-06-14 Angex Pharmaceutical, Inc. Carbamate and urea compounds as multikinase inhibitors
US11369623B2 (en) 2015-06-16 2022-06-28 Prism Pharma Co., Ltd. Anticancer combination of a CBP/catenin inhibitor and an immune checkpoint inhibitor
US11407760B2 (en) 2018-02-11 2022-08-09 Beijing Scitech-Mq Pharmaceuticals Limited Dioxinoquinoline compounds, preparation method and uses thereof
US11414406B2 (en) 2018-02-02 2022-08-16 Vanderbilt University Antagonists of the muscarinic acetylcholine receptor M4
WO2022201079A1 (en) * 2021-03-24 2022-09-29 Biocon Limited Process for preparation of cabozantinib
US11498902B2 (en) 2018-06-05 2022-11-15 Natco Pharma Limited Process for the preparation of Cabozantinib and its pharmaceutically acceptable salts thereof
US11542259B2 (en) 2018-01-26 2023-01-03 Exelixis, Inc. Compounds for the treatment of kinase-dependent disorders
US11547705B2 (en) 2015-03-04 2023-01-10 Merck Sharp & Dohme Llc Combination of a PD-1 antagonist and a VEGF-R/FGFR/RET tyrosine kinase inhibitor for treating cancer
RU2789405C2 (en) * 2018-12-12 2023-02-02 Тарапьютикс Сайенс Инк. Pan-kit kinase inhibitor having quinoline structure and its use
WO2023098853A1 (en) * 2021-12-03 2023-06-08 湖南湘源美东医药科技有限公司 Cocrystal of cabozantinib, preparation method therefor and application thereof as drug or in pharmaceutical preparation
WO2023165948A1 (en) 2022-03-01 2023-09-07 Synthon B.V. Cabozantinib salt with l-(+)-tartaric acid and solid forms thereof
WO2023222946A1 (en) 2022-05-18 2023-11-23 Fermion Oy Process for the preparation of cabozantinib

Families Citing this family (67)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7994159B2 (en) 2003-03-10 2011-08-09 Eisai R&D Management Co., Ltd. c-Kit kinase inhibitor
AU2005207946A1 (en) * 2004-01-23 2005-08-11 Amgen Inc. Quinoline quinazoline pyridine and pyrimidine counds and their use in the treatment of inflammation angiogenesis and cancer
KR20080026562A (en) * 2005-06-02 2008-03-25 갤럭시 바이오테크, 엘엘씨 Methods of treating brain tumors with antibodies
WO2007015578A1 (en) 2005-08-02 2007-02-08 Eisai R & D Management Co., Ltd. Method for assay on the effect of vascularization inhibitor
AR059246A1 (en) * 2006-01-30 2008-03-19 Array Biopharma Inc THIOPHENIC HETEROBICICLIC COMPOUNDS AND METHODS OF USE
AR059922A1 (en) * 2006-04-01 2008-05-07 Galaxy Biotech Llc HUMANIZED MONOCLONAL ANTIBODIES FOR THE GROWTH FACTOR OF HEPATOCITS
CA2652442C (en) 2006-05-18 2014-12-09 Eisai R & D Management Co., Ltd. Antitumor agent for thyroid cancer
WO2008035209A2 (en) * 2006-05-30 2008-03-27 Methylgene Inc. Inhibitors of protein tyrosine kinase activity
KR101472600B1 (en) 2006-08-28 2014-12-15 에자이 알앤드디 매니지먼트 가부시키가이샤 Antitumor agent for undifferentiated gastric cancer
CA2676796C (en) 2007-01-29 2016-02-23 Eisai R & D Management Co., Ltd. Composition for treatment of undifferentiated gastric cancer
CA2685967A1 (en) * 2007-05-21 2008-11-21 Novartis Ag Csf-1r inhibitors, compositions, and methods of use
WO2009060945A1 (en) 2007-11-09 2009-05-14 Eisai R & D Management Co., Ltd. Combination of anti-angiogenic substance and anti-tumor platinum complex
CN101977905B (en) 2008-01-23 2014-07-02 百时美施贵宝公司 4-pyridinone compounds and their use for cancer
EP2149565A1 (en) * 2008-07-24 2010-02-03 Bayer Schering Pharma AG Sulfoximine substituted chinazoline derivatives as immune modulators for the treatment of inflammatory and allergic diseases
BRPI0920765A2 (en) * 2008-10-29 2015-08-18 Deciphera Pharmaceuticals Llc Cilopropane amides and analogs that exhibit anticancer and antiproliferative activities
EP2210891A1 (en) * 2009-01-26 2010-07-28 Domain Therapeutics New adenosine receptor ligands and uses thereof
BR112012010524A2 (en) * 2009-11-03 2017-12-12 Glaxosmithkline Llc quinazoline derivatives
JP2013518810A (en) * 2009-11-27 2013-05-23 プロテオロジクス リミテッド Quinazolin-4 (3A) -one derivatives and methods of use thereof
WO2011117741A2 (en) * 2010-03-26 2011-09-29 EastNets Mobile remittance computer system and method
US20120070368A1 (en) * 2010-04-16 2012-03-22 Exelixis, Inc. Methods of Using C-Met Modulators
EP2586443B1 (en) 2010-06-25 2016-03-16 Eisai R&D Management Co., Ltd. Antitumor agent using compounds having kinase inhibitory effect in combination
CN103327979A (en) * 2010-11-22 2013-09-25 葛兰素史密斯克莱知识产权(第2号)有限公司 Method of treating cancer
CA2829131C (en) 2011-03-04 2018-11-20 Glaxosmithkline Intellectual Property (No.2) Limited Amino-quinolines as kinase inhibitors
US20120252840A1 (en) * 2011-04-04 2012-10-04 Exelixis, Inc. Method of Treating Cancer
CA2828946C (en) 2011-04-18 2016-06-21 Eisai R&D Management Co., Ltd. Therapeutic agent for tumor
US8759380B2 (en) 2011-04-22 2014-06-24 Cytokinetics, Inc. Certain heterocycles, compositions thereof, and methods for their use
TWI547494B (en) 2011-08-18 2016-09-01 葛蘭素史克智慧財產發展有限公司 Amino quinazolines as kinase inhibitors
US8843421B2 (en) 2011-11-01 2014-09-23 Accenture Global Services Limited Identification of entities likely to engage in a behavior
TWI577671B (en) 2011-11-14 2017-04-11 Sunshine Lake Pharma Co Ltd Aminoquinazoline derivatives and salts thereof and methods of use thereof
EP2620869B1 (en) 2012-01-27 2017-11-01 Optim Corporation Portable terminal, method, and program of changing user interface
US9358235B2 (en) * 2012-03-19 2016-06-07 Plexxikon Inc. Kinase modulation, and indications therefor
WO2013180128A1 (en) * 2012-05-28 2013-12-05 協和発酵キリン株式会社 4-[4-(5-membered aromatic heterocyclic carbonyl ureide)phenyloxy]quinoline derivative
TW201425307A (en) 2012-09-13 2014-07-01 Glaxosmithkline Llc Amino-quinolines as kinase inhibitors
TWI592417B (en) 2012-09-13 2017-07-21 葛蘭素史克智慧財產發展有限公司 Prodrugs of amino quinazoline kinase inhibitor
CN103804305A (en) * 2012-11-05 2014-05-21 韩文毅 Compound for treating eczema and application thereof
CN103804306A (en) * 2012-11-12 2014-05-21 韩文毅 Compounds for treating diabetes and application thereof
CN104755463A (en) 2012-12-21 2015-07-01 卫材R&D管理有限公司 Amorphous form of quinoline derivative, and method for producing same
WO2014128622A1 (en) 2013-02-21 2014-08-28 Glaxosmithkline Intellectual Property Development Limited Quinazolines as kinase inhibitors
CN105121404A (en) * 2013-03-15 2015-12-02 豪夫迈·罗氏有限公司 Aryl sulfamide and sulfamate derivatives as RORc modulators
ES2813877T3 (en) 2013-08-28 2021-03-25 Crown Bioscience Inc Taicang Gene expression flags predictive of a subject's response to a multikinase inhibitor and methods of using the same
TWI672141B (en) 2014-02-20 2019-09-21 美商醫科泰生技 Molecules for administration to ros1 mutant cancer cells
AU2015223835B2 (en) * 2014-02-28 2019-07-11 Fuso Pharmaceutical Industries, Ltd. Amide derivatives
US9815789B2 (en) 2014-05-23 2017-11-14 Mylan Laboratories, Ltd. Polymorphs of cabozantinib (S)-malate and cabozantinib free base
KR102595599B1 (en) 2014-12-02 2023-11-02 이그니타, 인코포레이티드 Combinations for the treatment of neuroblastoma
US20170020856A1 (en) * 2015-06-29 2017-01-26 Ontogenesis, Llc N-Acylalkyl Prodrugs of Multi-Tyrosine Kinase Inhibitors and Methods of Use
KR20180096621A (en) 2015-12-18 2018-08-29 이그니타, 인코포레이티드 Combination for the treatment of cancer
CN107556238A (en) * 2016-06-30 2018-01-09 深圳万乐药业有限公司 It is a kind of to block the rich synthetic method for Buddhist nun
US9957233B1 (en) 2016-08-05 2018-05-01 Calitor Sciences, Llc Process for preparing substituted quinolin-4-ol compounds
CN109922790B (en) * 2016-09-12 2022-06-14 珠海贝海生物技术有限公司 Cabotinib formulations
WO2018197643A1 (en) * 2017-04-27 2018-11-01 Astrazeneca Ab Phenoxyquinazoline compounds and their use in treating cancer
IL271759B2 (en) 2017-07-19 2024-01-01 Ignyta Inc Pharmaceutical compositions comprising entrectinib
US10180422B1 (en) 2017-08-22 2019-01-15 Scripps Health Methods of treating a neuroendocrine tumor
PE20210128A1 (en) * 2017-08-31 2021-01-19 Abbvie Inc ECTONUCLEOTIDE PYROPHOSPHATASE-PHOSPHODIESTERASE 1 (ENPP-1) INHIBITORS AND USES OF THEM
JP7311498B2 (en) 2017-10-17 2023-07-19 イグナイタ インコーポレイテッド Pharmaceutical compositions and dosage forms
US20190262330A1 (en) * 2017-12-21 2019-08-29 Exelixis, Inc. Method of Treating Hepatocellular Carcinoma Using N-(4-(6,7-dimethoxyquinolin-4-yloxy)phenyl)-N′-(4-fluorophenyl)cyclopropane-1,1-dicarboxamide, (2S)-hydroxybutanedioate
EP3806858A4 (en) 2018-06-15 2022-03-09 Handa Pharmaceuticals, Inc. Kinase inhibitor salts and compositions thereof
WO2020075196A1 (en) 2018-10-11 2020-04-16 Cipla Limited Polymorphs of n-(4-(6,7-dimethoxyquinolin-4-yloxy) phenyl)-n'-(4-fluorophenyl)cyclopropane-1, 1-dicarboxamide, (s)- malate, methods of production and pharmaceutical uses thereof
BR112021011894A2 (en) 2018-12-21 2021-09-08 Daiichi Sankyo Company, Limited PHARMACEUTICAL COMPOSITION
CN114025755A (en) * 2019-04-12 2022-02-08 台湾卫生研究院 Therapeutic use of heterocyclic compounds as kinase inhibitors
CN112778217B (en) * 2019-11-08 2024-01-26 沈阳化工研究院有限公司 Quinazoline compound and application thereof
CN111393363B (en) * 2020-04-27 2022-12-20 威海海洋生物医药产业技术研究院有限公司 4-phenoxy quinoline and N-sulfonyl amidine compound and preparation method and application thereof
CA3202761A1 (en) 2020-11-25 2022-06-02 Nanocopoeia, Llc Amorphous cabozantinib particles and uses thereof
IL303655A (en) * 2020-12-17 2023-08-01 Astrazeneca Ab N-(2-(4-cyanothiazolidin-3-yl)-2-oxoethyl)- quinoline-4-carboxamides
WO2023081923A1 (en) 2021-11-08 2023-05-11 Frequency Therapeutics, Inc. Platelet-derived growth factor receptor (pdgfr) alpha inhibitors and uses thereof
CN114605391B (en) * 2022-02-21 2024-01-26 广州六顺生物科技股份有限公司 Quinoxaline derivative, preparation method and application thereof
WO2023228095A1 (en) 2022-05-24 2023-11-30 Daiichi Sankyo Company, Limited Dosage regimen of an anti-cdh6 antibody-drug conjugate
WO2024008929A1 (en) 2022-07-08 2024-01-11 Astrazeneca Ab Epidermal growth factor receptor tyrosine kinase inhibitors in combination with hgf-receptor inhibitors for the treatment of cancer

Family Cites Families (170)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US506181A (en) 1893-10-03 Folding crate
GB2160201B (en) * 1984-06-14 1988-05-11 Wyeth John & Brother Ltd Quinazoline and cinnoline derivatives
US5123951A (en) * 1986-03-31 1992-06-23 Rhone-Poulenc Nederland B.V. Synergistic plant growth regulator compositions
JPS646261A (en) * 1987-03-31 1989-01-10 Nisshin Flour Milling Co 4-thioquinazoline derivative, its production and antiulcer agent containing said derivative as active component
US5143854A (en) * 1989-06-07 1992-09-01 Affymax Technologies N.V. Large scale photolithographic solid phase synthesis of polypeptides and receptor binding screening thereof
US5034393A (en) * 1989-07-27 1991-07-23 Dowelanco Fungicidal use of pyridopyrimidine, pteridine, pyrimidopyrimidine, pyrimidopyridazine, and pyrimido-1,2,4-triazine derivatives
US5238951A (en) 1991-02-01 1993-08-24 E. R. Squibb & Sons, Inc. Heterocyclic amido prostaglandin analogs
DE4114733A1 (en) * 1991-05-06 1992-11-12 Huels Chemische Werke Ag METHOD FOR PRODUCING SUBSTITUTED MALONESTERANILIDES AND MALONIC ACID MONOANILIDES
US5480883A (en) * 1991-05-10 1996-01-02 Rhone-Poulenc Rorer Pharmaceuticals Inc. Bis mono- and bicyclic aryl and heteroaryl compounds which inhibit EGF and/or PDGF receptor tyrosine kinase
US5710158A (en) * 1991-05-10 1998-01-20 Rhone-Poulenc Rorer Pharmaceuticals Inc. Aryl and heteroaryl quinazoline compounds which inhibit EGF and/or PDGF receptor tyrosine kinase
US5358941A (en) 1992-12-02 1994-10-25 Merck & Co., Inc. Dry mix formulation for bisphosphonic acids with lactose
US6498144B1 (en) 1993-10-18 2002-12-24 North Shore - Long Island Jewish Research Institute Use of scatter factor to enhance angiogenesis
IL112249A (en) 1994-01-25 2001-11-25 Warner Lambert Co Pharmaceutical compositions containing di and tricyclic pyrimidine derivatives for inhibiting tyrosine kinases of the epidermal growth factor receptor family and some new such compounds
GB9510757D0 (en) 1994-09-19 1995-07-19 Wellcome Found Therapeuticaly active compounds
TW321649B (en) 1994-11-12 1997-12-01 Zeneca Ltd
GB9508538D0 (en) 1995-04-27 1995-06-14 Zeneca Ltd Quinazoline derivatives
US5747498A (en) 1996-05-28 1998-05-05 Pfizer Inc. Alkynyl and azido-substituted 4-anilinoquinazolines
CA2222545A1 (en) 1995-06-07 1996-12-19 Sugen, Inc. Quinazolines and pharmaceutical compositions
ES2308962T3 (en) 1995-06-07 2008-12-16 Glaxosmithkline Biologicals S.A. VACCINES UNDERSTANDING A CONJUGATE OF ANTIGEN POLISACARIDO-PROTEINA TRANSPORADORA AND A PROTEIN FREE CARRIER.
US5650415A (en) 1995-06-07 1997-07-22 Sugen, Inc. Quinoline compounds
GB9514265D0 (en) 1995-07-13 1995-09-13 Wellcome Found Hetrocyclic compounds
WO1997017329A1 (en) * 1995-11-07 1997-05-15 Kirin Beer Kabushiki Kaisha Quinoline derivatives and quinazoline derivatives inhibiting autophosphorylation of growth factor receptor originating in platelet and pharmaceutical compositions containing the same
GB9523675D0 (en) 1995-11-20 1996-01-24 Celltech Therapeutics Ltd Chemical compounds
GB9624482D0 (en) 1995-12-18 1997-01-15 Zeneca Phaema S A Chemical compounds
NZ325248A (en) 1995-12-23 1999-09-29 Pfizer Res & Dev Quinoline and quinazoline compounds useful in therapy
TR199801530T2 (en) 1996-02-13 1998-11-23 Zeneca Limited Quinazoline derivatives as VEGF inhibitors.
GB9603095D0 (en) 1996-02-14 1996-04-10 Zeneca Ltd Quinazoline derivatives
JP4464466B2 (en) 1996-03-05 2010-05-19 アストラゼネカ・ユーケイ・リミテッド 4-anilinoquinazoline derivatives
EP0888310B1 (en) 1996-03-15 2005-09-07 AstraZeneca AB Cinnoline derivatives and use as medicine
US6107300A (en) 1996-03-27 2000-08-22 Dupont Pharmaceuticals Arylamino fused pyrimidines
PL190489B1 (en) 1996-04-12 2005-12-30 Warner Lambert Co Irreversible inhibitors of tyrosine kinases
ES2186908T3 (en) 1996-07-13 2003-05-16 Glaxo Group Ltd HETEROCICICLES CONDENSED COMPOUNDS AS INHIBITORS OF PPROTEINA-TIROSINA-QUINASAS.
HRP970371A2 (en) 1996-07-13 1998-08-31 Kathryn Jane Smith Heterocyclic compounds
GB9718972D0 (en) 1996-09-25 1997-11-12 Zeneca Ltd Chemical compounds
NZ334125A (en) 1996-09-25 2000-10-27 Zeneca Ltd Quinoline derivatives inhibiting the effect of growth factors such as VEGF
CA2239227C (en) 1996-10-01 2007-10-30 Kenji Matsuno Nitrogen-containing heterocyclic compounds
GB9700504D0 (en) 1997-01-11 1997-02-26 Pfizer Ltd Pharmaceutical compounds
GB9705361D0 (en) 1997-03-14 1997-04-30 Celltech Therapeutics Ltd Chemical compounds
UA73073C2 (en) 1997-04-03 2005-06-15 Уайт Холдінгз Корпорейшн Substituted 3-cyan chinolines
EP0990647B1 (en) 1997-04-18 2003-07-02 Kirin Beer Kabushiki Kaisha Process for producing quinolone derivatives
AU7644698A (en) 1997-04-22 1998-11-13 Janssen Pharmaceutica N.V. Crf antagonistic quino- and quinazolines
GB9708917D0 (en) 1997-05-01 1997-06-25 Pfizer Ltd Compounds useful in therapy
AR012634A1 (en) 1997-05-02 2000-11-08 Sugen Inc QUINAZOLINE BASED COMPOUND, FAMACEUTICAL COMPOSITION THAT UNDERSTANDS IT, METHOD TO SYNTHESIZE IT, ITS USE, METHODS OF MODULATION OF THE DESERINE / TREONIN PROTEIN-KINASE FUNCTION AND IN VITRO METHOD TO IDENTIFY COMPOUNDS THAT MODULATE
ZA986732B (en) 1997-07-29 1999-02-02 Warner Lambert Co Irreversible inhibitiors of tyrosine kinases
ATE368665T1 (en) 1997-08-22 2007-08-15 Astrazeneca Ab OXINDOLYLQUINAZOLINE DERIVATIVES AS ANGIOGENESIS INHIBITORS
JP4194678B2 (en) * 1997-11-28 2008-12-10 キリンファーマ株式会社 Quinoline derivative and pharmaceutical composition containing the same
GB9800569D0 (en) 1998-01-12 1998-03-11 Glaxo Group Ltd Heterocyclic compounds
AU4317399A (en) 1998-05-28 1999-12-13 Parker Hughes Institute Quinazolines for treating brain tumor
ATE459616T1 (en) 1998-08-11 2010-03-15 Novartis Ag ISOCHINOLINE DERIVATIVES WITH ANGIOGENESIS-INHIBITING EFFECT
WO2000010981A1 (en) 1998-08-21 2000-03-02 Parker Hughes Institute Quinazoline derivatives
US6184226B1 (en) 1998-08-28 2001-02-06 Scios Inc. Quinazoline derivatives as inhibitors of P-38 α
EP1143950B1 (en) 1998-09-10 2005-03-09 BioEqual AG Topical application products against onychomycoses
BR9914167B1 (en) 1998-09-29 2011-03-09 pharmaceutical compounds and compositions comprising substituted 3-cyano quinolines.
US6288082B1 (en) * 1998-09-29 2001-09-11 American Cyanamid Company Substituted 3-cyanoquinolines
IL142257A0 (en) 1998-10-01 2002-03-10 Astrazeneca Ab Amide derivatives, process for their preparation, compositions containing them and use thereof in the manufacture of a medicament for the treatment of cytokine-mediated diseases
US7262201B1 (en) 1998-10-08 2007-08-28 Astrazeneca Ab Quinazoline derivatives
ES2188254T3 (en) 1998-11-19 2003-06-16 Warner Lambert Co N- (4- (3-CHLORO-4-FLUORO-PHENYLAMINE) -7- (3-MORFOLIN-4-IL-PROPOXI) -QUIN AZOLIN-6-IL) -ACRILAMADA, AN IRREVERSIBLE THYROSINE KINASE INHIBITOR.
ATE253051T1 (en) * 1999-01-22 2003-11-15 Kirin Brewery QUINOLINE DERIVATIVES AND QUINAZOLINE DERIVATIVES
CZ306810B6 (en) 1999-02-10 2017-07-19 Astrazeneca Ab The use of a quinazoline derivative as an inhibitor of angiogenesis
GB9904103D0 (en) 1999-02-24 1999-04-14 Zeneca Ltd Quinoline derivatives
US6080747A (en) 1999-03-05 2000-06-27 Hughes Institute JAK-3 inhibitors for treating allergic disorders
DE19911509A1 (en) 1999-03-15 2000-09-21 Boehringer Ingelheim Pharma Bicyclic heterocycles, medicaments containing these compounds, their use and processes for their preparation
JP2002539262A (en) 1999-03-19 2002-11-19 パーカー ヒューズ インスティテュート Quinazoline compound formulations and their use in therapy
US6258820B1 (en) 1999-03-19 2001-07-10 Parker Hughes Institute Synthesis and anti-tumor activity of 6,7-dialkoxy-4-phenylamino-quinazolines
YU13200A (en) 1999-03-31 2002-10-18 Pfizer Products Inc. Process and intermediates for preparing anti-cancer compounds
GB9910577D0 (en) 1999-05-08 1999-07-07 Zeneca Ltd Chemical compounds
GB9910580D0 (en) * 1999-05-08 1999-07-07 Zeneca Ltd Chemical compounds
US6126917A (en) 1999-06-01 2000-10-03 Hadasit Medical Research Services And Development Ltd. Epidermal growth factor receptor binding compounds for positron emission tomography
YU90901A (en) 1999-06-21 2004-07-15 Boehringer Ingelheim Pharma Gmbh. & Co.Kg. Bicyclic heterocycles, medicaments containing these compounds, their use and methods for the production thereof
GB9922171D0 (en) 1999-09-21 1999-11-17 Zeneca Ltd Chemical compounds
RU2002110461A (en) * 1999-09-21 2004-03-10 Астразенека Аб (Se) Quinazoline derivatives and their use as pharmaceutical substances
US6759410B1 (en) 1999-11-23 2004-07-06 Smithline Beecham Corporation 3,4-dihydro-(1H)-quinazolin-2-ones and their use as CSBP/p38 kinase inhibitors
JP2003518023A (en) 1999-11-30 2003-06-03 パーカー ヒューズ インスティテュート Inhibitors of thrombin-induced platelet aggregation
US20020002169A1 (en) 1999-12-08 2002-01-03 Griffin John H. Protein kinase inhibitors
WO2001047890A1 (en) * 1999-12-24 2001-07-05 Kirin Beer Kabushiki Kaisha Quinoline and quinazoline derivatives and drugs containing the same
US6525046B1 (en) 2000-01-18 2003-02-25 Boehringer Ingelheim Pharmaceuticals, Inc. Aromatic heterocyclic compounds as antiinflammatory agents
WO2001055116A2 (en) * 2000-01-28 2001-08-02 Astrazeneca Ab Quinoline derivatives and their use as aurora 2 kinase inhibitors
US6664390B2 (en) 2000-02-02 2003-12-16 Warner-Lambert Company Llc Method for the simplified production of (3-chloro-4-fluorophenyl)-[7-(3-morpholin-4-yl-propoxy)-6-nitro-quinazoline-4-yl]-amine or (3-chloro-4-fluorophenyl)-[7-(3-morpholin-4-yl-propoxy)-6-amino-quinazoline-4-yl]-amine
AU2001236698A1 (en) 2000-02-07 2001-08-14 Abbott Gesellschaft Mit Beschrankter Haftung & Company Kommanditgesellschaft 2-benzothiazolyl urea derivatives and their use as protein kinase inhibitors
JP2003526686A (en) 2000-03-13 2003-09-09 アメリカン・サイアナミド・カンパニー Use of cyanoquinoline to treat or inhibit colon polyps
US6608048B2 (en) 2000-03-28 2003-08-19 Wyeth Holdings Tricyclic protein kinase inhibitors
US6521618B2 (en) 2000-03-28 2003-02-18 Wyeth 3-cyanoquinolines, 3-cyano-1,6-naphthyridines, and 3-cyano-1,7-naphthyridines as protein kinase inhibitors
US6627634B2 (en) 2000-04-08 2003-09-30 Boehringer Ingelheim Pharma Kg Bicyclic heterocycles, pharmaceutical compositions containing them, their use, and processes for preparing them
UA73993C2 (en) 2000-06-06 2005-10-17 Астразенека Аб Quinazoline derivatives for the treatment of tumours and a pharmaceutical composition
CA2413424C (en) 2000-06-22 2007-10-02 Pfizer Products Inc. Substituted bicyclic derivatives for the treatment of abnormal cell growth
SK18102002A3 (en) 2000-06-28 2003-07-01 Astrazeneca Ab Substituted quinazoline derivatives, their use and a composition comprising said derivatives
EP1174118A1 (en) 2000-06-28 2002-01-23 Cognis France S.A. Use of inulin and derivatives thereof
FR2811658B1 (en) 2000-07-17 2004-07-02 Cfpi Nufarm BIOLOGICAL REACTOR WITH SUBMERGED FIXED BED AND METHOD FOR TREATING LIQUID EFFLUENTS
JP2002030083A (en) * 2000-07-18 2002-01-29 Kirin Brewery Co Ltd N-(2-chloro-4-{[6-methoxy-7-(3-pyridylmethoxy)-4- quinolyl]oxy}phenyl)-n'-propylurea dihydrochloride
US7427689B2 (en) * 2000-07-28 2008-09-23 Georgetown University ErbB-2 selective small molecule kinase inhibitors
CZ2003486A3 (en) 2000-08-21 2003-05-14 Astrazeneca Ab Quinazoline derivatives, process of their preparation and pharmaceutical preparation in which the derivatives are comprised
US6617329B2 (en) 2000-08-26 2003-09-09 Boehringer Ingelheim Pharma Kg Aminoquinazolines and their use as medicaments
US6653305B2 (en) 2000-08-26 2003-11-25 Boehringer Ingelheim Pharma Kg Bicyclic heterocycles, pharmaceutical compositions containing them, their use, and processes for preparing them
US6403580B1 (en) 2000-08-26 2002-06-11 Boehringer Ingelheim Pharma Kg Quinazolines, pharmaceutical compositions containing these compounds, their use and processes for preparing them
US6656946B2 (en) 2000-08-26 2003-12-02 Boehringer Ingelheim Pharma Kg Aminoquinazolines which inhibit signal transduction mediated by tyrosine kinases
US6740651B2 (en) 2000-08-26 2004-05-25 Boehringer Ingelheim Pharma Kg Aminoquinazolines which inhibit signal transduction mediated by tyrosine kinases
DE10042058A1 (en) 2000-08-26 2002-03-07 Boehringer Ingelheim Pharma Bicyclic heterocycles, medicaments containing these compounds, their use and processes for their preparation
JP2004511479A (en) 2000-10-13 2004-04-15 アストラゼネカ アクチボラグ Quinazoline derivatives
US6939866B2 (en) 2000-10-13 2005-09-06 Astrazeneca Ab Quinazoline derivatives
ES2318649T3 (en) * 2000-10-20 2009-05-01 EISAI R&D MANAGEMENT CO., LTD. PROCEDURE FOR PREPARATION OF DERIVATIVES OF 4-FENOXI QUINOLINAS.
WO2002034744A1 (en) 2000-10-25 2002-05-02 Astrazeneca Ab Quinazoline derivatives
US7253184B2 (en) 2000-11-02 2007-08-07 Astrazeneca Ab 4-Substituted quinolines as antitumor agents
US7067532B2 (en) 2000-11-02 2006-06-27 Astrazeneca Substituted quinolines as antitumor agents
ATE383860T1 (en) 2000-11-02 2008-02-15 Nippon Shinyaku Co Ltd CHINAZOLINE DERIVATIVES AND DRUGS
US7019012B2 (en) 2000-12-20 2006-03-28 Boehringer Ingelheim International Pharma Gmbh & Co. Kg Quinazoline derivatives and pharmaceutical compositions containing them
US6900220B2 (en) 2001-01-02 2005-05-31 Syntex (U.S.A.) Llc Quinazolone derivatives as alpha 1A/B adrenergic receptor antagonists
ES2312557T3 (en) 2001-04-19 2009-03-01 Astrazeneca Ab DERIVATIVES OF QUINAZOLINA.
ES2256466T3 (en) 2001-04-27 2006-07-16 Kirin Beer Kabushiki Kaisha DERIVATIVE OF QUINOLINA THAT HAVE GRUPO AZOLILO AND DERIVATIVES OF QUINAZOLINA.
SE0101675D0 (en) 2001-05-11 2001-05-11 Astrazeneca Ab Novel composition
WO2002092577A1 (en) 2001-05-14 2002-11-21 Astrazeneca Ab Quinazoline derivatives
WO2002092579A1 (en) 2001-05-14 2002-11-21 Astrazeneca Ab 4-anilinoquinazoline derivatives
WO2002092578A1 (en) 2001-05-14 2002-11-21 Astrazeneca Ab Quinazoline derivatives
US6734303B2 (en) 2001-05-18 2004-05-11 Pfizer Inc. Process for the production of quinazolines
DE10125432A1 (en) * 2001-05-25 2002-11-28 Bayer Ag New benzoyl-substituted aliphatic ketones or aldehydes, useful as herbicides or fungicides, especially as pre- and post-emergence selective herbicides against a broad spectrum of weeds
AU2002350105A1 (en) 2001-06-21 2003-01-08 Ariad Pharmaceuticals, Inc. Novel quinazolines and uses thereof
CN100415720C (en) * 2001-06-22 2008-09-03 麒麟医药株式会社 Quinoline derivative and quinazoline derivative inhibiting self-phosphorylation of hepatocytus proliferator receptor and medicinal composition containing the same
KR100397792B1 (en) 2001-06-28 2003-09-13 한국과학기술연구원 4-(phenylamino)-[1,4]dioxano[2,3-g]quinazoline Derivatives and Process for Preparing the Same
GB0118752D0 (en) 2001-08-01 2001-09-26 Pfizer Ltd Process for the production of quinazolines
US7229774B2 (en) 2001-08-02 2007-06-12 Regents Of The University Of Michigan Expression profile of prostate cancer
US20030066060A1 (en) 2001-09-28 2003-04-03 Ford Richard L. Cross profile guided optimization of program execution
JP4383870B2 (en) * 2001-10-17 2009-12-16 協和発酵キリン株式会社 Quinoline derivatives and quinazoline derivatives that inhibit fibroblast growth factor receptor autophosphorylation and pharmaceutical compositions containing them
US7169788B2 (en) 2001-10-30 2007-01-30 Merck & Co., Inc. Tyrosine kinase inhibitors
GB0126433D0 (en) 2001-11-03 2002-01-02 Astrazeneca Ab Compounds
GB0128108D0 (en) 2001-11-23 2002-01-16 Astrazeneca Ab Therapeutic use
PL370137A1 (en) 2001-11-27 2005-05-16 Wyeth Holdings Corporation 3-cyanoquinolines as inhibitors of egf-r and her2 kinases
AU2002347336A1 (en) 2001-12-05 2003-06-17 Astrazeneca Ab Quinoline derivatives
GB0129099D0 (en) 2001-12-05 2002-01-23 Astrazeneca Ab Chemical compounds
WO2003047584A1 (en) 2001-12-05 2003-06-12 Astrazeneca Ab Quinoline derivatives
GEP20063872B (en) 2001-12-12 2006-07-10 Pfizer Prod Inc Salt forms of e-2-methoxy-n-(3-{4-[3-methyl-4-(6-methyl-pyridin-3-yloxy)-phenylamino]-quina-zolin-6-yl}-allyl)-acetamide and method of production
CN1602195A (en) 2001-12-12 2005-03-30 辉瑞产品公司 Quinazoline derivatives for the treatment of abnormal cell growth
AU2002361846A1 (en) 2001-12-21 2003-07-15 Bayer Pharmaceuticals Corporation Quinazoline and quinoline derivative compounds as inhibitors of prolylpeptidase, inducers of apoptosis and cancer treatment agents
TW200301123A (en) 2001-12-21 2003-07-01 Astrazeneca Uk Ltd New use
HU229477B1 (en) 2001-12-24 2014-01-28 Astrazeneca Ab Substituted quinazoline derivatives as inhibitors of aurora kinases
JP4445753B2 (en) 2002-01-29 2010-04-07 グラクソ グループ リミテッド Aminopiperidine derivatives
WO2003064431A2 (en) 2002-01-29 2003-08-07 Glaxo Group Limited Aminopiperidine compounds, process for their preparation, and pharmaceutical compositions containing them
US7268230B2 (en) 2002-02-01 2007-09-11 Astrazeneca Ab Quinazoline compounds
DE10204462A1 (en) 2002-02-05 2003-08-07 Boehringer Ingelheim Pharma Use of tyrosine kinase inhibitors for the treatment of inflammatory processes
AU2003225668A1 (en) * 2002-03-01 2003-09-16 Pintex Pharmaceutical, Inc. Pin1-modulating compounds and methods of use thereof
DE10217689A1 (en) 2002-04-19 2003-11-13 Boehringer Ingelheim Pharma Bicyclic heterocycles, medicaments containing these compounds, their use and processes for their preparation
US7598258B2 (en) * 2002-05-01 2009-10-06 Kirin Beer Kabushiki Kaisha Quinoline derivatives and quinazoline derivatives inhibiting autophosphorylation of macrophage colony stimulating factor receptor
EP1521747B1 (en) 2002-07-15 2018-09-05 Symphony Evolution, Inc. Receptor-type kinase modulators and methods of use
AU2003257666A1 (en) * 2002-08-23 2004-03-11 Kirin Beer Kabushiki Kaisha COMPOUND HAVING TGFss INHIBITORY ACTIVITY AND MEDICINAL COMPOSITION CONTAINING THE SAME
GB0219746D0 (en) 2002-08-23 2002-10-02 Inst Of Ex Botany Ascr Azapurine derivatives
PT1559715E (en) 2002-10-21 2007-10-24 Kirin Pharma Kk N-{2-chloro-4-[(6,7-dimethoxy-4-quinolyl)oxy]phenyl}-n`-(5-methyl-3-isoxazolyl)urea salt in crystalline form
JPWO2004039782A1 (en) * 2002-10-29 2006-03-02 麒麟麦酒株式会社 Quinoline derivatives and quinazoline derivatives that inhibit Flt3 autophosphorylation and pharmaceutical compositions containing them
PT1562955E (en) 2002-11-04 2008-05-05 Astrazeneca Ab Quinazoline derivatives as src tyrosine kinase inhibitors
JP2006515846A (en) 2002-12-13 2006-06-08 ニューロジェン・コーポレーション 2-Substituted quinazolin-4-ylamine analogues as capsaicin receptor modulators
CA2510323A1 (en) 2002-12-18 2004-07-01 Pfizer Products Inc. 4-anilino quinazoline derivatives for the treatment of abnormal cell growth
US7238679B2 (en) 2002-12-23 2007-07-03 Ariad Pharmaceuticals, Inc. Heterocycles and uses thereof
AU2003292838A1 (en) * 2002-12-27 2004-07-29 Kirin Beer Kabushiki Kaisha Therapeutic agent for wet age-related macular degeneration
US8176532B1 (en) * 2003-03-17 2012-05-08 Sprint Communications Company L.P. Secure access point for scada devices
KR100559180B1 (en) * 2003-05-20 2006-03-14 김민서 Electronic settlement method and server according to conditional trade
WO2005003140A1 (en) 2003-07-02 2005-01-13 Pharmacia & Upjohn Company Llc 4-oxo-4,7-dihydrothieno[2,3-b]pyridine-5-carboxamides as antiviral agents
WO2005005389A2 (en) 2003-07-07 2005-01-20 Merck Patent Gmbh Malonamide derivatives
EP2210607B1 (en) * 2003-09-26 2011-08-17 Exelixis Inc. N-[3-fluoro-4-({6-(methyloxy)-7-[(3-morpholin-4-ylpropyl)oxy]quinolin-4-yl}oxy)phenyl]-N'-(4-fluorophenyl)cyclopropane-1,1-dicarboxamide for the treatment of cancer
AU2005207946A1 (en) 2004-01-23 2005-08-11 Amgen Inc. Quinoline quinazoline pyridine and pyrimidine counds and their use in the treatment of inflammation angiogenesis and cancer
US20050288290A1 (en) * 2004-06-28 2005-12-29 Borzilleri Robert M Fused heterocyclic kinase inhibitors
WO2006014325A2 (en) * 2004-07-02 2006-02-09 Exelixis, Inc. C-met modulators and method of use
WO2006108059A1 (en) * 2005-04-06 2006-10-12 Exelixis, Inc. C-met modulators and methods of use
US7594258B2 (en) * 2005-06-27 2009-09-22 Yahoo! Inc. Access control systems and methods using visibility tokens with automatic propagation
WO2008035209A2 (en) * 2006-05-30 2008-03-27 Methylgene Inc. Inhibitors of protein tyrosine kinase activity
EP2349328A1 (en) 2008-10-01 2011-08-03 Ludwig Institute For Cancer Research Methods for the treatment of cancer
JP2012511017A (en) 2008-12-04 2012-05-17 エグゼリクシス, インコーポレイテッド Method for preparing quinoline derivatives
KR20200137052A (en) 2009-01-16 2020-12-08 엑셀리시스, 인코포레이티드 Malate salt of n-(4-{[6,7-bis(methyloxy)quinolin-4-yl]oxy}phenyl)-n'-(4-fluorophenyl)cyclopropane-1,1-dicarboxamide, and crystalline forms thereof for the treatment of cancer
KR20120051702A (en) 2009-07-17 2012-05-22 엑셀리시스, 인코포레이티드 Crystalline forms of n-[3-fluoro-4-({6-(methyloxy)-7-[(3-morpholin-4-ylpropyl)oxy]-quinolin-4-yl}oxy)phenyl]-n'-(4-fluorophenyl)cyclopropane-1,1-dicarboxamide
UA108618C2 (en) 2009-08-07 2015-05-25 APPLICATION OF C-MET-MODULATORS IN COMBINATION WITH THEMOSOLOMID AND / OR RADIATION THERAPY FOR CANCER TREATMENT
EP2475390A4 (en) 2009-09-09 2014-01-01 Quintiles Transnat Corp Methods and compositions for the treatment of receptor tyrosine kinase mediated diseases or disorders
JP5461213B2 (en) 2010-01-29 2014-04-02 三洋電機株式会社 Air conditioner outdoor unit
US20120070368A1 (en) 2010-04-16 2012-03-22 Exelixis, Inc. Methods of Using C-Met Modulators
US20120252840A1 (en) 2011-04-04 2012-10-04 Exelixis, Inc. Method of Treating Cancer

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
S. M. BERGE ET AL.: "Pharmaceutical Salts", J. PHARM. SCI., vol. 66, 1977, pages 1 - 19

Cited By (451)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8372981B2 (en) 2000-10-20 2013-02-12 Eisai R&D Management Co., Ltd. Nitrogen-containing aromatic derivatives
US7973160B2 (en) 2000-10-20 2011-07-05 Eisai R&D Management Co., Ltd. Nitrogen-containing aromatic derivatives
US8318752B2 (en) 2003-09-19 2012-11-27 Astrazeneca Ab 4-(3-chloro-2-fluoroanilino)-7-methoxy-6-{[1-(N-methylcarbamoyl-methyl)piperidin-4-yl]oxy}quinazoline, its pharmaceutically acceptable salts, and pharmaceutical compositions comprising the same
US11124482B2 (en) 2003-09-26 2021-09-21 Exelixis, Inc. C-met modulators and methods of use
US8058474B2 (en) 2003-11-11 2011-11-15 Eisai R&D Management Co., Ltd. Urea derivative and process for preparing the same
US7767670B2 (en) 2003-11-13 2010-08-03 Ambit Biosciences Corporation Substituted 3-carboxamido isoxazoles as kinase modulators
WO2005082855A1 (en) * 2004-02-27 2005-09-09 Eisai Co., Ltd. Novel pyridine derivative and pyrimidine derivative (2)
US7531532B2 (en) 2004-02-27 2009-05-12 Eisai R&D Management Co., Ltd. Pyridine derivative and pyrimidine derivative
AU2005217328B2 (en) * 2004-02-27 2007-10-04 Eisai R & D Management Co., Ltd. Novel pyridine derivative and pyrimidine derivative (2)
WO2005082854A1 (en) * 2004-02-27 2005-09-09 Eisai Co., Ltd. Novel pyridine derivative and pyrimidine derivative (1)
US8481536B2 (en) 2004-04-08 2013-07-09 Targegen, Inc. Benzotriazine inhibitors of kinases
JP2008510839A (en) * 2004-08-25 2008-04-10 ターゲジェン インコーポレーティッド Heterocyclic compounds and methods of use
US8372971B2 (en) 2004-08-25 2013-02-12 Targegen, Inc. Heterocyclic compounds and methods of use
US8969379B2 (en) 2004-09-17 2015-03-03 Eisai R&D Management Co., Ltd. Pharmaceutical compositions of 4-(3-chloro-4-(cyclopropylaminocarbonyl)aminophenoxy)-7=methoxy-6-quinolinecarboxide
US7951805B2 (en) 2004-09-20 2011-05-31 Xenon Pharmaceuticals Inc. Heterocyclic derivatives and their use as mediators of stearoyl-CoA desaturase
US7767677B2 (en) 2004-09-20 2010-08-03 Xenon Pharmaceuticals Inc. Heterocyclic derivatives and their use as stearoyl-CoA desaturase inhibitors
US8026360B2 (en) 2004-09-20 2011-09-27 Xenon Pharmaceuticals Inc. Substituted pyridazines as stearoyl-CoA desaturase inhibitors
US7919496B2 (en) 2004-09-20 2011-04-05 Xenon Pharmaceuticals Inc. Heterocyclic derivatives for the treatment of diseases mediated by stearoyl-CoA desaturase enzymes
US7777036B2 (en) 2004-09-20 2010-08-17 Xenon Pharmaceuticals Inc. Heterocyclic derivatives and their use as therapeutic agents
US7829712B2 (en) 2004-09-20 2010-11-09 Xenon Pharmaceuticals Inc. Pyridazine derivatives for inhibiting human stearoyl-CoA-desaturase
US8071603B2 (en) 2004-09-20 2011-12-06 Xenon Pharmaceuticals Inc. Heterocyclic derivatives and their use as stearoyl-CoA desaturase inhibitors
US8153643B2 (en) 2004-10-12 2012-04-10 Astrazeneca Ab Quinazoline derivatives
US7652009B2 (en) 2004-11-30 2010-01-26 Amgem Inc. Substituted heterocycles and methods of use
WO2006060318A3 (en) * 2004-11-30 2006-07-20 Amgen Inc Quinolines and quinazoline analogs and their use as medicaments for treating cancer
EA013863B1 (en) * 2004-12-27 2010-08-30 4-Эс-Ц АГ 2,5 and 2,6-substituted benzazole analogues useful as protein kinase inhibitors
AU2005321517B2 (en) * 2004-12-27 2011-09-29 4Sc Ag 2, 5 and 2, 6-disubstituted benzazole analogues useful as protein kinase inhibitors
US7576090B2 (en) * 2004-12-27 2009-08-18 4Sc Ag Benzazole analogues and uses thereof
WO2006069740A1 (en) * 2004-12-27 2006-07-06 4Sc Ag 2, 5 and 2, 6-disubstituted benzazole analogues useful as protein kinase inhibitors
EP1674466A1 (en) * 2004-12-27 2006-06-28 4Sc Ag 2,5- and 2,6-disubstituted benzazole analogues useful as protein kinase inhibitors
US8088794B2 (en) 2005-04-27 2012-01-03 Amgen Inc. Substituted amide derivatives and methods of use
US7858623B2 (en) 2005-04-27 2010-12-28 Amgen Inc. Substituted amide derivatives and methods of use
JP2009504804A (en) * 2005-05-20 2009-02-05 メチルジーン インコーポレイテッド Inhibitors of VEGF receptor and HGF receptor signaling
US8541457B2 (en) 2005-06-03 2013-09-24 Xenon Pharmaceuticals Inc. Aminothiazole derivatives as human stearoyl-CoA desaturase inhibitors
US8288538B2 (en) 2005-08-24 2012-10-16 Eisai R&D Management Co., Ltd. Pyridine derivatives and pyrimidine derivatives (3)
US7855290B2 (en) 2005-08-24 2010-12-21 Eisai R&D Management Co., Ltd. Pyridine derivatives and pyrimidine derivatives (3)
US7732465B2 (en) 2005-08-30 2010-06-08 Novartis Vaccines And Diagnostics, Inc. Substituted benzimidazoles and methods of their use
US7767820B2 (en) 2005-08-30 2010-08-03 Novartis Vaccines And Diagnostics, Inc. Substituted benzimidazoles and methods of preparation
US8592459B2 (en) 2005-08-30 2013-11-26 Novartis Ag Substituted benzimidazoles and methods of their use
US7732613B2 (en) 2005-09-14 2010-06-08 Bristol-Myers Squibb Company Met kinase inhibitors
EP1964837A1 (en) * 2005-11-22 2008-09-03 Eisai R&D Management Co., Ltd. Anti-tumor agent for multiple myeloma
EP1964837A4 (en) * 2005-11-22 2010-12-22 Eisai R&D Man Co Ltd Anti-tumor agent for multiple myeloma
US7973164B2 (en) 2006-03-02 2011-07-05 Astrazeneca Ab Quinoline derivatives
WO2007103308A2 (en) 2006-03-07 2007-09-13 Array Biopharma Inc. Heterobicyclic pyrazole compounds and methods of use
US8710048B2 (en) 2006-04-19 2014-04-29 Novartis Ag 6-O-substituted benzoxazole and benzothiazole compounds and methods of inhibiting CSF-1R signaling
JP2009534410A (en) * 2006-04-19 2009-09-24 ノバルティス アクチエンゲゼルシャフト 6-O-substituted benzoxazole and benzothiazole compounds and methods for inhibiting CSF-1R signaling
US8173689B2 (en) 2006-04-19 2012-05-08 Novartis Ag 6-O-substituted benzoxazole and benzothiazole compounds and methods of inhibiting CSF-1R signaling
US7872017B2 (en) 2006-05-19 2011-01-18 Abbott Laboratories Fused bicycloheterocycle substituted azabicyclic alkane derivatives
JP2009539878A (en) * 2006-06-08 2009-11-19 アレイ バイオファーマ、インコーポレイテッド Quinoline compounds and methods of use
US7683060B2 (en) 2006-08-07 2010-03-23 Incyte Corporation Triazolotriazines as kinase inhibitors
US7915408B2 (en) 2006-08-07 2011-03-29 Incyte Corporation Triazolotriazines as kinase inhibitors
US8143251B2 (en) 2006-08-07 2012-03-27 Incyte Corporation Triazolotriazines as kinase inhibitors
WO2008023698A1 (en) * 2006-08-23 2008-02-28 Eisai R & D Management Co., Ltd. Salt of phenoxypyridine derivative or crystal thereof and process for producing the same
JP5190365B2 (en) * 2006-08-23 2013-04-24 エーザイ・アール・アンド・ディー・マネジメント株式会社 Salt of phenoxypyridine derivative or crystal thereof and method for producing the same
US7790885B2 (en) 2006-08-31 2010-09-07 Eisai R&D Management Co., Ltd. Process for preparing phenoxypyridine derivatives
EP2684874A1 (en) 2006-10-23 2014-01-15 Cephalon, Inc. Fused bicyclic derivatives of 2,4-diaminopyrimidine as alk and C-met inhibitors
WO2008049855A3 (en) * 2006-10-27 2008-08-14 Glaxo Group Ltd 7-azaindole derivatives as c-met kinase inhibitors
WO2008049855A2 (en) * 2006-10-27 2008-05-02 Glaxo Group Limited 7-azaindole derivatives as c-met kinase inhibitors
EP3443958A1 (en) 2006-11-22 2019-02-20 Incyte Holdings Corporation Imidazotriazines and imidazopyrimidines as kinase inhibitors
EP2497470A1 (en) 2006-11-22 2012-09-12 Incyte Corporation Imidazotriazines and imidazopyrimidines as kinase inhibitors
US8461330B2 (en) 2006-11-22 2013-06-11 Incyte Corporation Imidazotriazines and imidazopyrimidines as kinase inhibitors
US9944645B2 (en) 2006-11-22 2018-04-17 Incyte Corporation Imidazotriazines and imidazopyrimidines as kinase inhibitors
EP3034075A1 (en) 2006-11-22 2016-06-22 Incyte Holdings Corporation Imidazotriazines and imidazopyrimidines as kinase inhibitors
US10738052B2 (en) 2006-11-22 2020-08-11 Incyte Holdings Corporation Imidazotriaines and imidazopyrimidines as kinase inhibitors
US7767675B2 (en) 2006-11-22 2010-08-03 Incyte Corporation Imidazotriazines and imidazopyrimidines as kinase inhibitors
US11261191B2 (en) 2006-11-22 2022-03-01 Incyte Holdings Corporation Imidazotriaines and imidazopyrimidines as kinase inhibitors
WO2008079291A3 (en) * 2006-12-20 2008-11-06 Amgen Inc Substituted heterocycles and methods of use
US7687522B2 (en) 2006-12-20 2010-03-30 Amgen Inc. Substituted pyridines and pyrimidines and their use in treatment of cancer
EP2125780B1 (en) * 2006-12-20 2012-08-29 Amgen Inc. Substituted heterocycles and methods of use
JP2010516680A (en) * 2007-01-19 2010-05-20 エックスカバリー,インコーポレイテッド Kinase inhibitor compounds
US8377938B2 (en) 2007-02-16 2013-02-19 Eisai R&D Management Co., Ltd. Phenoxypyridine derivative salts and crystals thereof, and process for preparing the same
DE102007026341A1 (en) 2007-06-06 2008-12-11 Merck Patent Gmbh Benzoxazolonderivate
WO2008148449A1 (en) 2007-06-06 2008-12-11 Merck Patent Gmbh 2-oxo-3-benzyl-benzoxazol-2-one derivatives and related compounds as met kinase inhibitors for the treatment of tumors
US7884114B2 (en) 2007-08-15 2011-02-08 Glaxo Group Limited Compounds
US8846927B2 (en) 2007-08-29 2014-09-30 Methylgene Inc. Inhibitors of protein tyrosine kinase activity
US8404846B2 (en) 2007-08-29 2013-03-26 Methylgene Inc. Inhibitors of protein tyrosine kinase activity
US8389541B2 (en) 2007-08-29 2013-03-05 Methylgene Inc. Inhibitors of protein tyrosine kinase activity
US7998948B2 (en) 2007-11-30 2011-08-16 Eisai R&D Management Co., Ltd. Pharmaceutical composition for treating esophageal cancer
US8969587B2 (en) 2008-03-17 2015-03-03 Ambit Biosciences Corporation RAF kinase modulator compounds and methods of use thereof
US10053430B2 (en) 2008-03-17 2018-08-21 Ambit Biosciences Corp. RAF kinase modulator compounds and methods of use thereof
US8618289B2 (en) 2008-03-17 2013-12-31 Ambit Biosciences Corporation RAF kinase modulator compounds and methods of use thereof
US9320739B2 (en) 2008-03-17 2016-04-26 Ambit Biosciences Corporation RAF kinase modulator compounds and methods of use thereof
US9730937B2 (en) 2008-03-17 2017-08-15 Ambit Biosciences Corporation RAF kinase modulator compounds and methods of use thereof
US8304427B2 (en) 2008-04-10 2012-11-06 Taiho Pharmaceutical Co., Ltd. Acylthiourea compound or salt thereof, and use thereof
WO2009125597A1 (en) 2008-04-10 2009-10-15 大鵬薬品工業株式会社 Acylthiourea compound or salt thereof, and use of the compound or the salt
US20110092503A1 (en) * 2008-04-16 2011-04-21 Axel Ullrich Quinoline derivatives as axl kinase inhibitors
US9206130B2 (en) * 2008-04-16 2015-12-08 MAX-PLANCK-Gesellschaft zur Förderung der Wissenschaften e.V. Quinoline derivatives as AXL kinase inhibitors
KR101325237B1 (en) * 2008-04-16 2013-11-04 막스-플랑크-게젤샤프트 츄어 푀르더룽 데어 비쎈샤프텐 에.파우. Quinoline derivatives as axl kinase inhibitors
WO2009127417A1 (en) 2008-04-16 2009-10-22 MAX-PLANCK-Gesellschaft zur Förderung der Wissenschaften e.V. Quinoline derivatives as axl kinase inhibitors
CN102083824A (en) * 2008-05-05 2011-06-01 葛兰素史密斯克莱有限责任公司 Method of treating cancer using a cMET and AXL inhibitor and an ErbB inhibitor
AU2009244453B2 (en) * 2008-05-05 2012-07-19 Glaxosmithkline Llc Method of treating cancer using a cMET and AXL inhibitor and an erbB inhibitor
US20130150363A1 (en) * 2008-05-05 2013-06-13 Glaxosmithkline Method of Treating Cancer Using a cMET and AXL Inhibitor and an ErbB Inhibitor
EA020779B1 (en) * 2008-05-05 2015-01-30 ГЛЭКСОСМИТКЛАЙН ЭлЭлСи METHOD OF TREATING CANCER USING A cMET AND AXL INHIBITOR AND AN ErbB INHIBITOR
US20130142790A1 (en) * 2008-05-05 2013-06-06 Glaxosmithkline Method of Treating Cancer Using a cMET and AXL Inhibitor and an ErbB Inhibitor
EP3287449A2 (en) 2008-05-21 2018-02-28 Incyte Holdings Corporation Salts of 2-fluoro-n-methyl-4-[7-(quinolin-6-yl-methyl)- imidazo[1,2-b][1,2,4]triazin-2-yl]benzamide and processes related to preparing the same
US8901123B2 (en) 2008-05-21 2014-12-02 Incyte Corporation Salts of 2-fluoro-N-methyl-4-[7-(quinolin-6-yl-methyl)-imidazo[1,2-B][1,2,4]triazin-2-yl]benzamide and processes related to preparing the same
US8420645B2 (en) 2008-05-21 2013-04-16 Incyte Corporation Salts of 2-fluoro-N-methyl-4-[7-(quinolin-6-yl-methyl)-imidazo[1,2-b][1,2,4]triazin-2-yl]benzamide and processes related to preparing the same
US10799509B2 (en) 2008-05-21 2020-10-13 Incyte Corporation Salts of 2-fluoro-N-methyl-4-[7-(quinolin-6-yl-methyl)-imidazo[1,2-B][1,2,4]triazin-2-yl]benzamide and processes related to preparing the same
US10245265B2 (en) 2008-05-21 2019-04-02 Incyte Incorporation Salts of 2-fluoro-N-methyl-4-[7-(quinolin-6-yl-methyl)-imidazo[1,2-B][1,2,4]triazin-2-yl]benzamide and processes related to preparing the same
US11452726B2 (en) 2008-05-21 2022-09-27 Incyte Corporation Salts of 2-fluoro-N-methyl-4-[7-(quinolin-6-yl-methyl)-imidazo[1,2-b][1,2,4]triazin-2-yl]benzamide and processes related to preparing the same
US8344018B2 (en) 2008-07-14 2013-01-01 Gilead Sciences, Inc. Oxindolyl inhibitor compounds
US8134000B2 (en) 2008-07-14 2012-03-13 Gilead Sciences, Inc. Imidazolyl pyrimidine inhibitor compounds
US8124764B2 (en) 2008-07-14 2012-02-28 Gilead Sciences, Inc. Fused heterocyclyc inhibitor compounds
US8088771B2 (en) 2008-07-28 2012-01-03 Gilead Sciences, Inc. Cycloalkylidene and heterocycloalkylidene inhibitor compounds
JP2012503671A (en) * 2008-09-26 2012-02-09 グラクソスミスクライン・リミテッド・ライアビリティ・カンパニー Preparation of quinolinyloxydiphenylcyclopropanedicarboxamide
EP2339918A4 (en) * 2008-09-26 2012-06-06 Glaxosmithkline Llc Preparation of a quinolinyloxydiphenylcyclopropanedicarboxamide
US20100081805A1 (en) * 2008-09-26 2010-04-01 Nicole Marie Deschamps Preparation of a Quinolinyloxydiphenylcyclopropanedicarboxamide
US8314232B2 (en) 2008-09-26 2012-11-20 Glaxosmithkline Llc Preparation of a quinolinyloxydiphenylcyclopropanedicarboxamide
EP2339918A1 (en) * 2008-09-26 2011-07-06 GlaxoSmithKline LLC Preparation of a quinolinyloxydiphenylcyclopropanedicarboxamide
US8293897B2 (en) * 2008-10-14 2012-10-23 Ning Xi Compounds comprising a spiro-ring and methods of use
US8426585B2 (en) 2008-10-14 2013-04-23 Ning Xi Compounds comprising a spiro-ring
US20100093727A1 (en) * 2008-10-14 2010-04-15 Ning Xi Compounds and methods of use
US20130197230A1 (en) * 2008-11-13 2013-08-01 Jo Ann Wilson Methods of preparing quinoline derivatives
WO2010056960A1 (en) * 2008-11-13 2010-05-20 Exelixis Inc. Methods of preparing quinoline derivatives
EA019247B1 (en) * 2008-11-13 2014-02-28 Экселиксис, Инк. Methods of preparing quinoline derivatives
WO2010065838A1 (en) 2008-12-04 2010-06-10 Exelixis, Inc. Methods of preparing quinoline derivatives
CN102239148A (en) * 2008-12-04 2011-11-09 埃克塞里艾克西斯公司 Methods of preparing quinoline derivatives
US20130030172A1 (en) * 2008-12-04 2013-01-31 Exelixis, Inc. Methods of Preparing Quinoline Derivatives
JP2012511017A (en) * 2008-12-04 2012-05-17 エグゼリクシス, インコーポレイテッド Method for preparing quinoline derivatives
WO2010078905A1 (en) 2009-01-07 2010-07-15 Merck Patent Gmbh Benzothiazolone derivative
DE102009003975A1 (en) 2009-01-07 2010-07-08 Merck Patent Gmbh Benzothiazolonderivate
WO2010083414A1 (en) 2009-01-16 2010-07-22 Exelixis, Inc. Malate salt of n- (4- { [ 6, 7-bis (methyloxy) quin0lin-4-yl] oxy}phenyl-n' - (4 -fluorophenyl) cyclopropane-1-dicarboxamide, and crystalline forms therof for the treatment of cancer
US11091439B2 (en) 2009-01-16 2021-08-17 Exelixis, Inc. Malate salt of N-(4-{[6,7-bis(methyloxy) quinolin-4-yl]oxy}phenyl)-N′-(4-fluorophenyl)cyclopropane-1,1-dicarboxamide, and crystalline forms therof for the treatment of cancer
CN110818633A (en) * 2009-01-16 2020-02-21 埃克塞里艾克西斯公司 Malate and crystal form thereof
TWI511956B (en) * 2009-01-16 2015-12-11 Exelixis Inc Method for preparing n-(4-{(6,7-bis(methyloxy)quinolin-4-yl)oxy}phenyl)-n'-(4-fluorophenyl)cyclopropane-1,1-dicarboxamide
CN106083714B (en) * 2009-01-16 2019-11-26 埃克塞里艾克西斯公司 N- (4- { [bis- (methyl oxygroup) quinolyl-4s of 6,7-] oxygroup } phenyl)-N '-(4- fluorophenyl) cyclopropane -1,1- diformamide malate and its crystal type
US8877776B2 (en) 2009-01-16 2014-11-04 Exelixis, Inc. (L)-malate salt of N-(4-{[6,7-bis(methyloxy) quinolin-4-yl]oxy}phenyl)-N'-(4-fluorophenyl)cyclopropane-1,1-dicarboxamide
KR101733773B1 (en) * 2009-01-16 2017-05-10 엑셀리시스, 인코포레이티드 ----4-----11- malate salt of n-4-[67-bismethyloxyquinolin-4-yl]oxyphenyl-n'-4-fluorophenylcyclopropane-11-dicarboxamide and crystalline forms thereof for the treatment of cancer
AU2016262732B2 (en) * 2009-01-16 2018-04-26 Exelixis, Inc. Malate salt of n-(4-{[6,7-bis(methyloxy)quinolin-4-yl]oxy}phenyl)-n'-(4 -fluorophenyl) cyclopropane-1,1-dicarboxamide, and crystalline forms thereof for the treatment of cancer
CN102388024A (en) * 2009-01-16 2012-03-21 埃克塞里艾克西斯公司 Malate salt of n- (4- { [ 6, 7-bis (methyloxy) quin0lin-4-yl] oxy}phenyl-n' - (4 -fluorophenyl) cyclopropane-1-dicarboxamide, and crystalline forms therof for the treatment of cancer
EA038195B1 (en) * 2009-01-16 2021-07-22 Экселиксис, Инк. Malate salt of n-(4-{[6,7-bis(methyloxy)quinolin-4-yl]oxy}phenyl-n'-(4-fluorophenyl)cyclopropane-1,1-dicarboxamide and use thereof for the treatment of kidney and liver cancer
US11091440B2 (en) 2009-01-16 2021-08-17 Exelixis, Inc. Malate salt of N-(4-{[6,7-bis(methyloxy) quinolin-4-yl]oxy}phenyl)- N′-(4-fluorophenyl)cyclopropane-1,1 -dicarboxamide, and crystalline forms thereof for the treatment of cancer
EA026425B1 (en) * 2009-01-16 2017-04-28 Экселиксис, Инк. Malate salt of n-(4-{[6,7-bis(methyloxy)quinolin-4-yl]oxy}phenyl)-n'-(4-fluorophenyl)cyclopropane-1,1-dicarboxamide, and crystalline forms thereof for the treatment of cancer
AU2010204619B2 (en) * 2009-01-16 2016-08-11 Exelixis, Inc. Malate salt of N-(4-{ [ 6, 7-bis (methyloxy)-quinolin-4-yl] oxy}phenyl)-N'-(4-fluorophenyl) cyclopropane-1,1-dicarboxamide, and crystalline forms thereof for the treatment of cancer
TWI577664B (en) * 2009-01-16 2017-04-11 艾克塞里克斯公司 Malate salt of n-(4-{[6,7-bis(methyloxy)quinolin-4-yl]oxy}phenyl)-n'-(4-fluorophenyl)cyclopropane-1,1-dicarboxamide, pharmaceutical composition comprising the same and use thereof
CN106083714A (en) * 2009-01-16 2016-11-09 埃克塞里艾克西斯公司 N (4 { [6,7 pairs of (methyl epoxide) quinoline 4 bases] epoxide } phenyl) N ' (4 fluorophenyl) cyclopropane 1,1 diformamide malate and crystal type thereof
KR102187034B1 (en) * 2009-01-16 2020-12-04 엑셀리시스, 인코포레이티드 Malate salt of n-(4-{[6,7-bis(methyloxy)quinolin-4-yl]oxy}phenyl)-n'-(4-fluorophenyl)cyclopropane-1,1-dicarboxamide, and crystalline forms thereof for the treatment of cancer
AU2013203780B2 (en) * 2009-01-16 2016-09-01 Exelixis, Inc. Malate salt of n-(4-{[6,7-bis (methyloxy) quinolin-4-yl]oxy}phenyl)-n'-(4-fluorophenyl) cyclopropane-1,1-dicarboxamide, and crystalline forms thereof for the treatment of cancer
US11098015B2 (en) 2009-01-16 2021-08-24 Exelixis, Inc. Malate salt of N-(4-{[6,7-bis(methyloxy) quinolin-4-yl]oxy}phenyl)-N′-(4-fluorophenyl)cyclopropane-1,1-dicarboxamide, and crystalline forms thereof for the treatment of cancer
TWI447108B (en) * 2009-01-16 2014-08-01 Exelixis Inc Malate salts of n-(4-{[6,7-bis(methyloxy)quinolin-4-yl]oxy}phenyl)-n'-(4-fluorophenyl)cyclopropane-1,1-dicarboxamide, and crystalline forms thereof
EA019959B1 (en) * 2009-01-16 2014-07-30 Экселиксис, Инк. Malate salt of n-(4-{[6,7-bis(methyloxy)quin0lin-4-yl]oxy}phenyl)-n'-(4-fluorophenyl)cyclopropane-1,1-dicarboxamide, and crystalline forms thereof for the treatment of cancer
US9809549B2 (en) 2009-01-16 2017-11-07 Exelixis, Inc. Malate salt of N-(4-{[6,7-bis(methyloxy)quinolin-4-yl]oxy}phenyl)-N′(4-fluorophenyl)cyclopropane-1,1-dicarboxamide, and crystalline forms therof for the treatment of cancer
KR20200027580A (en) * 2009-01-16 2020-03-12 엑셀리시스, 인코포레이티드 Malate salt of n-(4-{[6,7-bis(methyloxy)quinolin-4-yl]oxy}phenyl)-n'-(4-fluorophenyl)cyclopropane-1,1-dicarboxamide, and crystalline forms thereof for the treatment of cancer
TWI641593B (en) * 2009-01-16 2018-11-21 美商艾克塞里克斯公司 Pharmaceutical composition comprising malate salt of n-(4-{[6,7-bis(methyloxy)quinolin-4-yl]oxy}phenyl)-n'-(4-fluorophenyl)cyclopropane-1,1-dicarboxamide and use thereof
EP2408300A4 (en) * 2009-03-21 2012-12-12 Sunshine Lake Pharma Co Ltd Amino ester derivatives, salts thereof and methods of use
WO2010111063A1 (en) 2009-03-21 2010-09-30 Ning Xi Amino ester derivatives, salts thereof and methods of use
EP2408300A1 (en) * 2009-03-21 2012-01-25 Sunshine Lake Pharma Co., Ltd Amino ester derivatives, salts thereof and methods of use
JP2012521348A (en) * 2009-03-21 2012-09-13 クイ ニング Amino ester derivatives, salts thereof, and methods of use
WO2010140092A1 (en) 2009-06-05 2010-12-09 Pfizer Inc. L- ( piperidin-4-yl) -pyrazole derivatives as gpr 119 modulators
US8258316B2 (en) 2009-06-08 2012-09-04 Gilead Sciences, Inc. Alkanoylamino benzamide aniline HDAC inhibitor compounds
US8283357B2 (en) 2009-06-08 2012-10-09 Gilead Sciences, Inc. Cycloalkylcarbamate benzamide aniline HDAC inhibitor compounds
CN102510855B (en) * 2009-07-17 2015-11-25 埃克塞里艾克西斯公司 The crystalline form of N-[the fluoro-4-of 3-({ 6-(methyl oxygen base)-7-[(3-morpholine-4-base propyl group) oxygen base]-quinolyl-4 } oxygen base) phenyl]-N '-(4-fluorophenyl) cyclopropane-1,1-diformamide
US8673912B2 (en) 2009-07-17 2014-03-18 Exelixis, Inc. Crystalline Forms on N-[3-fluoro-4-({6-(methyloxy)-7-[(3-morpholin-4-ylpropyl)oxy]-quinolin-4-yl}oxy)phenyl]-N′-(4-fluorophenyl)cyclopropane-1,1-dicarboxamide
WO2011009095A1 (en) 2009-07-17 2011-01-20 Exelixis, Inc. Crystalline forms of n-[3-fluoro-4-({6-(methyloxy)-7-[(3-morpholin-4-ylpropyl)oxy]-quin0lin-4-yl}oxy)phenyl]-n'-(4-fluorophenyl)cyclopropane-1, 1-dicarboxamide
CN102510855A (en) * 2009-07-17 2012-06-20 埃克塞里艾克西斯公司 Crystalline forms of n-[3-fluoro-4-({6-(methyloxy)-7-[(3-morpholin-4-ylpropyl)oxy]-quin0lin-4-yl}oxy)phenyl]-n'-(4-fluorophenyl)cyclopropane-1, 1-dicarboxamide
US10736886B2 (en) 2009-08-07 2020-08-11 Exelixis, Inc. Methods of using c-Met modulators
EA024563B1 (en) * 2009-08-07 2016-09-30 Экселиксис, Инк. Use of c-met modulators in combined cancer treatment
US11433064B2 (en) 2009-08-07 2022-09-06 Exelixis, Inc. Methods of using c-Met modulators
CN102647985A (en) * 2009-08-07 2012-08-22 埃克塞里艾克西斯公司 Methods of using c-Met modulators
KR101954322B1 (en) * 2009-08-07 2019-03-05 엑셀리시스, 인코포레이티드 Methods of using c-met modulators
KR101761380B1 (en) * 2009-08-07 2017-07-25 엑셀리시스, 인코포레이티드 Methods of using c-met modulators
AU2010279234B2 (en) * 2009-08-07 2016-10-20 Exelixis, Inc. Methods of using c-Met modulators
CN107325048A (en) * 2009-08-07 2017-11-07 埃克塞里艾克西斯公司 Use the method for c Met conditioning agents
EA029585B1 (en) * 2009-08-07 2018-04-30 Экселиксис, Инк. METHOD OF PRODUCING c-Met MODULATORS
KR20170087966A (en) * 2009-08-07 2017-07-31 엑셀리시스, 인코포레이티드 Methods of using c-met modulators
AU2017200269B2 (en) * 2009-08-07 2019-02-07 Exelixis, Inc. Methods of using c-Met modulators
WO2011017639A1 (en) 2009-08-07 2011-02-10 Exelixis,Inc Methods of using c-met modulators
CN102030705B (en) * 2009-09-30 2012-12-19 上海睿智化学研究有限公司 Synthesis method of 7- benzyloxy-6-methoxyl-4-hydroxyquinoline
WO2011045084A1 (en) * 2009-10-16 2011-04-21 MAX-PLANCK-Gesellschaft zur Förderung der Wissenschaften e.V. Quinolinyloxyphenylsulfonamides
CN102574804A (en) * 2009-10-16 2012-07-11 马克斯普朗克科学发展组织 Quinolinyloxyphenylsulfonamides
EP2311809A1 (en) * 2009-10-16 2011-04-20 Max-Planck-Gesellschaft zur Förderung der Wissenschaften e.V. Quinolinyloxyphenylsulfonamides
DE102009056886A1 (en) 2009-12-03 2011-06-09 Bayer Schering Pharma Aktiengesellschaft cMet inhibitors for the treatment of endometriosis
WO2011067189A2 (en) 2009-12-03 2011-06-09 Bayer Schering Pharma Aktiengesellschaft Cmet inhibitors for treating endometriosis
CN102086211A (en) * 2009-12-08 2011-06-08 深圳市东阳光实业发展有限公司 Aromatic heterocyclic compounds serving as protein kinase inhibitor
CN102086211B (en) * 2009-12-08 2013-09-11 广东东阳光药业有限公司 Aromatic heterocyclic compounds serving as protein kinase inhibitor
US10919901B2 (en) 2010-02-03 2021-02-16 Incyte Holdings Corporation Imidazo[1,2-B][1,2,4]triazines as c-Met inhibitors
US9988387B2 (en) 2010-02-03 2018-06-05 Incyte Holdings Corporation Imidazo[1,2-B][1,2,4]triazines as c-Met inhibitors
US9221824B2 (en) 2010-02-03 2015-12-29 Incyte Holdings Corporation Imidazo[1,2-B][1,2,4]triazines as c-Met inhibitors
US8487096B2 (en) 2010-02-03 2013-07-16 Incyte Corporation Imidazo[1,2-B][1,2,4]triazines as C-MET inhibitors
WO2011162835A1 (en) 2010-02-03 2011-12-29 Incyte Corporation Imidazo[1,2-b][1,2,4]triazines as c-met inhibitors
US10472367B2 (en) 2010-02-03 2019-11-12 Incyte Incorporation Imidazo[1,2-B][1,2,4]triazines as c-Met inhibitors
CN102933551A (en) * 2010-03-12 2013-02-13 埃克塞里艾克西斯公司 Hydrated crystalline forms of n-[3-fluoro-4-({6-(methyloxy)-7-[(3-morpholin-4-ylpropyl)oxy]-quinolin-4-yl}oxy)phenyl]-n'-(4-fluorophenyl)cyclopropane-1,1-dicarboxamide
WO2011112896A1 (en) 2010-03-12 2011-09-15 Exelixis, Inc Hydrated crystalline forms of n-[3-fluoro-4-({6-(methyloxy)-7-[(3-morpholin-4-ylpropyl)oxy]-quinolin-4-yl}oxy)phenyl]-n'-(4-fluorophenyl)cyclopropane-1,1-dicarboxamide
CN102212062A (en) * 2010-04-02 2011-10-12 东莞市长安东阳光新药研发有限公司 Derivative of amino ester, salt thereof and using method
US9730886B2 (en) 2010-04-23 2017-08-15 Cytokinetics, Inc. Amino-pyrimidine skeletal muscle modulators
US9604965B2 (en) 2010-04-23 2017-03-28 Cytokinetics, Inc. Substituted pyridazines as skeletal muscle modulators
US10076519B2 (en) 2010-04-23 2018-09-18 Cytokinetics, Inc. Substituted pyridazines as skeletal muscle modulators
US10765624B2 (en) 2010-04-23 2020-09-08 Cytokinetics, Inc. Amino-pyrimidine skeletal muscle modulators
US9133123B2 (en) 2010-04-23 2015-09-15 Cytokinetics, Inc. Certain amino-pyridines and amino-triazines, compositions thereof, and methods for their use
US11369565B2 (en) 2010-04-23 2022-06-28 Cytokinetics, Inc. Amino-pyrimidine skeletal muscle modulators
US10272030B2 (en) 2010-04-23 2019-04-30 Cytokinetics, Inc. Amino-pyrimidine skeletal muscle modulators
US9994528B2 (en) 2010-04-23 2018-06-12 Cytokinetics, Inc. Certain amino-pyridines and amino-triazines, compositions thereof, and methods for their use
US8569319B2 (en) 2010-04-29 2013-10-29 Deciphera Pharmaceuticals, LLS Pyridone amides and analogs exhibiting anti-cancer and anti-proliferative activities
US8481739B2 (en) 2010-05-17 2013-07-09 Incozen Therapeutics Pvt. Ltd. 3,5-Disubstituted-3H-imidazo[4,5-b]pyridine and 3,5-disubstituted-3H[1,2,3]triazolo [4,5-b] Pyridine Compounds as Modulators of protein kinases
WO2011145035A1 (en) 2010-05-17 2011-11-24 Indian Incozen Therapeutics Pvt. Ltd. Novel 3,5-disubstitued-3h-imidazo[4,5-b]pyridine and 3,5- disubstitued -3h-[1,2,3]triazolo[4,5-b] pyridine compounds as modulators of protein kinases
US8912331B2 (en) 2010-05-17 2014-12-16 Rhizen Pharmaceuticals Sa 3,5-disubstituted-3H-imidazo[4,5-B]pyridine and 3,5-disubstituted-3H-[1,2,3]triazolo[4,5-B] pyridine compounds as modulators of protein kinases
US10087182B2 (en) 2010-05-17 2018-10-02 Incozen Therapeutics Pvt. Ltd. 3,5-disubstituted-3H-imidazo[4,5-B]pyridine and 3,5-disubstituted-3H-[1,2,3]triazolo[4,5-B] pyridine compounds as modulators of protein kinases
EP3450432A1 (en) 2010-05-17 2019-03-06 Incozen Therapeutics Pvt. Ltd. Novel 3,5-disubstitued-3h-imidazo[4,5-b]pyridine and 3,5- disubstitued - 3h-[1,2,3]triazolo[4,5-b] pyridine compounds as modulators of protein kinases
US10590129B2 (en) 2010-05-17 2020-03-17 Rhizen Pharmaceuticals Sa 3,5-disubstituted-3H-imidazo[4,5-B]pyridine and 3,5-disubstituted-3H-[1,2,3]triazolo[4,5-B] pyridine compounds as modulators of protein kinases
WO2012003338A1 (en) 2010-07-01 2012-01-05 Takeda Pharmaceutical Company Limited COMBINATION OF A cMET INHIBITOR AND AN ANTIBODY TO HGF AND/OR cMET
EP4014971A1 (en) 2010-07-16 2022-06-22 Exelixis, Inc. C-met modulator pharmaceutical compositions
CN106420743A (en) * 2010-07-16 2017-02-22 埃克塞里艾克西斯公司 C-MET modulator pharmaceutical compositions
CN103221035A (en) * 2010-07-16 2013-07-24 埃克塞里艾克西斯公司 C-MET modulator pharmaceutical compositions
JP2013531045A (en) * 2010-07-16 2013-08-01 エクセリクシス, インク. Pharmaceutical composition of C-MET modulator
EP2593090B1 (en) 2010-07-16 2021-10-13 Exelixis, Inc. C-met modulator pharmaceutical compositions
CN114209699A (en) * 2010-07-16 2022-03-22 埃克塞里艾克西斯公司 C-MET modulator pharmaceutical compositions
AU2011278950C1 (en) * 2010-07-16 2018-11-22 Exelixis, Inc. c-Met modulator pharmaceutical compositions
US10548888B2 (en) 2010-07-16 2020-02-04 Exelixis C-Met modulator pharmaceutical compositions
EA030435B1 (en) * 2010-07-16 2018-08-31 Экселиксис, Инк. TABLET COMPRISING c-MET MODULATOR IN THE FORM OF CRYSTALLINE L-MALATE SALT (EMBODIMENTS), PROCESS FOR MANUFACTURING SAME AND METHOD FOR TREATING CANCER USING SAME
US11123338B2 (en) 2010-07-16 2021-09-21 Exelixis, Inc. C-met modulator pharmaceutical compositions
AU2011278950B2 (en) * 2010-07-16 2016-07-21 Exelixis, Inc. c-Met modulator pharmaceutical compositions
AU2018220045B2 (en) * 2010-07-16 2020-03-05 Exelixis, Inc. C-met modulator pharmaceutical compositions
WO2012009723A1 (en) 2010-07-16 2012-01-19 Exelixis, Inc. C-met modulator pharmaceutical compositions
US10039757B2 (en) 2010-07-16 2018-08-07 Exelixis, Inc. C-Met modulator pharmaceutical compositions
US10034873B2 (en) 2010-07-16 2018-07-31 Exelixis, Inc. C-met modulator pharmaceutical compositions
WO2012009722A1 (en) 2010-07-16 2012-01-19 Exelixis, Inc. C-met modulator pharmaceutical compositions
US9724342B2 (en) 2010-07-16 2017-08-08 Exelixis, Inc. C-met modulator pharmaceutical compositions
KR101862324B1 (en) * 2010-07-16 2018-05-29 엑셀리시스, 인코포레이티드 C-met modulator pharmaceutical compositions
US8999982B2 (en) 2010-08-28 2015-04-07 Lead Discovery Center Gmbh Pharmaceutically active compounds as Axl inhibitors
CN105153028A (en) * 2010-09-12 2015-12-16 南京爱德程医药科技有限公司 Compounds as c-Met kinase inhibitors
AU2011299082B2 (en) * 2010-09-12 2016-09-29 Advenchen Laboratories, Llc Compounds as c-Met kinase inhibitors
EP2621904A2 (en) * 2010-09-12 2013-08-07 Advenchen Laboratories, LLC Compounds as c-met kinase inhibitors
EP2621904A4 (en) * 2010-09-12 2014-03-26 Advenchen Lab Llc Compounds as c-met kinase inhibitors
EA027476B1 (en) * 2010-09-27 2017-07-31 Экселиксис, Инк. Use of n-(4-{[6,7-bis(methyloxy)quinolin-4-yl]oxy}phenyl)-n'-(4-fluorophenyl)cyclopropane-1,1-dicarboxamide for the treatment of castration-resistant prostate cancer and osteoblastic bone metastases
KR20140025304A (en) * 2010-09-27 2014-03-04 엑셀리시스, 인코포레이티드 Dual inhibitors of met and vegf for the treatment of castration-resistant prostate cancer and osteoblastic bone metastases
WO2012044572A1 (en) 2010-09-27 2012-04-05 Exelixis, Inc. Dual inhibitors of met and vegf for the treatment of castration- resistant prostate cancer and osteoblastic bone metastases
WO2012044574A1 (en) 2010-09-27 2012-04-05 Exelixis, Inc. Dual inhibitors of met and vegf for the treatment of castration resistant prostate cancer and osteoblastic bone metastases
AU2011307304C1 (en) * 2010-09-27 2016-10-20 Exelixis, Inc. Dual inhibitors of MET and VEGF for the treatment of castration- resistant prostate cancer and osteoblastic bone metastases
CN103391772A (en) * 2010-09-27 2013-11-13 埃克塞里艾克西斯公司 Dual inhibitors of met and vegf for the treatment of castration resistant prostate cancer and osteoblastic bone metastases
WO2012044577A1 (en) 2010-09-27 2012-04-05 Exelixis, Inc. Dual inhibitors of met and vegf for the treatment of castration resistant prostate cancer and osteoblastic bone metastases
AU2011307304B2 (en) * 2010-09-27 2016-04-14 Exelixis, Inc. Dual inhibitors of MET and VEGF for the treatment of castration- resistant prostate cancer and osteoblastic bone metastases
US11612597B2 (en) 2010-09-27 2023-03-28 Exelixis, Inc. Method of treating cancer
CN103391773A (en) * 2010-09-27 2013-11-13 埃克塞里艾克西斯公司 Dual inhibitors of met and vegf for the treatment of castration resistant prostate cancer and osteoblastic bone metastases
KR101882328B1 (en) * 2010-09-27 2018-07-27 엑셀리시스, 인코포레이티드 Dual inhibitors of met and vegf for the treatment of castration-resistant prostate cancer and osteoblastic bone metastases
CN103402505A (en) * 2010-09-27 2013-11-20 埃克塞里艾克西斯公司 Dual inhibitors of MET and VEGF for the treatment of castration- resistant prostate cancer and osteoblastic bone metastases
WO2012042421A1 (en) 2010-09-29 2012-04-05 Pfizer Inc. Method of treating abnormal cell growth
CN102093421A (en) * 2011-01-28 2011-06-15 广州盈升生物科技有限公司 Phosphorus substituent group-containing quinoline compound and preparation method of quinoline compound as well as pharmaceutical composition containing quinoline compound and application of pharmaceutical composition
WO2012100459A1 (en) 2011-01-28 2012-08-02 广州盈升生物科技有限公司 Phosphorus substituted group-containing quinoline-like compound, its preparation process, medical composition containing the compound and application
US10543206B2 (en) 2011-02-10 2020-01-28 Exelixis, Inc. Processes for preparing quinoline compounds and pharmaceutical compositions containing such compounds
KR20140044782A (en) * 2011-02-10 2014-04-15 엑셀리시스, 인코포레이티드 Processes for preparing quinoline compounds and pharmaceutical compositions containing such compounds
EP2673262B1 (en) 2011-02-10 2021-11-03 Exelixis, Inc. Processes for preparing quinoline compounds and pharmaceutical compositions containing such compounds
US9717720B2 (en) 2011-02-10 2017-08-01 Exelixis, Inc. Processes for preparing quinoline compounds and pharmaceutical compositions containing such compounds
US11298349B2 (en) 2011-02-10 2022-04-12 Exelixis, Inc. Processes for preparing quinoline compounds and pharmaceutical compositions containing such compounds
WO2012109510A1 (en) 2011-02-10 2012-08-16 Exelixis, Inc. Processes for preparing quinoline compounds and pharmaceutical compositions containing such compounds
EP4019498A1 (en) 2011-02-10 2022-06-29 Exelixis, Inc. Processes for preparing quinoline compounds and pharmaceutical compositions containing such compounds
KR102030447B1 (en) 2011-02-10 2019-11-08 엑셀리시스, 인코포레이티드 Processes for preparing quinoline compounds and pharmaceutical compositions containing such compounds
US10123999B2 (en) 2011-02-10 2018-11-13 Exelixis, Inc. Processes for preparing quinoline compounds and pharmaceutical compositions containing such compounds
AU2012250759B2 (en) * 2011-05-02 2017-06-15 Exelixis, Inc. Method of treating cancer and bone cancer pain
CN103717221A (en) * 2011-05-02 2014-04-09 埃克塞里艾克西斯公司 Method of treating cancer and bone cancer pain
WO2012151326A1 (en) * 2011-05-02 2012-11-08 Exelixis, Inc. Method of treating cancer and bone cancer pain
EA029506B1 (en) * 2011-05-02 2018-04-30 Экселиксис, Инк. Method of treating cancer and bone cancer pain
US9945862B2 (en) 2011-06-03 2018-04-17 Eisai R&D Management Co., Ltd. Biomarkers for predicting and assessing responsiveness of thyroid and kidney cancer subjects to lenvatinib compounds
US11598776B2 (en) 2011-06-03 2023-03-07 Eisai R&D Management Co., Ltd. Biomarkers for predicting and assessing responsiveness of thyroid and kidney cancer subjects to lenvatinib compounds
WO2013036676A1 (en) 2011-09-06 2013-03-14 New York Blood Center, Inc. Hiv inhibitors
US9309237B2 (en) 2011-09-06 2016-04-12 New York Blood Center, Inc. HIV inhibitors
EP2753176A4 (en) * 2011-09-06 2015-10-21 New York Blood Ct Inc Hiv inhibitors
KR101879908B1 (en) * 2011-09-19 2018-07-18 베이징 콘런스 파마슈티칼 컴퍼니 리미티드 Hydroxamic acid compound containing quinolyl and preparation method thereof, and pharmaceutical composition containing this compound and use thereof
EP2769976A4 (en) * 2011-09-19 2015-06-03 Beijing Konruns Pharmaceutical Co Ltd Hydroxamic acid compound containing quinolyl and preparation method thereof, and pharmaceutical composition containing this compound and use thereof
WO2013040801A1 (en) 2011-09-19 2013-03-28 广州盈升生物科技有限公司 Hydroxamic acid compound containing quinolyl and preparation method thereof, and pharmaceutical composition containing this compound and use thereof
AU2011377440B2 (en) * 2011-09-19 2017-01-05 Beijing Konruns Pharmaceutical Co., Ltd. Hydroxamic acid compound containing quinolyl and preparation method thereof, and pharmaceutical composition containing this compound and use thereof
WO2013043840A1 (en) 2011-09-22 2013-03-28 Exelixis, Inc. Method for treating osteoporosis
EP3275443A1 (en) 2011-09-22 2018-01-31 Exelixis, Inc. Method for treating osteoporosis
CN104395284A (en) * 2011-10-20 2015-03-04 埃克塞里艾克西斯公司 Process for preparing quinoline derivatives
WO2013059788A1 (en) 2011-10-20 2013-04-25 Exelixis, Inc. Process for preparing quinoline derivatives
CN110511158A (en) * 2011-10-20 2019-11-29 埃克塞里艾克西斯公司 The method for preparing quinoline
US20160229805A1 (en) * 2011-10-20 2016-08-11 Exelixis, Inc. Process for Preparing Quinoline Derivatives
US9969692B2 (en) * 2011-10-20 2018-05-15 Exelixis, Inc. Process for preparing quinoline derivatives
WO2013070903A1 (en) 2011-11-08 2013-05-16 Exelixis, Inc. Method of quantifying cancer treatment
WO2013070890A1 (en) 2011-11-08 2013-05-16 Exelixis, Inc. Dual inhibitor of met and vegf for treating cancer
CN103172641A (en) * 2011-12-20 2013-06-26 钱卫 Heterocyclic amino and alkoxy-replaced quinazoline derivative and application thereof
US9382232B2 (en) 2011-12-30 2016-07-05 Shenyang Pharmaceutical University Quinoline and cinnoline derivatives and their applications
EP2799437A4 (en) * 2011-12-30 2015-08-19 Univ Shenyang Pharmaceutical Quinoline and cinnoline compounds and use thereof
US20140364431A1 (en) * 2011-12-30 2014-12-11 Shenyang Pharmaceutical University Quinoline and cinnoline derivatives and their applications
WO2013144737A2 (en) 2012-03-30 2013-10-03 Rhizen Pharmaceuticals Sa Novel 3,5-disubstitued-3h-imidazo[4,5-b]pyridine and 3,5- disubstitued -3h-[1,2,3]triazolo[4,5-b] pyridine compounds as modulators of c-met protein kinases
US9815831B2 (en) 2012-03-30 2017-11-14 Rhizen Pharmaceuticals Sa 3,5-disubstituted-3H-imidazo[4,5-B]pyridine and 3,5-disubstituted-3H-[1,2,3]triazolo[4,5-B] pyridine compounds as modulators of c-Met protein kinases
US11066402B2 (en) 2012-03-30 2021-07-20 Rhizen Pharmaceuticals Sa 3,5-disubstituted-3H-imidazo[4,5-b]pyridine and 3,5-disubstituted-3H-[1,2,3]triazolo[4,5-B] pyridine compounds as modulators of c-Met protein kinases
WO2013152252A1 (en) 2012-04-06 2013-10-10 OSI Pharmaceuticals, LLC Combination anti-cancer therapy
WO2013166296A1 (en) 2012-05-02 2013-11-07 Exelixis, Inc. A dual met - vegf modulator for treating osteolytic bone metastases
WO2014000686A1 (en) * 2012-06-28 2014-01-03 辰欣药业股份有限公司 Pyrimidobenzoazepine compound and use thereof as antitumour drug
CN103509024A (en) * 2012-06-28 2014-01-15 辰欣药业股份有限公司 Pyrimidobenzoazepine compound and application thereof as anti-tumor medicine
CN103509024B (en) * 2012-06-28 2015-10-28 上海医药工业研究院 Kui Linpyrimido quinoline benzazepine compounds and the application as antitumor drug thereof
WO2014029251A1 (en) * 2012-08-24 2014-02-27 辰欣药业股份有限公司 Benazepine compound and application thereof
WO2014029250A1 (en) * 2012-08-24 2014-02-27 辰欣药业股份有限公司 Benazepine compound and application thereof
CN103664776B (en) * 2012-09-26 2016-05-04 正大天晴药业集团股份有限公司 The preparation method of a kind of tyrosine kinase inhibitor and intermediate thereof
CN103664776A (en) * 2012-09-26 2014-03-26 正大天晴药业集团股份有限公司 Preparation method for tyrosine kinase inhibitor and midbody thereof
CN103705521A (en) * 2012-09-28 2014-04-09 韩冰 Compound for treating cerebral infarction and application thereof
EP2915806A4 (en) * 2012-11-05 2016-05-18 Univ Shenyang Pharmaceutical Novel quinoline compound and use thereof
CN103804382A (en) * 2012-11-05 2014-05-21 韩文毅 Compound for treating eczema and application thereof
KR20150095652A (en) * 2012-11-05 2015-08-21 션양 파마슈티컬 유니버시티 Novel quinoline derivatives and their applications
US9783499B2 (en) 2012-11-05 2017-10-10 Shenyang Pharmaceutical University Quinoline derivatives and their applications
KR101716068B1 (en) 2012-11-05 2017-03-13 션양 파마슈티컬 유니버시티 Novel quinoline derivatives and their applications
CN103804381A (en) * 2012-11-06 2014-05-21 韩冰 Compound for treatment of ischemic brain damage and application thereof
CN103800328A (en) * 2012-11-07 2014-05-21 韩冰 Compound for treating neurodegenerative diseases and application thereof
CN103804361A (en) * 2012-11-07 2014-05-21 韩冰 Compound for treating neurodegenerative diseases and application thereof
CN103804291A (en) * 2012-11-07 2014-05-21 韩冰 Compound for treating neurodegenerative diseases and application thereof
CN103804290A (en) * 2012-11-07 2014-05-21 韩冰 Compound for treating neurodegenerative diseases and application thereof
CN103804298A (en) * 2012-11-09 2014-05-21 韩冰 Compounds for treating glaucoma and application thereof
CN103800340A (en) * 2012-11-09 2014-05-21 韩冰 Compounds for treating glaucoma and application thereof
CN103804362A (en) * 2012-11-12 2014-05-21 韩文毅 Compounds for treating diabetes and application thereof
CN103804363A (en) * 2012-11-14 2014-05-21 韩冰 Compound with neuroprotective effect and use thereof
CN102964308A (en) * 2012-11-30 2013-03-13 中国药科大学 Novel pyrimidine compound, preparation method thereof, pharmaceutical composition containing novel pyrimidine compound and application of novel pyrimidine compound
CN103965104B (en) * 2013-01-29 2017-09-29 正大天晴药业集团股份有限公司 A kind of preparation method of tyrosine kinase inhibitor and its intermediate
CN103965104A (en) * 2013-01-29 2014-08-06 正大天晴药业集团股份有限公司 Preparation methods of tyrosine kinase inhibitor and intermediates thereof
US20160031818A1 (en) * 2013-03-15 2016-02-04 Exelixis, Inc. Metabolites of N-(4-phenyl)-N'-(4-fluorophenyl)cyclopropane-1,1-dicarboxamide
WO2014145715A1 (en) 2013-03-15 2014-09-18 Exelixis, Inc. Metabolites of n-(4-{[6,7-bis(methyloxy)quinolin-4-yl]oxy}phenyl)-n'-(4-fluorophenyl) cyclopropane-1,1-dicarboxamide
KR20150130376A (en) * 2013-03-15 2015-11-23 엑셀리시스, 인코포레이티드 Metabolites of n-(4-[[6,7??bis(methyloxy)quinolin-4-yl]oxy]phenyl)-n'-(4??fluorophenyl)cyclopropane-1, 1-dicarboxamide
KR102276348B1 (en) * 2013-03-15 2021-07-12 엑셀리시스, 인코포레이티드 Metabolites of n­(4­〔[6,7­bis(methyloxy)quinolin­4­yl]oxy〕phenyl)­n′­(4­fluorophenyl) cyclopropane­1,1­dicarboxamide
CN105121412A (en) * 2013-03-15 2015-12-02 埃克塞里艾克西斯公司 Metabolites of n-(4-{[6,7-bis(methyloxy)quinolin-4-yl]oxy}phenyl)-n'-(4-fluorophenyl) cyclopropane-1,1-dicarboxamide
CN105121412B (en) * 2013-03-15 2019-07-12 埃克塞里艾克西斯公司 N- (4- { [6,7- bis- (methyl oxygroup) quinolyl-4s] oxygroup } phenyl)-N′The metabolin of (4- fluorophenyl) cyclopropane -1,1- diformamide
AU2014232714B2 (en) * 2013-03-15 2018-07-19 Exelixis, Inc. Metabolites of N-(4-{[6,7-bis(methyloxy)quinolin-4-yl]oxy}phenyl)-n'-(4-fluorophenyl) cyclopropane-1,1-dicarboxamide
EA033786B1 (en) * 2013-03-15 2019-11-26 Exelixis Inc Metabolites of n-(4-{[6,7-bis-(methyloxy)quinolin-4-yl]oxy}phenyl)-n'-(4-fluorophenyl)cyclopropane-1,1-dicarboxamide
US10273211B2 (en) 2013-03-15 2019-04-30 Exelixis, Inc. Metabolites of N-{4-([6,7-bis(methyloxy)quinolin-4-yl]oxy}phenyl)-N′-(4-fluorophenyl)cyclopropane-1,1-dicarboxamide
WO2014145693A1 (en) * 2013-03-15 2014-09-18 Exelixis, Inc. Metabolites of n-[3-fluoro-4-({ 6-(methyloxy)-7-[(3-morpholin-4-ylpropyl)oxy]quinolin-4-yl}oxy)phenyl]-n'-(4-fluorophenyl)cyclopropane-1,1-dicarboxamide
JP2016516074A (en) * 2013-03-15 2016-06-02 エグゼリクシス, インコーポレイテッド N- (4-{[6,7-bis (menthyloxy) quinolin-4-yl] oxy} phenyl) -N '-(4-fluorophenyl) cyclopropane-1,1-dicarboxamide metabolite
WO2014165779A1 (en) 2013-04-04 2014-10-09 Exelixis, Inc. Drug combinations to treat cancer
WO2014165786A1 (en) 2013-04-04 2014-10-09 Exelixis, Inc. Cabozantinib dosage form and use in the treatment of cancer
CN104109128A (en) * 2013-04-19 2014-10-22 连云港润众制药有限公司 Cabozantinib malate and preparation method thereof
CN104109124B (en) * 2013-04-19 2016-08-31 正大天晴药业集团股份有限公司 The rich crystal for Buddhist nun 0.5 malate of card
CN104109124A (en) * 2013-04-19 2014-10-22 正大天晴药业集团股份有限公司 Crystal of cabozantinib*0.5 malate
US10517861B2 (en) 2013-05-14 2019-12-31 Eisai R&D Management Co., Ltd. Biomarkers for predicting and assessing responsiveness of endometrial cancer subjects to lenvatinib compounds
CN104370811B (en) * 2013-08-15 2019-02-12 广东东阳光药业有限公司 A kind of novel crystal forms of quinoline compound and preparation method thereof
CN104370811A (en) * 2013-08-15 2015-02-25 广东东阳光药业有限公司 New quinoline compound crystal form and preparation method thereof
CN104628657A (en) * 2013-11-06 2015-05-20 韩冰 Class of compounds for treating ischemic brain damage and purpose thereof
WO2015123639A1 (en) 2014-02-14 2015-08-20 Exelixis, Inc. Crystalline solid forms of n-{4-[(6,7-dimethoxyquinolin-4-yl)oxy]phenyl}-n'-(4-fluorophenyl) cyclopropane-1, 1-dicarboxamide, processes for making, and methods of use
KR102354963B1 (en) 2014-02-14 2022-01-21 엑셀리시스, 인코포레이티드 Crystalline solid forms of n-[4-[(6,7-dimethoxyquinolin-4-yl)oxy]phenyl]-n'-[4-fluorophenyl) cyclopropane-1,1-dicarboxamide, processes for making, and methods of use
KR20160121544A (en) * 2014-02-14 2016-10-19 엑셀리시스, 인코포레이티드 N-4-[67--4-]-n'-4- -11- crystalline solid forms of n-[-[-dimethoxyquinolin--yloxy]phenyl]-n'-[-fluorophenyl cyclopropane--dicarboxamide processes for making and methods of use
EP3738952A1 (en) 2014-02-14 2020-11-18 Exelixis, Inc. Crystalline solid forms of n-{4-[(6,7-dimethoxyquinolin-4-yl)oxy]phenyl}-n'-(4-fluorophenyl) cyclopropane-1,1-dicarboxamide, processes for making, and methods of use
WO2015142928A1 (en) 2014-03-17 2015-09-24 Exelixis, Inc. Dosing of cabozantinib formulations
US10240207B2 (en) 2014-03-24 2019-03-26 Genentech, Inc. Cancer treatment with c-met antagonists and correlation of the latter with HGF expression
WO2015164869A1 (en) 2014-04-25 2015-10-29 Exelixis, Inc. Method of treating lung adenocarcinoma
EP3906921A1 (en) 2014-04-25 2021-11-10 Exelixis, Inc. Method of treating lung adenocarcinoma
CN104788372A (en) * 2014-07-25 2015-07-22 上海圣考医药科技有限公司 Deuterated cabozantinib derivative, preparation method and application thereof, and intermediate of deuterated cabozantinib derivative
CN104788372B (en) * 2014-07-25 2018-01-30 上海圣考医药科技有限公司 A kind of deuterated card is rich to replace Buddhist nun's derivative, its preparation method, application and its intermediate
US20170217896A1 (en) * 2014-07-31 2017-08-03 Exelixis, Inc. Method of Preparing Fluorine-18 Labeled Cabozantinib and Its Analogs
CN106715397A (en) * 2014-07-31 2017-05-24 埃克塞里艾克西斯公司 Method of preparing fluorine-18 labeled cabozantinib and its analogs
US11124481B2 (en) * 2014-07-31 2021-09-21 Exelixis, Inc. Method of preparing fluorine-18 labeled Cabozantinib and its analogs
CN106715397B (en) * 2014-07-31 2021-07-23 埃克塞里艾克西斯公司 Method for preparing fluorine-18 labeled cabozantinib and analogs thereof
EA033834B1 (en) * 2014-07-31 2019-11-29 Exelixis Inc Method of preparing fluorine-18 labeled cabozantinib and its analogs
WO2016019285A1 (en) 2014-07-31 2016-02-04 Exelixis, Inc. Method of preparing fluorine-18 labeled cabozantinib and its analogs
WO2016022697A1 (en) 2014-08-05 2016-02-11 Exelixis, Inc. Drug combinations to treat multiple myeloma
US10407393B2 (en) 2014-08-28 2019-09-10 Eisai R&D Management Co., Ltd. High-purity quinoline derivative and method for manufacturing same
US10822307B2 (en) 2014-08-28 2020-11-03 Eisai R&D Management Co., Ltd. High-purity quinoline derivative and method for manufacturing same
US11186547B2 (en) 2014-08-28 2021-11-30 Eisai R&D Management Co., Ltd. High-purity quinoline derivative and method for manufacturing same
US10259791B2 (en) 2014-08-28 2019-04-16 Eisai R&D Management Co., Ltd. High-purity quinoline derivative and method for manufacturing same
US11059808B2 (en) 2014-09-19 2021-07-13 New York Blood Center, Inc. Substituted phenylpyrrolecarboxamides with therapeutic activity in HIV
US11571409B2 (en) 2014-09-19 2023-02-07 New York Blood Center, Inc. Substituted phenylpyrrolecarboxamides with therapeutic activity in HIV
WO2016091891A1 (en) 2014-12-09 2016-06-16 INSERM (Institut National de la Santé et de la Recherche Médicale) Human monoclonal antibodies against axl
US11090386B2 (en) 2015-02-25 2021-08-17 Eisai R&D Management Co., Ltd. Method for suppressing bitterness of quinoline derivative
WO2016135066A1 (en) 2015-02-26 2016-09-01 INSERM (Institut National de la Santé et de la Recherche Médicale) Fusion proteins and antibodies comprising thereof for promoting apoptosis
US11547705B2 (en) 2015-03-04 2023-01-10 Merck Sharp & Dohme Llc Combination of a PD-1 antagonist and a VEGF-R/FGFR/RET tyrosine kinase inhibitor for treating cancer
WO2016150966A1 (en) 2015-03-25 2016-09-29 Sandoz Ag Crystalline forms of cabozantinib phosphate and cabozantinib hydrochloride
CN107709320A (en) * 2015-05-21 2018-02-16 中国科学院上海药物研究所 A kind of pyrido nitrogen heterocyclic and its production and use
US11369623B2 (en) 2015-06-16 2022-06-28 Prism Pharma Co., Ltd. Anticancer combination of a CBP/catenin inhibitor and an immune checkpoint inhibitor
WO2017029362A1 (en) 2015-08-19 2017-02-23 Sandoz Ag Asymmetric bisamidation of malonic ester derivatives
US20180237378A1 (en) * 2015-08-19 2018-08-23 Sandoz Ag Asymmetric Bisamidation of Malonic Ester Derivatives
CN105218445A (en) * 2015-08-25 2016-01-06 江苏中邦制药有限公司 The preparation method of a kind of TYR enzyme inhibitors Foretinib
CN105218445B (en) * 2015-08-25 2018-05-22 江苏中邦制药有限公司 A kind of preparation method of tyrosine kinase inhibitor Foretinib
CN106543143B (en) * 2015-09-22 2019-03-22 合肥中科普瑞昇生物医药科技有限公司 A kind of novel FLT3 kinase inhibitor and application thereof
CN106543143A (en) * 2015-09-22 2017-03-29 合肥中科普瑞昇生物医药科技有限公司 New FLT3 kinase inhibitors of one class and application thereof
CN105541798A (en) * 2016-02-03 2016-05-04 中国人民解放军第二军医大学 Quinoline multi-target kinase inhibitor with antitumor activity and preparation method thereof
CN107129465B (en) * 2016-02-26 2020-06-19 中国科学院上海药物研究所 Aryl guanidine compound and preparation method and application thereof
CN107129465A (en) * 2016-02-26 2017-09-05 中国科学院上海药物研究所 Aryl guanidine compound and its production and use
WO2017173049A1 (en) * 2016-03-31 2017-10-05 Merck Patent Gmbh Compounds for the inhibition of cyclophilins and uses thereof
US11141413B2 (en) 2016-04-15 2021-10-12 Exelixis, Inc. Method of treating renal cell carcinoma using N-(4-(6,7-dimethoxyquinolin-4-yloxy)phenyl)-N′-(4-fluorophenyl)cyclopropane-1,1-dicarboxamide, (2S)-hydroxybutanedioate
CN105906568A (en) * 2016-04-28 2016-08-31 西安交通大学 Cyclopropane diamide compound with antitumor activity and preparation method and application thereof
CN106083715A (en) * 2016-06-01 2016-11-09 谢阳 A kind of quinoline, quinazoline compounds and pharmaceutical composition thereof and application
CN106008371A (en) * 2016-06-24 2016-10-12 谢阳 1-aryl ureido naphthenic base-1-formamide compound and pharmaceutical composition and application thereof
CN107235896A (en) * 2016-09-13 2017-10-10 上海翔锦生物科技有限公司 Tyrosine kinase inhibitor and its application
CN110437145A (en) * 2016-09-13 2019-11-12 上海翔锦生物科技有限公司 Tyrosine kinase inhibitor and its application
CN107235896B (en) * 2016-09-13 2019-11-05 上海翔锦生物科技有限公司 Tyrosine kinase inhibitor and its application
US20190248772A1 (en) * 2016-09-13 2019-08-15 Shanghai Xiangjin Biotechnology Co., Ltd. Tyrosine kinase inhibitor and application thereof
AU2017325641B2 (en) * 2016-09-13 2021-08-26 LaNova Medicines Limited Tyrosine kinase inhibitor and application thereof
US10882853B2 (en) * 2016-09-13 2021-01-05 LaNova Medicines Limited Tyrosine kinase inhibitor and application thereof
WO2018064191A1 (en) 2016-09-27 2018-04-05 The Usa Of America, As Represented By The Secretary, Department Of Health & Human Services Method of treating urothelial carcinoma and other genitourinary malignancies using n-(4-(6,7-dimethoxyquinolin-4-yloxy)-phenyl)-n'(4-fluorophenyl)cyclopropane-1,1-dicarboxamide
TWI742181B (en) * 2016-10-18 2021-10-11 大陸商北京康辰藥業股份有限公司 Quinolinyl substituted carboxylic acid compound or its pharmaceutically acceptable salt, its racemate or enantiomer, its preparation method, its pharmaceutical composition and application
CN109496212A (en) * 2016-10-18 2019-03-19 北京康辰药业股份有限公司 A kind of carboxylic acid compound or its pharmaceutically acceptable salt, its pharmaceutical composition and application that quinolyl replaces
US10723701B2 (en) 2016-10-18 2020-07-28 Beijing Konruns Pharmaceutical Co., Ltd. Quinolyl-substituted carboxylic acid compound or pharmaceutically acceptable salt thereof, pharmaceutical composition of the same, and use of the same
KR20190066626A (en) * 2016-10-18 2019-06-13 베이징 콘런스 파마슈티칼 컴퍼니 리미티드 A carboxylic acid compound substituted with a quinoline group or a pharmaceutically acceptable salt thereof, a pharmaceutical composition thereof and a use thereof
CN109496212B (en) * 2016-10-18 2020-08-14 北京康辰药业股份有限公司 Quinolyl-substituted carboxylic acid compound or pharmaceutically acceptable salt thereof, and pharmaceutical composition and application thereof
WO2018072614A1 (en) * 2016-10-18 2018-04-26 北京康辰药业股份有限公司 Quinolinyl-substituted carboxylic acid compound or pharmaceutically acceptable salt thereof, pharmaceutical composition thereof, and use thereof
KR102262280B1 (en) 2016-10-18 2021-06-09 베이징 콘런스 파마슈티칼 컴퍼니 리미티드 Carboxylic acid compound substituted with quinoline group or pharmaceutically acceptable salt thereof, pharmaceutical composition and use thereof
AU2017346104B2 (en) * 2016-10-18 2020-06-11 Beijing Konruns Pharmaceutical Co., Ltd. Quinolinyl-substituted carboxylic acid compound or pharmaceutically acceptable salt thereof, pharmaceutical composition thereof, and use thereof
US10799503B2 (en) 2016-12-01 2020-10-13 Ignyta, Inc. Methods for the treatment of cancer
CN106632028A (en) * 2016-12-22 2017-05-10 上海再启生物技术有限公司 Cabozantinib preparation method
CN106632028B (en) * 2016-12-22 2019-01-22 上海再启生物技术有限公司 A kind of rich preparation method for Buddhist nun of card
CN108329298A (en) * 2017-01-17 2018-07-27 南昌弘益药业有限公司 A kind of quinolines noval chemical compound preparation method
WO2018133159A1 (en) * 2017-01-17 2018-07-26 南昌弘益药业有限公司 Method for preparing novel quinoline compound
WO2018136796A1 (en) 2017-01-20 2018-07-26 Exelixis, Inc. Combinations of cabozantinib and atezolizumab to treat cancer
US11110062B2 (en) 2017-02-15 2021-09-07 Taiho Pharmaceutical Co., Ltd. Pharmaceutical composition
CN111788207A (en) * 2017-02-27 2020-10-16 北京赛特明强医药科技有限公司 Dioxane quinoline compound and preparation method and application thereof
CN110156803A (en) * 2017-02-27 2019-08-23 北京赛特明强医药科技有限公司 Dioxanes and quinolines and the preparation method and application thereof
AU2019218187B2 (en) * 2017-02-27 2021-10-14 Beijing Scitech-Mq Pharmaceuticals Limited Dioxinoquinoline compounds, preparation method and uses thereof
CN111788207B (en) * 2017-02-27 2021-03-23 北京赛特明强医药科技有限公司 Dioxane quinoline compound and preparation method and application thereof
US11279675B2 (en) 2017-05-26 2022-03-22 Exelixis, Inc. Crystalline solid forms of salts of N-{4-[(6,7-dimethoxyquinolin-4-yl)oxy]phenyl}-N′-(4-fluorophenyl) cyclopropane-1, 1-dicarboxamide, processes for making, and methods of use
AU2018272088C1 (en) * 2017-05-26 2022-09-22 Exelixis, Inc. Crystalline solid forms of salts of N-{4-[(6,7-dimethoxyquinolin-4-yl) oxy]phenyl}-N'-(4-fluorphenyl) cyclopropane-1,1-dicarboxamide, processes for making, and methods of use
CN110621662A (en) * 2017-05-26 2019-12-27 埃克塞里艾克西斯公司 Crystalline solid form of a salt, process for preparation and method of use
WO2018218233A1 (en) 2017-05-26 2018-11-29 Exelixis, Inc. Crystalline solid forms of salts of n-{4-[(6,7-dimethoxyquinolin-4-yl) oxy]phenyl}-n'-(4-fluorphenyl) cyclopropane-1,1-dicarboxamide, processes for making, and methods of use
EA039654B1 (en) * 2017-05-26 2022-02-22 Экселиксис, Инк. Crystalline solid forms of salts of n-{4-[(6,7-dimethoxyquinolin-4-yl)oxy]phenyl}-n'-(4-fluorphenyl)cyclopropane-1,1-dicarboxamide, processes for making and methods of use
US11731941B2 (en) 2017-05-26 2023-08-22 Exelixis, Inc. Crystalline solid forms of salts of N-{4-[(6,7-dimethoxyquinolin-4-yl)oxy]phenyl}-N′-(4-fluorophenyl) cyclopropane-1,1-dicarboxamide, processes for making, and methods of use
AU2018272088B2 (en) * 2017-05-26 2022-06-16 Exelixis, Inc. Crystalline solid forms of salts of N-{4-[(6,7-dimethoxyquinolin-4-yl) oxy]phenyl}-N'-(4-fluorphenyl) cyclopropane-1,1-dicarboxamide, processes for making, and methods of use
EP3630726B1 (en) 2017-05-26 2021-12-22 Exelixis, Inc. Crystalline solid forms of salts of n-{4-[(6,7-dimethoxyquinolin-4-yl) oxy]phenyl}-n'-(4-fluorphenyl) cyclopropane-1,1-dicarboxamide, processes for making, and methods of use
CN110621662B (en) * 2017-05-26 2023-06-23 埃克塞里艾克西斯公司 Crystalline solid form of salt, preparation process and use method
WO2018227119A1 (en) 2017-06-09 2018-12-13 Exelixis, Inc. Liquid dosage forms to treat cancer
CN111793060B (en) * 2017-07-14 2023-06-06 先天肿瘤免疫公司 NLRP3 modulators
CN111793060A (en) * 2017-07-14 2020-10-20 先天肿瘤免疫公司 NLRP3 modulators
US11344543B2 (en) 2017-07-14 2022-05-31 Innate Tumor Immunity, Inc. NLRP3 modulators
WO2019014402A1 (en) * 2017-07-14 2019-01-17 Innate Tumor Immunity, Inc. Nlrp3 modulators
US11299481B2 (en) 2017-10-20 2022-04-12 Vanderbilt University Antagonists of the muscarinic acetylcholine receptor M4
CN111491920A (en) * 2017-10-20 2020-08-04 范德比尔特大学 Antagonists of muscarinic acetylcholine receptor M4
WO2019079783A1 (en) 2017-10-20 2019-04-25 Vanderbilt University Antagonists of the muscarinic acetylcholine receptor m4
EP3697759A4 (en) * 2017-10-20 2021-05-12 Vanderbilt University Antagonists of the muscarinic acetylcholine receptor m4
US11820757B2 (en) 2017-10-20 2023-11-21 Vanderbilt University Antagonists of the muscarinic acetylcholine receptor M4
CN111491920B (en) * 2017-10-20 2024-01-30 范德比尔特大学 Antagonists of muscarinic acetylcholine receptor M4
US11352344B2 (en) 2017-10-31 2022-06-07 Vanderbilt University Antagonists of the muscarinic acetylcholine receptor M4
CN109824587A (en) * 2017-11-23 2019-05-31 上海翔锦生物科技有限公司 The preparation method of tyrosine kinase inhibitor XJF007 and its intermediate
CN109896997A (en) * 2017-12-08 2019-06-18 中国药科大学 The preparation method and its usage of N- anilid class c-Met kinase inhibitor
US11358949B2 (en) 2017-12-20 2022-06-14 Angex Pharmaceutical, Inc. Carbamate and urea compounds as multikinase inhibitors
US11542259B2 (en) 2018-01-26 2023-01-03 Exelixis, Inc. Compounds for the treatment of kinase-dependent disorders
US11673897B2 (en) 2018-01-26 2023-06-13 Exelixis, Inc. Compounds for the treatment of kinase-dependent disorders
CN111936487A (en) * 2018-01-26 2020-11-13 埃克塞里艾克西斯公司 Compounds for the treatment of kinase-dependent disorders
US11708367B2 (en) 2018-01-26 2023-07-25 Exelixis, Inc. Compounds for the treatment of kinase-dependent disorders
CN112312909A (en) * 2018-01-26 2021-02-02 埃克塞里艾克西斯公司 Compounds for the treatment of kinase-dependent disorders
WO2019148043A1 (en) * 2018-01-26 2019-08-01 Exelixis, Inc. Compounds for the treatment of kinase-dependent disorders
US11414406B2 (en) 2018-02-02 2022-08-16 Vanderbilt University Antagonists of the muscarinic acetylcholine receptor M4
CN108264482A (en) * 2018-02-05 2018-07-10 南京法恩化学有限公司 It is a kind of to block the rich preparation method for Buddhist nun
US11407760B2 (en) 2018-02-11 2022-08-09 Beijing Scitech-Mq Pharmaceuticals Limited Dioxinoquinoline compounds, preparation method and uses thereof
WO2019180141A1 (en) 2018-03-23 2019-09-26 Bayer Aktiengesellschaft Combinations of rogaratinib
US11498902B2 (en) 2018-06-05 2022-11-15 Natco Pharma Limited Process for the preparation of Cabozantinib and its pharmaceutically acceptable salts thereof
US20210309668A1 (en) * 2018-08-01 2021-10-07 Agency For Science, Technology And Research Bicyclic compounds as kinase modulators, methods and uses thereof
US11702425B2 (en) * 2018-08-01 2023-07-18 Agency For Science, Technology And Research Bicyclic compounds as kinase modulators, methods and uses thereof
CN110862398B (en) * 2018-08-27 2021-04-06 北京赛特明强医药科技有限公司 Urea substituted aromatic ring-linked dioxane quinazoline or quinoline compound, composition and application thereof
CN110862398A (en) * 2018-08-27 2020-03-06 北京赛特明强医药科技有限公司 Urea substituted aromatic ring-linked dioxane quinazoline or quinoline compound, composition and application thereof
CN111303024B (en) * 2018-12-12 2023-03-28 安徽中科拓苒药物科学研究有限公司 Quinoline-structured pan-KIT kinase inhibitor and application thereof
WO2020118753A1 (en) * 2018-12-12 2020-06-18 安徽中科拓苒药物科学研究有限公司 Pan-kit kinase inhibitor having quinoline structure and application thereof
RU2789405C2 (en) * 2018-12-12 2023-02-02 Тарапьютикс Сайенс Инк. Pan-kit kinase inhibitor having quinoline structure and its use
AU2018453128B2 (en) * 2018-12-12 2022-03-17 Tarapeutics Science Inc. Pan-KIT kinase inhibitor having quinoline structure and application thereof
CN111303024A (en) * 2018-12-12 2020-06-19 安徽中科拓苒药物科学研究有限公司 Quinoline-structured pan-KIT kinase inhibitor and application thereof
EP3896059A4 (en) * 2018-12-12 2022-08-17 Tarapeutics Science Inc. Pan-kit kinase inhibitor having quinoline structure and application thereof
WO2020123800A1 (en) * 2018-12-13 2020-06-18 Exelixis, Inc. Crystalline forms and salt forms of a kinase inhibitor
WO2020216188A1 (en) * 2019-04-22 2020-10-29 北京康辰药业股份有限公司 Crystal forms of compound, preparation method therefor, pharmaceutical composition and application thereof
CN112119062B (en) * 2019-04-22 2023-09-05 北京康辰药业股份有限公司 Compound crystal form, preparation method thereof, pharmaceutical composition and application
CN112119062A (en) * 2019-04-22 2020-12-22 北京康辰药业股份有限公司 Compound crystal form, preparation method, pharmaceutical composition and application thereof
WO2020247019A1 (en) * 2019-06-03 2020-12-10 Exelixis, Inc. Crystalline salt forms of a kinase inhibitor
CN110117254B (en) * 2019-06-20 2022-05-13 江苏君若药业有限公司 Preparation method of cabozantinib
CN110117254A (en) * 2019-06-20 2019-08-13 江苏君若医药有限公司 Preparation method of cabozantinib
CN113577066A (en) * 2020-04-30 2021-11-02 中国科学院上海药物研究所 Use of arylguanidine compounds or pharmaceutically acceptable salts thereof
WO2021218884A1 (en) * 2020-04-30 2021-11-04 中国科学院上海药物研究所 Use of aryl guanidine compound or pharmaceutically acceptable salt thereof
IT202000027678A1 (en) 2020-11-18 2022-05-18 Indena Spa CABOZANTINIB-(S)-MALATO AMORPHOUS SOLID DISPERSIONS AND PROCESSES FOR THEIR PREPARATION
WO2022201079A1 (en) * 2021-03-24 2022-09-29 Biocon Limited Process for preparation of cabozantinib
WO2023098853A1 (en) * 2021-12-03 2023-06-08 湖南湘源美东医药科技有限公司 Cocrystal of cabozantinib, preparation method therefor and application thereof as drug or in pharmaceutical preparation
WO2023165948A1 (en) 2022-03-01 2023-09-07 Synthon B.V. Cabozantinib salt with l-(+)-tartaric acid and solid forms thereof
WO2023222946A1 (en) 2022-05-18 2023-11-23 Fermion Oy Process for the preparation of cabozantinib

Also Published As

Publication number Publication date
HK1164312A1 (en) 2012-09-21
US20070225307A1 (en) 2007-09-27
DK2392564T3 (en) 2014-01-13
US20090170896A1 (en) 2009-07-02
US20090105299A1 (en) 2009-04-23
EP2210607B1 (en) 2011-08-17
JP5325172B2 (en) 2013-10-23
AU2010204461B2 (en) 2013-06-20
PL2213661T3 (en) 2011-12-30
DK2392565T3 (en) 2014-05-05
US11124482B2 (en) 2021-09-21
DK2213661T3 (en) 2011-09-19
US8497284B2 (en) 2013-07-30
US20140155378A9 (en) 2014-06-05
SI2213661T1 (en) 2011-11-30
JP5185331B2 (en) 2013-04-17
PL2210607T3 (en) 2012-01-31
CY1111956T1 (en) 2015-11-04
JP2019070015A (en) 2019-05-09
JP2014055190A (en) 2014-03-27
PT2213661E (en) 2011-12-15
HRP20140012T1 (en) 2014-02-14
CY2014037I1 (en) 2015-11-04
JP2014074059A (en) 2014-04-24
ES2651730T3 (en) 2018-01-29
US20130252940A1 (en) 2013-09-26
PT2210607E (en) 2012-01-31
BE2014C055I2 (en) 2023-12-14
HK1256476A1 (en) 2019-09-27
SI2392564T1 (en) 2014-02-28
US20070244116A1 (en) 2007-10-18
EP3329918B1 (en) 2022-05-25
AU2010204461A1 (en) 2010-08-19
ATE517091T1 (en) 2011-08-15
JP5325173B2 (en) 2013-10-23
JP2010280674A (en) 2010-12-16
JP5694501B2 (en) 2015-04-01
US20150376133A1 (en) 2015-12-31
US20110077233A1 (en) 2011-03-31
AU2010204459B2 (en) 2013-06-20
LU92508I2 (en) 2015-11-02
US8178532B2 (en) 2012-05-15
CY2014037I2 (en) 2019-07-10
PL2392565T3 (en) 2014-08-29
EP2609919A2 (en) 2013-07-03
US7579473B2 (en) 2009-08-25
US9174947B2 (en) 2015-11-03
JP2017160270A (en) 2017-09-14
US20120184523A1 (en) 2012-07-19
EP2409704A2 (en) 2012-01-25
AU2004275842A1 (en) 2005-04-07
JP2010235631A (en) 2010-10-21
EP2392564A1 (en) 2011-12-07
DK2210607T3 (en) 2011-12-12
ES2369652T3 (en) 2011-12-02
EP2409704B1 (en) 2017-09-20
EP2609919A3 (en) 2014-02-26
JP2021050233A (en) 2021-04-01
SI2210607T1 (en) 2011-11-30
US8476298B2 (en) 2013-07-02
CA2744997A1 (en) 2005-04-07
JP2024023447A (en) 2024-02-21
EP1673085B1 (en) 2011-11-09
US20200331860A1 (en) 2020-10-22
US20160185725A1 (en) 2016-06-30
CA2537812C (en) 2013-01-22
EP3329918A3 (en) 2018-08-15
US20140155396A1 (en) 2014-06-05
SI2392565T1 (en) 2014-05-30
CY1112056T1 (en) 2015-11-04
ES2436888T3 (en) 2014-01-07
WO2005030140A3 (en) 2005-05-19
EP2213661A1 (en) 2010-08-04
HRP20110810T1 (en) 2011-12-31
JP2010235632A (en) 2010-10-21
EP2392564B1 (en) 2013-10-23
JP2010265302A (en) 2010-11-25
US20120022065A1 (en) 2012-01-26
NL300678I2 (en) 2017-01-31
JP2011042686A (en) 2011-03-03
HUS1400052I1 (en) 2022-02-28
EP1673085A4 (en) 2008-12-10
JP2014055191A (en) 2014-03-27
HRP20110707T1 (en) 2011-10-31
JP2007506777A (en) 2007-03-22
EP2409704A3 (en) 2013-02-13
EP2612853A1 (en) 2013-07-10
EP1673085A2 (en) 2006-06-28
US20210253530A9 (en) 2021-08-19
ES2466818T3 (en) 2014-06-11
ATE520403T1 (en) 2011-09-15
EP2392565B1 (en) 2014-03-19
HK1143068A1 (en) 2010-12-24
AU2010204459A1 (en) 2010-08-19
JP2015131859A (en) 2015-07-23
ATE532782T1 (en) 2011-11-15
US20070054928A1 (en) 2007-03-08
CA2537812A1 (en) 2005-04-07
US8067436B2 (en) 2011-11-29
EP2213661B1 (en) 2011-07-20
AU2004275842B2 (en) 2010-09-02
HK1164313A1 (en) 2012-09-21
EP2392565A1 (en) 2011-12-07
ES2371383T3 (en) 2011-12-30
ES2925655T3 (en) 2022-10-19
EP3329918A2 (en) 2018-06-06
EP2210607A1 (en) 2010-07-28
JP4638436B2 (en) 2011-02-23
HRP20140563T1 (en) 2014-07-18
HK1141799A1 (en) 2010-11-19
US20170355678A1 (en) 2017-12-14
JP5185332B2 (en) 2013-04-17

Similar Documents

Publication Publication Date Title
US11124482B2 (en) C-met modulators and methods of use
JP2007506777A5 (en)
EP1874759A1 (en) C-met modulators and methods of use
AU2020201638A1 (en) c-Met modulators and methods of use

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

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

AL Designated countries for regional patents

Kind code of ref document: A2

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

121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 2537812

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 2004275842

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2006528265

Country of ref document: JP

ENP Entry into the national phase

Ref document number: 2004275842

Country of ref document: AU

Date of ref document: 20040924

Kind code of ref document: A

WWP Wipo information: published in national office

Ref document number: 2004275842

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2004789057

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 2004789057

Country of ref document: EP