US20030134836A1 - Substituted arylamine derivatives and methods of use - Google Patents

Substituted arylamine derivatives and methods of use Download PDF

Info

Publication number
US20030134836A1
US20030134836A1 US10/197,960 US19796002A US2003134836A1 US 20030134836 A1 US20030134836 A1 US 20030134836A1 US 19796002 A US19796002 A US 19796002A US 2003134836 A1 US2003134836 A1 US 2003134836A1
Authority
US
United States
Prior art keywords
methyl
optionally substituted
phenyl
boc
amino
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US10/197,960
Other languages
English (en)
Inventor
Daniel Elbaum
Benny Askew
Shon Booker
Julie Germain
Gregory Habgood
Michael Handley
Tae-Seong Kim
Aiwen Li
Nobuko Nishimura
Vinod Patel
Chester Yuan
Joseph Kim
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Amgen Inc
Original Assignee
Amgen 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
Priority claimed from US10/046,526 external-priority patent/US20020147198A1/en
Application filed by Amgen Inc filed Critical Amgen Inc
Priority to US10/197,960 priority Critical patent/US20030134836A1/en
Assigned to AMGEN INC. reassignment AMGEN INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: ASKEW, BENNY, NISHIMURA, NOBUKO, YUAN, CHESTER CHENGUANG, BOOKER, SHON, ELBAUM, DANIEL, GERMAIN, JULIE, HABGOOD, GREGORY J., KIM, JOSEPH L., KIM, TAE-SEONG, PATEL, VINOD F., LI, AIWEN
Priority to CA002492164A priority patent/CA2492164A1/en
Priority to PL376532A priority patent/PL376532A1/pl
Priority to MXPA05000651A priority patent/MXPA05000651A/es
Priority to JP2004521923A priority patent/JP2006505511A/ja
Priority to EP03764756A priority patent/EP1583744A2/en
Priority to AU2003256577A priority patent/AU2003256577A1/en
Priority to PCT/US2003/022276 priority patent/WO2004007457A2/en
Publication of US20030134836A1 publication Critical patent/US20030134836A1/en
Priority to US10/823,809 priority patent/US7101868B2/en
Priority to US10/996,035 priority patent/US20050153960A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/60Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom with 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
    • C07D213/78Carbon atoms having three bonds to hetero atoms, with at the most one bond to halogen, e.g. ester or nitrile radicals
    • C07D213/81Amides; Imides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • A61P15/08Drugs for genital or sexual disorders; Contraceptives for gonadal disorders or for enhancing fertility, e.g. inducers of ovulation or of spermatogenesis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/02Drugs for dermatological disorders for treating wounds, ulcers, burns, scars, keloids, or the like
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • A61P27/06Antiglaucoma agents or miotics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/04Anorexiants; Antiobesity agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/20Antivirals for DNA viruses
    • A61P31/22Antivirals for DNA viruses for herpes viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic 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
    • C04CEMENTS; CONCRETE; ARTIFICIAL STONE; CERAMICS; REFRACTORIES
    • C04BLIME, MAGNESIA; SLAG; CEMENTS; COMPOSITIONS THEREOF, e.g. MORTARS, CONCRETE OR LIKE BUILDING MATERIALS; ARTIFICIAL STONE; CERAMICS; REFRACTORIES; TREATMENT OF NATURAL STONE
    • C04B35/00Shaped ceramic products characterised by their composition; Ceramics compositions; Processing powders of inorganic compounds preparatory to the manufacturing of ceramic products
    • C04B35/622Forming processes; Processing powders of inorganic compounds preparatory to the manufacturing of ceramic products
    • C04B35/626Preparing or treating the powders individually or as batches ; preparing or treating macroscopic reinforcing agents for ceramic products, e.g. fibres; mechanical aspects section B
    • C04B35/63Preparing or treating the powders individually or as batches ; preparing or treating macroscopic reinforcing agents for ceramic products, e.g. fibres; mechanical aspects section B using additives specially adapted for forming the products, e.g.. binder binders
    • C04B35/632Organic additives
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/60Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom with 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
    • C07D213/78Carbon atoms having three bonds to hetero atoms, with at the most one bond to halogen, e.g. ester or nitrile radicals
    • C07D213/81Amides; Imides
    • C07D213/82Amides; Imides in position 3
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D237/00Heterocyclic compounds containing 1,2-diazine or hydrogenated 1,2-diazine rings
    • C07D237/02Heterocyclic compounds containing 1,2-diazine or hydrogenated 1,2-diazine rings not condensed with other rings
    • C07D237/04Heterocyclic compounds containing 1,2-diazine or hydrogenated 1,2-diazine rings not condensed with other rings having less than three double bonds between ring members or between ring members and non-ring members
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D237/00Heterocyclic compounds containing 1,2-diazine or hydrogenated 1,2-diazine rings
    • C07D237/02Heterocyclic compounds containing 1,2-diazine or hydrogenated 1,2-diazine rings not condensed with other rings
    • C07D237/06Heterocyclic compounds containing 1,2-diazine or hydrogenated 1,2-diazine rings not condensed with other rings having three double bonds between ring members or between ring members and non-ring members
    • C07D237/10Heterocyclic compounds containing 1,2-diazine or hydrogenated 1,2-diazine rings not condensed with other rings having three double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D237/24Carbon atoms having three bonds to hetero atoms with at the most one bond to halogen
    • 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
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/02Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings
    • C07D405/12Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D409/00Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms
    • C07D409/02Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings
    • C07D409/12Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/12Heterocyclic 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
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems

Definitions

  • This invention is in the field of pharmaceutical agents and specifically relates to compounds, compositions, uses and methods for treating cancer and angiogenesis-related disorders.
  • Protein kinases represent a large family of proteins which play a central role in the regulation of a wide variety of cellular processes, maintaining control over cellular function.
  • a partial list of such kinases includes abl, Akt, bcr-ab1, Blk, Brk, Btk, c-kit, c-met, c-src, CDK1, CDK2, CDK3, CDK4, CDK5, CDK6, CDK7, CDK8, CDK9, CDK10, cRaf1, CSF1R, CSK, EGFR, ErbB2, ErbB3, ErbB4, Erk, Fak, fes, FGFR1, FGFR2, FGFR3, FGFR4, FGFR5, Fgr, flt-1, Fps, Frk, Fyn, Hck, IGF-1R, INS-R, Jak, KDR, Lck, Lyn, MEK, p38, PDGFR, PIK, PKC, PYK
  • Certain diseases are known to be associated with deregulated angiogenesis, for example ocular neovascularisation, such as retinopathies (including diabetic retinopathy), age-related macular degeneration, psoriasis, hemangioblastoma, hemangioma, arteriosclerosis, inflammatory disease, such as a rheumatoid or rheumatic inflammatory disease, especially arthritis (including rheumatoid arthritis), or other chronic inflammatory disorders, such as chronic asthma, arterial or post-transplantational atherosclerosis, endometriosis, and neoplastic diseases, for example so-called solid tumors and liquid tumors (such as leukemias).
  • retinopathies including diabetic retinopathy
  • age-related macular degeneration psoriasis
  • hemangioblastoma hemangioma
  • arteriosclerosis arteriosclerosis
  • inflammatory disease such as a rheumatoid or
  • VEGF Vascular Endothelial Growth Factor
  • VPF Vascular Permeability Factor
  • VEGF is a dimeric, disulfide-linked 46-kDa glycoprotein related to “Platelet-Derived Growth Factor” (PDGF); it is produced by normal cell lines and tumor cell lines; is an endothelial cell-specific mitogen; shows angiogenic activity in in vivo test systems (e.g. rabbit cornea); is chemotactic for endothelial cells and monocytes; and induces plasminogen activators in endothelial cells, which are involved in the proteolytic degradation of extracellular matrix during the formation of capillaries.
  • PDGF Platinum-Derived Growth Factor
  • VEGF receptors are transmembranous receptor tyrosine kinases. They are characterized by an extracellular domain with seven immunoglobulin-like domains and an intracellular tyrosine kinase domain.
  • Various types of VEGF receptor are known, e.g. VEGFR-1 (also known as flt-1), VEGFR-2 (also known as KDR), and VEGFR-3.
  • VEGF vascular endothelium
  • VEGF expression could explain the occurrence of cerebral edema in patients with glioma.
  • Direct evidence of the role of VEGF as a tumor angiogenesis factor in vivo is shown in studies in which VEGF expression or VEGF activity was inhibited. This was achieved with anti-VEGF antibodies, with dominant-negative VEGFR-2 mutants which inhibited signal transduction, and with antisense-VEGF RNA techniques. All approaches led to a reduction in the growth of glioma cell lines or other tumor cell lines in vivo as a result of inhibited tumor angiogenesis.
  • Angiogenesis is regarded as an absolute prerequisite for tumors which grow beyond a diameter of about 1-2 mm; up to this limit, oxygen and nutrients may be supplied to the tumor cells by diffusion. Every tumor, regardless of its origin and its cause, is thus dependent on angiogenesis for its growth after it has reached a certain size.
  • VEGF's are unique in that they are the only angiogenic growth factors known to contribute to vascular hyperpermeability and the formation of edema. Indeed, vascular hyperpermeability and edema that is associated with the expression or administration of many other growth factors appears to be mediated via VEGF production.
  • Inflammatory cytokines stimulate VEGF production.
  • Hypoxia results in a marked upregulation of VEGF in numerous tissues, hence situations involving infarct, occlusion, ischemia, anemia, or circulatory impairment typically invoke VEGF/VPF-mediated responses.
  • Vascular hyperpermeability, associated edema, altered transendothelial exchange and macromolecular extravasation, which is often accompanied by diapedesis, can result in excessive matrix deposition, aberrant stromal proliferation, fibrosis, etc.
  • VEGF-mediated hyperpermeability can significantly contribute to disorders with these etiologic features. As such, regulators of angiogenesis have become an important therapeutic target.
  • Japanese patent JP2000256358 describes pyrazole derivatives that block the calcium release-activated calcium channel.
  • WO01/29009 describes aminopyridines as KDR inhibitors.
  • WO01/30745 describes anthranilic acids as CGMP phosphodiesterase inhibitors.
  • WO00/02851 published Jan.
  • WO98/45268 describes nicotinamide derivatives as PDE4 inhibitors.
  • WO98/24771 describes benzamides as vasopressin antagonists.
  • Triazine-substituted amines are described for their aggregating ability (J. Amer. Chem. Soc., 115, 905-16 (1993).
  • Substituted imidazolines were tested for their antidepressant activity in Ind. J. Het. Chem., 2, 129-32 (1992).
  • N-(4-Pyridyl)anthranilic amides were described in Chem Abstr. 97:109837 (1981).
  • PCT publication WO99/32477 published Jul. 1, 1999, describes anthranilamides as anti-coagulants.
  • U.S. Pat. No. 6,140,351 describes anthranilamides as anti-coagulants.
  • PCT publication WO99/62885 published Dec. 9, 1999, describes 1-(4-aminophenyl)pyrazoles as antiinflammatories.
  • PCT publication WO00/39111 published Jul. 6, 2000, describes amides as factor Xa inhibitors.
  • PCT publication WO00/39117 published Jul. 6, 2000, describes heteroaromatic amides as factor Xa inhibitors.
  • PCT publication WO00/27819 published May 18, 2000, describes anthranilic acid amides as VEGF inhibitors.
  • PCT publication WO00/27820 published May 18, 2000 describes N-aryl anthranilic acid amides as VEGF inhibitors. 7-Chloroquinolinylamines are described in FR2168227 as antiinflammatories.
  • WO01/55114 published Aug. 2, 2001, describes nicotinamides for the treatment of cancer.
  • WO01/55115 published Aug. 2, 2001, describes nicotinamides for the treatment of apoptosis.
  • WO01/85715, published Nov. 15, 2001 describes substituted pyridines and pyrimidines as anti-angiogenesis agents.
  • PCT publication WO01/85691 published Nov. 15, 2001 describes anthranilic amides as VEGF inhibitors.
  • PCT publication WO01/85671 published Nov. 15, 2001 describes anthranyl amides as VEGF inhibitors.
  • PCT publication WO01/81311 published Nov. 1, 2001 describes anthranilic amides as VEGF inhibitors.
  • compounds of the current invention have not been described as inhibitors of angiogenesis such as for the treatment of cancer.
  • a class of compounds useful in treating cancer and angiogenesis is defined by Formula I
  • each of A 1 and A 2 is independently C or N;
  • a 1 -A 2 together are part of a ring A selected from 5- or 6-membered heteroaryl, more preferably 5-membered heteroaryl selected from thienyl, oxazolyl, imidazolyl, pyrrolyl, pyrazolyl, isoxazolyl, triazolyl, isothiazolyl, and 6-membered heteroaryl selected from pyridyl, pyrazinyl, pyrimidinyl and pyridazinyl, even more preferably pyridyl or pyrimidinyl, most preferably pyridyl;
  • Z is oxygen or sulfur
  • R a and R b are independently selected from H, halo, and C 1-4 -alkyl substituted with R 1 , or wherein R a and R b together form C 3 -C 4 cycloalkyl, preferably H, halo, and C 1-2 -alkyl substituted with R 1 , or
  • R a and R b together form C 3 -C 4 cycloalkyl, more preferably H, halo and C 1 -C 2 -alkyl, even more preferably H;
  • R c is C 1 -C 4 alkylenyl, where one of the CH 2 groups may be substituted with an oxygen atom or an —NH—, preferably C 1 -C 2 alkylenyl, where one of the CH 2 groups may be substituted with an oxygen atom or an —NH—, more preferably —CH 2 —;
  • R 1 is one or more substituents independently selected from H, halo, —OR 7 , oxo, —SR 7 , —CO 2 R 7 , —COR 7 , —CONR 7 R 7 , —NR 7 R 7 , —SO 2 NR 7 R 7 , —NR 7 C(O)OR 7 , —NR 7 C(O)R 7 , cycloalkyl, optionally substituted phenylalkylenyl, optionally substituted 5-6 membered heterocyclyl, optionally substituted heteroarylalkylenyl, optionally substituted phenyl, lower alkyl, cyano, lower hydroxyalkyl, lower carboxyalkyl, nitro, lower alkenyl, lower alkynyl, lower aminoalkyl, lower alkylaminoalkyl and lower haloalkyl;
  • R 2 is selected from
  • aryl selected from phenyl, naphthyl, indenyl and tetrahydronaphthyl, substituted or unsubstituted 5-6 membered heteroaryl, and substituted or unsubstituted 9-10 membered fused heteroaryl,
  • substituted R 2 is substituted with one or more substituents independently selected from halo, —OR 7 , —SR 7 , —SO 2 R 7 , —CO 2 R 7 , —CONR 7 R 7 , —COR 7 , —NR 7 R 7 , —SO 2 NR 7 R 7 , —NR 7 C(O)OR 7 , —NR 7 C(O)R 7 , —NH (C 1 -C 4 alkylenylR 7 ), optionally substituted cycloalkyl, optionally substituted 5-6 membered heterocyclyl, optionally substituted phenyl, lower alkyl substituted with R 1 , cyano, nitro, lower alkenyl and lower alkynyl, preferably halo, —OR 7 , —SR 7 , —SO 2 R 7 , —CO 2 R 7 , —CONR 7 R 7 , —COR 7 , —NR 7 R 7
  • R 3 is selected from aryl, preferably phenyl
  • R 3 is substituted with one or more substituents independently selected from halo, —OR 7 , —SR 7 , —CO 2 R 7 , —CONR 7 R 7 , —COR 7 , —NR 7 R 7 , —SO 2 NR 7 R 7 , —NR 7 C(O)OR 7 , —NR 7 C (O)R 7 , cycloalkyl, optionally substituted 5-6 membered heterocyclyl, optionally substituted heteroarylalkylenyl, optionally substituted phenyl, lower alkyl substituted with R 1 , cyano, nitro, lower alkenyl and lower alkynyl, preferably halo, —OR 7 , —SR 7 , —CO 2 R 7 , —CONR 7 R 7 , —COR 7 , —NR 7 R 7 , —SO 2 NR 7 R 7 , —NR 7 C(O)OR 7 , —NR
  • R 4 is independently selected from C 2 -C 4 alkylenyl, C 2 -C 4 alkenylenyl and C 2 -C 4 alkynylenyl, where one of the CH 2 groups may be substituted with an oxygen atom or an —NH—, preferably C 2-3 -alkylenyl, where one of the CH 2 groups may be substituted with an oxygen atom or an —NH—;
  • R 5 is selected from H, lower alkyl, phenyl and lower aralkyl, preferably H or C 1-2 -alkyl;
  • R 6 is selected from H or C 1-6 -alkyl
  • R 7 is selected from H, lower alkyl, phenyl, 5-6 membered heterocyclyl, C 3 -C 6 cycloalkyl, and lower haloalkyl, preferably H, C 1-2 -alkyl, phenyl, C 3 -C 6 cycloalkyl and C 1-2 -haloalkyl, more preferably H, methyl, ethyl, cyclopropyl, cyclohexyl and trifluoromethyl;
  • R 3 is substituted with one or more radicals selected from —OR 7 , —SR 7 , —CO 2 R 7 , —CONR 7 R 7 , —COR 7 , —NR 7 R 7 , lower aminoalkyl, lower alkylaminoalkyl, —SO 2 NR 7 R 7 , —NR 7 C(O)OR 7 , —NR 7 C(O)R 7 , cyano or lower hydroxyalkyl.
  • the invention also relates to compounds of Formula II
  • each of A 3 and A 4 is independently C or N, provided at least one of A 3 and A 4 is N; wherein n is 1-2; wherein R 1 is one or more substituents independently selected from H, chloro, fluoro, bromo, amino, hydroxy, methyl, ethyl, propyl, trifluoromethyl, methoxy, ethoxy, trifluoromethoxy, carboxymethyl, unsubstituted or substituted phenyl and unsubstituted or substituted heteroaryl selected from thienyl, furanyl, pyridyl, imidazolyl and pyrazolyl; wherein R 2 is selected from phenyl, isoquinolyl and quinolyl, where R 2 is unsubstituted or substituted with one or more substituents selected from chloro, fluoro, amino, hydroxy, cyclohexyl, phenylmethyl, morpholinylmethyl, methylpiperdinylmethyl,
  • a class of compounds useful in treating cancer and angiogenesis is defined by Formula I′
  • each of A 1 and A 2 is independently C or N;
  • a 1 -A 2 form part of a ring A selected from 5- or 6-membered heteroaryl, preferably
  • Z is oxygen or sulfur
  • Y is selected from
  • p is 0 to 2, preferably 2;
  • R a and R b are independently selected from H, halo, cyano, —NHR 6 and C 1-4 -alkyl substituted with R 1 , or wherein R a and R b together form C 3 -C 6 cycloalkyl;
  • R a and R b together form C 3 -C 4 cycloalkyl, more preferably H, chloro, fluoro and C 1 -C 2 -alkyl, even more preferably H;
  • R z is selected from C 2 -C 6 -alkylenyl, where one of the CH 2 groups may be replaced with an oxygen atom or an —NH— group; wherein one of the CH 2 groups may be substituted with one or two radicals selected from halo, cyano, —NHR 6 and C 1-4 -alkyl substituted with R 1 ;
  • R d is optionally substituted cycloalkyl, preferably C 3-6 -cycloalkyl
  • R 1 is one or more substituents independently selected from H, halo, —OR 7 , oxo, —SR 7 , —CO 2 R 7 , —COR 7 , —CONR R 7 R 7 , —NR 7 R 7 , —SO 2 NR 7 R 7 , —NR 7 C(O)OR 7 , —NR 7 C(O)R 7 , optionally substituted cycloalkyl, optionally substituted phenylalkyl, optionally substituted heterocyclyl, optionally substituted heterocyclylalkyl, optionally substituted phenyl, lower alkyl, cyano, lower hydroxyalkyl, lower carboxyalkyl, nitro, lower alkenyl, lower alkynyl, lower aminoalkyl, lower alkylaminoalkyl and lower haloalkyl,
  • R 2 is selected from
  • aryl preferably phenyl, naphthyl, benzodioxolyl, indanyl, indenyl and tetrahydronaphthyl, more preferably phenyl, indanyl, tetrahydronaphthyl, and naphthyl,
  • cycloalkyl preferably C 3-6 -cycloalkyl, more preferably cyclohexyl
  • R 2 is substituted with one or more substituents independently selected from halo, —OR 7 , oxo, —SR 7 , —CO 2 R 7 , —CONR 7 R 7 , —COR 7 , —NR 7 R 7 , —NH (C 1 -C 4 alkylenylR 9 ), —SO 2 R 7 , —SO 2 NR 7 R 7 , —NR 7 C(O)OR 7 , —NR 7 C(O)R 7 , —NR 7 C(O) NR 7 R 7 , optionally substituted cycloalkyl, optionally substituted heterocyclyl, optionally substituted phenyl, halosulfonyl, cyano, alkylaminoalkoxy, alkylaminoalkoxyalkoxy, nitro, lower alkyl substituted with R 1 , lower alkenyl substituted with R 1 , and lower
  • C 1-4 -alkoxy even more preferably bromo, chloro, fluoro, iodo, nitro, amino, cyano, aminoethyl, Boc-aminoethyl, hydroxy, aminosulfonyl, 4-methylpiperazinylsulfonyl, cyclohexyl, phenyl, phenylmethyl, morpholinylmethyl, methylpiperazinylmethyl, morpholinylethyl, methylpiperazinylpropyl, 1-(4-morpholinyl)-2,2-dimethylpropyl, piperidinylmethyl, morpholinylpropyl, methylpiperidinylmethyl, piperidinylethyl, piperidinylpropyl, pyrrolidinylpropyl, pyrrolidinylpropenyl, pyrrolidinylbutenyl, fluorosulfonyl, methylsulfonyl,
  • R 3 is selected from unsubstituted or substituted aryl, preferably substituted phenyl,
  • substituted R 3 is substituted with one or more substituents independently selected from halo, —OR 7 , —SR 7 , —SO 2 R 7 , —CO 2 R 7 , —CONR 7 R 7 , —COR 7 , —NR 7 R 7 , —SO 2 NR 7 R 7 , —NR 7 C(O)OR 7 , —NR 7 C(O)R 7 , cycloalkyl, optionally substituted heterocyclyl, optionally substituted phenyl, nitro, alkylaminoalkoxyalkoxy, cyano, alkylaminoalkoxy, lower alkyl substituted with R 1 , lower alkenyl substituted with R 1 , and lower alkynyl substituted with R 1 ;
  • R 4 is independently selected from a direct bond, C 2-4 -alkylenyl, C 2-4 -alkenylenyl and C 2-4 -alkynylenyl, where one of the CH 2 groups may be substituted with an oxygen atom or —NH—, wherein R 4 is optionally substituted with hydroxy, preferably a direct bond or R 4a ;
  • R 4a is selected from C 2-4 -alkylenyl where one of the CH 2 groups may be replaced with an oxygen atom or —NH—,
  • R 4a is optionally substituted with hydroxy, preferably ethyl, butyl, and
  • R 5 is selected from H, lower alkyl, phenyl and lower aralkyl, preferably H, methyl or ethyl, more preferably H;
  • R 5a is selected from H, lower alkyl, phenyl and lower aralkyl, preferably H, methyl or ethyl, more preferably H;
  • R 6 is selected from H or C 1-6 -alkyl, preferably H or C 1-2 alkyl;
  • R 7 is selected from H, lower alkyl, optionally substituted phenyl, optionally substituted heterocyclyl, optionally substituted C 3 -C 6 -cycloalkyl, optionally substituted phenyl-C 1-6 -alkyl, optionally substituted heterocyclyl-C 1-6 -alkyl, optionally substituted C 3 -C 6 cycloalkyl-C 1-6 -alkyl, lower alkylaminoalkyl, and lower haloalkyl, preferably H, C 1-4 -alkyl, optionally substituted phenyl, optionally substituted phenyl-C 1-4 -alkyl, optionally substituted 4-6 membered heterocyclyl, optionally substituted 4-6 membered heterocyclyl-C 1-4 -alkyl, optionally substituted C 3 -C 6 cycloalkyl, C 1-2 -alkylamino-C 1-4 -alkyl and C 1-2 -haloalkyl,
  • R c is selected from H, methyl and optionally substituted phenyl
  • R e and R f are independently selected from H and C 1-2 -haloalkyl, preferably —CF 3 ;
  • R g is selected from H, C 1-6 -alkyl, optionally substituted phenyl-C 1-6 -alkyl, optionally substituted 4-6 membered heterocyclyl, optionally substituted 4-6 membered heterocyclyl-C 1 -C 6 -alkyl, C 1-4 -alkoxy-C 1-4 -alkyl and C 1-4 -alkoxy-C 1-4 -alkoxy-C 1-4 -alkyl, preferably H, C 1-3 -alkyl, optionally substituted phenyl-C 1-3 -alkyl, optionally substituted 4-6 membered heterocyclyl-C 1 -C 3 -alkyl, C 1-3 -alkoxy-C 1-3 -alkyl and C 1-3 -alkoxy-C 13 -alkoxy-C 1-3 -alkyl; and
  • R 9 is selected from H, optionally substituted phenyl, optionally substituted 4-6 membered heterocyclyl and C 3 -C 6 cycloalkyl;
  • R 2 is not 3-trifluoromethylphenyl when A is pyridyl, when X is —C(O)NH—, when Y is —NH—CH 2 —, when R 1 is H and R 3 is 3-(N-methylamino-carbonyl)phenyl, 4-hydroxyphenyl, 3-hydroxyphenyl or phenyl;
  • R 2 is not substituted with —SO 2 NR 7 R 7 when Y is —NHSO 2 —;
  • R 2 is not 3-trifluoromethylphenyl when A is pyridyl, when X is —C(O)NH—, when Y is —N(benzyl)-CH 2 —, when R 1 is H and when R 3 is phenyl;
  • R 2 is not cyclohexyl when A is pyridyl, when X is —C(O)NH—, when Y is —NH—CH 2 —, when R 1 is H and when R 3 is 2-methoxyphenyl or 3-methoxyphenyl;
  • R 1 is not 2-hydroxymethylpyrrol-5-yl when A is pyridyl;
  • R 1 is not 4-(methoxyaminocarbonylamino)phenyl when A is thienyl;
  • R 1 is not 2-pyridylmethoxy when A is pyrimidyl, when X is —C(O)NH—, and when Y is —NH—CH 2 —;
  • R 1 is not 4-methylpiperidyl when A is pyrimidyl, when X is —C(O)NH—, when Y is —NH—CH 2 —, and when R 3 is 3-chloro-4-methoxyphenyl;
  • R 1 is not bromo when A is pyrimidyl, when X is —C(O)NH—CH 2 —, when Y is —NH—CH 2 —, and when R 3 is 3-chloro-4-methoxyphenyl;
  • R 2 is not 2-chloro-3-pyridyl when A is pyridyl;
  • R 2 is not 2-methoxyphenyl when A is pyridyl, when X is —C(O)NH—, when Y is —NH—CH 2 —, when R 1 is H and R 3 is phenyl.
  • the invention also relates to compounds of Formula II′
  • each of A 3 and A 4 is independently CH or N, provided at least one of A 3 and A 4 is N;
  • n 1-2;
  • R 1 is one or more substituents independently selected from H, chloro, fluoro, bromo, hydroxy, methoxy, ethoxy, trifluoromethoxy, oxo, amino, dimethylamino, aminosulfonyl, carboxymethyl, cyclopropyl, optionally substituted phenyl, methyl, ethyl, propyl, cyano, hydroxymethyl, nitro, propenyl, propynyl, morpholinylethylamino, trifluoromethyl and unsubstituted or substituted heteroaryl selected from thienyl, furanyl, pyridyl, imidazolyl and pyrazolyl;
  • R 2 is a substituted or unsubstituted ring selected from phenyl, tetrahydronaphthyl, indanyl, benzodioxolyl, indenyl, naphthyl, isoxazolyl, pyrazolyl, thiazolyl, thiadiazolyl, thienyl, pyridyl, pyrimidinyl, pyridazinyl, 1,2-dihydroquinolyl, 1,2,3,4-tetrahydro-isoquinolyl, 1,2,3,4-tetrahydro-quinolyl, isoquinolyl, quinolyl, indolyl, isoindolyl, 2,3-dihydro-1H-indolyl, naphthyridinyl, quinozalinyl, 2,3,4, 4a, 9, 9a-hexahydro-1H-3-aza-fluorenyl, 5,6,7-trione
  • substituted R 2 is substituted with one or more substituents independently selected from bromo, chloro, fluoro, iodo, nitro, amino, cyano, aminoethyl, Boc-aminoethyl, hydroxy, oxo, aminosulfonyl, 4-methylpiperazinylsulfonyl, cyclohexyl, phenyl, phenylmethyl, morpholinylmethyl, 1-methylpiperazin-4-ylmethyl, 1-methylpiperazin-4-ylpropyl, morpholinylpropyl, piperidin-1-ylmethyl, 1-methylpiperidin-4-ylmethyl, 2-methyl-2-(1-methylpiperidin-4-yl)ethyl, morpholinylethyl, 1-(4-morpholinyl)-2,2-dimethylpropyl, piperidin-4-ylethyl, 1-Boc-piperidin-4-ylethyl, piperidin-1-
  • R 8 is one or more substituents independently selected from H, chloro, fluoro, bromo, hydroxy, methoxy, ethoxy, —O—CH 2 —O—, trifluoromethoxy, 1-methylpiperidinylmethoxy, dimethylaminoethoxy, amino, dimethylamino, dimethylaminopropyl, diethylamino, aminosulfonyl, cyclohexyl, dimethylaminopropynyl, 3-(4-morpholinyl)propyn-1-yl, dimethylaminoethoxyethoxy, 3-(4-morpholinyl)propylamino, optionally substituted piperidinyl, morpholinyl, optionally substituted piperazinyl, optionally substituted phenyl, methyl, ethyl, propyl, cyano, hydroxymethyl, aminomethyl, nitro and trifluoromethyl;
  • R 2 is not 3-trifluoromethylphenyl when A 3 is N, when A 4 is CH, when n is 1, when R 1 is H and R 8 is 4-hydroxy, 3-hydroxy or H; further provided R 2 is not 2-chloro-3-pyridyl when A 3 is N, when A 4 is CH, when n is 1, when R 1 is H and R 8 is H or 4-methoxy; and further provided R 2 is not 2-methoxyphenyl when A 3 is N, when A 4 is CH, when n is 1, when R 1 is H and R 8 is H.
  • the invention also relates to compounds of Formula III
  • R 1 is one or more substituents independently selected from
  • R 2 is selected from unsubstituted or substituted phenyl, and 9-10 membered bicyclic and 13-14 membered tricyclic unsaturated or partially unsaturated heterocyclyl, preferably phenyl, 1,2-dihydroquinolyl, 1,2,3,4-tetrahydro-isoquinolyl, 1,2,3,4-tetrahydro-quinolyl, 2,3-dihydro-1H-indolyl, 2,3,4,4a,9,9a-hexahydro-1H-3-aza-fluorenyl, 5,6,7-trihydro-1,2,4-triazolo[3,4-a]isoquinolyl, 3,4-dihydro-2H-benzo[1,4]oxazinyl, and benzo[l,4]dioxanyl, more preferably phenyl, 1,2,3,4-tetrahydro-isoquinolyl, 1,2,3,4-tetrahydro-quinolyl,
  • substituted R 2 is substituted with one or more substituents selected from halo, C 1-6 -alkyl, optionally substituted C 3-6 -cycloalkyl, optionally substituted phenyl, optionally substituted phenyl-C 1 -C 4 -alkylenyl, C 1-2 -haloalkoxy, optionally substituted phenyloxy, optionally substituted 4-6 membered heterocyclyl-C 1 -C 4 -alkyl, optionally substituted 4-6 membered heterocyclyl-C 2 -C 4 -alkenyl, optionally substituted 4-6 membered heterocyclyl, optionally substituted 4-6 membered heterocyclyloxy, optionally substituted 4-6 membered heterocyclyl-C 1 -C 4 -alkoxy, optionally substituted 4-6 membered heterocyclylsulfonyl, optionally substituted 4-6 membered heterocyclylamino, optionally substituted 4-6 member
  • C 1-4 -alkoxy preferably bromo, chloro, fluoro, iodo, nitro, amino, cyano, aminoethyl, Boc-aminoethyl, hydroxy, oxo, aminosulfonyl, 4-methylpiperazinylsulfonyl, cyclohexyl, phenyl, phenylmethyl, morpholinylmethyl, 1-methylpiperazin-4-ylmethyl, 1-methylpiperazin-4-ylpropyl, morpholinylpropyl, piperidin-1-ylmethyl, 1-methylpiperidin-4-ylmethyl, 2-methyl-2-(1-methylpiperidin-4-yl)ethyl, morpholinylethyl, 1-(4-morpholinyl)-2,2-dimethylpropyl, piperidin-4-ylethyl, 1-Boc-piperidin-4-ylethyl, piperidin-1-ylethyl, 1-Boc-pipe
  • R 2 is phenyl, it has a substituent selected from optionally substituted 4-6 membered heterocyclyl-C 1 -C 4 -alkyl, optionally substituted 4-6 membered heterocyclyl-C 2 -C 4 -alkenyl, optionally substituted 4-6 membered heterocyclyl, optionally substituted 4-6 membered heterocyclyloxy, optionally substituted 4-6 membered heterocyclyl-C 1 -C 4 -alkoxy, optionally substituted 4-6 membered heterocyclylsulfonyl, optionally substituted 4-6 membered heterocyclylamino, optionally substituted 4-6 membered heterocyclylcarbonyl, optionally substituted 4-6 membered heterocyclylcarbonyl-C 1-4 -alkyl, optionally substituted 4-6 membered heterocyclyl-C 1-4 -alkylcarbonyl;
  • R 7 is selected from H, C 1-3 -alkyl, optionally substituted phenyl-C 1-3 -alkyl, 4-6 membered heterocyclyl, and optionally substituted 4-6 membered heterocyclyl-C 1 -C 3 -alkyl;
  • R e and R f are independently selected from H and C 1-2 -haloalkyl, preferably —CF 3 ;
  • R g is selected from H, C 1-3 -alkyl, optionally substituted phenyl-C 1-3 -alkyl, 4-6 membered heterocyclyl, and optionally substituted 4-6 membered heterocyclyl-C 1 -C 3 -alkyl, C 1-3 -alkoxy-C 1-2 -alkyl and C 1-3 -alkoxy-C 1-3 -alkoxy-C 1-3 -alkyl; and
  • R 8 is one or more substituents selected from H, halo, amino, hydroxy, C 1-6 -alkyl, C 1-6 -haloalkyl, C 1-6 -alkoxy, C 1-6 -haloalkoxy, C 1-6 -aminoalkyl, C 1-6 -hydroxyalkyl, optionally substituted phenyl, optionally substituted heterocyclyl, optionally substituted heterocyclyl-C 1-6 -alkoxy, aminosulfonyl, C 3-6 -cycloalkyl, C 1-6 -alkylamino, C 1-6 -alkylamino-C 1-6 -alkyl, optionally substituted heterocyclyl-C 1-6 -alkylamino, optionally substituted heterocyclyl-C 1-6 -alkyl, C 1-6 -alkylamino-C 2-4 -alkynyl, C 1-6 -alkylamino-C 1-6 -alkoxy,
  • R 2 is not 3-trifluoromethylphenyl when R 1 is H and R 8 is 4-hydroxy, 3-hydroxy or H; and further provided R 2 is not 2-methoxyphenyl when R 1 is H and R 8 is H.
  • the invention also relates to compounds of Formula IV
  • R 2 is selected from unsubstituted or substituted phenyl, and 9-10 membered bicyclic and 11-14 membered tricyclic unsaturated or partially unsaturated heterocyclyl, preferably phenyl, 1,2-dihydroquinolyl, 1,2,3,4-tetrahydro-isoquinolyl, 1′,2′-dihydro-spiro[cyclopropane-1,3′-[3H]indol]-6′-yl, isoquinolyl, quinolyl, indolyl, isoindolyl, 2,3-dihydro-1H-indolyl, naphthyridinyl, 1,2,3,4-tetrahydro-[1,8]naphthyridinyl, quinozalinyl, benzo[d]isothiazolyl, 3,4-dihydro-quinazolinyl, 3,4-dihydro-2H-benzo[
  • substituted R 2 is substituted with one or more substituents selected from halo, C 1-6 -alkyl, optionally substituted C 3-6 -cycloalkyl, optionally substituted phenyl, optionally substituted phenyl-C 1 -C 4 -alkylenyl, C 1-2 -haloalkoxy, optionally substituted phenyloxy, optionally substituted 4-6 membered heterocyclyl-C 1 -C 6 -alkyl, optionally substituted 4-6 membered heterocyclyl-C 2 -C 4 -alkenyl, optionally substituted 4-6 membered heterocyclyl, optionally substituted 4-6 membered heterocyclyloxy, optionally substituted 4-6 membered heterocyclyl-C 1-4 -alkoxy, optionally substituted 4-6 membered heterocyclylsulfonyl, optionally substituted 4-6 membered heterocyclylamino, optionally substituted 4-6 membered heterocyclylamino
  • C 1-4 -alkoxy preferably bromo, chloro, fluoro, iodo, nitro, amino, cyano, Boc-aminoethyl, hydroxy, oxo, fluorosulfonyl, methylsulfonyl, aminosulfonyl, 4-methylpiperazinylsulfonyl, cyclohexyl, phenyl, phenylmethyl, 4-pyridylmethyl, 4-morpholinylmethyl, 1-methylpiperazin-4-ylmethyl, 1-methylpiperazin-4-ylpropyl, morpholinylpropyl, piperidin-1-ylmethyl, 1-methylpiperidin-4-ylmethyl, 2-methyl-2-(1-methylpiperidin-4-yl)ethyl, 2-methyl-2-(4-pyrimidinyl)ethyl, 2-methyl-2-(5-methyloxadiazol-2-yl)ethyl, 2-methyl-2-(pyra
  • R e and R f are independently selected from H and C 1-2 -haloalkyl, preferably trifluoromethyl;
  • R g is selected from H, C 1-3 -alkyl, optionally substituted phenyl-C 1-3 -alkyl, 4-6 membered heterocyclyl, and optionally substituted 4-6 membered heterocyclyl-C 1 -C 3 -alkyl, C 1-3 -alkoxy-C 1-2 -alkyl and C 1-3 -alkoxy-C 1-3 -alkoxy-C 1-3 -alkyl; and
  • R 8 is one or more substituents selected from halo, amino, nitro, hydroxy, C 1-6 -alkyl, C 1-6 -haloalkyl, C 1-6 -alkoxy, C 1-6 -haloalkoxy, C 1-6 -aminoalkyl, C 1-6 -hydroxyalkyl, optionally substituted phenyl, optionally substituted heterocyclyl, optionally substituted heterocyclyl-C 1-6 -alkoxy, aminosulfonyl, C 3-6 -cycloalkyl, C 1-6 -alkylamino, C 1-6 -alkylamino-C 1-6 -alkyl, optionally substituted heterocyclyl-C 1-6 -alkylamino, optionally substituted heterocyclyl-C 1-6 -alkyl, C 1-6 -alkylamino-C 2-4 -alkynyl, C 1-6 -alkylamino-C 1-6 -alkoxy
  • a family of specific compounds of particular interest within Formula I consists of compounds and pharmaceutically-acceptable derivatives thereof as follows:
  • Compounds of the present invention would be useful for, but not limited to, the prevention or treatment of angiogenesis-related diseases.
  • the compounds of the invention have kinase inhibitory activity, such as VEGFR/KDR inhibitory activity.
  • the compounds of the invention are useful in therapy as antineoplasia agents or to minimize deleterious effects of VEGF.
  • neoplasia including cancer and metastasis, including, but not limited to: carcinoma such as cancer of the bladder, breast, colon, kidney, liver, lung (including small cell lung cancer), esophagus, gall-bladder, ovary, pancreas, stomach, cervix, thyroid, prostate, and skin (including squamous cell carcinoma); hematopoietic tumors of lymphoid lineage (including leukemia, acute lymphocitic leukemia, acute lymphoblastic leukemia, B-cell lymphoma, T-cell-lymphoma, Hodgkin's lymphoma, non-Hodgkin's lymphoma, hairy cell lymphoma and Burkett's lymphoma); hematopoietic tumors of myeloid lineage (including acute and chronic myelogenous leukemias, myelodysplastic syndrome and promyelocytic leukemia);
  • carcinoma such as cancer of the bladder, breast, colon, kidney,
  • tumors of the central and peripheral nervous system including astrocytoma, neuroblastoma, glioma and schwannomas); and other tumors (including melanoma, seminoma, teratocarcinoma, osteosarcoma, xenoderoma pigmentosum, keratoctanthoma, thyroid follicular cancer and Kaposi's sarcoma).
  • the compounds are useful for the treatment of neoplasia selected from lung cancer, colon cancer and breast cancer.
  • the compounds also would be useful for treatment of ophthalmological conditions such as corneal graft rejection, ocular neovascularization, retinal neovascularization including neovascularization following injury or infection, diabetic retinopathy, retrolental fibroplasia and neovascular glaucoma; retinal ischemia; vitreous hemorrhage; ulcerative diseases such as gastric ulcer; pathological, but non-malignant, conditions such as hemangiomas, including infantile hemaginomas, angiofibroma of the nasopharynx and avascular necrosis of bone; and disorders of the female reproductive system such as endometriosis.
  • the compounds are also useful for the treatment of edema, and conditions of vascular hyperpermeability.
  • the compounds of the invention are useful in therapy of proliferative diseases. These compounds can be used for the treatment of an inflammatory rheumatoid or rheumatic disease, especially of manifestations at the locomotor apparatus, such as various inflammatory rheumatoid diseases, especially chronic polyarthritis including rheumatoid arthritis, juvenile arthritis or psoriasis arthropathy; paraneoplastic syndrome or tumor-induced inflammatory diseases, turbid effusions, collagenosis, such as systemic Lupus erythematosus, poly-myositis, dermato-myositis, systemic sclerodermia or mixed collagenosis; postinfectious arthritis (where no living pathogenic organism can be found at or in the affected part of the body), seronegative spondylarthritis, such as spondylitis ankylosans; vasculitis, sarcoidosis, or arthrosis; or further any combinations thereof.
  • synovial inflammation for example, synovitis, including any of the particular forms of synovitis, in particular bursal synovitis and purulent synovitis, as far as it is not crystal-induced.
  • synovial inflammation may for example, be consequential to or associated with disease, e.g. arthritis, e.g. osteoarthritis, rheumatoid arthritis or arthritis deformans.
  • the present invention is further applicable to the systemic treatment of inflammation, e.g. inflammatory diseases or conditions, of the joints or locomotor apparatus in the region of the tendon insertions and tendon sheaths.
  • Such inflammation may be, for example, be consequential to or associated with disease or further (in a broader sense of the invention) with surgical intervention, including, in particular conditions such as insertion endopathy, myofasciale syndrome and tendomyosis.
  • the present invention is further especially applicable to the treatment of inflammation, e.g. inflammatory disease or condition, of connective tissues including dermatomyositis and myositis.
  • These compounds can be used as active agents against such disease states as arthritis, atherosclerosis, psoriasis, hemangiomas, myocardial angiogenesis, coronary and cerebral collaterals, ischemic limb angiogenesis, wound healing, peptic ulcer Helicobacter related diseases, fractures, cat scratch fever, rubeosis, neovascular glaucoma and retinopathies such as those associated with diabetic retinopathy or macular degeneration.
  • diseases as arthritis, atherosclerosis, psoriasis, hemangiomas, myocardial angiogenesis, coronary and cerebral collaterals, ischemic limb angiogenesis, wound healing, peptic ulcer Helicobacter related diseases, fractures, cat scratch fever, rubeosis, neovascular glaucoma and retinopathies such as those associated with diabetic retinopathy or macular degeneration.
  • some of these compounds can be used as active agents against solid tumors, malignant ascites, hematopoietic cancers and hyperproliferative disorders such as thyroid hyperplasia (especially Grave's disease), and cysts (such as hypervascularity of ovarian stroma, characteristic of polycystic ovarian syndrome (Stein-Leventhal syndrome)) since such diseases require a proliferation of blood vessel cells for growth and/or metastasis.
  • thyroid hyperplasia especially Grave's disease
  • cysts such as hypervascularity of ovarian stroma, characteristic of polycystic ovarian syndrome (Stein-Leventhal syndrome)
  • some of these compounds can be used as active agents against burns, chronic lung disease, stroke, polyps, anaphylaxis, chronic and allergic inflammation, ovarian hyperstimulation syndrome, brain tumor-associated cerebral edema, high-altitude, trauma or hypoxia induced cerebral or pulmonary edema, ocular and macular edema, ascites, and other diseases where vascular hyperpermeability, effusions, exudates, protein extravasation, or edema is a manifestation of the disease.
  • the compounds will also be useful in treating disorders in which protein extravasation leads to the deposition of fibrin and extracellular matrix, promoting stromal proliferation (e.g. fibrosis, cirrhosis and carpal tunnel syndrome).
  • the compounds of the present invention are also useful in the treatment of ulcers including bacterial, fungal, Mooren ulcers and ulcerative colitis.
  • the compounds of the present invention are also useful in the treatment of conditions wherein undesired angiogenesis, edema, or stromal deposition occurs in viral infections such as Herpes simplex, Herpes Zoster, AIDS, Kaposi's sarcoma, protozoan infections and toxoplasmosis, following trauma, radiation, stroke, endometriosis, ovarian hyperstimulation syndrome, systemic lupus, sarcoidosis, synovitis, Crohn's disease, sickle cell anaemia, Lyme disease, pemphigoid, Paget's disease, hyperviscosity syndrome, Osler-Weber-Rendu disease, chronic inflammation, chronic occlusive pulmonary disease, asthma, and inflammatory rheumatoid or rheumatic disease.
  • the compounds are also useful in the reduction of sub-cutaneous fat and for the treatment of obesity.
  • the compounds of the present invention are also useful in the treatment of ocular conditions such as ocular and macular edema, glaucoma, ocular neovascular disease, scleritis, radial keratotomy, uveitis, vitritis, myopia, optic pits, chronic retinal detachment, post-laser complications, conjunctivitis, Stargardt's disease and Eales disease in addition to retinopathy and macular degeneration.
  • ocular conditions such as ocular and macular edema, glaucoma, ocular neovascular disease, scleritis, radial keratotomy, uveitis, vitritis, myopia, optic pits, chronic retinal detachment, post-laser complications, conjunctivitis, Stargardt's disease and Eales disease in addition to retinopathy and macular degeneration.
  • the compounds of the present invention are also useful in the treatment of cardiovascular conditions such as atherosclerosis, restenosis, arteriosclerosis, vascular occlusion and carotid obstructive disease.
  • the compounds of the present invention are also useful in the treatment of cancer related indications such as solid tumors, sarcomas (especially Ewing's sarcoma and osteosarcoma), retinoblastoma, rhabdomyosarcomas, neuroblastoma, hematopoietic malignancies, including leukemia and lymphoma, tumor-induced pleural or pericardial effusions, and malignant ascites.
  • cancer related indications such as solid tumors, sarcomas (especially Ewing's sarcoma and osteosarcoma), retinoblastoma, rhabdomyosarcomas, neuroblastoma, hematopoietic malignancies, including leukemia and lymphoma, tumor-induced pleural or pericardial effusions, and malignant ascites.
  • the compounds of the present invention are also useful in the treatment of diabetic conditions such as diabetic retinopathy and microangiopathy.
  • the compounds of this invention may also act as inhibitors of other protein kinases, e.g. p38, EGFR, CDK-2, CDK-5, IKK, JNK3, bFGFR, PDGFR and RAF and thus be effective in the treatment of diseases associated with other protein kinases.
  • protein kinases e.g. p38, EGFR, CDK-2, CDK-5, IKK, JNK3, bFGFR, PDGFR and RAF
  • these compounds are also useful for veterinary treatment of companion animals, exotic animals and farm animals, including mammals, rodents, and the like. More preferred animals include horses, dogs, and cats.
  • the compounds of the present invention include the pharmaceutically acceptable derivatives thereof.
  • a “pharmaceutically-acceptable derivative” denotes any salt, ester of a compound of this invention, or any other compound which upon administration to a patient is capable of providing (directly or indirectly) a compound of this invention, or a metabolite or residue thereof, characterized by the ability to inhibit angiogenesis.
  • treatment includes therapeutic treatment as well as prophylactic treatment (either preventing the onset of disorders altogether or delaying the onset of a preclinically evident stage of disorders in individuals).
  • neoplastic therapeutic agents prolong the survivability of the patient, inhibit the rapidly-proliferating cell growth associated with the neoplasm, or effect a regression of the neoplasm.
  • prevention includes either preventing the onset of disorders altogether or delaying the onset of a preclinically evident stage of disorders in individuals. This includes prophylactic treatment of those at risk of developing a disease, such as a cancer, for example. “Prophylaxis” is another term for prevention.
  • H denotes a single hydrogen atom. This radical may be attached, for example, to an oxygen atom to form a hydroxyl radical.
  • alkyl is used, either alone or within other terms such as “haloalkyl” and “alkylamino”, it embraces linear or branched radicals having one to about twelve carbon atoms. More preferred alkyl radicals are “lower alkyl” radicals having one to about six carbon atoms. Examples of such radicals include methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, pentyl, isoamyl, hexyl and the like. Even more preferred are lower alkyl radicals having one or two carbon atoms.
  • alkylenyl embraces bridging divalent alkyl radicals such as methylenyl and ethylenyl.
  • lower alkyl substituted with R 1 does not include an acetal moiety.
  • alkenyl embraces linear or branched radicals having at least one carbon-carbon double bond of two to about twelve carbon atoms. More preferred alkenyl radicals are “lower alkenyl” radicals having two to about six carbon atoms. Most preferred lower alkenyl radicals are radicals having two to about four carbon atoms. Examples of alkenyl radicals include ethenyl, propenyl, allyl, propenyl, butenyl and 4-methylbutenyl.
  • alkenyl and “lower alkenyl” embrace radicals having “cis” and “trans” orientations, or alternatively, “E” and “Z” orientations.
  • alkynyl denotes linear or branched radicals having at least one carbon-carbon triple bond and having two to about twelve carbon atoms. More preferred alkynyl radicals are “lower alkynyl” radicals having two to about six carbon atoms. Most preferred are lower alkynyl radicals having two to about four carbon atoms. Examples of such radicals include propargyl, butynyl, and the like.
  • halo means halogens such as fluorine, chlorine, bromine or iodine atoms.
  • haloalkyl embraces radicals wherein any one or more of the alkyl carbon atoms is substituted with halo as defined above. Specifically embraced are monohaloalkyl, dihaloalkyl and polyhaloalkyl radicals including perhaloalkyl.
  • a monohaloalkyl radical for one example, may have either an iodo, bromo, chloro or fluoro atom within the radical.
  • Dihalo and polyhaloalkyl radicals may have two or more of the same halo atoms or a combination of different halo radicals.
  • “Lower haloalkyl” embraces radicals having 1-6 carbon atoms.
  • haloalkyl radicals having one to three carbon atoms.
  • haloalkyl radicals include fluoromethyl, difluoromethyl, trifluoromethyl, chloromethyl, dichloromethyl, trichloromethyl, pentafluoroethyl, heptafluoropropyl, difluorochloromethyl, dichlorofluoromethyl, difluoroethyl, difluoropropyl, dichloroethyl and dichloropropyl.
  • Perfluoroalkyl means alkyl radicals having all hydrogen atoms replaced with fluoro atoms. Examples include trifluoromethyl and pentafluoroethyl.
  • hydroxyalkyl embraces linear or branched alkyl radicals having one to about ten carbon atoms any one of which may be substituted with one or more hydroxyl radicals. More preferred hydroxyalkyl radicals are “lower hydroxyalkyl” radicals having one to six carbon atoms and one or more hydroxyl radicals. Examples of such radicals include hydroxymethyl, hydroxyethyl, hydroxypropyl, hydroxybutyl and hydroxyhexyl. Even more preferred are lower hydroxyalkyl radicals having one to three carbon atoms.
  • alkoxy embrace linear or branched oxy-containing radicals each having alkyl portions of one to about ten carbon atoms. More preferred alkoxy radicals are “lower alkoxy” radicals having one to six carbon atoms. Examples of such radicals include methoxy, ethoxy, propoxy, butoxy and tert-butoxy. Even more preferred are lower alkoxy radicals having one to three carbon atoms. Alkoxy radicals may be further substituted with one or more halo atoms, such as fluoro, chloro or bromo, to provide “haloalkoxy” radicals. Even more preferred are lower haloalkoxy radicals having one to three carbon atoms. Examples of such radicals include fluoromethoxy, chloromethoxy, trifluoromethoxy, trifluoroethoxy, fluoroethoxy and fluoropropoxy.
  • aryl alone or in combination, means a carbocyclic aromatic system containing one or two rings wherein such rings may be attached together in a fused manner.
  • aryl embraces aromatic radicals such as phenyl, naphthyl, indenyl, tetrahydronaphthyl, and indanyl. More preferred aryl is phenyl.
  • Said “aryl” group may have 1 to 3 substituents such as lower alkyl, hydroxyl, halo, haloalkyl, nitro, cyano, alkoxy and lower alkylamino. Phenyl substituted with —O—CH 2 —O— forms the aryl benzodioxolyl substituent.
  • heterocyclyl embraces saturated, partially saturated and unsaturated heteroatom-containing ring radicals, where the heteroatoms may be selected from nitrogen, sulfur and oxygen. It does not include rings containing —O—O—, —O—S— or —S—S— portions.
  • Said “heterocyclyl” group may have 1 to 3 substituents such as hydroxyl, Boc, halo, haloalkyl, cyano, lower alkyl, lower aralkyl, oxo, lower alkoxy, amino and lower alkylamino.
  • saturated heterocyclic radicals include saturated 3 to 6-membered heteromonocyclic groups containing 1 to 4 nitrogen atoms [e.g. pyrrolidinyl, imidazolidinyl, piperidinyl, pyrrolinyl, piperazinyl]; saturated 3 to 6-membered heteromonocyclic group containing 1 to 2 oxygen atoms and 1 to 3 nitrogen atoms [e.g. morpholinyl]; saturated 3 to 6-membered heteromonocyclic group containing 1 to 2 sulfur atoms and 1 to 3 nitrogen atoms [e.g., thiazolidinyl].
  • partially saturated heterocyclyl radicals include dihydrothienyl, dihydropyranyl, dihydrofuryl and dihydrothiazolyl.
  • Examples of unsaturated heterocyclic radicals include unsaturated 5 to 6 membered heteromonocyclyl group containing 1 to 4 nitrogen atoms, for example, pyrrolyl, imidazolyl, pyrazolyl, 2-pyridyl, 3-pyridyl, 4-pyridyl, pyrimidyl, pyrazinyl, pyridazinyl, triazolyl [e.g., 4H-1,2,4-triazolyl, 1H-1,2,3-triazolyl, 2H-1,2,3-triazolyl]; unsaturated 5- to 6-membered heteromonocyclic group containing an oxygen atom, for example, pyranyl, 2-furyl, 3-furyl, etc.; unsaturated 5 to 6-membered heteromonocyclic group containing a sulfur atom, for example, 2-thienyl, 3-thienyl, etc.; unsaturated 5-
  • the term also embraces radicals where heterocyclic radicals are fused/condensed with aryl radicals: unsaturated condensed heterocyclic group containing 1 to 5 nitrogen atoms, for example, indolyl, isoindolyl, indolizinyl, benzimidazolyl, quinolyl, isoquinolyl, indazolyl, benzotriazolyl, tetrazolopyridazinyl [e.g., tetrazolo [1,5-b]pyridazinyl]; unsaturated condensed heterocyclic group containing 1 to 2 oxygen atoms and 1 to 3 nitrogen atoms [e.g.
  • heterocyclic radicals include five to ten membered fused or unfused radicals.
  • heteroaryl radicals include quinolyl, isoquinolyl, imidazolyl, pyridyl, thienyl, thiazolyl, oxazolyl, furyl, and pyrazinyl.
  • Other preferred heteroaryl radicals are 5- or 6-membered heteroaryl, containing one or two heteroatoms selected from sulfur, nitrogen and oxygen, selected from thienyl, furyl, pyrrolyl, indazolyl, pyrazolyl, oxazolyl, triazolyl, imidazolyl, pyrazolyl, isoxazolyl, isothiazolyl, pyridyl, piperidinyl and pyrazinyl.
  • non-nitrogen containing heteroaryl include pyranyl, 2-furyl, 3-furyl, 2-thienyl, 3-thienyl, benzofuryl, benzothienyl, and the like.
  • Particular examples of partially saturated and saturated heterocyclyl include pyrrolidinyl, imidazolidinyl, piperidinyl, pyrrolinyl, pyrazolidinyl, piperazinyl, morpholinyl, tetrahydropyranyl, thiazolidinyl, dihydrothienyl, 2,3-dihydro-benzo[1,4]dioxanyl, indolinyl, isoindolinyl, dihydrobenzothienyl, dihydrobenzofuryl, isochromanyl, chromanyl, 1,2-dihydroquinolyl, 1,2,3,4-tetrahydro-isoquinolyl, 1,2,3,4-tetrahydro-quinolyl, 2,3,4,4a,9,9a-hexahydro-1H-3-aza-fluorenyl, 5,6,7-trihydro-1,2,4-triazolo[3,4-a]isoquinolyl,
  • sulfonyl whether used alone or linked to other terms such as alkylsulfonyl, denotes respectively divalent radicals —SO 2 —.
  • sulfamyl denotes a sulfonyl radical substituted with an amine radical, forming a sulfonamide (—SO 2 NH 2 )
  • alkylaminosulfonyl includes “N-alkylaminosulfonyl” where sulfamyl radicals are independently substituted with one or two alkyl radical(s). More preferred alkylaminosulfonyl radicals are “lower alkylaminosulfonyl” radicals having one to six carbon atoms. Even more preferred are lower alkylaminosulfonyl radicals having one to three carbon atoms. Examples of such lower alkylaminosulfonyl radicals include N-methylaminosulfonyl, and N-ethylaminosulfonyl.
  • carbonyl whether used alone or with other terms, such as “aminocarbonyl”, denotes —(C ⁇ O)—.
  • aminocarbonyl denotes an amide group of the formula —C( ⁇ O)NH 2 .
  • N-alkylaminocarbonyl and “N,N-dialkylaminocarbonyl” denote aminocarbonyl radicals independently substituted with one or two alkyl radicals, respectively. More preferred are “lower alkylaminocarbonyl” having lower alkyl radicals as described above attached to an aminocarbonyl radical.
  • N-arylaminocarbonyl and “N-alkyl-N-arylaminocarbonyl” denote aminocarbonyl radicals substituted, respectively, with one aryl radical, or one alkyl and one aryl radical.
  • heterocyclylcarbonylalkyl denotes alkyl groups which have been substituted with a heterocyclylcarbonyl radical. More preferred are contain 4-6 membered heterocyclyl groups and C 1 -C 6 -alkyl radicals, such as 4-methylpiperazinylcarbonylethyl.
  • heterocyclylalkylcarbonyl denotes carbonyl groups which have been substituted with a heterocyclylalkyl radical. More preferred are contain 4-6 membered heterocyclyl groups and C 1 -C 6 -alkyl radicals, such as piperidinylmethylcarbonyl.
  • alkoxycarbonylaminoalkyl denotes an aminoalkyl group, which is substituted with an alkoxycarbonyl radical. More preferred are “lower alkoxycarbonylaminoalkyl” having C 1 -C 6 -alkyl radicals.
  • heterocyclylalkylenyl embraces heterocyclic-substituted alkyl radicals. More preferred heterocyclylalkylenyl radicals are “5- or 6-membered heteroarylalkylenyl” radicals having alkyl portions of one to six carbon atoms and a 5- or 6-membered heteroaryl radical. Even more preferred are lower heteroarylalkylenyl radicals having alkyl portions of one to four carbon atoms. Examples include such radicals as pyridylmethyl and thienylmethyl.
  • aralkyl embraces aryl-substituted alkyl radicals.
  • Preferable aralkyl radicals are “lower aralkyl” radicals having aryl radicals attached to alkyl radicals having one to six carbon atoms. Even more preferred are “phenylalkylenyl” attached to alkyl portions having one to three carbon atoms. Examples of such radicals include benzyl, diphenylmethyl and phenylethyl.
  • the aryl in said aralkyl may be additionally substituted with halo, alkyl, alkoxy, halkoalkyl and haloalkoxy.
  • alkylthio embraces radicals containing a linear or branched alkyl radical, of one to ten carbon atoms, attached to a divalent sulfur atom. Even more preferred are lower alkylthio radicals having one to three carbon atoms.
  • An example of “alkylthio” is methylthio, (CH 3 S—).
  • haloalkylthio embraces radicals containing a haloalkyl radical, of one to ten carbon atoms, attached to a divalent sulfur atom. Even more preferred are lower haloalkylthio radicals having one to three carbon atoms. An example of “haloalkylthio” is trifluoromethylthio.
  • alkylamino embraces “N-alkylamino” and “N,N-dialkylamino” where amino groups are independently substituted with one alkyl radical and with two alkyl radicals, respectively. More preferred alkylamino radicals are “lower alkylamino” radicals having one or two alkyl radicals of one to six carbon atoms, attached to a nitrogen atom. Even more preferred are lower alkylamino radicals having one to three carbon atoms. Suitable alkylamino radicals may be mono or dialkylamino such as N-methylamino, N-ethylamino, N,N-dimethylamino, N,N-diethylamino and the like.
  • arylamino denotes amino groups which have been substituted with one or two aryl radicals, such as N-phenylamino.
  • the arylamino radicals may be further substituted on the aryl ring portion of the radical.
  • heteroarylamino denotes amino groups which have been substituted with one or two heteroaryl radicals, such as N-thienylamino.
  • heteroarylamino radicals may be further substituted on the heteroaryl ring portion of the radical.
  • aralkylamino denotes amino groups which have been independently substituted with one or two aralkyl radicals. More preferred are phenyl-C 1 -C 3 -alkylamino radicals, such as N-benzylamino. The aralkylamino radicals may be further substituted on the aryl ring portion.
  • N-alkyl-N-arylamino and “N-aralkyl-N-alkylamino” denote amino groups which are independently substituted with one aralkyl and one alkyl radical, or one aryl and one alkyl radical, respectively, to an amino group.
  • aminoalkyl embraces linear or branched alkyl radicals having one to about ten carbon atoms any one of which may be substituted with one or more amino radicals. More preferred aminoalkyl radicals are “lower aminoalkyl” radicals having one to six carbon atoms and one or more amino radicals. Examples of such radicals include aminomethyl, aminoethyl, aminopropyl, aminobutyl and aminohexyl. Even more preferred are lower aminoalkyl radicals having one to three carbon atoms.
  • alkylaminoalkyl embraces alkyl radicals substituted with alkylamino radicals. More preferred alkylaminoalkyl radicals are “lower alkylaminoalkyl” radicals having alkyl radicals of one to six carbon atoms. Even more preferred are lower alkylaminoalkyl radicals having alkyl radicals of one to three carbon atoms. Suitable alkylaminoalkyl radicals may be mono or dialkyl substituted, such as N-methylaminomethyl, N,N-dimethyl-aminoethyl, N,N-diethylaminomethyl and the like.
  • alkylaminoalkoxy embraces alkoxy radicals substituted with alkylamino radicals. More preferred alkylaminoalkoxy radicals are “lower alkylaminoalkoxy” radicals having alkoxy radicals of one to six carbon atoms. Even more preferred are lower alkylaminoalkoxy radicals having alkyl radicals of one to three carbon atoms. Suitable alkylaminoalkoxy radicals may be mono or dialkyl substituted, such as N-methylaminoethoxy, N,N-dimethylaminoethoxy, N,N-diethylaminoethoxy and the like.
  • alkylaminoalkoxyalkoxy embraces alkoxy radicals substituted with alkylaminoalkoxy radicals. More preferred alkylaminoalkoxyalkoxy radicals are “lower alkylaminoalkoxyalkoxy” radicals having alkoxy radicals of one to six carbon atoms. Even more preferred are lower alkylaminoalkoxyalkoxy radicals having alkyl radicals of one to three carbon atoms.
  • Suitable alkylaminoalkoxyalkoxy radicals may be mono or dialkyl substituted, such as N-methylaminoethoxyethoxy, N-methylaminomethoxyethoxy, N,N-dimethylaminoethoxyethoxy, N,N-diethylaminomethoxymethoxy and the like.
  • carboxyalkyl embraces linear or branched alkyl radicals having one to about ten carbon atoms any one of which may be substituted with one or more carboxy radicals. More preferred carboxyalkyl radicals are “lower carboxyalkyl” radicals having one to six carbon atoms and one carboxy radical. Examples of such radicals include carboxymethyl, carboxypropyl, and the like. Even more preferred are lower carboxyalkyl radicals having one to three CH 2 groups.
  • halosulfonyl embraces sulfonyl radicals substituted with a halogen radical. Examples of such halosulfonyl radicals include chlorosulfonyl and fluorosulfonyl.
  • arylthio embraces aryl radicals of six to ten carbon atoms, attached to a divalent sulfur atom.
  • An example of “arylthio” is phenylthio.
  • aralkylthio embraces aralkyl radicals as described above, attached to a divalent sulfur atom. More preferred are phenyl-C 1 -C 3 -alkylthio radicals. An example of “aralkylthio” is benzylthio.
  • aryloxy embraces optionally substituted aryl radicals, as defined above, attached to an oxygen atom. Examples of such radicals include phenoxy.
  • aralkoxy embraces oxy-containing aralkyl radicals attached through an oxygen atom to other radicals. More preferred aralkoxy radicals are “lower aralkoxy” radicals having optionally substituted phenyl radicals attached to lower alkoxy radical as described above.
  • heteroaryloxy embraces optionally substituted heteroaryl radicals, as defined above, attached to an oxygen atom.
  • heteroarylalkoxy embraces oxy-containing heteroarylalkyl radicals attached through an oxygen atom to other radicals. More preferred heteroarylalkoxy radicals are “lower heteroarylalkoxy” radicals having optionally substituted heteroaryl radicals attached to lower alkoxy radical as described above.
  • cycloalkyl includes saturated carbocyclic groups.
  • Preferred cycloalkyl groups include C 3 -C 6 rings. More preferred compounds include, cyclopentyl, cyclopropyl, and cyclohexyl.
  • cycloalkylalkyl embraces cycloalkyl-substituted alkyl radicals.
  • Preferable cycloalkylalkyl radicals are “lower cycloalkylalkyl” radicals having C 3-6 cycloalkyl radicals attached to alkyl radicals having one to six carbon atoms.
  • cycloalkenyl includes carbocyclic groups having one or more carbon-carbon double bonds including “cycloalkyldienyl” compounds.
  • Preferred cycloalkenyl groups include C 3 -C 6 rings. More preferred compounds include, for example, cyclopentenyl, cyclopentadienyl, cyclohexenyl and cycloheptadienyl.
  • the compounds of the invention are endowed with kinase inhibitory activity, such as KDR inhibitory activity.
  • the present invention also comprises the use of a compound of the invention, or pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment either acutely or chronically of an angiogenesis mediated disease state, including those described previously.
  • the compounds of the present invention are useful in the manufacture of an anti-cancer medicament.
  • the compounds of the present invention are also useful in the manufacture of a medicament to attenuate or prevent disorders through inhibition of KDR.
  • the present invention comprises a pharmaceutical composition
  • a pharmaceutical composition comprising a therapeutically-effective amount of a compound of Formulas I-IV in association with a least one pharmaceutically-acceptable carrier, adjuvant or diluent.
  • the present invention also comprises a method of treating angiogenesis related disorders in a subject having or susceptible to such disorder, the method comprising treating the subject with a therapeutically-effective amount of a compound of Formula I
  • each of A 1 and A 2 is independently C or N;
  • a 1 -A 2 form part of a ring A selected from 5- or 6-membered heteroaryl
  • Z is oxygen or sulfur
  • Y is selected from
  • R a and R b are independently selected from H, halo, cyano, —NHR 6 and C 1-4 -alkyl substituted with R 1 , or wherein R a and R b together form C 3 -C 6 cycloalkyl;
  • R z is selected from C 2 -C 6 -alkylenyl, where one of the CH 2 groups may be replaced with an oxygen atom or an —NH—; wherein one of the CH 2 groups may be substituted with one or two radicals selected from halo, cyano, —NHR 6 and C 1-4 -alkyl substituted with R 1 ;
  • R d is cycloalkyl
  • R 1 is one or more substituents independently selected from H, halo, —OR 7 , oxo, —SR 7 , —CO 2 R 7 , —COR 7 , —CONR 7 R 7 , —NR 7 R 7 , —SO 2 NR 7 R 7 , —NR 7 C(O)OR 7 , —NR 7 C(O)R 7 , —NR 7 C(O)NR 7 R 7 , cycloalkyl, optionally substituted phenylalkylenyl, optionally substituted 5-6 membered heterocyclyl, optionally substituted heteroarylalkylenyl, optionally substituted phenyl, lower alkyl, cyano, lower hydroxyalkyl, lower carboxyalkyl, nitro, lower alkenyl, lower alkynyl, lower aminoalkyl, lower alkylaminoalkyl and lower haloalkyl;
  • R 2 is selected from
  • substituted R 2 is substituted with one or more substituents independently selected from halo, —OR 7 , —SR 7 , —CO 2 R 7 —CONR 7 R 7 , —COR 7 , —NR 7 R 7 , —NH(C 1 —C 4 alkylenylR 9 ), —SO 2 R 7 , —SO 2 NR 7 R 7 , —NR 7 C(O)OR 7 , —NR 7 C(O)R 7 , optionally substituted cycloalkyl, optionally substituted 5-6 membered heterocyclyl, optionally substituted phenyl, halosulfonyl, cyano, alkylaminoalkoxy, alkylaminoalkoxyalkoxy, nitro, lower alkyl substituted with R 1 , lower alkenyl substituted with R 1 , and lower alkynyl substituted with Rl;
  • R 3 is selected from aryl substituted with one or more substituents independently selected from halo, —OR 7 , —SR 7 , —SO 2 R 7 , —CO 2 R 7 , —CONR 7 R 7 , —COR 7 , —NR 7 R 7 , —SO 2 NR 7 R 7 , —NR 7 C(O)OR 7 , —NR 7 C(O)R 7 , cycloalkyl, optionally substituted 5-6 membered heterocyclyl, optionally substituted phenyl, nitro, alkylaminoalkoxyalkoxy, cyano, alkylaminoalkoxy, lower alkyl substituted with R 1 , lower alkenyl substituted with R 1 , and lower alkynyl substituted with R 1 ;
  • R 4 is selected from a direct bond, C 2-4 -alkylenyl, C 2-4 -alkenylenyl and C 2-4 -alkynylenyl, where one of the CH 2 groups may be substituted with an oxygen atom or an —NH—,
  • R 4 is optionally substituted with hydroxy
  • R 5 is selected from H, lower alkyl, phenyl and lower aralkyl
  • R 5a is selected from H, lower alkyl, phenyl and lower aralkyl
  • R 6 is selected from H or C 1-6 -alkyl
  • R 7 is selected from H, lower alkyl, phenyl, 5-6 membered heterocyclyl, C 3 -C 6 -cycloalkyl, phenylalkyl, 5-6 membered heterocyclylalkyl, C 3 -C 6 cycloalkylalkyl, and lower haloalkyl;
  • R 9 is selected from H, phenyl, 5-6 membered heterocyclyl and C 3 -C 6 cycloalkyl.
  • the compounds of the invention can be administered as the sole active pharmaceutical agent, they can also be used in combination with one or more compounds of the invention or other agents.
  • the therapeutic agents can be formulated as separate compositions that are administered at the same time or sequentially at different times, or the therapeutic agents can be given as a single composition.
  • co-therapy in defining use of a compound of the present invention and another pharmaceutical agent, is intended to embrace administration of each agent in a sequential manner in a regimen that will provide beneficial effects of the drug combination, and is intended as well to embrace co-administration of these agents in a substantially simultaneous manner, such as in a single capsule having a fixed ratio of these active agents or in multiple, separate capsules for each agent.
  • the administration of compounds of the present invention may be in conjunction with additional therapies known to those skilled in the art in the prevention or treatment of neoplasia, such as with radiation therapy or with cytostatic or cytotoxic agents.
  • antineoplastic agents available in commercial use, in clinical evaluation and in pre-clinical development, which would be selected for treatment of neoplasia by combination drug chemotherapy.
  • Such antineoplastic agents fall into several major categories, namely, antibiotic-type agents, alkylating agents, antimetabolite agents, hormonal agents, immunological agents, interferon-type agents and a category of miscellaneous agents.
  • a first family of antineoplastic agents which may be used in combination with compounds of the present invention consists of antimetabolite-type/thymidilate synthase inhibitor antineoplastic agents.
  • Suitable antimetabolite antineoplastic agents may be selected from but not limited to the group consisting of 5-FU-fibrinogen, acanthifolic acid, aminothiadiazole, brequinar sodium, carmofur, Ciba-Geigy CGP-30694, cyclopentyl cytosine, cytarabine phosphate stearate, cytarabine conjugates, Lilly DATHF, Merrel Dow DDFC, dezaguanine, dideoxycytidine, dideoxyguanosine, didox, Yoshitomi DMDC, doxifluridine, Wellcome EHNA, Merck & Co.
  • EX-015 benzrabine, floxuridine, fludarabine phosphate, 5-fluorouracil, N-(2′-furanidyl)-5-fluorouracil, Daiichi Seiyaku FO-152, isopropyl pyrrolizine, Lilly LY-188011, Lilly LY-264618, methobenzaprim, methotrexate, Wellcome MZPES, norspermidine, NCI NSC-127716, NCI NSC-264880, NCI NSC-39661, NCI NSC-612567, Warner-Lambert PALA, pentostatin, piritrexim, plicamycin, Asahi Chemical PL-AC, Takeda TAC-788, thioguanine, tiazofurin, Erbamont TIF, trimetrexate, tyrosine kinase inhibitors, Taiho UFT and uricytin.
  • a second family of antineoplastic agents which may be used in combination with compounds of the present invention consists of alkylating-type antineoplastic agents.
  • Suitable alkylating-type antineoplastic agents may be selected from but not limited to the group consisting of Shionogi 254-S, aldo-phosphamide analogues, altretamine, anaxirone, Boehringer Mannheim BBR-2207, bestrabucil, budotitane, Wakunaga CA-102, carboplatin, carmustine, Chinoin-139, Chinoin-153, chlorambucil, cisplatin, cyclophosphamide, American Cyanamid CL-286558, Sanofi CY-233, cyplatate, Degussa D-19-384, Sumimoto DACHP(Myr)2, diphenylspiromustine, diplatinum cytostatic, Erba distamycin derivatives, Chugai DWA-2114R, ITI
  • a third family of antineoplastic agents which may be used in combination with compounds of the present invention consists of antibiotic-type antineoplastic agents.
  • Suitable antibiotic-type antineoplastic agents may be selected from but not limited to the group consisting of Taiho 4181-A, aclarubicin, actinomycin D, actinoplanone, Erbamont ADR-456, aeroplysinin derivative, Ajinomoto AN-201-II, Ajinomoto AN-3, Nippon Soda anisomycins, anthracycline, azino-mycin-A, bisucaberin, Bristol-Myers BL-6859, Bristol-Myers BMY-25067, Bristol-Myers BMY-25551, Bristol-Myers BMY-26605, Bristol-Myers BMY-27557, Bristol-Myers BMY-28438, bleomycin sulfate, bryostatin-1, Taiho C-1027, calichemycin, chrom
  • a fourth family of antineoplastic agents which may be used in combination with compounds of the present invention consists of a miscellaneous family of antineoplastic agents, including tubulin interacting agents, topoisomerase II inhibitors, topoisomerase I inhibitors and hormonal agents, selected from but not limited to the group consisting of ⁇ -carotene, ⁇ -difluoromethyl-arginine, acitretin, Biotec AD-5, Kyorin AHC-52, alstonine, amonafide, amphethinile, amsacrine, Angiostat, ankinomycin, anti-neoplaston A10, antineoplaston A2, antineoplaston A3, antineoplaston A5, antineoplaston AS2-1, Henkel APD, aphidicolin glycinate, asparaginase, Avarol, baccharin, batracylin, benfluron, benzotript, Ipsen-Beaufour BIM-23015
  • the present compounds may also be used in co-therapies with other anti-neoplastic agents, such as acemannan, aclarubicin, aldesleukin, alemtuzumab, alitretinoin, altretamine, amifostine, aminolevulinic acid, amrubicin, amsacrine, anagrelide, anastrozole, ANCER, ancestim, ARGLABIN, arsenic trioxide, BAM 002 (Novelos), bexarotene, bicalutamide, broxuridine, capecitabine, celmoleukin, cetrorelix, cladribine, clotrimazole, cytarabine ocfosfate, DA 3030 (Dong-A), daclizumab, denileukin diftitox, deslorelin, dexrazoxane, dilazep, docetaxel, docos
  • the present compounds may also be used in co-therapies with other anti-neoplastic agents, such as other kinase inhibitors including p38 inhibitors and CDK inhibitors, TNF inhibitors, metallomatrix proteases inhibitors (MMP), COX-2 inhibitors including celecoxib, rofecoxib, parecoxib, valdecoxib, and etoricoxib, NSAID's, SOD mimics or ⁇ v ⁇ 3 inhibitors.
  • other anti-neoplastic agents such as other kinase inhibitors including p38 inhibitors and CDK inhibitors, TNF inhibitors, metallomatrix proteases inhibitors (MMP), COX-2 inhibitors including celecoxib, rofecoxib, parecoxib, valdecoxib, and etoricoxib, NSAID's, SOD mimics or ⁇ v ⁇ 3 inhibitors.
  • the present invention comprises processes for the preparation of a compound of Formulas I-IV.
  • compositions of Formulas I-IV are also included in the family of compounds of Formulas I-IV.
  • pharmaceutically-acceptable salts embraces salts commonly used to form alkali metal salts and to form addition salts of free acids or free bases. The nature of the salt is not critical, provided that it is pharmaceutically-acceptable.
  • Suitable pharmaceutically-acceptable acid addition salts of compounds of Formulas I-IV may be prepared from an inorganic acid or from an organic acid. Examples of such inorganic acids are hydrochloric, hydrobromic, hydroiodic, nitric, carbonic, sulfuric and phosphoric acid.
  • organic acids may be selected from aliphatic, cycloaliphatic, aromatic, arylaliphatic, heterocyclic, carboxylic and sulfonic classes of organic acids, example of which are formic, acetic, adipic, butyric, propionic, succinic, glycolic, gluconic, lactic, malic, tartaric, citric, ascorbic, glucuronic, maleic, fumaric, pyruvic, aspartic, glutamic, benzoic, anthranilic, mesylic, 4-hydroxybenzoic, phenylacetic, mandelic, embonic (pamoic), methanesulfonic, ethanesulfonic, ethanedisulfonic, benzenesulfonic, pantothenic, 2-hydroxyethanesulfonic, toluenesulfonic, sulfanilic, cyclohexylaminosulfonic, camphoric, camphorsulfonic,
  • Suitable pharmaceutically-acceptable base addition salts of compounds of Formulas I-IV include metallic salts, such as salts made from aluminum, calcium, lithium, magnesium, potassium, sodium and zinc, or salts made from organic bases including primary, secondary and tertiary amines, substituted amines including cyclic amines, such as caffeine, arginine, diethylamine, N-ethyl piperidine, aistidine, glucamine, isopropylamine, lysine, morpholine, N-ethyl morpholine, piperazine, piperidine, triethylamine, trimethylamine. All of these salts may be prepared by conventional means from the corresponding compound of the invention by reacting, for example, the appropriate acid or base with the compound of Formulas I-IV
  • the basic nitrogen-containing groups can be quaternized with such agents as lower alkyl halides, such as methyl, ethyl, propyl, and butyl chloride, bromides and iodides; dialkyl sulfates like dimethyl, diethyl, dibutyl, and diamyl sulfates, long chain halides such as decyl, lauryl, myristyl and stearyl chlorides, bromides and iodides, aralkyl halides like benzyl and phenethyl bromides, and others. Water or oil-soluble or dispersible products are thereby obtained.
  • lower alkyl halides such as methyl, ethyl, propyl, and butyl chloride, bromides and iodides
  • dialkyl sulfates like dimethyl, diethyl, dibutyl, and diamyl sulfates
  • long chain halides such as de
  • Other examples include salts with alkali metals or alkaline earth metals, such as sodium, potassium, calcium or magnesium or with organic bases.
  • Preferred salts include hydrochloride, phosphate and edisylate.
  • Cyclic amides can be prepared according to the method set out in Scheme 1.
  • the amino group of compound 1 (where R o is alkyl, aryl, and the like) is protected, such as with Boc anhydride, followed by treatment, to remove the ester, such as with base, forming the protected amine/free acid 2.
  • R o is alkyl, aryl, and the like
  • Substitued amines are coupled with the free acid, such as with EDC, to form the protected amine/amide 3.
  • the protected amine moiety is removed, such as with acid, and reacted via one step reductive alkylation with carbonyl-containing compounds to form the 1-amido-2-substituted amino-compounds 4.
  • the amination is in an alcohol, such as MeOH, EtOH or propanol, and at a temperature between about 0-50° C., such as RT.
  • Aldehydes or ketones are preferred carbonyl-containing compounds.
  • Alternative carbonyl-containing compounds are, for example, bisulfite adducts or semiacetals, acetals, semiketals or ketals of compounds with alcohols, for example lower hydroxyalkyl compounds; or thioacetals or thioketals of compounds with mercaptans, for example lower alkylthio compounds.
  • the reductive alkylation is preferably carried out with hydrogenation in the presence of a catalyst, such as platinum or especially palladium, which is preferably bonded to a carrier material, such as carbon, or a heavy metal catalyst, such as Raney nickel, at normal pressure or at pressures of from 0.1 to 10 MegaPascal (MPa), or with reduction by means of complex hydrides, such as borohydrides, especially alkali metal cyanoborohydrides, for example sodium cyanoborohydride, in the presence of a suitable acid, preferably relatively weak acids, such as lower alkylcarboxylic acids, especially acetic acid, or a sulfonic acid, such as p-toluenesulfonic acid; in customary solvents, for example alcohols, such as methanol or ethanol, or ethers, for example cyclic ethers, such as tetrahydrofuran, in the presence or absence of water.
  • a catalyst such as platinum or especially palladium
  • a carrier material such
  • compounds 4 can be prepared from mixed acid/amines 5 as shown in Scheme 2.
  • Substituted amines are coupled with the mixed acid/amines 5 such as with a coupling reagent, for example EDC, to form the mixed amine/amide 6.
  • a coupling reagent for example EDC
  • Substituted carbonyl compounds such as acid halides, anhydrides, carboxylic acids, esters, ketones, aldehydes and the like, are added to the mixed amine/amide 6 followed with reduction to give the substituted amide/substituted amine compounds 4.
  • Imino compounds 7 can be formed from the mixed amine/amides 6, such as by reacting with a substituted carbonyl compound.
  • Substituted cyclic carboxamides can be prepared from the corresponding imino analogs by the process outlined in Scheme 4. Treatment of the imino compound 7 with a reducing agent yields compound 4. Reagents which can be used to add hydrogen to an imine double bond include borane in THF, LiAlH 4 , NaBH 4 , sodium in EtOH and hydrogen in the presence of a catalyst, among others.
  • Substituted carboxamides 4 can be prepared from the corresponding halo analogs 8 by the process outlined in Schemne 5.
  • Substituted amino acids 9 are prepared from the corresponding chioro compounds 8 such as by reacting with an amine at a suitable temperature, such as about 80° C. The acid 9 is coupled with an amine, preferably in the presence of a coupling agent such as EDC, to form the corresponding amide 4.
  • the amination process can be carried out as an Ullmann type reaction using a copper catalyst, such as copper[0] or a copper[I] compound such as copper[I]oxide, copper[I]bromide or copper[I]iodide in the presence of a suitable base (such as a metal carbonate, for example K 2 CO 3 ) to neutralize the acid generated in the reaction.
  • a suitable base such as a metal carbonate, for example K 2 CO 3
  • the amount of catalyst is typically in the range of 1 to 20 mole percent.
  • reaction is carried out in an inert, aprotic solvent such as an ether (for example dimethoxyethane or dioxane) or an amide (for example dimethylformamide or N-methylpyrrolidone), under an inert atmosphere in the temperature range of 60-180° C.
  • aprotic solvent such as an ether (for example dimethoxyethane or dioxane) or an amide (for example dimethylformamide or N-methylpyrrolidone
  • An alternative amination process involves using a Group VIII element, where the metal core of the catalyst should be a zero-valent transition metal, such as palladium or nickel, which has the ability to undergo oxidative addition to the aryl-halogen bond.
  • the zero valent state of the metal may be generated in situ from the M[II] state.
  • the catalyst complexes may include chelating ligands, such as alkyl, aryl or heteroaryl derivatives of phosphines or biphosphines, imines or arsines.
  • Preferred catalysts contain palladium or nickel.
  • Such catalysts include palladium[II]chloride, palladium[II]acetate, tetrakis(triphenyl-phosphine)palladium[0] and nickel[II]acetylacetonate.
  • the metal catalyst is typically in the range of 0.1 to 10 mole percent.
  • the chelating ligands may be either monodentate, as in the case for example of trialkyphosphines, such as tributylphosphine, triarylphosphines, such as tri-(ortho-tolyl)phosphine, and triheteroaryl phosphines, such as tri-2-furylphosphine; or they may be bidentate such as in the case of 2,2′-bis(diphenylphosphino)-1,1′binaphthyl, 1,2-bis(diphenylphosphino)ethane, 1,1′-bis(diphenylphosphino)ferrocene and 1-(N,N-dimethyl-amino)-1′-(dicyclohexylphosphino)biphenyl.
  • trialkyphosphines such as tributylphosphine
  • triarylphosphines such as tri-(ortho-tolyl)phos
  • the supporting ligand may be complexed to the metal center in the form of a metal complex prior to being added to the reaction mixture or may be added to the reaction mixture as a separate compound.
  • the supporting ligand is typically present in the range 0.01 to 20 mole percent. It is often necessary to add a suitable base to the reaction mixture, such as a trialkylamine (for example DIEA or 1,5-diazabicyclo[5,4,O]undec-5-ene), a Group I alkali metal alkoxide (for example potassium tert-butoxide) or carbonate (for example cesium carbonate) or potassium phosphate.
  • a suitable base such as a trialkylamine (for example DIEA or 1,5-diazabicyclo[5,4,O]undec-5-ene), a Group I alkali metal alkoxide (for example potassium tert-butoxide) or carbonate (for example cesium carbonate) or potassium phosphate.
  • the reaction is typically carried out in an inert aprotic solvent such as an ether (for example dimethoxyethane or dioxane) or an amide (for example DMF or N-methylpyrrolidone), under an inert atmosphere in the temperature range of 60-180° C.
  • aprotic solvent such as an ether (for example dimethoxyethane or dioxane) or an amide (for example DMF or N-methylpyrrolidone)
  • the amination is preferably carried out in an inert, aprotic, preferably anhydrous, solvent or solvent mixture, for example in a carboxylic acid amide, for example dimethylformamide or dimethylacetamide, a cyclic ether, for example THF or dioxane, or a nitrile, for example CH 3 CN, or in a mixture thereof, at an appropriate temperature, for example in a temperature range of from about 40° C. to about 180° C., and if necessary under an inert gas atmosphere, for example a nitrogen or argon atmosphere.
  • an inert, aprotic preferably anhydrous, solvent or solvent mixture
  • a carboxylic acid amide for example dimethylformamide or dimethylacetamide
  • a cyclic ether for example THF or dioxane
  • a nitrile for example CH 3 CN
  • Substituted carboxamides 4 can be prepared from the corresponding halo analogs 8 by the process outlined in Scheme 6.
  • the chloro acid 8 is coupled with an amine, preferably in the presence of a coupling agent such as EDC, to form the corresponding chloro amide 10.
  • Substituted amino-amides 4 are prepared from the corresponding chloro compounds 10 such as by reacting with an amine at a suitable temperature, such as about 80° C.
  • the amination reaction can be run in the presence of an appropriate catalyst such as a palladium catalyst, in the presence of an aprotic base such as sodium t-butoxide or cesium carbonate, or a nickel catalyst, or a copper catalyst.
  • Substituted carboxamides 4 can be prepared from the corresponding bromo/chloro analogs 11 by the process outlined in Scheme 7.
  • the bromo/chloro acid 11 is coupled with an amine, preferably in the presence of a coupling agent such as EDC, to form the corresponding bromo substituted amide 12.
  • Suzuki coupling with the bromo amide 12 and suitable boronic acids provides the substituted amide 10.
  • Substituted amino-amides 4 are prepared from the corresponding chloro compounds 10 as described in Scheme 6.
  • Substituted pyridines can be prepared such as by the method found in Scheme 8.
  • 2-Aminonicotinic acid 13 is coupled with a substituted amine at a suitable temperature, nonprotic solvent such as CH 2 Cl 2 , such as with EDC and HOBt, to form the nicotinamide 14.
  • the nicotinamide 14 is reductively alkylated such as with substituted 4-benzaldehydes and NaBH(OAc) 3 , to yield the 2-substituted amino-pyridyl carboxamides 15.
  • Substituted pyridines may be prepared by the method found in Scheme 9.
  • 2-Chloro-nicotinic acid 16 (where LG is OH) is coupled with an amine at a suitable temperature, such as a temperature over about 100° C. to give the 2-substituted amino-nicotinic acid 17.
  • the 2-substituted amino-nicotinic acid 17 is reacted with a substituted amine in the presence of a coupling reagent, such as BOP-Cl and base, such as TEA to form the 2-substituted amino-nicotinamide 15.
  • a coupling reagent such as BOP-Cl and base, such as TEA
  • 2-chloro-nicotinoyl chloride (LG is Cl) is coupled first with R 2 -NH 2 , such as in the presence of base, e.g., NaHCO 3 , in a suitable solvent, such as CH 2 Cl 2 , to form the amide 16A, then coupled with a benzylamine to yield the 2-substituted amino-nicotinamide 15.
  • Imino-substituted pyridines may be prepared by the method found in Scheme 10. (2-Amino-(4-pyridyl))-carboxamide 18 is reacted with substituted 4-benzaldehydes, such as in the presence of p-toluenesulfonic acid monohydrate to yield the imino compound 19.
  • Substituted pyridines alternatively may be prepared by the method found in Scheme 11.
  • the imino compound 19 is reduced, such as with NaBH 4 , to form the substituted amine 20.
  • Substituted pyridines can be prepared by the process outlined in Scheme 12.
  • a solution of sodium hypobromite is freshly prepared and added to 2-hydroxynicotinic acid 21 and heated, preferably at a temperature at about 50° C. Additional hypobromite solution may be needed to form the bromo compound 22.
  • the 5-bromo-2-hydroxynicotinic acid 22 is reacted with thionyl chloride, preferably at a temperature >RT, more preferably at about 80° C. to form the 2-chloro-nicotinic acid analog 23.
  • the acid is coupled with an amine, preferably in the presence of EDC, HOBT, and DIEA to form the corresponding substituted amide 24.
  • 2-Amino-nicotinamides 26 are prepared from the corresponding chloro compounds 25 such as by reacting with substituted amines at a suitable temperature, such as about 80° C.
  • sulfonamides 27 can be prepared from amines 6 as shown in Scheme 13. Substituted sulfonyl compounds, such as sulfonyl halides, preferably chloro or bromo, sulfonic acids, an activated ester or reactive anhydride, or in the form of a cyclic amide, and the like, are added to the amine 6 to give the sulfonamide compounds 27.
  • sulfonyl compounds such as sulfonyl halides, preferably chloro or bromo, sulfonic acids, an activated ester or reactive anhydride, or in the form of a cyclic amide, and the like.
  • reaction is carried out in a suitable solvent, such as CH 2 Cl 2 , at a temperature between about RT to about the reflux temperature of the solvent, in the presence of a suitable base, such as DIEA or DMAP.
  • a suitable solvent such as CH 2 Cl 2
  • a suitable base such as DIEA or DMAP.
  • the amino group of compounds 6 is preferably in free form, especially when the sulfonyl group reacting therewith is present in reactive form.
  • the amino group may, however, itself be a derivative, for example by reaction with a phosphite, such as diethylchlorophosphite, 1,2-phenylene chlorophosphite, ethyldichlorophosphite, ethylene chlorophosphite or tetraethylpyrophosphite.
  • a derivative of such a compound having an amino group also can be a carbamic acid halide or an isocyanate.
  • the condensation of activated sulfonic esters, reactive anhydrides or reactive cyclic amides with the corresponding amines is customarily carried out in the presence of an inorganic base, such as an alkaline metal hydrogen carbonate of carbonate, or especially an organic base, for example simple lower (alkyl) 3 -amines, for example TEA or tributylamine, or one of the above-mentioned organic bases.
  • an inorganic base such as an alkaline metal hydrogen carbonate of carbonate, or especially an organic base, for example simple lower (alkyl) 3 -amines, for example TEA or tributylamine, or one of the above-mentioned organic bases.
  • a condensation agent is additionally used, for example as described for free carboxylic acids.
  • the condensation is preferably carried out in an inert, aprotic, preferably anhydrous, solvent or solvent mixture, for example in a carboxylic acid amide, for example formamide or DMF, a halogenated hydrocarbon, for example CH 2 Cl 2 , CCl 4 or chlorobenzene, a ketone, for example acetone, a cyclic ether, for example THF or dioxane, an ester, for example EtOAc, or a nitrile, for example CH 3 CN, or in a mixture thereof, as appropriate at reduced or elevated temperature, for example in a temperature range of from about ⁇ 40° C. to about 100° C., preferably from about ⁇ 10° C. to about 70° C., and when arylsulfonyl esters are used also at temperatures of from about 10-30° C., and if necessary under an inert gas atmosphere, for example a nitrogen or argon atmosphere.
  • an inert gas atmosphere for example a nitrogen or arg
  • Alcoholic solvents for example EtOH, or aromatic solvents, for example benzene or toluene, may also be used.
  • alkali metal hydroxides are present as bases, acetone may also be added where appropriate.
  • Substituted pyridines can be prepared by the process outlined in Scheme 14. 2-Chloronicotinic acid 28 and substituted amine are coupled under conditions similar to that described in the previous schemes to give the amide 29. 6-Chloro-2-aminopyridines 30 are prepared from the amide 29, such as by reacting with substituted amines at a suitable temperature, such as above about 80° C., preferably above about 100° C., more preferably at about 130° C., neat. 6-Chloro-2-aminopyridines 30 are de-chlorinated such as by hydrogenation, for example by treatment with H 2 in the presence of Pd/C, to yield other compounds of the present invention 31.
  • 1,2,3,6-Tetrahydro-pyridyl substituted anilines (where R x is a substituent selected from those available for substituted R 2 ) are prepared such as by the procedure described in Scheme 15.
  • Nitrobenzenes 32 are brominated, such as with bromine in the presence of acid, H 2 SO 4 for example, or with NBS to yield the 3-bromo derivative 33.
  • Alkylation of the nitrophenyl-pyridine 34 such as by treatment with iodomethane, preferably above about 50° C., and more preferably at about 80° C., yields the pyridinium compound 35, which upon reduction, such as by NaBH 4 , yields the tetrahydyropyridine 36.
  • 6-Amino substituted pyridines are prepared such as by the procedure described in Scheme 16. Similar to the method of Scheme 13, chloropyridine 37 and is reacted with an amine, preferably above about 50° C., and more preferably at about 80° C., to yield the 6-aminopyridines 38.
  • a series of substituted anilines are prepared such as by the procedure described in Scheme 17.
  • a nitrobenzyl bromide 39 is coupled with morpholine, such as at a temperature at about RT, to yield the heterocyclylmethyl nitrobenzene derivative.
  • Protected alkylamine substituted anilines can be prepared from the nitro free amines 41, such as with standard protecting agents and chemistry known in the art, such as BOC chemistry. Reduction of the protected nitro compound, such as with iron powder, preferably above about 50° C., and more preferably at about 80° C., yields the aniline 42.
  • Sulfonamide substituted anilines can be prepared from nitrobenzenesulfonyl chlorides 43. Coupling of nitrobenzenesulfonyl chlorides 43 with reactive heterocyclic compounds, such as substituted piperazines, piperidines, and the like, in a protic solvent such as EtOH, such as at a temperature about RT, yields the nitrobenzenesulfonamides 43. Reduction of the nitro benzenesulfonamide, such as with iron powder, preferably above about 50° C., and more preferably at about 80° C., yields the aniline 44.
  • reactive heterocyclic compounds such as substituted piperazines, piperidines, and the like
  • EtOH such as at a temperature about RT
  • a series of perhaloalkyl-substituted anilines 47, where R y represents perhaloalkyl radicals, are prepared such as by the procedure described in Scheme 18.
  • 1-Nitro-4-(perfluoroethyl)benzene can be synthesized by the method described in the reference [John N. Freskos, Synthetic Communications, 18(9), 965-972 (1988)].
  • 1-Nitro-4-(perfluoroalkyl)benzene can be synthesized from the nitro compound, where LG is a leaving group, such as iodo, by the method described by W. A. Gregory, et al. [J. Med. Chem., 1990, 33, 2569-2578].
  • piperazinyl substituted anilines 53 can be prepared by the treatment of an aniline 51 with an N-substituted-bis(2-chloroethyl)amine, base, such as K 2 CO 3 and NaI, at a temperature above about 50° C., preferably above about 100° C., and more preferably at about 170° C., to give the piperazinylbenzene compound 52. Nitration, such as with H 2 SO 4 and KNO 3 , at a temperature above 0° C., and preferably at about RT, followed by hydrogenation, such as with H 2 atmosphere gives the substituted aniline 53.
  • an aniline 51 with an N-substituted-bis(2-chloroethyl)amine, base, such as K 2 CO 3 and NaI
  • piperazinyl substituted anilines 56 can be prepared by the treatment of a fluoro-nitro-substituted aryl compounds 54.
  • the fluoro-nitro-substituted aryl compounds 54 and 1-substituted piperazines are heated, preferably neat, at a temperature above about 50° C., and preferably at about 90° C., to yield the piperazinyl-nitroaryl compounds 55.
  • Hydrogenation such as with H 2 atmosphere in the presence of a catalyst, such as 10% Pd/C, gives the substituted aniline 56.
  • Substituted indolines are prepared such as by the procedures described in Scheme 20.
  • Substituted amino-indolines 59 are prepared from the nitroindoline 57 and a ketone in the presence of NaHB(OAc) 3 to form the 1-substituted indoline 58.
  • the nitroindoline 58 is hydrogenated, such as with H 2 in the presence of a catalyst, such as Pd/C, to yield the amino-indoline 59.
  • substituted amino-indolines 62 are prepared from the nitroindoline 57.
  • Nitroindoline 57 is reacted with an acid chloride to form an amide.
  • Further treatment with a primary or secondary amine, preferably a secondary amine, such as in the presence of NaI, at a temperature above about 50° C., and preferably at about 70° C. yields the nitroindoline 60.
  • the nitro compound 60 is hydrogenated, such as with H 2 in the presence of a catalyst, such as Pd/C, to yield the amino-indoline 61.
  • the carbonyl is reduced, such as with BH 3 -THF, to yield 1-aminoalkyl-indolines 62.
  • Substituted indolines are prepared such as by the procedures described in Scheme 21.
  • Substituted acetamides 64 are prepared from the coupling of halo-5-nitroanilines 63 (where LG is bromo or chloro, preferably chloro) and an acylating agent, such as acetyl chloride or acetic anhydride, under standard coupling chemistry, such as with DIEA, and DMAP, at a temperature of about RT, in a suitable solvent, such as CH 2 Cl 2 , DMF and/or DMAC.
  • the N-(2-methylprop-2-enyl)acetamide 65 is prepared from the acetamide 64, such as by the treatment of base, such as NaH in a suitable solvent such as NMP or anhydrous DMF and a 3-halo-2-methylpropene such as 3-bromo-2-methylpropene or 3-chloro-2-methylpropene, at a temperature between about 0° C. and RT, and preferably at about RT; or with CsCO 3 at a temperature above RT, preferably above about 50° C. and more preferably above about 60° C.
  • base such as NaH
  • a suitable solvent such as NMP or anhydrous DMF
  • a 3-halo-2-methylpropene such as 3-bromo-2-methylpropene or 3-chloro-2-methylpropene
  • Deprotection such as with strong acid such as AcOH, or HCl at a temperature above about 50° C., and preferably at about 70-80° C., yields the 3,3-dimethyl-6-nitro-2,3-dihydro-indol-1-yl 67.
  • the protected dihydro-6-nitro indoline 66 can be reduced, such as with Fe, or with 10% Pd/C in the presence of an excess of NH 4 CO 2 H, or with H 2 in the presence of a catalyst to form the protected dihydro-6-amino indoline 66a.
  • Substituted anilines are prepared such as by the procedures described in Scheme 22.
  • Nitrophenyl esters 69 are formed from the acid 68, such as by treatment with MeOH and acid.
  • Alkylation of the ester 69 such as by treatment with base, such as Nail, followed by alkyl halide, yields the branched alkyl compounds 70.
  • Reduction of the ester 70 such as with BH 3 , yields the alcohol 71.
  • the aldehyde 72 is prepared from the alcohol 71, such as by treatment with TPAP in the presence of N-methylmorpholine-N-oxide. Subsequent treatment with methoxymethyltriphenylphosphonium chloride and KHMDS yields 72.
  • Substituted aniline compounds (where R x is a substituent selected those available for substituted R 2 , preferably haloalkyl and alkyl) are prepared such as by the procedure described in Scheme 23.
  • Alkynyl-aniline 81 prepared similar to that described in Scheme 23, is hydrogenated such as with H 2 in the presence of a catalyst, such as Pd(OH) 2 , to yield the substituted alkyl 82.
  • Substituted bromophenyl compounds are prepared such as by the procedure described in Scheme 24. Bromine is added to a optionally substituted nitrobenzene 83, AgSO 4 and acid, such as H 2 SO 4 , to provide the bromo derivative 84.
  • Substituted anilines are prepared such as by the procedure described in Scheme 25 (where R t and R v are alkyl, or together with the nitrogen atom form a 4-6 membered heterocyclic ring).
  • Acryloyl chloride 85 is reacted with an amine, preferably a secondary amine, such as at a temperature between about 0° C. and about RT, to form the amide 86.
  • a bromo-nitrobenzene 84 is reacted with the amide 88, such as in the presence of base, for example TEA, together with Pd(OAc) 2 and Pd(PPh 3 ) 4 , at a temperature above about 50° C., and preferably at about 120° C., such as in a sealed container, to form the substituted alkene 87.
  • Hydrogenation of the alkene 87, such as with H 2 in the presence of a catalyst, for example Pd/C catalyst yields the substituted aniline 88.
  • Reduction of the amide 88, such as with LiALH 4 at a temperature above about 50° C., and preferably at about 80° C. yields the aniline 89.
  • Substituted indoles are prepared such as by the procedure described in Scheme 26.
  • a nitroindole 90 is coupled with a halo compound, in the presence of base, for example K 2 CO 3 .
  • Heating at a temperature above about 50° C., and preferably at about reflux yields the substituted-nitro-1H-indole 91.
  • Hydrogenation similar to conditions described above yieldS the amino derivative 92.
  • Substituted pyrimidines are prepared such as by the procedure described in Scheme 27.
  • 2-Methylthio-5-pyrimidyl acids 95 are prepared from the corresponding esters 93 similar to procedures described above.
  • the amides 96 are formed from the acids 95 by coupling with the amine such as in the presence of HATU and base, TEA for example.
  • the methylthio group can be removed, such as with Raney-Ni and heat, preferably at about reflux temperature, to form the pyrimidine 97.
  • Substituted anilines are prepared such as by the procedure described in Scheme 28. Treatment with the haloalkyl alcohol 101 with an alcohol, such as in the presence of DEAD and PPh 3 yields the ether 102 or 103.
  • Functionalized pyridines are prepared such as by the procedure described in Scheme 29.
  • 2-Fluoropyridine 104 is treated with base, such as LDA, at a temperature below about 0° C., and preferably at about ⁇ 78° C., and quenched with a stream of dry CO 2 to form the nicotinic acid 105.
  • base such as LDA
  • solid CO 2 dry ice
  • the acid 105 is converted to the acid halide 106, such as by treatment with thionyl chloride and heating at a temperature above about 50° C., and preferably at about reflux.
  • Chloro-substituted pyridines 107 are prepared such as by the procedure described in Scheme 30. 2-Chloronicotinic acid is activated with ethyl chloroformate, in the presence of base, such as TEA, at a temperature of about RT. Reaction with an amine produces amide 107. Alternatively, the amine can be coupled with the acid chloride 108, such as with polymer-supported DIEA. Excess acid chloride is removed by treating the reaction mixture with polymer-supported trisamine resin, to form amide 107.
  • base such as TEA
  • Amino-substituted indoles 111 are prepared such as by the procedure described in Scheme 31. Nitroindoline 109 is reacted with N-methyl-4-piperidone in the presence of NaOMe at a temperature above about 50° C., and preferably at about reflux, to form the 3-substituted indole 110. Hydrogenation as previously discussed yields the amino indole 111 .
  • Alkylated indazoles can be prepared by the process outlined in Scheme 32.
  • strong base such as NaH at a temperature below RT, preferably at about 0° C.
  • Alkylhalides such as where R′′ is methyl, are added and reacted at a temperature about RT to give 1-alkyl-6-nitro-1H-indazole 113.
  • the nitro indazole 113 is hydrogenated, such as with an H 2 atmosphere in the presence of a catalyst, such as Pd/C to give the 1-substituted-6-amino-1-indazole 114.
  • Brominated indazoles can be prepared by the process outlined in Scheme 33. NBS is slowly added to an acidic solution, such as a mixture of TFA:H 2 SO 4 (5:1) and tert-butyl-4-nitrobenzene 115 at a temperature of about RT to yield the brominated compound 116.
  • an acidic solution such as a mixture of TFA:H 2 SO 4 (5:1) and tert-butyl-4-nitrobenzene 115 at a temperature of about RT to yield the brominated compound 116.
  • Substituted anilines (where R x is a substituent selected those available for substituted R 2 ) can be prepared by the process outlined in Scheme 34.
  • a mixture of 1-(substituted)-2-bromo-4-nitrobenzene 117 and N-methylpiperazine is heated, such as with or without solvent, preferably without solvent, at a temperature above RT, preferably at a temperature above about 100° C., and more preferably at a temperature at about 130° C. to give the 1-[5-(substituted)-2-nitrophenyl]-4-methylpiperazine 118.
  • the nitro compound 118 is hydrogenated, such as with an H 2 atmosphere in the presence of a catalyst, such as Pd/C to furnish 4-(substituted)-2-(4-methylpiperazinyl)phenylamine 119.
  • a catalyst such as Pd/C
  • Tricyclic heterocycles can be prepared by the process outlined in Scheme 35.
  • 7-Nitro-2,3,4-trihydroisoquinolin-1-one 120 is heated in POC1 3 at a temperature above RT, preferably at a temperature sufficient for reflux, to form the 1-chloro-7-nitro-3,4-dihydroisoquinoline 121.
  • the 1-chloro-7-nitro-3,4-dihydroisoquinoline 121 is dissolved in a solvent, such as THF, and H 2 NNH 2 is added.
  • the reaction is heated with HC(OEt) 3 at a temperature above RT, preferably at a temperature above about 75° C., and more preferably at a temperature at about 115° C.
  • Indolinyl substituted carboxamides can be prepared from the corresponding nitro indoline 123 by the process outlined in Scheme 36.
  • 3,3-dimethyl-6-nitroindoline 123 is alkylated, such as with N-protected-4-formylpiperidine in the presence of NaHB(OAc) 3 and acid, such as glacial AcOH, and solvent, such as dichloromethane, at a temperature of about RT, to afford the alkylated indane 124.
  • Substituted anilines can be prepared by the process outlined in Scheme 37 (where R x is a substituent selected those available for substituted R 2 , preferably haloalkyl and alkyl).
  • R x is a substituent selected those available for substituted R 2 , preferably haloalkyl and alkyl.
  • 1-Methyl-4-piperidinone 129 is added to a solution of strong base such as LiHMDS, in a solvent such as THF, at a temperature below RT, preferably lower than about ⁇ 50° C., more preferably at about ⁇ 78° C.
  • Tf 2 NPh is reacted with the enolate at a temperature of about RT, to give l-methyl-4-(1,2,5,6-tetrahydro)pyridyl-(trifluoromethyl)sulfonate.
  • a mixture of the triflate intermediate, bis(pinacolato)diboron, potassium acetate, PdCl 2 dppf, and dppf in a solvent such as dioxane is heated at a temperature above RT, preferably at a temperature above about 50° C., and more preferably at a temperature at about 80° C. to give 4,4,5,5-tetramethyl-2-(1-methyl(4-1,2,5,6-tetrahydropyridyl))-1,3,2-dioxaborolane 130.
  • the substituted aniline 131 is formed from the 1,3,2-dioxaborolane 130 such as with treatment with an amine in the presence of PdCl 2 dppf and base, such as K 2 CO 3 , in a solvent such as DMF at a temperature above RT, preferably at a temperature above about 50° C., and more preferably at a temperature at about 80° C.
  • Substituted anilines can be prepared by the process outlined in Scheme 38.
  • 4-Cyano-4-phenylpiperidine hydrochloride 132 is treated with base, such as KOH, at a temperature above RT, preferably at a temperature above about 100° C., and more preferably at a temperature at about 160° C., to provide the phenyl piperidine 133.
  • base such as KOH
  • Alkylation of the phenyl piperidine 133 such as with formaldehyde and NaCNBH 3 in a solvent such as CH 3 CN, with sufficient acid to maintain the reaction pH near 7, to provide the alkylated piperidine 134.
  • Nitration of the phenylpiperidine 134 such as with H 2 SO 4 and fuming HNO 3 at a temperature below RT, and preferably at about 0° C., gives the nitro intermediate 135.
  • Substituted amides can be prepared by the process outlined in Scheme 39. 3-Nitrocinnamic acid 137 is coupled with 1-methylpiperazine in the presence of EDC and a solvent such as CH 2 Cl 2 , at a temperature of about RT gives the carboxamide 138.
  • Substituted benzylamines can be prepared by the process outlined in Scheme 40.
  • a substituted bromobenzylamine 139 where R 2a is a substituent described for R 2 is protected such as with Boc 2 O in the presence of base, such as TEA in an appropriate solvent such as CH 2 Cl 2 .
  • the protected bromobenzylamine 140 is alkylated, such as with 1-dimethylamino-2-propyne in the presence of catalyst, such as PdCl 2 (PPh 3 ) 2 bis(triphenyphosphino)-palladium chloride, and CuI, in the presence of base, such as TEA, at a temperature above RT, preferably at a temperature above about 50° C., and more preferably at a temperature at about 100° C., such as in a sealed tube, to form the propynylbenzylamine 141.
  • the propynylbenzylamine is hydrogenated such as with H 2 in the presence of Pd(OH) 2 and MeOH to provide the propylbenzylamine 142.
  • Deprotection, such as with strong acid, such as TFA, for removal of a Boc protecting group yields the propylbenzylamine 143.
  • Substituted benzylamines can be prepared by the process outlined in Scheme 41.
  • the protected bromobenzylamine 140 is alkylated, such as with propargyl alcohol in the presence of catalyst, such as PdCl 2 (PPh 3 ), and CuI, in the presence of base, such as TEA, at a temperature above RT, preferably at a temperature above about 50° C., and more preferably at a temperature at about 100° C., such as in a sealed tube, to form the protected hydroxypropynylbenzylamine 144.
  • the protected hydroxypropynylbenzylamine is treated with N-methylmorpholine oxide in the presence of a catalyst, such as tetrapropylammonium perruthenate, to form the aldehyde intermediate.
  • a catalyst such as tetrapropylammonium perruthenate
  • Reductive amination such as with the addition of morpholine and NaBH(OAc) 3 provides the morpholinyl derivative.
  • Deprotection such as with strong acid, such as TFA, for removal of a Boc protecting group, yields the propylbenzylamine 145.
  • Substituted heterocycles may be prepared by the method found in Scheme 42. Chloro-heterocycles 146 (where LG is OH) is coupled with an amine 147 at a suitable temperature, such as a temperature over about 100° C. to give the 2-substituted amino-nicotinic acid 148. The 2-substituted amino-nicotinic acid 148 is reacted with a substituted amine in the presence of a coupling reagent, such as BOP-Cl and base, such as TEA to form the 2-substituted amino-nicotinamide 149.
  • a coupling reagent such as BOP-Cl and base, such as TEA
  • 2-chloro-nicotinoyl chloride 146 (where LG is Cl) is coupled first with R 2 —NH 2 , such as in the presence of base, e.g., NaHCO 3 , in a suitable solvent, such as IpOH or CH 2 Cl 2 , to form the amide 150, then coupled with a benzylamine 147 to yield the 2-substituted amino-nicotinamide 149.
  • A is a pi-electron rich heterocycle
  • KF such as 40% KF on alumina in IpOH, at a temperature over about 100° C., preferably about 160° C., can be used in the formation of 149 from 150.
  • 2,3,4,4a,9,9a-Hexahydro-1H-3-aza-fluoren-6-ylamine may be prepared by the method found in Scheme 43.
  • Nitrobenzylpyridines 151 are alkylated, such as with MeI, in the presence of TBAI and base to form the pyridinium compound 152.
  • the pyridinium compounds 152 are halogenated, such as brominated with NBS, to form the brominated pyridinium compounds 153 which are reduced such as with NaBH 4 to form the tetrahydro-pyridines 154.
  • Palladium catalyzed intramolecular Heck coupling followed by hydrogenation forms the hexahydro-fluorenes 156.
  • the starting compounds defined in Schemes 1-43 may also be present with functional groups in protected form if necessary and/or in the form of salts, provided a salt-forming group is present and the reaction in salt form is possible. If so desired, one compound of Formula I-IV can be converted into another compound of Formula I-IV or an N-oxide thereof; a compound of Formula I-IV can be converted into a salt; a salt of a compound of Formula I-IV can be converted into the free compound or another salt; and/or a mixture of isomeric compounds of Formula I-IV can be separated into the individual isomers.
  • N-Oxides can be obtained in a known matter by reacting a compound of Formula I-IV with hydrogen peroxide or a peracid, e.g., 3-chloroperoxy-benzoic acid, in an inert solvent, e.g., CH 2 Cl 2 , at a temperature between about ⁇ 10-35° C., such as about 0° C.—RT.
  • a peracid e.g., 3-chloroperoxy-benzoic acid
  • one or more other functional groups for example carboxy, hydroxy, amino, or mercapto, are or need to be protected in a compound of Formula I-IV or in the synthesis of a compound of Formula I-IV, because they should not take part in the reaction, these are such groups as are usually used in the synthesis of peptide compounds, and also of cephalosporins and penicillins, as well as nucleic acid derivatives and sugars.
  • the protecting groups may already be present in precursors and should protect the functional groups concerned against unwanted secondary reactions, such as acylations, etherifications, esterifications, oxidations, solvolysis, and similar reactions. It is a characteristic of protecting groups that they lend themselves readily, i.e. without undesired secondary reactions, to removal, typically by solvolysis, reduction, photolysis or also by enzyme activity, for example under conditions analogous to physiological conditions, and that they are not present in the end-products. The specialist knows, or can easily establish, which protecting groups are suitable with the reactions mentioned above and hereinafter.
  • functional groups of the starting compounds which should not take part in the reaction may be present in unprotected form or may be protected for example by one or more of the protecting groups mentioned above under “protecting groups”.
  • the protecting groups are then wholly or partly removed according to one of the methods described there.
  • Salts of a compound of Formula I-IV with a salt-forming group may be prepared in a manner known per se. Acid addition salts of compounds of Formula I-IV may thus be obtained by treatment with an acid or with a suitable anion exchange reagent.
  • a salt with two acid molecules for example a dihalogenide of a compound of formula I
  • Salts can usually be converted to free compounds, e.g., by treating with suitable basic agents, for example with alkali metal carbonates, alkali metal hydrogen carbonates, or alkali metal hydroxides, typically potassium carbonate or sodium hydroxide.
  • suitable basic agents for example with alkali metal carbonates, alkali metal hydrogen carbonates, or alkali metal hydroxides, typically potassium carbonate or sodium hydroxide.
  • a compound of formula I, wherein Z is oxygen can be converted into the respective compound wherein Z is sulfur, for example, by using an appropriate sulfur compound, e. g. using reaction with Lawesson's reagent (2,4-bis-(4-methoxyphenyl)-1,3-dithia-2,4-diphosphetane-2,4-disulfide) in a halogenated hydrocarbon, such as CH 2 Cl 2 , or an aprotic solvent, such as toluene or xylene, at temperatures from about 30° C. to reflux.
  • Lawesson's reagent 2,4-bis-(4-methoxyphenyl)-1,3-dithia-2,4-diphosphetane-2,4-disulfide
  • a halogenated hydrocarbon such as CH 2 Cl 2
  • an aprotic solvent such as toluene or xylene
  • All process steps described here can be carried out under known reaction conditions, preferably under those specifically mentioned, in the absence of or usually in the presence of solvents or diluents, preferably such as are inert to the reagents used and able to dissolve these, in the absence or presence of catalysts, condensing agents or neutralizing agents, for example ion exchangers, typically cation exchangers, for example in the H+ form, depending on the type of reaction and/or reactants at reduced, normal, or elevated temperature, for example in the range from about ⁇ 100° C. to about 190° C., preferably from about ⁇ 80° C.
  • solvents or diluents preferably such as are inert to the reagents used and able to dissolve these, in the absence or presence of catalysts, condensing agents or neutralizing agents, for example ion exchangers, typically cation exchangers, for example in the H+ form, depending on the type of reaction and/or reactants at reduced, normal, or elevated temperature
  • Salts may be present in all starting compounds and transients, if these contain salt-forming groups. Salts may also be present during the reaction of such compounds, provided the reaction is not thereby disturbed.
  • the solvents from which those can be selected which are suitable for the reaction in question include for example water, esters, typically lower alkyl-lower alkanoates, e.g., ethyl acetate, ethers, typically aliphatic ethers, e.g., diethyl ether, or cyclic ethers, e.g., THF, liquid aromatic hydrocarbons, typically benzene or toluene, alcohols, typically MeOH, EtOH or 1-propanol, IpOH, nitrites, typically CH 3 CN, halogenated hydrocarbons, typically CH 2 Cl 2 , acid amides, typically DMF, bases, typically heterocyclic nitrogen bases, e.g.
  • carboxylic acids typically lower alkanecarboxylic acids, e.g., AcOH
  • carboxylic acid anhydrides typically lower alkane acid anhydrides, e.g., acetic anhydride
  • cyclic linear, or branched hydrocarbons, typically cyclohexane, hexane, or isopentane, or mixtures of these solvents, e.g., aqueous solutions, unless otherwise stated in the description of the process.
  • solvent mixtures may also be used in processing, for example in chromatography.
  • the invention relates also to those forms of the process in which one starts from a compound obtainable at any stage as a transient and carries out the missing steps, or breaks off the process at any stage, or forms a starting material under the reaction conditions, or uses said starting material in the form of a reactive derivative or salt, or produces a compound obtainable by means of the process according to the invention and processes the said compound in situ.
  • the compounds of Formula I-IV, including their salts, are also obtainable in the form of hydrates, or their crystals can include for example the solvent used for crystallization (present as solvates).
  • amine 1 can be prepared by reduction of the corresponding nitro.
  • the reduction preferably takes place in the presence of a suitable reducing agent, such as tin(II) chloride or hydrogen in the presence of an appropriate catalyst, such as Raney nickel (then preferably the hydrogen is used under pressure, e.g. between 2 and 20 bar) or PtO 2 , in an appropriate solvent, e.g. an alcohol, such as MeOH.
  • a suitable reducing agent such as tin(II) chloride or hydrogen
  • an appropriate catalyst such as Raney nickel (then preferably the hydrogen is used under pressure, e.g. between 2 and 20 bar) or PtO 2
  • an appropriate solvent e.g. an alcohol, such as MeOH.
  • the reaction temperature is preferably between about 0° C. and about 80° C., especially about 15° C. to about 30° C.
  • Compounds of the present invention can possess, in general, one or more asymmetric carbon atoms and are thus capable of existing in the form of optical isomers as well as in the form of racemic or non-racemic mixtures thereof.
  • the optical isomers can be obtained by resolution of the racemic mixtures according to conventional processes, e.g., by formation of diastereoisomeric salts, by treatment with an optically active acid or base.
  • appropriate acids are tartaric, diacetyltartaric, dibenzoyltartaric, ditoluoyltartaric, and camphorsulfonic acid and then separation of the mixture of diastereoisomers by crystallization followed by liberation of the optically active bases from these salts.
  • a different process for separation of optical isomers involves the use of a chiral chromatography column optimally chosen to maximize the separation of the enantiomers.
  • Still another available method involves synthesis of covalent diastereoisomeric molecules by reacting compounds of the invention with an optically pure acid in an activated form or an optically pure isocyanate.
  • the synthesized diastereoisomers can be separated by conventional means such as chromatography, distillation, crystallization or sublimation, and then hydrolyzed to deliver the enantiomerically pure compound.
  • the optically active compounds of the invention can likewise be obtained by using optically active starting materials. These isomers may be in the form of a free acid, a free base, an ester or a salt.
  • the compounds of this invention may contain one or more asymmetric centers and thus occur as racemates and racemic mixtures, scalemic mixtures, single enantiomers, individual diastereomers and diastereomeric mixtures. All such isomeric forms of these compounds are expressly included in the present invention.
  • the invention expressly includes all tautomeric forms of the compounds described herein.
  • the compounds may also occur in cis- or trans- or E- or Z-double bond isomeric forms. All such isomeric forms of such compounds are expressly included in the present invention. All crystal forms of the compounds described herein are expressly included in the present invention.
  • Substituents on ring moieties may be attached to specific atoms, whereby they are intended to be fixed to that atom, or they may be drawn unattached to a specific atom, whereby they are intended to be attached at any available atom that is not already substituted by an atom other than H (hydrogen).
  • the compounds of this invention may contain heterocyclic ring systems attached to another ring system.
  • Such heterocyclic ring systems may be attached through a carbon atom or a heteroatom in the ring system.
  • a compound of any of the formulas delineated herein may be synthesized according to any of the processes delineated herein.
  • the steps may be performed in an alternate order and may be preceded, or followed, by additional protection/deprotection steps as necessary.
  • the processes may further comprise use of appropriate reaction conditions, including inert solvents, additional reagents, such as bases (e.g., LDA, DIEA, pyridine, K 2 CO 3 , and the like), catalysts, and salt forms of the above.
  • bases e.g., LDA, DIEA, pyridine, K 2 CO 3 , and the like
  • Purification methods include, for example, crystallization, chromatography (liquid and gas phase, simulated moving bed (“SMB”)), extraction, distillation, trituration, reverse phase HPLC and the like Reactions conditions such as temperature, duration, pressure, and atmosphere (inert gas, ambient) are known in the art and may be adjusted as appropriate for the reaction.
  • SMB simulated moving bed
  • Reactions conditions such as temperature, duration, pressure, and atmosphere (inert gas, ambient) are known in the art and may be adjusted as appropriate for the reaction.
  • the compounds of this invention may be modified by appending appropriate functionalities to enhance selective biological properties.
  • modifications are known in the art and include those which increase biological penetration into a given biological compartment (e.g., blood, lymphatic system, central nervous system), increase oral availability, increase solubility to allow administration by injection, alter metabolism and alter rate of excretion.
  • Preparative HPLC was conducted on Beckman or Waters HPLC system with 0.1% TFA/H 2 O and 0.1% TFA/CH 3 CN as mobile phase. The flow rate was at 20 ml/min. and gradient method was used. 1 H NMR spectra were determined with super conducting FT NMR spectrometers operating at 400 MHz or a Varian 300 MHz instrument. Chemical shifts are expressed in ppm downfield from internal standard tetramethylsilane. All compounds showed NMR spectra consistent with their assigned structures.
  • Mass spectra were determined on a Perkin Elmer-SCIEX API 165 electrospray mass spectrometer (positive and, or negative) or an HP 1100 MSD LC-MS with eletrospray ionization and quadrupole detection. All parts are by weight and temperatures are in Degrees centigrade unless otherwise indicated.
  • 1-Boc-4- ⁇ 3-[(2-chloro-pyridine-3-carbonyl)-amino]-5-trifluoromethyl-phenoxy ⁇ -piperidine was prepared from 1-Boc-4-(3-amino-5-trifluoromethyl-phenoxy)-piperidine and 2-chloropyridine-3-carbonyl chloride by a procedure similar to that described in the preparation of 1-Boc-4- ⁇ 3-[(2-fluoro-pyridine-3-carbonyl)-amino]-5-trifluoromethyl-phenoxy ⁇ -piperidine.
  • 1-Boc-2- ⁇ 3-[(2-Fluoro-pyridine-3-carbonyl)-amino]-5-trifluoromethyl-phenoxymethyl ⁇ -pyrrolidine was prepared from 1-Boc-2-(3-amino-5-trifluoromethyl-phenoxymethyl)-pyrrolidine by a procedure similar to that described in the preparation of 1-Boc-4- ⁇ 3-[(2-fluoro-pyridine-3-carbonyl)-amino]-5-trifluoromethyl-phenoxy ⁇ -piperidine.
  • N-[3-(3-Piperidin-1-yl-propyl)-5-trifluoromethyl-phenyl]-acetamide (1.33 g) was dissolved in EtOH (40 ml) and 12 N HCl (40 ml) was added. After stirring overnight at 70° C. and RT, the mixture was concentrated in vacuo, affording 3-(3-piperidin-1-yl-propyl)-5-trifluoromethyl-phenylamine as a brown oil.
  • N-[3-(3-Morpholin-4-yl-propyl)-5-trifluoromethyl-phenyl]-acetamide was prepared from allyl morpholine and N-(3-bromo-5-trifluoromethyl-phenyl)-acetamide similar to that described in the preparation of N [3-(3-piperidin-1-yl-propyl)-5-trifluoromethyl-phenyl]-acetamide.
  • 3-(3-Morpholin-4-yl-propyl)-5-trifluoromethyl-phenylamine was prepared from N-[3-(3-morpholin-4-yl-propyl)-5-trifluoromethyl-phenyl]-acetamide similar to that described in the preparation of 3-(3-piperidin-1-yl-propyl)-5-trifluoromethyl-phenylamine.
  • Ph 3 PCH 3 I 50 g, Aldrich
  • Et 2 O 20 ml
  • butyllithium 77.3 ml, 1.6 M in hexanes, Aldrich
  • 1-methylpiperidone (12.3 ml, Aldrich) was added slowly.
  • the mixture was stirred at RT overnight.
  • the solid was removed by filtration, the volume was reduced to ⁇ 400 ml and additional solid was removed by filtration.
  • the Et 2 O was washed with H20 (2 ⁇ ) and 2N HCl (4 ⁇ ).
  • the pH of the acid washings was adjusted to ⁇ 11 with 6 N NaOH, then they were extracted with CH 2 Cl 2 (4 ⁇ ).
  • the CH2Cl 2 washings were dried over Na 2 SO 4 and concentrated cold in vacuo to provide 1-methyl-4-methylene-piperidine which was used as is.
  • N-[3-(1-Methylpiperidin-4-yl)-5-trifluoromethyl-phenyl]-acetamide was prepared from 1-methyl-4-methylene-piperidine and N-(3-bromo-5-trifluoromethyl-phenyl)-acetamide similar to that described in the preparation of N-[3-(3-piperidin-1-yl-propyl)-5-trifluoromethyl-phenyl]-acetamide.
  • 3-(1-Methylpiperidin-4-yl)-5-trifluoromethyl-phenylamine was prepared from N-[3-(1-methylpiperidin-4-yl)-5-trifluoromethyl-phenyl]-acetamide similar to the procedure described in the preparation of 3-(3-piperidin-1-yl-propyl)-5-trifluoromethyl-phenylamine.
  • 2-(2-Morpholin-4-yl-ethoxy)-4-pyridylcarbonitrile was prepared from 2-chloro-4-cyanopyridine and 2-morpholin-4-yl-ethanol by a procedure similar to that described in the preparation of 2-(1-methylpiperidin-4-yloxy)-4-pyridylcarbonitrile.
  • the HCl salt was prepared similar to that described for [2-(1-methylpiperidin-4-yloxy)-pyridin-4-yl]methylamine bis hydrochloride.
  • 2-(2-Morpholin-4-yl-propoxy)-4-pyridylcarbonitrile was prepared from 2-chloro-4-cyanopyridine and 2-morpholin-4-yl-propanol by a procedure similar to that described in the preparation of 2-(1-methylpiperidin-4-yloxy)-4-pyridylcarbonitrile.
  • N-[4-tert-Butyl-3-(1-methyl-piperidin-4-ylmethoxy)-phenyl]-2-chloro-nicotinamide was prepared from 4-tert-butyl-3-(1-methyl-piperidin-4-ylmethoxy)-phenylamine by a procedure similar to that described in the preparation of 1-Boc-4- ⁇ 3-[(2-chloro-pyridine-3-carbonyl)-amino]-5-trifluoromethyl-phenoxy ⁇ -piperidine.
  • 1-Boc-4-Hydroxymethyl-piperidine was prepared from 1-Boc-piperidine-4-carboxylic acid ethyl ester by a procedure similar to that described in the preparation of 2-morpholin-4-yl-propanol.
  • N-Boc-(2-chloropyrimidine)-methylamine (663 mg) and 4-(aminopropyl)morpholine (786 mg) were dissolved in MEOH and concentrated in vacuo. The residue was heated at 100° C. for 15 min, forming a solid which was dissolved in CH 2 Cl 2 /MeOH then concentrated again and heated 15 min more. Concentrated in vacuo and dried under high vacuum. Triturated with a small amount of IpOH and allowed to settle over a weekend. Filtered, rinsing with a small amount of IpOH to provide the compound as a white solid.
  • N 2 was bubbled through a solution of 3-bromo-5-trifluoromethyl-phenylamine (2.38 g), 5,5,5′,5′-tetramethyl-[2,2′]bi[[1,3,2]dioxaborinanyl] (2.24 g, Frontier Scientific) and KOAc (2.92 g), dppf (165 mg, Aldrich) in anhydrous dioxane (50 ml) for 2 min. PdCl 2 (dppf) (243 mg, Aldrich) was added and the reaction was heated to 80° C. for 4 h.
  • the acidic aqueous layer was basified to pH>12 with 6 N NaOH, extracted with Et 2 O, brine-washed, dried over Na 2 SO 4 , filtered, and concentrated in vacuo to provide 1-[4-(2-tert-butyl-5-nitro-phenyl)-but-3-enyl]-pyrrolidine as a orange-brown oil.
  • N-(2-Bromo-5-nitrophenyl)-N-(2-methylprop-2-enyl)acetamide (4.5 g) was dissolved in anhydrous DMF (50 mL), tetraethyl-ammonium chloride (2.5 g), sodium formate (1.2 g), NaOAc (3 g) were added, and the resulting mixture was bubbled with N 2 gas for 10 min.
  • Pd(OAc) 2 350 mg was added and the mixture was heated at 80° C. under N 2 atmosphere overnight. After the mixture was concentrated in vacuo, it was partitioned between saturated NaHCO 3 solution and EtOAc, the resulting organic layer was dried over MgSO 4 , filtered and concentrated in vacuo.
  • NBS (125.0 g, 697.5 mmol, 1.5 eq) was slowly added to a solution of TFA:H 2 SO 4 (5:1, 750 mL) and tert-butyl-4-nitrobenzene (100.0 g, 558.0 mmol) at RT. The solution was stirred for 24 h and poured over 5 kg of ice. The resulting suspension was filtered and washed with a 1:1 MeOH:H 2 O solution (200 mL) and dried in a vacuum oven. MS (ES+): 258.1, 260.1 (M+H) + . Calc'd for C 10 H 12 BrNO 2 : 257.0.
  • 6-Nitroindoline (5 g) was dissolved in 200 mL of dichloroethane. N-Methyl-4-piperidone (5 g) was added to the mixture, followed by NaHB(OAc) 3 (12 g) and 1 mL of glacial AcOH. The mixture was stirred at RT overnight. A saturated NaHCO 3 (200 mL) solution was added to the reaction mixture and stirred for 1 h. The resulting mixture was separated by separation funnel. The organic layer was extracted once with saturated NaHCO 3 solution and once with brine. The resulting organic layer was dried over MgSO 4 , filtered and concentrated in vacuo. The crude material was purified by flash chromatography on silica gel with 2:1 EtOAc:MeOH to afford orange oil. MS: 262 (M+1). Calc'd. for C 14 H 19 N 3 O 2 -261.3.
  • N-(2-Bromo-5-nitrophenyl)-N-(2-methylprop-2-enyl)acetamide (4.5 g) was dissolved in 50 mL anhydrous DMF, 2.5 g tetraethyl-ammonium chloride, 1.2 g sodium formate, 3 g sodium acetate were added, the resulting mixture was bubbled with N 2 gas for 10 min. Pd(OAc) 2 (350 mg) was added and the mixture was heated at 80° C. under N 2 overnight. After the mixture was concentrated in vacuo, it was extracted between saturated NaHCO 3 solution and EtOAc, the resulting organic layer was dried over MgSO 4 , filtered and concentrated in vacuo. The crude material was purified by flash chromatography on silica gel with 2:1 Hexane:EtOAc to afford a yellow gum. MS: 235 (M+1). Calc'd. for C 12 H 14 N 2 O 3 -234.2.
  • K 2 CO 3 (5.08 g, 36.726 mmol) was added to a slurry of 6-nitroindole (1.985 g, 12.242 mmol), 4-(2-chloroethyl) morpholine HCl (2.278 g, 12.242 mmol), and CH 3 CN (100 ml). The mix was heated to reflux for 18 h, then cooled to RT, filtered, and concentrated in vacuo. The crude was eluted through a silica gel column with a gradient of 3:97 to 5:95 and finally 8:92 MeOH/CH 2 Cl 2 , to yield upon drying the desired intermediate which was hydrogenated under conditions previously described.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Pharmacology & Pharmacy (AREA)
  • General Chemical & Material Sciences (AREA)
  • Veterinary Medicine (AREA)
  • General Health & Medical Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Immunology (AREA)
  • Virology (AREA)
  • Oncology (AREA)
  • Diabetes (AREA)
  • Hematology (AREA)
  • Manufacturing & Machinery (AREA)
  • Pulmonology (AREA)
  • Ceramic Engineering (AREA)
  • Reproductive Health (AREA)
  • Dermatology (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Endocrinology (AREA)
  • Obesity (AREA)
  • Ophthalmology & Optometry (AREA)
  • Neurology (AREA)
  • Communicable Diseases (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Structural Engineering (AREA)
  • Molecular Biology (AREA)
  • Biomedical Technology (AREA)
  • Materials Engineering (AREA)
  • Inorganic Chemistry (AREA)
  • Neurosurgery (AREA)
  • Cardiology (AREA)
  • Heart & Thoracic Surgery (AREA)
US10/197,960 2001-01-12 2002-07-17 Substituted arylamine derivatives and methods of use Abandoned US20030134836A1 (en)

Priority Applications (10)

Application Number Priority Date Filing Date Title
US10/197,960 US20030134836A1 (en) 2001-01-12 2002-07-17 Substituted arylamine derivatives and methods of use
PCT/US2003/022276 WO2004007457A2 (en) 2002-07-17 2003-07-15 Substituted benzylamine derivatives and methods of use
AU2003256577A AU2003256577A1 (en) 2002-07-17 2003-07-15 Substituted benzylamine derivatives and methods of use
MXPA05000651A MXPA05000651A (es) 2002-07-17 2003-07-15 Derivados de bencilamina substituida y metodos de uso.
PL376532A PL376532A1 (pl) 2002-07-17 2003-07-15 Pochodne podstawionych benzyloamin oraz sposoby wykorzystania
CA002492164A CA2492164A1 (en) 2002-07-17 2003-07-15 Substituted benzylamine derivatives and methods of use
JP2004521923A JP2006505511A (ja) 2002-07-17 2003-07-15 置換ベンジルアミン誘導体および使用方法
EP03764756A EP1583744A2 (en) 2002-07-17 2003-07-15 Substituted benzylamine derivatives and methods of use
US10/823,809 US7101868B2 (en) 2001-01-12 2004-04-12 Substituted arylamine derivatives and methods of use
US10/996,035 US20050153960A1 (en) 2001-01-12 2004-11-22 Substituted arylamine derivatives and methods of use

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US26136001P 2001-01-12 2001-01-12
US32368601P 2001-09-19 2001-09-19
US10/046,526 US20020147198A1 (en) 2001-01-12 2002-01-10 Substituted arylamine derivatives and methods of use
US10/197,960 US20030134836A1 (en) 2001-01-12 2002-07-17 Substituted arylamine derivatives and methods of use

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US10/046,526 Continuation-In-Part US20020147198A1 (en) 2001-01-12 2002-01-10 Substituted arylamine derivatives and methods of use

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US10/823,809 Continuation US7101868B2 (en) 2001-01-12 2004-04-12 Substituted arylamine derivatives and methods of use

Publications (1)

Publication Number Publication Date
US20030134836A1 true US20030134836A1 (en) 2003-07-17

Family

ID=30115147

Family Applications (3)

Application Number Title Priority Date Filing Date
US10/197,960 Abandoned US20030134836A1 (en) 2001-01-12 2002-07-17 Substituted arylamine derivatives and methods of use
US10/823,809 Expired - Fee Related US7101868B2 (en) 2001-01-12 2004-04-12 Substituted arylamine derivatives and methods of use
US10/996,035 Abandoned US20050153960A1 (en) 2001-01-12 2004-11-22 Substituted arylamine derivatives and methods of use

Family Applications After (2)

Application Number Title Priority Date Filing Date
US10/823,809 Expired - Fee Related US7101868B2 (en) 2001-01-12 2004-04-12 Substituted arylamine derivatives and methods of use
US10/996,035 Abandoned US20050153960A1 (en) 2001-01-12 2004-11-22 Substituted arylamine derivatives and methods of use

Country Status (8)

Country Link
US (3) US20030134836A1 (pl)
EP (1) EP1583744A2 (pl)
JP (1) JP2006505511A (pl)
AU (1) AU2003256577A1 (pl)
CA (1) CA2492164A1 (pl)
MX (1) MXPA05000651A (pl)
PL (1) PL376532A1 (pl)
WO (1) WO2004007457A2 (pl)

Cited By (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050261343A1 (en) * 2000-05-09 2005-11-24 Martin Krueger Anthranilamides and their use as pharmaceutical agents
US7051310B2 (en) * 2003-05-08 2006-05-23 Cadence Design Systems, Inc. Two-stage clock tree synthesis with buffer distribution balancing
US20060116380A1 (en) * 2004-11-03 2006-06-01 Rolf Bohlmann Novel anthranilamide pyridinureas as vascular endothelial growth factor (VEGF) receptor kinase inhibitors
US20060160861A1 (en) * 2004-11-03 2006-07-20 Rolf Bohlmann Novel nicotinamide pyridinureas as vascular endothelial growth factor (VEGF) receptor kinase inhibitors
US20060264425A1 (en) * 2004-03-11 2006-11-23 Rolf Bohlmann Novel anthranilamide pyridinureas as vascular endothelial growth factor (VEGF) receptor kinase inhibitors
US20070015794A1 (en) * 2002-07-31 2007-01-18 Andreas Huth VEGFR-2 and VEGFR-3 inhibitory anthranilamide pyridines
US20070135489A1 (en) * 2003-06-13 2007-06-14 Andreas Huth Vegfr-2 and vegfr-3 inhibitory anthranilamide pyridones
US20070208044A1 (en) * 2003-07-03 2007-09-06 Myriad Genetics, Incorporated Compounds and therapeutical use thereof
US20070259891A1 (en) * 2004-07-27 2007-11-08 Aventis Pharma S.A. Heterocycle-Substituted Cyclic Urea Derivatives, Preparation Thereof And Pharmaceutical Use Thereof As Kinase Inhibitors
US7517894B2 (en) 2002-07-31 2009-04-14 Bayer Schering Pharma Ag VEGFR-2 and VEGFR-3 inhibitory anthranilamide pyridines
US20090233900A1 (en) * 2005-11-30 2009-09-17 Astellas Pharma Inc. 2-aminobenzamide derivative
DE102008041214A1 (de) 2008-08-13 2010-02-18 Bayer Cropscience Ag N-substituierte Azinylakyl-azincarboxamide und deren Analoge
US7989462B2 (en) 2003-07-03 2011-08-02 Myrexis, Inc. 4-arylamin-or-4-heteroarylamino-quinazolines and analogs as activators of caspases and inducers of apoptosis and the use thereof
US8258145B2 (en) 2005-01-03 2012-09-04 Myrexis, Inc. Method of treating brain cancer
US8309562B2 (en) 2003-07-03 2012-11-13 Myrexis, Inc. Compounds and therapeutical use thereof
US8853215B2 (en) 2009-04-16 2014-10-07 Takeda Pharmaceutical Company Limited Derivatives of N-acyl-N′-phenylpiperazine useful (inter alia) for the prophylaxis or treatment of diabetes
CN109988125A (zh) * 2018-10-25 2019-07-09 河南省锐达医药科技有限公司 一类新型苯甲酰苯胺化合物的制备方法和在肿瘤治疗上的应用
US11427558B1 (en) 2019-07-11 2022-08-30 ESCAPE Bio, Inc. Indazoles and azaindazoles as LRRK2 inhibitors
CN115010646A (zh) * 2022-07-21 2022-09-06 南京大学 一种吲哚酮类化合物的光诱导合成方法
WO2023158795A1 (en) * 2022-02-18 2023-08-24 Accent Therapeutics, Inc. Inhibitors of rna helicase dhx9 and uses thereof

Families Citing this family (25)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6878714B2 (en) * 2001-01-12 2005-04-12 Amgen Inc. Substituted alkylamine derivatives and methods of use
US6995162B2 (en) 2001-01-12 2006-02-07 Amgen Inc. Substituted alkylamine derivatives and methods of use
MXPA04012948A (es) * 2002-07-31 2005-09-12 Schering Ag Inhibidores antranilamidopiridinas vegfr-2 y vegfr-3.
KR20080044836A (ko) 2005-07-15 2008-05-21 라보라뚜와르 세로노 에스. 에이. 자궁내막증 치료용 jnk 억제제
BRPI0613042A2 (pt) * 2005-07-15 2010-12-14 Serono Lab inibidores de jnk para o tratamento de endometriose
WO2007030362A1 (en) * 2005-09-07 2007-03-15 Laboratoires Serono Sa. Ikk inhibitors for the treatment of endometriosis
US8247556B2 (en) * 2005-10-21 2012-08-21 Amgen Inc. Method for preparing 6-substituted-7-aza-indoles
AR059066A1 (es) * 2006-01-27 2008-03-12 Amgen Inc Combinaciones del inhibidor de la angiopoyetina -2 (ang2) y el inhibidor del factor de crecimiento endotelial vascular (vegf)
AU2008271051C1 (en) * 2007-06-29 2012-10-04 Acucela Inc. Alkynyl phenyl derivative compounds for treating ophthalmic diseases and disorders
WO2009051244A1 (ja) 2007-10-18 2009-04-23 Takeda Pharmaceutical Company Limited 複素環化合物
MX2011004315A (es) 2008-10-22 2011-08-24 Acucela Inc Compuestos para el tratamiento de enfermedades y transtornos oftalmologicos.
RU2612959C2 (ru) 2011-02-18 2017-03-14 АСАНА БАЙОСАЙЕНСИЗ, ЭлЭлСи Аминоиндановые соединения и их применение при лечении боли
CN103717757A (zh) * 2011-05-24 2014-04-09 威斯特研究所 用于调节爱泼斯坦-巴尔核抗原1的活性的组合物和方法
WO2014028675A1 (en) 2012-08-15 2014-02-20 Endo Pharmaceuticals Inc. Use of aminoindane compounds in treating overactive bladder and interstitial cystitis
DK3094329T3 (en) 2014-01-13 2018-11-19 Aurigene Discovery Tech Ltd BICYCLIC HETEROCYCLYL DERIVATIVES AS IRAC4 INHIBITORS
BR112016029853A2 (pt) * 2014-06-20 2017-08-22 Aurigene Discovery Tech Ltd compostos de indazol substituído como inibidores de irak4
EA034891B1 (ru) 2015-03-06 2020-04-02 Фармакеа, Инк. Фторированные ингибиторы лизилоксидазоподобного фермента-2 и их применение
WO2016144702A1 (en) 2015-03-06 2016-09-15 Pharmakea, Inc. Lysyl oxidase-like 2 inhibitors and uses thereof
WO2017151409A1 (en) 2016-02-29 2017-09-08 University Of Florida Research Foundation, Incorporated Chemotherapeutic methods
CN106496107A (zh) * 2016-08-31 2017-03-15 浙江永宁药业股份有限公司 Vegfr‑2抑制剂及其制备方法
US10774069B2 (en) 2016-09-07 2020-09-15 Pharmakea, Inc. Crystalline forms of a lysyl oxidase-like 2 inhibitor and methods of making
JP7097874B2 (ja) 2016-09-07 2022-07-08 ファーマケア,インク. リシルオキシダーゼ様2阻害剤の使用
MX2019011158A (es) 2017-03-31 2019-10-17 Aurigene Discovery Tech Ltd Compuestos y composiciones para tratar trastornos hematologicos.
FI3704108T3 (fi) 2017-10-31 2024-05-23 Curis Inc Irak4-estäjä yhdistelmässä bcl-2-estäjän kanssa käytettäväksi syövän hoitoon
CN111909053B (zh) * 2020-08-06 2022-12-06 湖北省生物农药工程研究中心 基于二芳胺单元的酰胺类衍生物及其制备方法和应用

Family Cites Families (57)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US27820A (en) * 1860-04-10 carville
CH438343A (de) * 1962-11-08 1967-06-30 Thomae Gmbh Dr K Verfahren zur Herstellung von 5,6-Dihydro-6-oxo-11H-pyrido (2,3-b) (1,4)-benzodiazepinen
US3226394A (en) * 1964-06-16 1965-12-28 Shulton Inc Pyridylethylated anthranilamides and derivatives thereof
US3822277A (en) * 1967-11-13 1974-07-02 C Dufour Certain pyridyl cyclopropylamides
BE794226A (fr) 1972-01-21 1973-07-18 Synthelabo Derives de la quinoleine, leur preparation et les medicaments qui en contiennent
DE2934543A1 (de) * 1979-08-27 1981-04-02 Basf Ag, 6700 Ludwigshafen Substituierte n-benzoylanthranilsaeurederivate und deren anydroverbindungen, verfahren zu ihrer herstellung und ihre verwendung als herbizide
DE3305755A1 (de) * 1983-02-19 1984-08-23 Gödecke AG, 1000 Berlin N-phenyl-benzamid-derivate, verfahren zu deren herstellung und deren verwendung bei der bekaempfung von erkranungen des immunsystems
US5571912A (en) * 1990-10-19 1996-11-05 Boehringer Ingelheim Pharmaceuticals, Inc. Method for the preparation of 5,11-dihydro-6h-dipyrido [3,2-b:2',3'-e][1,4]diazepines
EP1195372A1 (en) * 1994-04-18 2002-04-10 Mitsubishi Pharma Corporation N-heterocyclic substituted benzamide derivatives with antihypertensive activity
US5559135A (en) * 1994-09-14 1996-09-24 Merck & Co., Inc. Endothelin antagonists bearing pyridyl amides
US5532358A (en) * 1994-10-12 1996-07-02 Boehringer Ingelheim Pharmaceuticals, Inc. Method for preparing alkyl-5,11-dihydro-6h-dipyrido[3,2-B:2',3'-E] [1,4] diazepin-6-ones
GB9511694D0 (en) 1995-06-09 1995-08-02 Fujisawa Pharmaceutical Co Benzamide derivatives
US5770613A (en) * 1995-09-29 1998-06-23 Geron Corporation Telomerase inhibitors
US5663357A (en) * 1995-11-22 1997-09-02 Allergan Substituted heteroarylamides having retinoid-like biological activity
US6008234A (en) * 1996-09-12 1999-12-28 Berlex Laboratories, Inc. Benzamidine derivatives substituted by cyclic amino acid and cyclic hydroxy acid derivatives and their use as anti-coagulants
AUPO395396A0 (en) 1996-12-02 1997-01-02 Fujisawa Pharmaceutical Co., Ltd. Benzamide derivatives
TW523506B (en) 1996-12-18 2003-03-11 Ono Pharmaceutical Co Sulfonamide or carbamide derivatives and drugs containing the same as active ingredients
NZ337698A (en) 1997-04-04 2001-07-27 Pfizer Prod Inc Nicotinamide derivatives for selective inhibition of phosphodiesterase type 4 (PDE4) and the production of tumour necrosis factor (TNF) useful for the treatment of respiratory, rheumatoid and allergic diseases
US5919970A (en) * 1997-04-24 1999-07-06 Allergan Sales, Inc. Substituted diaryl or diheteroaryl methanes, ethers and amines having retinoid agonist, antagonist or inverse agonist type biological activity
IL133623A0 (en) * 1997-06-26 2001-04-30 Lilly Co Eli Antithrombotic agents
AU8270698A (en) * 1997-06-26 1999-01-19 Eli Lilly And Company Antithrombotic agents
CA2295153A1 (en) * 1997-06-26 1999-01-07 Gerald Floyd Smith Antithrombotic agents
IL133621A0 (en) * 1997-06-26 2001-04-30 Lilly Co Eli Antithrombotic agents
US6140351A (en) * 1997-12-19 2000-10-31 Berlex Laboratories, Inc. Ortho-anthranilamide derivatives as anti-coagulants
RU2226529C2 (ru) 1997-12-19 2004-04-10 Шеринг Акциенгезельшафт Производные ортоантраниламида в качестве антикоагулянтов, фармацевтическая композиция и способ лечения
US6271237B1 (en) * 1997-12-22 2001-08-07 Dupont Pharmaceuticals Company Nitrogen containing heteromatics with ortho-substituted P1s as factor Xa inhabitors
WO1999062885A1 (en) 1998-06-05 1999-12-09 Boehringer Ingelheim Pharmaceuticals, Inc. Substituted 1-(4-aminophenyl)pyrazoles and their use as anti-inflammatory agents
HU228111B1 (en) 1998-07-08 2012-11-28 Sanofi Aventis Deutschland Sulfur substituted sulfonylaminocarboxylic acid n-arylamides, their preparation, their use and pharmaceutical preparations comprising them
DE19830430A1 (de) 1998-07-08 2000-01-13 Hoechst Marion Roussel De Gmbh Schwefelsubstituierte Sulfonylamino-carbonsäure-N-arylamide, ihre Herstellung, ihre Verwendung und sie enthaltende pharmazeutische Präparate
UA71587C2 (uk) 1998-11-10 2004-12-15 Шерінг Акцієнгезелльшафт Аміди антранілової кислоти та їхнє застосування як лікарських засобів
GB9824579D0 (en) 1998-11-10 1999-01-06 Novartis Ag Organic compounds
EP1140903B1 (en) * 1998-12-23 2004-08-04 Eli Lilly And Company Aromatic amides
CA2358091A1 (en) * 1998-12-23 2000-07-06 Eli Lilly And Company Antithrombotic amides
AU2364600A (en) 1998-12-23 2000-07-31 Eli Lilly And Company Heteroroaromatic amides as inhibitor of factor xa
JP2000256358A (ja) 1999-03-10 2000-09-19 Yamanouchi Pharmaceut Co Ltd ピラゾール誘導体
PT1219609E (pt) 1999-09-16 2007-06-19 Tanabe Seiyaku Co ''compostos cíclicos aromático azotados de seis membros''
GB9924862D0 (en) 1999-10-20 1999-12-22 Celltech Therapeutics Ltd Chemical compounds
AUPQ365299A0 (en) 1999-10-25 1999-11-18 Fujisawa Pharmaceutical Co., Ltd. Anthranilic acid derivatives
GB0001930D0 (en) * 2000-01-27 2000-03-22 Novartis Ag Organic compounds
AU2001231143A1 (en) * 2000-01-27 2001-08-07 Cytovia, Inc. Substituted nicotinamides and analogs as activators of caspases and inducers of apoptosis and the use thereof
DE10021246A1 (de) 2000-04-25 2001-10-31 Schering Ag Substituierte Benzoesäureamide und deren Verwendung als Arzneimittel
DE10023485A1 (de) 2000-05-09 2001-11-22 Schering Ag Anthranylalkyl- und -cycloalkylamide und deren Verwendung als Arzneimittel
DE10023492A1 (de) * 2000-05-09 2001-11-22 Schering Ag Aza- und Polyazanthranylamide und deren Verwendung als Arzneimittel
DE10023484A1 (de) 2000-05-09 2001-11-22 Schering Ag Anthranylamide und deren Verwendung als Arzneimittel
DE10023486C1 (de) * 2000-05-09 2002-03-14 Schering Ag Ortho substituierte Anthranilsäureamide und deren Verwendung als Arzneimittel
DE10060809A1 (de) * 2000-12-07 2002-06-20 Aventis Pharma Gmbh Substituierte Anthranilsäuren, ihre Verwendung als Medikament oder Diagnostikum, sowie sie enthaltendes Medikament, sowie ein pharmazeutisches Kombinationspräparat mit einem Natrium/Wasserstoff-Austausch (NHE)-Blocker
US20020147198A1 (en) * 2001-01-12 2002-10-10 Guoqing Chen Substituted arylamine derivatives and methods of use
US6995162B2 (en) 2001-01-12 2006-02-07 Amgen Inc. Substituted alkylamine derivatives and methods of use
US7105682B2 (en) 2001-01-12 2006-09-12 Amgen Inc. Substituted amine derivatives and methods of use
AR035431A1 (es) 2001-02-13 2004-05-26 Aventis Pharma Gmbh 6,7,8,9-tetrahidro-5h-benzocicloheptenil aminas aciladas, un metodo para su sintesis, composiciones farmaceuticas y el uso de dichos compuestos para la fabricacion de medicamentos
EP1394154A4 (en) * 2001-03-23 2005-05-18 Takeda Pharmaceutical HETEROCYCLIC DERIVATIVE WITH FIVE MEMBERS OF ALKANOIC ACID
US7312235B2 (en) * 2001-03-30 2007-12-25 Millennium Pharmaceuticals, Inc. Benzamide inhibitors of factor Xa
JP2004528378A (ja) * 2001-05-08 2004-09-16 シエーリング アクチエンゲゼルシャフト N−オキシドアントラニルアミド誘導体と医薬製剤としての利用
PL364478A1 (pl) * 2001-05-08 2004-12-13 Schering Aktiengesellschaft Selektywne amidy antranyloamidopirydyny jako inhibitory VEGFR-2 i VEGFR-3
JP2005506983A (ja) * 2001-09-19 2005-03-10 ファルマシア・コーポレーション 炎症の治療のための置換ピラゾリル化合物
US20030195192A1 (en) * 2002-04-05 2003-10-16 Fortuna Haviv Nicotinamides having antiangiogenic activity
US7517894B2 (en) * 2002-07-31 2009-04-14 Bayer Schering Pharma Ag VEGFR-2 and VEGFR-3 inhibitory anthranilamide pyridines

Cited By (29)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050261343A1 (en) * 2000-05-09 2005-11-24 Martin Krueger Anthranilamides and their use as pharmaceutical agents
US7615565B2 (en) 2002-07-31 2009-11-10 Bayer Schering Pharma Aktiengesellschaft VEGFR-2 and VEGFR-3 inhibitory anthranilamide pyridines
US20070015794A1 (en) * 2002-07-31 2007-01-18 Andreas Huth VEGFR-2 and VEGFR-3 inhibitory anthranilamide pyridines
US7517894B2 (en) 2002-07-31 2009-04-14 Bayer Schering Pharma Ag VEGFR-2 and VEGFR-3 inhibitory anthranilamide pyridines
US7051310B2 (en) * 2003-05-08 2006-05-23 Cadence Design Systems, Inc. Two-stage clock tree synthesis with buffer distribution balancing
US20070135489A1 (en) * 2003-06-13 2007-06-14 Andreas Huth Vegfr-2 and vegfr-3 inhibitory anthranilamide pyridones
US8309562B2 (en) 2003-07-03 2012-11-13 Myrexis, Inc. Compounds and therapeutical use thereof
US20070208044A1 (en) * 2003-07-03 2007-09-06 Myriad Genetics, Incorporated Compounds and therapeutical use thereof
US20080039479A1 (en) * 2003-07-03 2008-02-14 Myriad Genetics, Incorporated Compounds and therapeutical use thereof
US7989462B2 (en) 2003-07-03 2011-08-02 Myrexis, Inc. 4-arylamin-or-4-heteroarylamino-quinazolines and analogs as activators of caspases and inducers of apoptosis and the use thereof
US20070249601A1 (en) * 2003-07-03 2007-10-25 Myriad Genetics, Incorporated Compounds and therapeutical use thereof
US20070244113A1 (en) * 2003-07-03 2007-10-18 Myriad Genetics, Incorporated Compounds and therapeutical use thereof
US20060264425A1 (en) * 2004-03-11 2006-11-23 Rolf Bohlmann Novel anthranilamide pyridinureas as vascular endothelial growth factor (VEGF) receptor kinase inhibitors
US20070259891A1 (en) * 2004-07-27 2007-11-08 Aventis Pharma S.A. Heterocycle-Substituted Cyclic Urea Derivatives, Preparation Thereof And Pharmaceutical Use Thereof As Kinase Inhibitors
US7572794B2 (en) 2004-11-03 2009-08-11 Bayer Schering Pharma Ag Anthranilamide pyridinureas as vascular endothelial growth factor (VEGF) receptor kinase inhibitors
US20060160861A1 (en) * 2004-11-03 2006-07-20 Rolf Bohlmann Novel nicotinamide pyridinureas as vascular endothelial growth factor (VEGF) receptor kinase inhibitors
US20060116380A1 (en) * 2004-11-03 2006-06-01 Rolf Bohlmann Novel anthranilamide pyridinureas as vascular endothelial growth factor (VEGF) receptor kinase inhibitors
US7902229B2 (en) 2004-11-03 2011-03-08 Bayer Schering Pharma Ag Anthranilamide pyridinureas as vascular endothelial growth factor (VEGF) receptor kinase inhibitors
US20110124680A1 (en) * 2004-11-03 2011-05-26 Rolf Bohlmann Novel anthranilamide pyridinureas as vascular endothelial growth factor (vegf) receptor kinase inhibitors
US7906533B2 (en) 2004-11-03 2011-03-15 Bayer Schering Pharma Ag Nicotinamide pyridinureas as vascular endothelial growth factor (VEGF) receptor kinase inhibitors
US8258145B2 (en) 2005-01-03 2012-09-04 Myrexis, Inc. Method of treating brain cancer
US20090233900A1 (en) * 2005-11-30 2009-09-17 Astellas Pharma Inc. 2-aminobenzamide derivative
US8106190B2 (en) * 2005-11-30 2012-01-31 Astellas Pharma Inc. 2-aminobenzamide derivatives
DE102008041214A1 (de) 2008-08-13 2010-02-18 Bayer Cropscience Ag N-substituierte Azinylakyl-azincarboxamide und deren Analoge
US8853215B2 (en) 2009-04-16 2014-10-07 Takeda Pharmaceutical Company Limited Derivatives of N-acyl-N′-phenylpiperazine useful (inter alia) for the prophylaxis or treatment of diabetes
CN109988125A (zh) * 2018-10-25 2019-07-09 河南省锐达医药科技有限公司 一类新型苯甲酰苯胺化合物的制备方法和在肿瘤治疗上的应用
US11427558B1 (en) 2019-07-11 2022-08-30 ESCAPE Bio, Inc. Indazoles and azaindazoles as LRRK2 inhibitors
WO2023158795A1 (en) * 2022-02-18 2023-08-24 Accent Therapeutics, Inc. Inhibitors of rna helicase dhx9 and uses thereof
CN115010646A (zh) * 2022-07-21 2022-09-06 南京大学 一种吲哚酮类化合物的光诱导合成方法

Also Published As

Publication number Publication date
PL376532A1 (pl) 2006-01-09
WO2004007457A3 (en) 2005-08-04
CA2492164A1 (en) 2004-01-22
US20050153960A1 (en) 2005-07-14
AU2003256577A1 (en) 2004-02-02
US20040204437A1 (en) 2004-10-14
MXPA05000651A (es) 2005-03-31
EP1583744A2 (en) 2005-10-12
WO2004007457A2 (en) 2004-01-22
US7101868B2 (en) 2006-09-05
JP2006505511A (ja) 2006-02-16

Similar Documents

Publication Publication Date Title
EP1562933B1 (en) Substituted amine derivatives and methods of use in the treatment of angiogenesis related disorders
EP1537084B1 (en) Substituted 2-alkylamine nicotinic amide derivatives and use there of
US7514564B2 (en) Substituted amine derivatives and methods of use
US7101868B2 (en) Substituted arylamine derivatives and methods of use
US7307088B2 (en) Substituted anthranilic amide derivatives and methods of use
US8058445B2 (en) Substituted pyridinecarboxamides for the treatment of cancer
AU2002248339B2 (en) Substituted Arylamine Derivatives and Methods of Use
AU2002248339A1 (en) Substituted Arylamine Derivatives and Methods of Use
AU2002253890A1 (en) Substituted Amine Derivatives and Methods of Use
AU2002248340A1 (en) Substituted alkylamine derivatives and methods of use

Legal Events

Date Code Title Description
AS Assignment

Owner name: AMGEN INC., CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:ELBAUM, DANIEL;ASKEW, BENNY;BOOKER, SHON;AND OTHERS;REEL/FRAME:013451/0372;SIGNING DATES FROM 20020924 TO 20021017

STCB Information on status: application discontinuation

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