WO2007146824A2 - Quinoline compounds and methods of use - Google Patents

Quinoline compounds and methods of use Download PDF

Info

Publication number
WO2007146824A2
WO2007146824A2 PCT/US2007/070787 US2007070787W WO2007146824A2 WO 2007146824 A2 WO2007146824 A2 WO 2007146824A2 US 2007070787 W US2007070787 W US 2007070787W WO 2007146824 A2 WO2007146824 A2 WO 2007146824A2
Authority
WO
WIPO (PCT)
Prior art keywords
fluorophenyl
yloxy
compound
methoxyquinolin
aryl
Prior art date
Application number
PCT/US2007/070787
Other languages
French (fr)
Other versions
WO2007146824A3 (en
Inventor
John Gaudino
Steven Armen Boyd
Allison L. Marlow
Tomas Kaplan
Kin Chiu Fong
Jeongbeob Seo
Hongqi Tian
James Blake
Kevin Koch
Original Assignee
Array Biopharma 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
Application filed by Array Biopharma Inc. filed Critical Array Biopharma Inc.
Priority to EP07798333A priority Critical patent/EP2032538A2/en
Priority to CA002655128A priority patent/CA2655128A1/en
Priority to US12/303,930 priority patent/US20110053931A1/en
Priority to JP2009514553A priority patent/JP2009539878A/en
Publication of WO2007146824A2 publication Critical patent/WO2007146824A2/en
Publication of WO2007146824A3 publication Critical patent/WO2007146824A3/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D215/00Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems
    • C07D215/02Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom
    • C07D215/16Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D215/20Oxygen atoms
    • C07D215/22Oxygen atoms attached in position 2 or 4
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • 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

Definitions

  • the invention relates to quinoline compounds having protein tyrosine kinase activity.
  • the quinoline compounds may be useful in the treatment of hyperproliferative disorders, such as cancer, in mammals.
  • the invention also relates to pharmaceutical compositions and formulations, methods of synthesis, and methods of use such as treating hyperproliferative disorders.
  • Met tyrosine kinase is a high-affinity transmembrane receptor for the hepatocyte growth factor (HGF, Bottaro et al (1991) Science 251:802-804). Met was cloned, named (Cooper et al (1984) 311:29-33) and identified as an oncogene (Park et al (1986) Cell 45:895-904). When deregulated by overexpression or mutations, Met receptor tyrosine kinase leads to tumor growth and invasion (Cristiani et al (2005) Biochem. 44:14110-14119).
  • HGF also known as Scatter Factor
  • Stimulation of Met by the ligand HGF initiates numerous physiological processes, including cell proliferation, scattering, morphogenic differentiation, angiogenesis, wound healing, tissue regeneration, and embryological development
  • Receptor c-Met is rapidly internalized via clathrin-coated vesicles and traffics through an early endosomal compartment after hepatocyte growth factor stimulation.
  • c-Met accumulates progressively in perinuclear compartments, which in part include the Golgi (Kermorgant et al (2003) J. of Biol. Chem. 278(31):28921-28929).
  • c-Met Antibodies specific for c-Met have been expressed to block binding of HGF to c-Met (US 2005/0037431; US 2004/0166544). c-Met is also over-expressed in both non-small cell lung cancer and small cell lung cancer cells, in lung, breast, colon and prostate tumors (Herynk et al (2003) Cancer Res. 63(l l):2990-2996; Maulik et al (2002) Clin. Cancer Res. 8:620-627). Since c-Met appears to play an important role in oncogenesis of a variety of tumors, various inhibition strategies have been employed to therapeutically target this receptor tyrosine kinase.
  • Protein kinases are enzymes that catalyze the phosphorylation of hydroxy groups on tyrosine, serine and threonine residues of proteins by transfer of the terminal (gamma) phosphate from ATP. Through signal transduction pathways, these enzymes modulate cell growth, differentiation and proliferation, i.e., virtually all aspects of cell life in one way or another depend on PK activity. Furthermore, abnormal PK activity has been related to a host of disorders, ranging from relatively non-life threatening diseases such as psoriasis to extremely virulent diseases such as glioblastoma (brain cancer). Protein kinases include two classes; protein tyrosine kinases (PTK) and serine-threonine kinases (STK).
  • PTK protein tyrosine kinases
  • STK serine-threonine kinases
  • PTK activity is their involvement with growth factor receptors which are cell-surface proteins. When bound by a growth factor ligand, growth factor receptors are converted to an active form which interacts with proteins on the inner surface of a cell membrane. This leads to phosphorylation on tyrosine residues of the receptor and other proteins and to the formation inside the cell of complexes with a variety of cytoplasmic signaling 111-03-PCT - P2338R1 - 02120.004WO1
  • HGF hepatocyte growth factor receptor tyrosine kinase
  • hHGFR human hepatocyte growth factor receptor tyrosine kinase
  • the zymogen-like form of HGF beta mutant was shown to bind Met with 14-fold lower affinity than the wild-type serine protease-like form, suggesting optimal interactions result from conformational changes upon cleavage of the single-chain form (US 2005/0037431).
  • Extensive mutagenesis of the HGF beta region corresponding to the active site and activation domain of serine proteases showed that 17 of the 38 purified two-chain HGF mutants resulted in impaired cell migration or Met phosphorylation but no loss in Met binding.
  • reduced biological activities were well correlated with reduced Met binding of corresponding mutants of HGF beta itself in assays eliminating dominant alpha-chain binding contributions.
  • PTK Protein-tyrosine kinases
  • RTK receptor tyrosine kinases
  • NRTK non-receptor tyrosine kinases
  • RTK span the plasma membrane and contain an extra-cellular domain, which binds ligand, and an intracellular portion, which possesses catalytic activity and regulatory sequences.
  • Most RTK like the hepatocyte growth factor receptor c-Met, possess a single polypeptide chain and are monomeric in the absence of ligand.
  • tyrosine autophosphorylation either stimulates the intrinsic catalytic kinase activity of the receptor or generates recruitment sites for downstream signaling 111-03-PCT- P2338R1 - 02120.004WO1
  • SH2 Src homology 2
  • PTB phosphotyrosine-binding
  • PHA-665752 is a small molecule, ATP-competitive, active-site inhibitor of the catalytic activity of c-Met, as well as phenotypes such as cell growth, cell motility, invasion, and morphology of a variety of tumor cells (Ma et al (2005) Clin. Cancer Res. 11:2312-2319; Christensen et al (2003) Cancer Res. 63:7345-7355).
  • the invention relates to quinoline compounds that are inhibitors of receptor tyrosine kinases (RTK), including c-Met.
  • RTK receptor tyrosine kinases
  • Certain hyperproliferative disorders are characterized by the overactivation of c-Met kinase function, for example by mutations or overexpression of the protein. Accordingly, the compounds of the invention are useful in the treatment of hyperproliferative disorders such as cancer.
  • one aspect of the invention provides quinoline compounds of
  • Another aspect of the invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising a quinoline compound of Formula I and a pharmaceutically acceptable carrier.
  • the pharmaceutical composition may further comprise one or more additional therapeutic agents selected from anti-proliferative agents, anti-inflammatory agents, immunomodulatory agents, neurotropic factors, agents for treating cardiovascular disease, agents for treating liver disease, 111-03-PCT - P2338R1 - 02120.004WO1
  • Another aspect of the invention provides methods of inhibiting c-Met kinase activity, comprising contacting a c-Met kinase with an effective inhibitory amount of a quinoline compound of Formula I, or a stereoisomer, geometric isomer, tautomer, solvate, metabolite, or pharmaceutically acceptable salt or prodrug thereof.
  • Another aspect of the invention provides methods of preventing or treating a disease or disorder modulated by c-Met kinases, comprising administering to a mammal in need of such treatment an effective amount of a compound of Formula I, or a stereoisomer, geometric isomer, tautomer, solvate, metabolite, or pharmaceutically acceptable salt or prodrug thereof.
  • diseases, conditions and disorders include, but are not limited to, hyperproliferative disorders (e.g., cancer, including melanoma and other cancers of the skin), neurodegeneration, cardiac hypertrophy, pain, migraine, neurotraumatic diseases, stroke, diabetes, hepatomegaly, cardiovascular disease, Alzheimer's disease, cystic fibrosis, viral diseases, autoimmune diseases, atherosclerosis, restenosis, psoriasis, allergic disorders, inflammation, neurological disorders, hormone-related diseases, conditions associated with organ transplantation, immunodeficiency disorders, destructive bone disorders, proliferative disorders, infectious diseases, conditions associated with cell death, thrombin-induced platelet aggregation, chronic myelogenous leukemia (CML), liver disease, pathologic immune conditions involving T cell activation, and CNS disorders.
  • hyperproliferative disorders e.g., cancer, including melanoma and other cancers of the skin
  • neurodegeneration e.g., cancer, including melanoma and other cancers of the skin
  • Another aspect of the invention provides methods of preventing or treating a hyperproliferative disorder, comprising administering to a mammal in need of such treatment an effective amount of a compound of Formula I, or a stereoisomer, geometric isomer, tautomer, solvate, metabolite, or pharmaceutically acceptable salt or prodrug thereof, alone or in combination with one or more additional compounds having anti-hyperproliferative properties.
  • the present invention provides a method of using a compound of this invention to treat a disease or condition modulated by c-Met in a mammal.
  • An additional aspect of the invention is the use of a compound of this invention in the preparation of a medicament for the treatment or prevention of a disease or condition modulated by c-Met in a mammal.
  • kits comprising a compound of Formula
  • Another aspect of the invention includes methods of preparing, methods of separating, and methods of purifying compounds of Formula I.
  • alkyl refers to a saturated linear or branched-chain monovalent hydrocarbon radical of one to twelve carbon atoms, wherein the alkyl radical may be optionally substituted independently with one or more substituents described below.
  • alkyl groups include, but are not limited to, methyl (Me, -CH 3 ), ethyl (Et, -CH 2 CH 3 ), 1 -propyl (n- Pr, n-propyl, -CH 2 CH 2 CH 3 ), 2-propyl (i-Pr, i-propyl, -CH(CH 3 ) 2 ), 1 -butyl (n-Bu, n-butyl, -CH 2 CH 2 CH 2 CH 3 ), 2-methyl-l -propyl (i-Bu, i-butyl, -CH 2 CH(CH 3 ) 2 ), 2-butyl (s-Bu, s-butyl, -CH(CH 3 )CH 2 CH 3 ), 2-methyl-2-propyl (t-Bu, t-butyl, -C(CH 3 ) 3 ), 1-pentyl (n-pentyl, -CH 2 CH 2 CH 2 CH 2 CH 3 ), 2-pentyl (
  • alkynyl refers to a linear or branched monovalent hydrocarbon radical of two to twelve carbon atoms with at least one site of unsaturation, i.e., a carbon-carbon, sp triple bond, wherein the alkynyl radical may be optionally substituted independently with one or more substituents described herein. Examples include, but are not limited to, ethynyl (-C ⁇ CH), propynyl (propargyl, -CH 2 C ⁇ CH), and the like.
  • carrier refers to a monovalent or multivalent non-aromatic, saturated or partially unsaturated ring having 3 to 12 carbon atoms as a monocyclic ring or 7 to 12 carbon atoms as a bicyclic ring.
  • Bicyclic carbocycles having 7 to 12 atoms can be arranged, for example, as a bicyclo [4,5], [5,5], [5,6] or [6,6] system, and bicyclic carbocycles having 9 or 10 ring atoms can be arranged as a bicyclo [5,6] or [6,6] system, or as bridged systems such as bicyclo[2.2.1]heptane, bicyclo[2.2.2]octane and bicyclo[3.2.2]nonane.
  • monocyclic carbocycles include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, 1-cyclopent-l-enyl, l-cyclopent-2-enyl, l-cyclopent-3-enyl, cyclohexyl, 1-cyclohex-l-enyl, l-cyclohex-2-enyl, l-cyclohex-3-enyl, cyclohexadienyl, cycloheptyl, cyclooctyl, cyclononyl, cyclodecyl, cycloundecyl, cyclododecyl, and the like.
  • Aryl means a monovalent or multivalent aromatic hydrocarbon radical of 6-20 carbon atoms derived by the removal of one or more hydrogen atoms from a carbon atom of a parent aromatic ring system. Some aryl groups are represented in the exemplary structures as "Ar”. Aryl includes bicyclic radicals comprising an aromatic ring fused to a saturated, partially unsaturated ring, or aromatic carbocyclic or heterocyclic ring.
  • Typical aryl groups include, but are not limited to, radicals derived from benzene, substituted benzenes, naphthalene, anthracene, biphenyl, indenyl, indanyl, 1,2-dihydronapthalene, 1,2,3,4-tetrahydronapthyl, and the like.
  • Examples of aryl fused to a heterocyclic ring include, but are not limited to, the structure: 111-03-PCT- P2338R1 - 02120.004WO1
  • n 0, 1 or 2.
  • aryl fused to a carbocylic ring include, but are not limited to, the structures:
  • R 5 is as defined herein.
  • heterocycle refers to a saturated or a partially unsaturated (i.e., having one or more double and/or triple bonds within the ring) carbocyclic, monovalent or multivalent radical of 3 to 20 carbon atoms, and in which at least one ring atom is a heteroatom selected from nitrogen, oxygen and sulfur, the remaining ring atoms being C, where one or more ring atoms is optionally substituted independently with one or more substituents described below.
  • a heterocycle may be a monocycle having 3 to 7 ring members (2 to 6 carbon atoms and 1 to 3 heteroatoms selected from N, O, P, and S, wherein the S is optionally substituted with one or more oxo to provide the group SO or SO 2 ) or a bicycle having 7 to 10 ring members (4 to 9 carbon atoms and 1 to 3 heteroatoms selected from N, O, P, and S, wherein the S is optionally substituted with one or more oxo to provide the group SO or SO 2 ), for example: a bicyclo [4,5], [5,5], [5,6], or [6,6] system. Heterocycles are described in Paquette, Leo A.; "Principles of Modern Heterocyclic Chemistry" (W.A.
  • heterocyclyl may be a carbon radical or heteroatom radical.
  • heterocycle includes heterocycloalkoxy.
  • Heterocyclyl also includes radicals where heterocycle radicals are fused with a saturated, partially unsaturated ring, or aromatic carbocyclic or heterocyclic ring. Examples of heterocyclic rings include, but are not limited to, pyrrolidinyl, 111-03-PCT - P2338R1 - 02120.004WO1
  • the heterocycle groups herein are optionally substituted independently with one or more substituents described herein.
  • heteroaryl refers to a monovalent or multivalent aromatic radical of 5-
  • heteroaryl groups include fused ring systems (at least one of which is aromatic) of 1 to 20 carbon atoms, and containing one or more heteroatoms independently selected from nitrogen, oxygen, and sulfur.
  • heteroaryl groups are pyridinyl (including, for example, 2-hydroxypyridinyl), imidazolyl, imidazopyridinyl, pyrimidinyl (including, for example, 4-hydroxypyrimidinyl), pyrazolyl, triazolyl, pyrazinyl, tetrazolyl, furyl, thienyl, isoxazolyl, thiazolyl, oxazolyl, isothiazolyl, pyrrolyl, quinolinyl, isoquinolinyl, indolyl, benzimidazolyl, benzofuranyl, cinnolinyl, indazolyl, indolizinyl, phthalazinyl, pyridaziny
  • R 5 and R 10 are as defined herein.
  • the heterocycle or heteroaryl groups may be C-attached or N-attached where such is possible.
  • carbon bonded heterocycles or heteroaryls are bonded at position 2, 3, 4, 5, or 6 of a pyridine, position 3, 4, 5, or 6 of a pyridazine, position 2, 4, 5, or 6 of a pyrimidine, position 2, 3, 5, or 6 of a pyrazine, position 2, 3, 4, or 5 of a furan, tetrahydrofuran, thiofuran, thiophene, pyrrole or tetrahydropyrrole, position 2, 4, or 5 of an oxazole, imidazole or thiazole, position 3, 4, or 5 of an isoxazole, pyrazole, or 111-03-PCT- P2338R1 - 02120.004WO1
  • nitrogen bonded heterocycles or heteroaryls are bonded at position 1 of an aziridine, azetidine, pyrrole, pyrrolidine, 2-pyrroline, 3- pyrroline, imidazole, imidazolidine, 2-imidazoline, 3-imidazoline, pyrazole, pyrazoline, 2- pyrazoline, 3-pyrazoline, piperidine, piperazine, indole, indoline, lH-indazole, position 2 of a isoindole, or isoindoline, position 4 of a morpholine, and position 9 of a carbazole, or ⁇ -carboline.
  • Substituted alkyl mean alkyl, alkenyl, alkynyl, aryl, heteroaryl, heterocyclyl and cycloalkyl, respectively, in which one or more hydrogen atoms are each independently replaced with a substituent.
  • Substituents may also be combinations of alkyl, alkenyl, alkynyl, carbocycle, aryl, and heteroaryl radicals, such as cyclopropylmethyl, cyclohexylethyl, benzyl, and N-ethylmorpholino, and substituted forms thereof.
  • beneficial or desired clinical results include, but are not limited to, alleviation of symptoms, diminishment of extent of disease, stabilized (i.e., not worsening) state of disease, delay or slowing of disease progression, amelioration or palliation of the disease state, and remission (whether partial or total), whether detectable or undetectable.
  • Treatment can also mean prolonging survival as compared to expected survival if not receiving treatment.
  • Those in need of treatment include those already with the condition or disorder as well as those prone to have the condition or disorder or those in which the condition or disorder is to be prevented.
  • the phrase "therapeutically effective amount” means an amount of a compound of the present invention that (i) treats or prevents the particular disease, condition, or disorder, (ii) attenuates, ameliorates, or eliminates one or more symptoms of the particular disease, condition, 111-03-PCT - P2338R1 - 02120.004WO1
  • the therapeutically effective amount of the drug may reduce the number of cancer cells; reduce the tumor size; inhibit (i.e., slow to some extent and preferably stop) cancer cell infiltration into peripheral organs; inhibit (i.e., slow to some extent and preferably stop) tumor metastasis; inhibit, to some extent, tumor growth; and/or relieve to some extent one or more of the symptoms associated with the cancer.
  • the drug may prevent growth and/or kill existing cancer cells, it may be cytostatic and/or cytotoxic.
  • cancer therapy efficacy can be measured, for example, by assessing the time to disease progression (TTP) and/or determining the response rate (RR).
  • TTP time to disease progression
  • RR response rate
  • cancer and “cancerous” refer to or describe the physiological condition in mammals that is typically characterized by unregulated cell growth.
  • a “tumor” comprises one or more cancerous cells. Examples of cancer include, but are not limited to, carcinoma, lymphoma, blastoma, sarcoma, and leukemia or lymphoid malignancies.
  • squamous cell cancer e.g., epithelial squamous cell cancer
  • lung cancer including small- cell lung cancer, non-small cell lung cancer ("NSCLC"), adenocarcinoma of the lung and squamous carcinoma of the lung, cancer of the peritoneum, hepatocellular cancer, gastric or stomach cancer including gastrointestinal cancer, pancreatic cancer, glioblastoma, cervical cancer, ovarian cancer, liver cancer, bladder cancer, hepatoma, breast cancer, colon cancer, rectal cancer, colorectal cancer, endometrial or uterine carcinoma, salivary gland carcinoma, kidney or renal cancer, prostate cancer, vulval cancer, thyroid cancer, hepatic carcinoma, anal carcinoma, penile carcinoma, as well as head and neck cancer.
  • NSCLC non-small cell lung cancer
  • adenocarcinoma of the lung and squamous carcinoma of the lung cancer of the peritoneum, hepatocellular cancer, gastric or stomach cancer including gastrointestinal cancer
  • a "chemotherapeutic agent” is a chemical compound useful in the treatment of cancer.
  • chemotherapeutic agents include Erlotinib (TARCEVA®, Genentech/OSI Pharm.), Bortezomib (VELCADE®, Millennium Pharm.), Fulvestrant (FASLODEX®, AstraZeneca), Sutent (SUl 1248, Pfizer), Letrozole (FEMARA®, Novartis), Imatinib mesylate (GLEEVEC®, Novartis), PTK787/ZK 222584 (Novartis), Oxaliplatin (Eloxatin®, Sanofi), 5-FU (5-fluorouracil), Leucovorin, Rapamycin (Sirolimus, RAPAMUNE®, Wyeth), lapatinib (TYKERB®, GlaxoSmithKline PLC), Lonafarnib (SCH 66336), Sorafenib (BAY43-9006, Bayer Labs), and
  • dynemicin including dynemicin A; bisphosphonates, such as clodronate; an esperamicin; as well as neocarzinostatin chromophore and related chromoprotein enediyne antibiotic chromophores), aclacinomysins, actinomycin, authramycin, azaserine, bleomycins, cactinomycin, carabicin, carminomycin, carzinophilin, chromomycinis, dactinomycin, daunorubicin, detorubicin, 6-diazo-5-oxo-L-norleucine, ADRIAMYCIN® (doxorubicin), morpholino-doxorubicin, cyanomorpholino-doxorubicin, 2-pyrrolino-doxorubicin and deoxydoxorubicin), epirubicin,
  • chemotherapeutic agent also included in the definition of "chemotherapeutic agent” are: (i) anti -hormonal agents that act to regulate or inhibit hormone action on tumors such as anti-estrogens and selective estrogen receptor modulators (SERMs), including, for example, tamoxifen (including NOLVADEX®; tamoxifen citrate), raloxifene, droloxifene, 4-hydroxytamoxifen, trioxifene, keoxifene, LYl 17018, onapristone, and FARESTON® (toremifine citrate); (ii) aromatase inhibitors that inhibit the enzyme aromatase, which regulates estrogen production in the adrenal glands, such as, for example, 4(5)-imidazoles, aminoglutethimide, MEGASE® (megestrol acetate), AROMASIN® (exemestane; Pfizer), formestanie, fadrozole, RIVISOR® (vorozole), F
  • prodrug refers to a precursor or derivative form of a compound of the invention that is less cytotoxic to cells compared to the parent compound or drug and is capable of being enzymatically or hydrolytically activated or converted into the more active parent form. See, e.g., Wilman, "Prodrugs in Cancer Chemotherapy” Biochemical Society Transactions, 14, pp. 375-382, 615th Meeting Harbor (1986) and Stella et al, "Prodrugs: A Chemical Approach to Targeted Drug Delivery,” Directed Drug Delivery, 111-03-PCT - P2338R1 - 02120.004WO1
  • the prodrugs of this invention include, but are not limited to, phosphate-containing prodrugs, thiophosphate-containing prodrugs, sulfate-containing prodrugs, peptide-containing prodrugs, D-amino acid-modified prodrugs, glycosylated prodrugs, ⁇ -lactam-containing prodrugs, optionally substituted phenoxyacetamide- containing prodrugs, optionally substituted phenylacetamide-containing prodrugs, 5- fluorocytosine and other 5-fluorouridine prodrugs which can be converted into the more active cytotoxic free drug.
  • cytotoxic drugs that can be derivatized into a prodrug form for use in this invention include, but are not limited to, compounds of the invention and chemotherapeutic agents such as described above.
  • a "metabolite” is a product produced through metabolism in the body of a specified compound or salt thereof. Metabolites of a compound may be identified using routine techniques known in the art and their activities determined using tests such as those described herein. Such products may result for example from the oxidation, reduction, hydrolysis, amidation, deamidation, esterification, deesterification, enzymatic cleavage, and the like, of the administered compound. Accordingly, the invention includes metabolites of compounds of the invention, including compounds produced by a process comprising contacting a compound of this invention with a mammal for a period of time sufficient to yield a metabolic product thereof.
  • a “liposome” is a small vesicle composed of various types of lipids, phospholipids and/or surfactant which is useful for delivery of a drug (such as the c-Met inhibitors disclosed herein and, optionally, a chemotherapeutic agent) to a mammal.
  • a drug such as the c-Met inhibitors disclosed herein and, optionally, a chemotherapeutic agent
  • the components of the liposome are commonly arranged in a bilayer formation, similar to the lipid arrangement of biological membranes.
  • package insert is used to refer to instructions customarily included in commercial packages of therapeutic products, that contain information about the indications, usage, dosage, administration, contraindications and/or warnings concerning the use of such therapeutic products.
  • chiral refers to molecules which have the property of non-superimposability of the mirror image partner, while the term “achiral” refers to molecules which are superimposable on their mirror image partner.
  • stereoisomers refers to compounds which have identical chemical constitution, but differ with regard to the arrangement of the atoms or groups in space.
  • “Diastereomer” refers to a stereoisomer with two or more centers of chirality and whose molecules are not mirror images of one another. Diastereomers have different physical properties, e.g. melting points, boiling points, spectral properties, and reactivities. Mixtures of 111-03-PCT - P2338R1 - 02120.004WO1
  • 15 diastereomers may separate under high resolution analytical procedures such as electrophoresis and chromatography.
  • Enantiomers refer to two stereoisomers of a compound which are non- superimposable mirror images of one another.
  • the compounds of the invention may contain asymmetric or chiral centers, and therefore exist in different stereoisomeric forms. It is intended that all stereoisomeric forms of the compounds of the invention, including but not limited to, diastereomers, enantiomers and atropisomers, as well as mixtures thereof such as racemic mixtures, form part of the present invention.
  • Many organic compounds exist in optically active forms, i.e., they have the ability to rotate the plane of plane-polarized light.
  • the prefixes D and L, or R and S are used to denote the absolute configuration of the molecule about its chiral center(s).
  • the prefixes d and 1 or (+) and (-) are employed to designate the sign of rotation of plane-polarized light by the compound, with (-) or 1 meaning that the compound is levorotatory.
  • a compound prefixed with (+) or d is dextrorotatory.
  • these stereoisomers are identical except that they are mirror images of one another.
  • a specific stereoisomer may also be referred to as an enantiomer, and a mixture of such isomers is often called an enantiomeric mixture.
  • a 50:50 mixture of enantiomers is referred to as a racemic mixture or a racemate, which may occur where there has been no stereoselection or stereospecificity in a chemical reaction or process.
  • the terms “racemic mixture” and “racemate” refer to an equimolar mixture of two enantiomeric species, devoid of optical activity.
  • the term “tautomer” or “tautomeric form” refers to structural isomers of different energies which are interconvertible via a low energy barrier.
  • proton tautomers also known as prototropic tautomers
  • Valence tautomers include interconversions by reorganization of some of the bonding electrons.
  • a "salt,” as used herein, refers to organic or inorganic salts of a compound of the invention.
  • Exemplary salts include, but are not limited, to sulfate, citrate, acetate, oxalate, chloride, bromide, iodide, nitrate, bisulfate, phosphate, acid phosphate, isonicotinate, lactate, salicylate, acid citrate, tartrate, oleate, tannate, pantothenate, bitartrate, ascorbate, succinate, maleate, gentisinate, fumarate, gluconate, glucuronate, saccharate, formate, benzoate, glutamate, methanesulfonate, ethanesulfonate, benzenesulfonate, /?-toluenesulfonate, and pamoate (i.e., 1,1'- 111-03-PCT - P2338R1
  • a salt may involve the inclusion of another molecule such as an acetate ion, a succinate ion or other counter ion.
  • the counter ion may be any organic or inorganic moiety that stabilizes the charge on the parent compound.
  • a salt may have more than one charged atom in its structure. Instances where multiple charged atoms are part of the salt can have multiple counter ions. Hence, a salt can have one or more charged atoms and/or one or more counter ion.
  • the desired salt may be prepared by any suitable method available in the art, for example, treatment of the free base with an inorganic acid, such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid and the like, or with an organic acid, such as acetic acid, maleic acid, succinic acid, mandelic acid, fumaric acid, malonic acid, pyruvic acid, oxalic acid, glycolic acid, salicylic acid, a pyranosidyl acid, such as glucuronic acid or galacturonic acid, an alpha hydroxy acid, such as citric acid or tartaric acid, an amino acid, such as aspartic acid or glutamic acid, an aromatic acid, such as benzoic acid or cinnamic acid, a sulfonic acid, such as p-toluenesulfonic acid or ethanesulfonic acid, or the like.
  • an inorganic acid such as hydrochloric acid, hydrobro
  • the desired salt may be prepared by any suitable method, for example, treatment of the free acid with an inorganic or organic base, such as an amine (primary, secondary or tertiary), an alkali metal hydroxide or alkaline earth metal hydroxide, or the like.
  • suitable salts include, but are not limited to, organic salts derived from amino acids, such as glycine and arginine, ammonia, primary, secondary, and tertiary amines, and cyclic amines, such as piperidine, morpholine and piperazine, and inorganic salts derived from sodium, calcium, potassium, magnesium, manganese, iron, copper, zinc, aluminum and lithium.
  • the salt is a pharmaceutically acceptable salt.
  • pharmaceutically acceptable indicates that the substance or composition must be compatible chemically and/or toxicologically, with the other ingredients comprising a formulation, and/or the mammal being treated therewith.
  • the compounds of Formula I also include salts of such compounds which are not necessarily pharmaceutically acceptable salts, and which may be useful as intermediates for preparing and/or purifying compounds of Formula I and/or for separating enantiomers of compounds of Formula I.
  • a “solvate” refers to an association or complex of one or more solvent molecules and a compound of the invention.
  • solvents that form solvates include, but are not 111-03-PCT - P2338R1 - 02120.004WO1
  • protecting group refers to a substituent that is commonly employed to block or protect a particular functionality while reacting other functional groups on the compound.
  • an "amino-protecting group” is a substituent attached to an amino group that blocks or protects the amino functionality in the compound.
  • Suitable amino-protecting groups include acetyl, trifluoroacetyl, t-butoxycarbonyl (BOC), benzyloxycarbonyl (CBZ) and 9- fluorenylmethylenoxycarbonyl (Fmoc).
  • a "hydroxy-protecting group” refers to a substituent of a hydroxy group that blocks or protects the hydroxy functionality.
  • Suitable protecting groups include acetyl and silyl.
  • a “carboxy-protecting group” refers to a substituent of the carboxy group that blocks or protects the carboxy functionality.
  • Common carboxy-protecting groups include -CH 2 CH 2 SO 2 Ph, cyanoethyl, 2-(trimethylsilyl)ethyl, 2- (trimethylsilyl)ethoxymethyl, 2-(p-toluenesulfonyl) ethyl, 2-(p-nitrophenylsulfenyl)ethyl, 2- (diphenylphosphino)-ethyl, nitroethyl and the like.
  • protecting groups and their use see T. W. Greene, Protective Groups in Organic Synthesis, John Wiley & Sons, New York, 1991.
  • compound of this invention and “compounds of the present invention” and “compounds of Formula I” include compounds of Formula I and stereoisomers, geometric isomers, tautomers, solvates, metabolites, salts and pharmaceutically acceptable prodrugs thereof.
  • mammal includes, but is not limited to, humans, dogs, cats, horses, cows, pigs, and sheep, and poultry.
  • the present invention provides quinoline compounds, and pharmaceutical formulations thereof, that are potentially useful in the treatment of diseases, conditions and/or disorders modulated by c-Met. More specifically, the present invention provides compounds of Formula I:
  • R 1 , R 2 and R 4 are independently selected from H, F, Cl, Br, I, CN,
  • R 13 is H, Ci-C 6 alkyl, -(CR 14 R 15 ) n -cycloalkyl, -(CR 14 R 15 ) n -heterocyclyl,
  • R 14 is null and R 10 and R 15 together with the atoms to which they are attached form a 5-6 membered heteroaryl ring,
  • R 12 and R 14 together with the atoms to which they are attached form a saturated or partially unsaturated C 2 -Cj 2 heteorcyclic ring;
  • R a and R b are independently H, C J -C J2 alkyl, C 2 -C 8 alkenyl, C 2 -C 8 alkynyl,
  • Ci-C 12 alkyl C 2 -Cg alkenyl, C 2 -Cg alkynyl, C 6 -C 2O aryl, or C 1 -C 2 0 heteroaryl; [0077] Y, Y 1 and Y 2 are independently O or S;
  • t is 1, 2, 3, 4, 5 or 6;
  • n and m are independently 0, 1, 2, 3, 4, 5 or 6.
  • R 1 and R 2 is -OR 10 where R 10 is Ci-Ci 2 alkyl.
  • R 1 and R 2 are methoxy.
  • R 10 is Ci-Ci 2 alkyl substituted with NR a R b or C 2 -C 2O heterocyclyl, wherein said heterocyclyl is optionally substituted with C 1 -C 6 alkyl, CH 2 OH or SO 2 Me.
  • Exemplary embodiments include the structures:
  • R 1 is methoxy and R 2 is selected from
  • R 1 and R 2 is -OR 10 wherein R 10 is C]-Ci 2 alkyl substituted with Ci-C 20 heteroaryl, wherein said heteroaryl is optionally substituted with C]-C 6 alkyl.
  • exemplary embodiments include the structures: 111-03-PCT - P2338R1 - 02120.004WO1
  • R 1 and R 2 are independently selected from
  • R 1 and R 2 are independently selected from optionally substituted aryl or heteroaryl, including the exemplary structures:
  • R ! and R 2 is independently alkyl optionally substituted with one or more groups independently selected from OR 10 , NR 10 R 11 , heterocyclyl and heteroaryl. Examples include, but are not limited to, methyl, -CH 2 OH, -CH 2 CH 2 OH,
  • R 1 and R 2 are independently -OR , including the exemplary structure:
  • each R 4 is H.
  • L-R 5 is (C 3 -Ci 2 carbocyclyO-R 5 , including the exemplary structures:
  • L-R 5 is (C 2 -C 20 heterocyclyl)-R 5 wherein said heterocyclyl is optionally substituted, including the exemplary structures: 111-03-PCT- P2338R1 - 02120.004WO1
  • L-R 5 is (C 6 -C 2 o aryl)-R 5 wherein said aryl is optionally substituted, including the exemplary structures:
  • L-R 5 is (C 6 -C 2O aryl)-R 5 include the structures:
  • L-R 5 is (C 6 -C 20 aryl)-R 5
  • L-R 5 includes the structures: 111-03-PCT- P2338R1 - 02120.004WO1
  • L-R 5 is (C 6 -C 2 O aryl)-R
  • R 5 is (C 6 -C 2 O aryl)-R
  • L-R 5 is (C 6 -C 20 aryl)-R 5 include the structures:
  • L-R 5 is (Ci-C 20 heteroaryl)-R 5 .
  • Exemplary embodiments include the structures:
  • L-R 5 is (C]-C 2O heteroaryl)-R 5 also include the structures:
  • R 13 is
  • -(CR 14 R 15 ) n -cycloalkyl -(CR 14 R 15 ) n -aryl, -(CR 14 R 15 ) n -O-(CR 14 R 15 ) m -aryl, or -(CR 14 R 15 V heterocyclyl-(CR 14 R 15 ) r aryl, wherein said heterocyclyl portion is optionally substituted with SO 2 R 0 or C]-Ci 2 alkyl.
  • Exemplary embodiments include the structures: 111-03-PCT - P2338R1 - 02120.004WO1
  • R 10 is H or Ci-Ci 2 alkyl
  • R 13 is H, Q-C 6 alkyl, -(CR 14 R 15 ) n -cycloalkyl, or -(CR 14 R 15 ) n -aryl, wherein said alkyl, cycloalkyl, and aryl portions are optionally substituted with F or 0R a .
  • Exemplary embdodiments of R 5 include the structures:
  • R 5 is -NR 10 R 13 .
  • R 10 is H or
  • Ci-Ci 2 alkyl, and R 13 is -(CR 14 R 15 ) n -heterocyclyl or -(CR 14 R 15 ) n -heteroaryl, wherein said heterocyclyl and heteroaryl are optionally substituted with OR a , SO 2 R 0 or O-(CH 2 )-aryl.
  • Exemplary embdodiments of R 5 include the structures:
  • R 15 and R 10 optionally together with the atoms to which they are attached form a 5-6 membered 111-03-PCT - P2338R1 - 02120.004WO1
  • R 5 include the structures:
  • exemplary embodiments of R 5 include the structures: 111-03-PCT - P2338R1 - 02120.004WO1
  • R 11 is aryl or a Ci-Ci 2 alkyl substituted with aryl, wherein said aryl portions are optionally substituted.
  • Particular embodiments include the structures:
  • R 12 and R 14 together with the atoms to which they are attached form a 5-6 membered heterocyclic ring.
  • exemplary embodiments include, but are not limited to 111-03-PCT - P2338R1 - 02120.004WO1
  • a particular example includes
  • R 13 is C 1 -
  • Exemplary embodiments of R 5 include the structures:
  • R 5 is -NR 12 SO 2 R 10 , including where R 10 is alkyl or optionally substituted aryl.
  • exemplary embodiments include the structures: 111-03-PCT- P2338R1 - 02120.004WO1
  • R 5 is an optionally substituted carbocyclyl, including the exemplary structures:
  • R 5 is an optionallysubstituted heterocyclyl, including the exemplary structures:
  • R 5 is an optionally substituted aryl, including the exemplary structures: 111-03-PCT - P2338R1 - 02120.004WO1
  • R 5 is an optionally substituted heteroaryl, including the exemplary structures:
  • R ⁇ is H, Ci-C 12 alkyl, C 3 -Ci 2 cycloalkyl, C 6 -C 20 aryl, or Ci-C 20 heteroaryl, and Ir 21 1 and
  • R are independently selected from H or Ci-Ci 2 alkyl, wherein said alkyl, cycloalkyl, aryl, heteroaryl are optionally substituted with one or more groups independently selected from F, Cl, Br, I and Ci-Ci 2 alkyl; and where the wavy line indicates the point of attachment to L.
  • R 20 is H. 111-03-PCT - P2338R1 - 02120.004WO1
  • R 5 is an optionally substituted heteroaryl, including the exemplary structures:
  • R 5 is an optionally substituted heteroaryl, including the exemplary structures: 111-03-PCT- P2338R1 - 02120.004WO1
  • the quinoline compounds of the invention may contain asymmetric or chiral centers, and therefore exist in different stereoisomeric forms. It is intended that all stereoisomeric forms of the compounds of the invention, including but not limited to, diastereomers, enantiomers and atropisomers, as well as mixtures thereof such as racemic mixtures, form part of the present invention.
  • the present invention embraces all geometric and positional isomers.
  • a quinoline compound of the present invention incorporates a double bond or a fused ring
  • the cis- and trans-forms, as well as mixtures thereof are embraced within the scope of the invention.
  • Both the single positional isomers and mixture of positional isomers, e.g., resulting from the N-oxidation of the pyrimidine and pyrazine rings, are also within the scope of the present invention.
  • stereochemistry of any particular chiral atom is not specified, then all stereoisomers are contemplated and included as the compounds of the invention. Where stereochemistry is specified by a solid wedge or dashed line representing a particular configuration, then that stereoisomer is so specified and defined.
  • the compounds of the present invention may exist in unsolvated as well as solvated forms with pharmaceutically acceptable solvents such as water, ethanol, and the like, and it is intended that the invention embrace both solvated and unsolvated forms.
  • tautomer or tautomeric form refers to structural isomers of different energies which are interconvertible via a low energy barrier.
  • proton tautomers also known as prototropic tautomers
  • Valence tautomers include interconversions by reorganization of some of the bonding electrons.
  • the present invention also embraces isotopically-labeled compounds of the present invention which are identical to those recited herein, but for the fact that one or more atoms are replaced by an atom having an atomic mass or mass number different from the atomic 111-03-PCT - P2338R1 - 02120.004WO1
  • isotopes of any particular atom or element as specified are contemplated within the scope of the compounds of the invention, and their uses.
  • Exemplary isotopes that can be incorporated into compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine, chlorine and iodine, such as 2 H, 3 H, 11 C, 13 C, 14 C, 13 N, 15 N, 15 O, 17 O, 18 0, 32 P, 33 P, 35 S, 18 F, 36 Cl, 123 I and 125 I.
  • Certain isotopically- labeled compounds of the present invention e.g., those labeled with 3 H and 14 C) are useful in compound and/or substrate tissue distribution assays.
  • Tritiated ( 3 H) and carbon- 14 ( 14 C) isotopes are useful for their ease of preparation and detectability. Further, substitution with heavier isotopes such as deuterium (i.e., 2 H) may afford certain therapeutic advantages resulting from greater metabolic stability (e.g., increased in vivo half-life or reduced dosage requirements) and hence may be preferred in some circumstances.
  • Positron emitting isotopes such as 15 O, 13 N, 11 C and 18 F are useful for positron emission tomography (PET) studies to examine substrate receptor occupancy.
  • Isotopically labeled compounds of the present invention can generally be prepared by following procedures analogous to those disclosed in the Schemes and/or in the Examples herein below, by substituting an isotopically labeled reagent for a non-isotopically labeled reagent.
  • Quinoline compounds of Formula I of the present invention may be synthesized by synthetic routes that include processes analogous to those well-known in the chemical arts, particularly in light of the description contained herein.
  • the starting materials are generally available from commercial sources such as Aldrich Chemicals (Milwaukee, WI) or are readily prepared using methods well known to those skilled in the art (e.g., prepared by methods generally described in Louis F. Fieser and Mary Fieser, Reagents for Organic Synthesis, v. 1-19, Wiley, N.Y. (1967-1999 ed.), or Beilsteins Handbuch der organischen Chemie, 4, Aufl. ed. Springer- Verlag, Berlin, including supplements (also available via the Beilstein online database).
  • compounds of Formula I may be readily prepared using procedures well-known to prepare quinoline compounds; and other heterocycles, which are described in: Comprehensive Heterocyclic Chemistry, Editors Katritzky and Rees, Pergamon Press, 1984; Klemm et al (1970) J. Hetero. Chem. 7(2):373-379; Klemm et al (1974) J. Hetero. Chem. 11(3): 355-361; Klemm et al (1976) J. Hetero. Chem. 13:273-275; Klemm et al (1985) J. Hetero. Chem. 22(5):1395-1396; Bisagni et al (1974) Bull. Soc. Chim. Fr.
  • Compounds of Formula I may be prepared singly or as compound libraries comprising at least 2, for example 5 to 1,000 compounds, or 10 to 100 compounds.
  • Libraries of compounds of Formula I may be prepared by a combinatorial 'split and mix' approach or by multiple parallel syntheses using either solution phase or solid phase chemistry, by procedures known to those skilled in the art.
  • a compound library comprising at least 2 compounds, or pharmaceutically acceptable salts thereof.
  • Schemes 1-20 show general methods for preparing the compounds of the present invention as well as key intermediates. For a more detailed description of the individual reaction steps, see the Examples section below. Those skilled in the art will appreciate that other synthetic routes may be used to synthesize the inventive compounds. Although specific starting materials and reagents are depicted in the Schemes and discussed below, other starting materials and reagents can be easily substituted to provide a variety of derivatives and/or reaction conditions. In addition, many of the compounds prepared by the methods described below can be further modified in light of this disclosure using conventional chemistry well known to those skilled in the art.
  • Suitable amino-protecting groups include acetyl, trifluoroacetyl, t- butoxycarbonyl (BOC), benzyloxycarbonyl (CBz) and 9-fluorenylmethyleneoxycarbonyl (Fmoc).
  • NH-Pg amino-protecting groups
  • BOC t- butoxycarbonyl
  • CBz benzyloxycarbonyl
  • Fmoc 9-fluorenylmethyleneoxycarbonyl
  • Scheme 1 shows a general method for the synthesis of intermediate compound 4, which is useful for the synthesis of compounds of Formula I. Syntheses of 4-phenoxy-6,7- dialkoxyquinolines have been previously reported in US 2004/0242603; US 2005/0002326; WO 2005/030140; J. Med. Chem. (2005) 48:1359-1366. As shown in Scheme 1, reaction of a 4- hydroxy-6,7-dialkoxyquinoline 1 with a variably substituted p-halonitroarene or heteroarene wherein X is F or Cl and Y is N or CH using an appropriate base (e.g. Cs 2 CO 3 , NaH, KOt-Bu, or the like) provides intermediate 2.
  • an appropriate base e.g. Cs 2 CO 3 , NaH, KOt-Bu, or the like
  • Scheme 2 shows a general synthetic route for the synthesis of compound 9, which is useful for the synthesis of compounds of Formula I.
  • a 4-chloro-6,7-dialkoxyquinoline, such as compound 6, can be prepared by chlorination of the corresponding hydroxyquinoline (compound 1, Scheme 1) typically using POCl 3 , MeSO 2 Cl and the like.
  • Compound 7 can then be reacted under basic conditions, typically DMAP in bromobenzene or Cs 2 C ⁇ 3 in DMF, with a functionalized phenol 8 (general schemes for synthesis of preferred functionalized phenols are shown below) to give rise to compound 9.
  • a functionalized phenol 8 generally schemes for synthesis of preferred functionalized phenols are shown below
  • Compound 9 can optionally be further manipulated depending on the phenol functionalization.
  • PG exchange for R 11 can be omitted from the sequence and thus provide intermediate 9 with the 6,7-alkoxy substituents originally contained in intermediate 6.
  • Scheme 3 shows a route for the preparation of phenol compound 14.
  • Scheme 4 shows a route for the preparation of the 1 -substituted pyrimidinone intermediate 20 (wherein R 10 is independently selected from H, alkyl, aryl and heteroaryl).
  • 5- bromo-2,4-dichloropyrimidine 15 is hydrolyzed with NaOH to give 5-bromo-2-chloropyrimidin- 4(3H)-one 16 as described in EP1506967 Al.
  • Alkylation of compound 16 to provide the 1- substituted pyrimidinone can be accomplished with an alkylation agent (e.g. iodomethane, or the like) mediated by an appropriate base (e.g. sodium alkoxides, lithium or sodium hydride, or the like) providing a mixture of isomers 17 and 18.
  • an alkylation agent e.g. iodomethane, or the like
  • an appropriate base e.g. sodium alkoxides, lithium or sodium hydride, or the like
  • Isomers 17 and 18 can be separated using purification techniques known to those skilled in the art (e.g. flash chromatography, reverse phase HPLC, or the like).
  • Compound 18 is reacted with the appropriate zinc reagent and palladium catalyst to give 2-substituted intermediate 19.
  • Suzuki coupling of compound 19 to an appropriate boronic acid followed by final deprotection of the phenol gives compound 20, which can be reacted with the appropriate core intermediate 7 as in Scheme 2 to provide compound 9.
  • Scheme 5 shows a method for preparing phenol intermediate 27 (wherein R 10 is independently selected from H, alkyl, aryl and heteroaryl).
  • Nucleophilic substitution of 2-chloro- 4-methoxypyrimidine 21 with a compound of the formula HY-R 10 , (wherein Y is O, N or S) can be accomplished in an appropriate solvent such as n-butanol, at refluxing temperature to give intermediate 22.
  • Deprotection of the methoxypyrimidine with HBr in acetic acid provides 2- substituted pyrimidinone 23.
  • Alkylation of 23 to provide the 1 -substituted pyrimidinone can be accomplished with an alkylation agent (e.g.
  • iodomethane or the like
  • an appropriate base e.g. sodium alkoxides, lithium or sodium hydride, or the like
  • Isomers 24 and 25 can be separated using purification techniques known to those skilled in the art (e.g. flash chromatography, reverse phase HPLC, or the like).
  • Bromination in the 5-position with a brominating agent such as Br 2 or NBS gives compound 26.
  • Suzuki coupling of compound 26 to an appropriate boronic acid gives a bicyclic intermediate which after final deprotection of the phenol gives compound 27.
  • Compound 27 can be reacted with the appropriate core intermediate 7 as in Scheme 2 to provide compound 9.
  • Scheme 6 shows an alternative route to compound 27 (wherein R 10 is independently selected from H, alkyl, aryl and heteroaryl).
  • Nucleophilic substitution of compound 28 with a compound of the formula HY-R 10 can be accomplished at elevated temperature with a base such as NaHCO 3 in an appropriate solvent such as n-butanol to give intermediate 26.
  • Suzuki coupling of compound 26 to an appropriate boronic acid gives a bicyclic intermediate which after final deprotection of the phenol gives compound 27.
  • Intermediate 27 can then be reacted with the appropriate core intermediate 7 as in Scheme 2 to give compound 9.
  • Scheme 7 shows a route to phenol intermediate 32 (wherein R 10 is independently selected from H, alkyl, aryl and heteroaryl).
  • Nucleophilic substitution of compound 29 with NaOMe can be accomplished at elevated temperature in an appropriate solvent such as methanol.
  • Nucleophilic substitution of compound 30 with a compound of the formula HY-R 10 , (wherein Y is O, N or S), to form the 5-substituted pyrimidinone 31 can be accomplished at elevated temperature with a base such as NaHCO 3 in an appropriate solvent such as n-butanol. Under these reaction conditions, deprotection of the methoxypyrimidine to the pyrimidinone can also be achieved.
  • deprotection of the methoxypyrimidine can be accomplished with HBr in acetic acid.
  • Copper (I)-mediated coupling of compound 31 to an appropriate halide provides compound 32.
  • the halide used in the coupling reaction contains a standard protecting group. In those cases, the protecting group can be removed by standard conditions known in the art.
  • Compound 32 can then be reacted with the appropriate core intermediate 7 as in Scheme 2 to provide compound 9. 111-03-PCT - P2338R1 - 02120.004WO1
  • Scheme 8 shows a route for the preparation of the 1 -substituted pyridone intermediate 38 (wherein R 10 is independently selected from H, alkyl, aryl and heteroaryl).
  • Alkylation of 6-chloropyridin-2-ol 33 to provide the 1 -substituted pyridone 35 can be accomplished with an alkylation agent (e.g. iodomethane, or the like) mediated by an appropriate base (e.g. potassium carbonate, sodium alkoxides, lithium or sodium hydride, or the like) providing a mixture of isomers 34 and 35.
  • alkylation agent e.g. iodomethane, or the like
  • an appropriate base e.g. potassium carbonate, sodium alkoxides, lithium or sodium hydride, or the like
  • Isomers 34 and 35 can be separated using purification techniques known to those skilled in the art (e.g.
  • Scheme 9 shows a method for preparing phenol intermediate 43 (wherein R 10 is independently selected from H, alkyl, aryl and heteroaryl). 1 -Substituted pyridone 35, which can 111-03-PCT- P2338R1 - 02120.004WO1
  • Scheme 10 shows a route for the preparation of the 6-acyl pyridin-2(lH)-one phenol compound 48.
  • Base-mediated halogen exchange of the commercially available bromopyridine 44 followed by quenching with an aldehyde gives the secondary alcohol compound 45.
  • Oxidation of the alcohol followed by demethylation gives compound 46.
  • Bromination of compound 46 followed by a Suzuki coupling with an appropriate boronic acid gives a coupling compound 47.
  • Final deprotection of the phenol gives compound 48, which can then be reacted with appropriate core intermediate 7 as in Scheme 2 to give compound 9.
  • Sodium borohydride reduction of this compound gives compound 49, and acetylation of compound 49 gives intermediate 50.
  • Compounds 49 and 50 can also be reacted with appropriate compound 7 as in Scheme 2 to provide compound 9.
  • Scheme 12 shows a route for the preparation of the 5-benzyl-3-(4- hydroxyphenyl)pyrimidin-4(3H)-one phenol intermediate 58 which is useful for the synthesis of compounds of Formula I.
  • Commercially available 4,6-dichloropyrimidine-5-carbaldehyde 54 is reacted with the appropriate substituted phenyl magnesium halide to give the secondary alcohol 55.
  • Monobenzylation gives compound 56, which is subjected to hydrogenation to provide compound 57.
  • Copper (I)-mediated coupling of compound 57 to an appropriate phenol provides the desired compound 58, which can be reacted with appropriate core intermediate 7 as in Scheme 2 to provide compound 9.
  • the pyridazino carboxylic acid compound 62 can be prepared using methods described by McNab H. et al (1982) J. Chem. Soc. Perkin Trans. 1:1845 as depicted in Scheme 13. Substituted hydrazine 59 can be converted to hydrazono acetaldehyde 60 with standard dehydrating conditions such as acetic acid at room temperature.
  • the carbonyl group condensation product 61 is prepared in a suitable organic solvent such as toluene, benzene or dioxane at room temperature using piperidinium acetate as catalyst.
  • Carboxylic acid pyridazinone 62 is prepared from hydrazono ethylidene 61 by cyclization under basic conditions (sodium methoxide in methanol) at 7O 0 C.
  • Compound 62 may then be used to acylate aniline intermediate 4 whose preparation is described in Scheme 1 to prepare compounds of Formula I.
  • Scheme 14 shows a route for the preparation of oxo-4-phenyl-3,4- dihydropyrazine-2 -carboxylic acids.
  • the pyrazine-2-carbonitrile 63 was prepared using methods described by Hoornaert, G., et al (1983) J. Heterocyclic Chem. 20:919 and Hoornaert, G., et al (1990) Tetrahedron 46:5715.
  • the pyrazine-2-carboxylic acid 64 can be prepared by hydrolysis to the carboxylic acid followed by removal of the chloro group under hydrogenolysis conditions to 111-03-PCT - P2338R1 - 02120.004WO1
  • Methyl 3-oxo-3,4-dihydropyrazine-2-carboxylate 65 can be converted to alkylpyrazino carboxylate 66 by standard basic alkylation conditions with alkyl halides. These conditions include but are not limited to treatment with K 2 CO 3 in acetone or DMF at room or elevated temperature, or NaH in THF at ambient or elevated temperature, followed by addition of the alkyl halide. In certain embodiments, this alkylation is achieved with LiH in DMF at O 0 C, followed by addition of alkyl chloride or alkyl bromide or alkyl iodide and warming to room temperature.
  • Carboxylic acid 67 can then be prepared using standard saponification conditions such as LiOH or NaOH in standard mixed aqueous/organic solvent systems. The acid 67 can then be coupled via standard amide bond forming techniques to an aniline bearing core 4 constructed according to Scheme 1 to provide final compound 5.
  • Scheme 16 shows a method for preparing pyrrolidin-2-one intermediate 70
  • Scheme 17 shows a method for the preparation of the fused bicyclic cyclopropane lactam ester 74.
  • An optionally substituted allylic amine 71 is acylated with a malonyl chloride ester under basic conditions to give the allylic amide intermediate 72.
  • Cyclization under conditions which generate the malonyl carbine (preferably manganese III acetate catalyzed) provide the fused cyclopropyllactam compound 73.
  • Deprotection under basic conditions typically LiOH or NaOH in an aqueous/organic solvent mixture
  • the acid 74 can then be coupled via standard amide bond forming techniques to aniline bearing cores such as 4 constructed according to Scheme 1 to provide final compound 5.
  • Scheme 18 shows a route for the preparation of (piperidin-l-yl)methanone phenol intermediate 78 which is useful for the synthesis of compounds of Formula I.
  • Substituted and O- protected benzoyl chlorides of type 76 are reacted with an appropriate amine 75 to form the corresponding amide 77, which after final deprotection of the phenol gives compound 78.
  • Compound 78 can then be reacted with the appropriate core intermediate 7 as in Scheme 2 to provide compound 9.
  • Scheme 19 shows a method for the preparation of phenolic intermediate 82.
  • 2,5- dibromopyridine 79 is treated with the appropriate boronic acid under Suzuki type reaction conditions to give selective coupling at the pyridine 2-position to provide compound 80.
  • Buchwald type palladium coupling of compound 80 with an appropriate heteroatom bearing an R 10 group gives the protected compound 81.
  • Final deprotection of compound 81 gives compound 82 which can be reacted with an appropriate core intermediate 7 as in Scheme 2 to provide compound 9.
  • Scheme 20 shows a method for the preparation of phenolic intermediate 86. 2,5-
  • Dibromopyrimidine 83 is treated with the appropriate heteroatom bearing an R 10 group with heating in an appropriate solvent such as 1-propanol. Reaction occurs selectively at the 2- position to give the bromopyrimidine intermediate 84. Suzuki coupling to the appropriately substituted boronic acid gives intermediate 85, which after deprotection gives the phenolic 111-03-PCT - P2338R1 - 02120.004WO1
  • reaction products from one another and/or from starting materials.
  • the desired products of each step or series of steps is separated and/or purified (hereinafter separated) to the desired degree of homogeneity by the techniques common in the art.
  • separations involve multiphase extraction, crystallization from a solvent or solvent mixture, distillation, sublimation, or chromatography.
  • Chromatography can involve any number of methods including, for example: reverse-phase and normal phase; size exclusion; ion exchange; high, medium and low pressure liquid chromatography methods and apparatus; small scale analytical; simulated moving bed (SMB) and preparative thin or thick layer chromatography, as well as techniques of small scale thin layer and flash chromatography.
  • SMB simulated moving bed
  • Another class of separation methods involves treatment of a mixture with a reagent selected to bind to or render otherwise separable a desired product, unreacted starting material, reaction by product, or the like.
  • reagents include adsorbents or absorbents such as activated carbon, molecular sieves, ion exchange media, or the like.
  • the reagents can be acids in the case of a basic material, bases in the case of an acidic material, binding reagents such as antibodies, binding proteins, selective chelators such as crown ethers, liquid/liquid ion extraction reagents (LIX), or the like.
  • Diastereomeric mixtures can be separated into their individual diastereomers on the basis of their physical chemical differences by methods well known to those skilled in the art, such as by chromatography and/or fractional crystallization.
  • Enantiomers can be separated by converting the enantiomeric mixture into a diastereomeric mixture by reaction with an appropriate optically active compound (e.g., chiral auxiliary such as a chiral alcohol or Mosher's acid chloride), separating the diastereomers and converting (e.g., hydrolyzing) the individual diastereoisomers to the corresponding pure enantiomers.
  • an appropriate optically active compound e.g., chiral auxiliary such as a chiral alcohol or Mosher's acid chloride
  • some of the compounds of the present invention may be atropisomers (e.g., substituted biaryls) and are considered as part of this invention. Enantiomers can also be separated by use of a chiral FIPLC column. 111-03-PCT- P2338R1 - 02120.004WO1
  • a single stereoisomer e.g., an enantiomer, substantially free of its stereoisomer may be obtained by resolution of the racemic mixture using a method such as formation of diastereomers using optically active resolving agents (Eliel, E. and Wilen, S. "Stereochemistry of Organic Compounds,” John Wiley & Sons, Inc., New York, 1994; Lochmuller, C. H., (1975) J. Chromatogr., 113(3):283-302).
  • Racemic mixtures of chiral compounds of the invention can be separated and isolated by any suitable method, including: (1) formation of ionic, diastereomeric salts with chiral compounds and separation by fractional crystallization or other methods, (2) formation of diastereomeric compounds with chiral derivatizing reagents, separation of the diastereomers, and conversion to the pure stereoisomers, and (3) separation of the substantially pure or enriched stereoisomers directly under chiral conditions. See: “Drug Stereochemistry, Analytical Methods and Pharmacology,” Irving W. Wainer, Ed., Marcel Dekker, Inc., New York (1993).
  • diastereomeric salts can be formed by reaction of enantiomerically pure chiral bases such as brucine, quinine, ephedrine, strychnine, ⁇ -methyl- ⁇ - phenylethylamine (amphetamine), and the like with asymmetric compounds bearing acidic functionality, such as carboxylic acid and sulfonic acid.
  • the diastereomeric salts may be induced to separate by fractional crystallization or ionic chromatography.
  • the substrate to be resolved is reacted with one enantiomer of a chiral compound to form a diastereomeric pair
  • a diastereomeric pair E. and Wilen, S. "Stereochemistry of Organic Compounds", John Wiley & Sons, Inc., 1994, p. 322
  • Diastereomeric compounds can be formed by reacting asymmetric compounds with enantiomerically pure chiral derivatizing reagents, such as menthyl derivatives, followed by separation of the diastereomers and hydrolysis to yield the pure or enriched enantiomer.
  • a method of determining optical purity involves making chiral esters, such as a menthyl ester, e.g., (-) menthyl chloroformate in the presence of base, or Mosher ester, oc-methoxy- ⁇ - (trifluoromethyl)phenyl acetate (Jacob III (1982) J. Org. Chem 47:4165), of the racemic mixture, and analyzing the 1 H NMR spectrum for the presence of the two atropisomeric enantiomers or diastereomers.
  • chiral esters such as a menthyl ester, e.g., (-) menthyl chloroformate in the presence of base, or Mosher ester, oc-methoxy- ⁇ - (trifluoromethyl)phenyl acetate (Jacob III (1982) J. Org. Chem 47:4165), of the racemic mixture, and analyzing the 1 H NMR spectrum for the presence of the two atropisomeric
  • Stable diastereomers of atropisomeric compounds can be separated and isolated by normal- and reverse-phase chromatography following methods for separation of atropisomeric naphthyl-isoquinolines (WO 96/15111).
  • method (3) a racemic mixture of two enantiomers can be separated by chromatography using a chiral stationary phase ("Chiral Liquid 111-03-PCT - P2338R1 - 02120.004WO1
  • Enriched or purified enantiomers can be distinguished by methods used to distinguish other chiral molecules with asymmetric carbon atoms, such as optical rotation and circular dichroism.
  • Exemplary compounds of this invention include compounds 101-205 as described in Examples 1-105. BIOLOGICAL EVALUATION
  • Determination of the activity of c-Met kinase activity of a compound of Formula I is possible by a number of direct and indirect detection methods.
  • One example of an assay used for the determination of c-Met kinase activity is based on an enzyme linked immunosorbant assay (ELISA).
  • the assay includes a compound of Formula I, c-Met (His-tagged recombinant human Met (amino acids 974-end), expressed by baculovirus), and ATP in assay buffer, as described in Example 106.
  • Exemplary compounds described herein were prepared, characterized, and assayed for their c-Met binding activity and in vitro activity against tumor cells.
  • the range of c- Met binding activities was less than 1 nM to about 10 ⁇ M.
  • Certain exemplary compounds of the invention had c-Met binding activity IC 50 values less than 10 nM.
  • Certain compounds of the invention had MKN45 cell-based activity IC 50 values less than 100 nM.
  • the compounds of the invention may be administered by any route appropriate to the condition to be treated. Suitable routes include oral, parenteral (including subcutaneous, intramuscular, intravenous, intraarterial, intradermal, intrathecal and epidural), transdermal, rectal, nasal, topical (including buccal and sublingual), vaginal, intraperitoneal, intrapulmonary and intranasal.
  • routes include oral, parenteral (including subcutaneous, intramuscular, intravenous, intraarterial, intradermal, intrathecal and epidural), transdermal, rectal, nasal, topical (including buccal and sublingual), vaginal, intraperitoneal, intrapulmonary and intranasal.
  • the compounds may be administered by intralesional administration, including perfusing or otherwise contacting the graft with the inhibitor before transplantation. It will be appreciated that the preferred route may vary with for example the condition of the recipient. Where the compound is administered orally, it may be formulated as a pill, capsule, tablet, etc. with a pharmaceutically acceptable carrier or
  • the compound may be formulated with a pharmaceutically acceptable parenteral vehicle and in a unit dosage injectable form, as detailed below.
  • Compounds of the present invention are useful for treating diseases, conditions and/or disorders including, but not limited to, those characterized by over expression of receptor tyrosine kinases (RTK), e.g. c-Met kinase.
  • RTK receptor tyrosine kinases
  • another aspect of this invention includes methods of treating or preventing diseases or conditions that can be treated or prevented by inhibiting receptor tyrosine kinases (RTK), including c-Met.
  • the method comprises administering to a mammal in need thereof a therapeutically effective amount of a compound of Formula I, or a stereoisomer, geometric isomer, tautomer, solvate, metabolite, or pharmaceutically acceptable salt or prodrug thereof.
  • Diseases and conditions treatable according to the methods of this invention include, but are not limited to, cancer, stroke, diabetes, hepatomegaly, cardiovascular disease, Alzheimer's disease, cystic fibrosis, viral disease, autoimmune diseases, atherosclerosis, restenosis, psoriasis, allergic disorders, inflammation, neurological disorders, a hormone-related disease, conditions associated with organ transplantation, immunodeficiency disorders, destructive bone disorders, proliferative disorders, infectious diseases, conditions associated with cell death, thrombin-induced platelet aggregation, chronic myelogenous leukemia (CML), liver disease, pathologic immune conditions involving T cell activation, and CNS disorders in a patient.
  • a human patient is treated with a compound of Formula I and a pharmaceutically acceptable carrier, adjuvant, or vehicle, wherein said compound of Formula I is present in an amount to detectably inhibit c-Met kinase activity.
  • Cancers which can be treated according to the methods of this invention include, but are not limited to, breast, ovary, cervix, prostate, testis, genitourinary tract, esophagus, larynx, glioblastoma, neuroblastoma, stomach, skin, keratoacanthoma, lung, epidermoid carcinoma, large cell carcinoma, non-small cell lung carcinoma (NSCLC), small cell carcinoma, lung adenocarcinoma, bone, colon, adenoma, pancreas, adenocarcinoma, thyroid, follicular carcinoma, undifferentiated carcinoma, papillary carcinoma, seminoma, melanoma, sarcoma, bladder carcinoma, liver carcinoma and biliary passages, kidney carcinoma, myeloid disorders, lymphoid disorders, hairy cells, buccal cavity and pharynx (oral), lip, tongue, mouth, pharynx, small intestine, colon-rectum, large intestine,
  • Cardiovascular diseases which can be treated according to the methods of this invention include, but are not limited to, restenosis, cardiomegaly, atherosclerosis, myocardial infarction, and congestive heart failure.
  • Neurodegenerative disease which can be treated according to the methods of this invention include, but are not limited to, Alzheimer's disease, Parkinson's disease, amyotrophic 111-03-PCT - P2338R1 - 02120.004WO1
  • Inflammatory diseases which can be treated according to the methods of this invention include, but are not limited to, rheumatoid arthritis, psoriasis, contact dermatitis, and delayed hypersensitivity reactions.
  • Another aspect of this invention provides a compound of this invention for use in the treatment of the diseases or conditions described herein in a mammal, for example, a human, suffering from such disease or condition. Also provided is the use of a compound of this invention in the preparation of a medicament for the treatment of the diseases and conditions described herein in a warm-blooded animal, such as a mammal, for example a human, suffering from such disorder.
  • a pharmaceutical composition comprising a compound of this invention in association with a pharmaceutically acceptable diluent or carrier.
  • a typical formulation is prepared by mixing a compound of the present invention and a carrier, diluent or excipient.
  • Suitable carriers, diluents and excipients are well known to those skilled in the art and include materials such as carbohydrates, waxes, water soluble and/or swellable polymers, hydrophilic or hydrophobic materials, gelatin, oils, solvents, water and the like.
  • Solvents are generally selected based on solvents recognized by persons skilled in the art as safe (GRAS) to be administered to a mammal.
  • safe solvents are non-toxic aqueous solvents such as water and other nontoxic solvents that are soluble or miscible in water.
  • Suitable aqueous solvents include water, ethanol, propylene glycol, polyethylene glycols (e.g., PEG 400, PEG 300), etc. and mixtures thereof.
  • the formulations may also include one or more buffers, stabilizing agents, surfactants, wetting agents, lubricating agents, emulsifiers, suspending agents, preservatives, antioxidants, opaquing agents, glidants, processing aids, colorants, sweeteners, perfuming agents, flavoring agents and other known additives to provide an elegant presentation of the drug (i.e., a compound of the present invention or pharmaceutical composition thereof) or aid in the manufacturing of the pharmaceutical product (i.e., medicament).
  • buffers stabilizing agents, surfactants, wetting agents, lubricating agents, emulsifiers, suspending agents, preservatives, antioxidants, opaquing agents, glidants, processing aids, colorants, sweeteners, perfuming agents, flavoring agents and other known additives to provide an elegant presentation of the drug (i.e., a compound of the present invention or pharmaceutical composition thereof) or aid in the manufacturing of the pharmaceutical product (i.e., medicament).
  • the formulations may be prepared using conventional dissolution and mixing procedures.
  • the bulk drug substance i.e., compound of the present invention or stabilized form of the compound (e.g., complex with a cyclodextrin derivative or other known complexation agent) is dissolved in a suitable solvent in the presence of one or more of the excipients described above.
  • the compound of the present invention is typically formulated into pharmaceutical dosage forms to provide an easily controllable dosage of the drug and to enable patient compliance with the prescribed regimen.
  • the pharmaceutical composition (or formulation) for application may be packaged in a variety of ways depending upon the method used for administering the drug.
  • an article for distribution includes a container having deposited therein the pharmaceutical formulation in an appropriate form.
  • Suitable containers are well known to those skilled in the art and include materials such as bottles (plastic and glass), sachets, ampoules, plastic bags, metal cylinders, and the like.
  • the container may also include a tamper-proof assemblage to prevent indiscreet access to the contents of the package.
  • the container has deposited thereon a label that describes the contents of the container. The label may also include appropriate warnings.
  • compositions of the compounds of the present invention may be prepared for various routes and types of administration.
  • a compound of Formula I having the desired degree of purity may optionally be mixed with pharmaceutically acceptable diluents, carriers, excipients or stabilizers (Remington's Pharmaceutical Sciences (1980) 16th edition, Osol, A. Ed.), in the form of a lyophilized formulation, milled powder, or an aqueous solution.
  • Formulation may be conducted by mixing at ambient temperature at the appropriate pH, and at the desired degree of purity, with physiologically acceptable carriers, i.e., carriers that are non-toxic to recipients at the dosages and concentrations employed.
  • the pH of the formulation depends mainly on the particular use and the concentration of compound, but may range from about 3 to about 8.
  • Formulation in an acetate buffer at pH 5 is a suitable embodiment.
  • the compound of this invention for use herein is preferably sterile.
  • formulations to be used for in vivo administration must be sterile. Such sterilization is readily accomplished by filtration through sterile filtration membranes.
  • the compound ordinarily can be stored as a solid composition, a lyophilized formulation or as an aqueous solution.
  • compositions of the invention will be formulated, dosed and administered in a fashion, i.e., amounts, concentrations, schedules, course, vehicles and route of administration, consistent with good medical practice.
  • Factors for consideration in this context 111-03-PCT- P2338R1 - 0212 0 .004WOl
  • the "therapeutically effective amount" of the compound to be administered will be governed by such considerations, and is the minimum amount necessary to prevent, ameliorate, or treat the coagulation factor mediated disorder. Such amount is preferably below the amount that is toxic to the host or renders the host significantly more susceptible to bleeding.
  • the initial pharmaceutically effective amount of the inhibitor administered parenterally per dose will be in the range of about 0.01-100 mg/kg, namely about 0.1 to 20 mg/kg of patient body weight per day, with the typical initial range of compound used being 0.3 to 15 mg/kg/day.
  • Acceptable diluents, carriers, excipients and stabilizers are nontoxic to recipients at the dosages and concentrations employed, and include buffers such as phosphate, citrate and other organic acids; antioxidants including ascorbic acid and methionine; preservatives (such as octadecyldimethylbenzyl ammonium chloride; hexamethonium chloride; benzalkonium chloride, benzethonium chloride; phenol, butyl or benzyl alcohol; alkyl parabens such as methyl or propyl paraben; catechol; resorcinol; cyclohexanol; 3-pentanol; and m-cresol); low molecular weight (less than about 10 residues) polypeptides; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, aspara
  • the active pharmaceutical ingredients may also be entrapped in microcapsules prepared, for example, by coacervation techniques or by interfacial polymerization, for example, hydroxymethylcellulose or gelatin-microcapsules and poly- (methylmethacylate) microcapsules, respectively, in colloidal drug delivery systems (for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules) or in macroemulsions.
  • colloidal drug delivery systems for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules
  • sustained-release preparations include semipermeable matrices of solid hydrophobic polymers containing a compound of Formula I, which matrices are in the form of shaped articles, e.g., films, or microcapsules.
  • sustained-release matrices include 111-03-PCT - P2338R1 - 02120.004WO1
  • polyesters for example, poly(2-hydroxyethyl-methacrylate), or poly(vinyl alcohol)
  • polylactides U.S. Patent No. 3,773,919
  • copolymers of L-glutamic acid and gamma-ethyl-L- glutamate non-degradable ethylene-vinyl acetate
  • degradable lactic acid-glycolic acid copolymers such as the LUPRON DEPOT® (injectable microspheres composed of lactic acid- glycolic acid copolymer and leuprolide acetate) and poly-D-(-)-3-hydroxybutyric acid.
  • the formulations include those suitable for the administration routes detailed herein.
  • formulations may conveniently be presented in unit dosage form and may be prepared by any of the methods well known in the art of pharmacy. Techniques and formulations generally are found in Remington's Pharmaceutical Sciences (Mack Publishing Co., Easton, PA). Such methods include the step of bringing into association the active ingredient with the carrier which constitutes one or more accessory ingredients. In general the formulations are prepared by uniformly and intimately bringing into association the active ingredient with liquid carriers or finely divided solid carriers or both, and then, if necessary, shaping the product. [00178] Formulations of a compound of Formula I suitable for oral administration may be prepared as discrete units such as pills, capsules, cachets or tablets each containing a predetermined amount of a compound of Formula I.
  • Compressed tablets may be prepared by compressing in a suitable machine the active ingredient in a free-flowing form such as a powder or granules, optionally mixed with a binder, lubricant, inert diluent, preservative, surface active or dispersing agent. Molded tablets may be made by molding in a suitable machine a mixture of the powdered active ingredient moistened with an inert liquid diluent. The tablets may optionally be coated or scored and optionally are formulated so as to provide slow or controlled release of the active ingredient therefrom.
  • Tablets, troches, lozenges, aqueous or oil suspensions, dispersible powders or granules, emulsions, hard or soft capsules, e.g., gelatin capsules, syrups or elixirs may be prepared for oral use.
  • Formulations of compounds of Formula I intended for oral use may be prepared according to any method known to the art for the manufacture of pharmaceutical compositions and such compositions may contain one or more agents including sweetening agents, flavoring agents, coloring agents and preserving agents, in order to provide a palatable preparation. Tablets containing the active ingredient in admixture with non-toxic pharmaceutically acceptable excipient which are suitable for manufacture of tablets are acceptable.
  • excipients may be, for example, inert diluents, such as calcium or sodium carbonate, lactose, calcium or sodium phosphate; granulating and disintegrating agents, such as maize starch, or alginic acid; binding agents, such as starch, gelatin or acacia; and lubricating 111-03-PCT - P2338R1 - 02120.004WO1
  • 57 agents such as magnesium stearate, stearic acid or talc. Tablets may be uncoated or may be coated by known techniques including microencapsulation to delay disintegration and adsorption in the gastrointestinal tract and thereby provide a sustained action over a longer period.
  • a time delay material such as glyceryl monostearate or glyceryl distearate alone or with a wax may be employed.
  • the formulations are preferably applied as a topical ointment or cream containing the active ingredient(s) in an amount of, for example, 0.075 to 20% w/w.
  • the active ingredients may be employed with either a paraffinic or a water-miscible ointment base.
  • the active ingredients may be formulated in a cream with an oil-in-water cream base.
  • the aqueous phase of the cream base may include a polyhydric alcohol, i.e., an alcohol having two or more hydroxyl groups such as propylene glycol, butane 1,3-diol, mannitol, sorbitol, glycerol and polyethylene glycol (including PEG 400) and mixtures thereof.
  • the topical formulations may desirably include a compound which enhances absorption or penetration of the active ingredient through the skin or other affected areas. Examples of such dermal penetration enhancers include dimethyl sulfoxide and related analogs.
  • the oily phase of the emulsions of this invention may be constituted from known ingredients in a known manner.
  • the phase may comprise merely an emulsifier, it desirably comprises a mixture of at least one emulsifier with a fat or an oil or with both a fat and an oil.
  • a hydrophilic emulsifier is included together with a lipophilic emulsifier which acts as a stabilizer. It is also preferred to include both an oil and a fat.
  • the emulsifier(s) with or without stabilizer(s) make up the so-called emulsifying wax, and the wax together with the oil and fat make up the so-called emulsifying ointment base which forms the oily dispersed phase of the cream formulations.
  • Emulsifiers and emulsion stabilizers suitable for use in the formulation of the invention include TWEEN® 60, Span® 80, cetostearyl alcohol, benzyl alcohol, myristyl alcohol, glyceryl mono-stearate and sodium lauryl sulfate.
  • Aqueous suspensions of Formula I compounds contain the active materials in admixture with excipients suitable for the manufacture of aqueous suspensions.
  • excipients include a suspending agent, such as sodium carboxymethylcellulose, croscarmellose, povidone, methylcellulose, hydroxypropyl methylcellulose, sodium alginate, polyvinylpyrrolidone, gum tragacanth and gum acacia, and dispersing or wetting agents such as a naturally occurring phosphatide (e.g., lecithin), a condensation product of an alkylene oxide with a fatty acid (e.g., polyoxyethylene stearate), a condensation product of ethylene oxide with a long chain aliphatic 111-03-PCT - P2338R1 - 02120.004WO1
  • a suspending agent such as sodium carboxymethylcellulose, croscarmellose, povidone, methylcellulose, hydroxypropyl methylcellulose, sodium alginate, polyvinylpyr
  • the aqueous suspension may also contain one or more preservatives such as ethyl or n-propyl p-hydroxybenzoate, one or more coloring agents, one or more flavoring agents and one or more sweetening agents, such as sucrose or saccharin.
  • preservatives such as ethyl or n-propyl p-hydroxybenzoate, one or more coloring agents, one or more flavoring agents and one or more sweetening agents, such as sucrose or saccharin.
  • compositions of compounds of Formula I may be in the form of a sterile injectable preparation, such as a sterile injectable aqueous or oleaginous suspension.
  • a sterile injectable preparation such as a sterile injectable aqueous or oleaginous suspension.
  • This suspension may be formulated according to the known art using those suitable dispersing or wetting agents and suspending agents which have been mentioned above.
  • the sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally acceptable diluent or solvent, such as a solution in 1,3-butanediol or prepared as a lyophilized powder.
  • a non-toxic parenterally acceptable diluent or solvent such as a solution in 1,3-butanediol or prepared as a lyophilized powder.
  • acceptable vehicles and solvents that may be employed are water, Ringer's solution and isotonic sodium chloride solution.
  • sterile fixed oils may conventionally be employed as a solvent or suspending medium.
  • any bland fixed oil may be employed including synthetic mono- or diglycerides.
  • fatty acids such as oleic acid may likewise be used in the preparation of injectables.
  • a time-release formulation intended for oral administration to humans may contain approximately 1 to 1000 mg of active material compounded with an appropriate and convenient amount of carrier material which may vary from about 5 to about 95% of the total compositions (weight:weight).
  • the pharmaceutical composition can be prepared to provide easily measurable amounts for administration.
  • an aqueous solution intended for intravenous infusion may contain from about 3 to 500 ⁇ g of the active ingredient per milliliter of solution in order that infusion of a suitable volume at a rate of about 30 mL/hr can occur.
  • Formulations suitable for parenteral administration include aqueous and nonaqueous sterile injection solutions which may contain anti-oxidants, buffers, bacteriostats and solutes which render the formulation isotonic with the blood of the intended recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents.
  • Formulations suitable for topical administration to the eye also include eye drops wherein the active ingredient is dissolved or suspended in a suitable carrier, especially an aqueous solvent for the active ingredient.
  • a suitable carrier especially an aqueous solvent for the active ingredient.
  • the active ingredient is preferably present in such formulations 111-03-PCT - P2338R1 - 02120.004WO1
  • 59 in a concentration of about 0.5 to 20% w/w, for example about 0.5 to 10% w/w, for example about 1.5% w/w.
  • Formulations suitable for topical administration in the mouth include lozenges comprising the active ingredient in a flavored basis, usually sucrose and acacia or tragacanth; pastilles comprising the active ingredient in an inert basis such as gelatin and glycerin, or sucrose and acacia; and mouthwashes comprising the active ingredient in a suitable liquid carrier.
  • Formulations for rectal administration may be presented as a suppository with a suitable base comprising for example cocoa butter or a salicylate.
  • Formulations suitable for intrapulmonary or nasal administration have a particle size for example in the range of 0.1 to 500 microns (including particle sizes in a range between 0.1 and 500 microns in increments microns such as 0.5, 1, 30 microns, 35 microns, etc.), which is administered by rapid inhalation through the nasal passage or by inhalation through the mouth so as to reach the alveolar sacs.
  • Suitable formulations include aqueous or oily solutions of the active ingredient.
  • Formulations suitable for aerosol or dry powder administration may be prepared according to conventional methods and may be delivered with other therapeutic agents such as compounds heretofore used in the treatment or prophylaxis disorders as described below.
  • Formulations suitable for vaginal administration may be presented as pessaries, tampons, creams, gels, pastes, foams or spray formulations containing in addition to the active ingredient such carriers as are known in the art to be appropriate.
  • the formulations may be packaged in unit-dose or multi-dose containers, for example sealed ampoules and vials, and may be stored in a f ⁇ eeze-dried (lyophilized) condition requiring only the addition of the sterile liquid carrier, for example water, for injection immediately prior to use.
  • sterile liquid carrier for example water
  • Extemporaneous injection solutions and suspensions are prepared from sterile powders, granules and tablets of the kind previously described.
  • Preferred unit dosage formulations are those containing a daily dose or unit daily sub-dose, as herein above recited, or an appropriate fraction thereof, of the active ingredient.
  • the invention further provides veterinary compositions comprising at least one active ingredient as above defined together with a veterinary carrier therefore.
  • Veterinary carriers are materials useful for the purpose of administering the composition and may be solid, liquid or gaseous materials which are otherwise inert or acceptable in the veterinary art and are compatible with the active ingredient. These veterinary compositions may be administered parenterally, orally or by any other desired route. COMBINATION THERAPY 111-03-PCT- P2338R1 - 02120.Q04WO1
  • the compounds of Formula I may be employed alone or in combination with other therapeutic agents for the treatment of a disease or disorder described herein, such as a hyperproliferative disorder (e.g., cancer).
  • a compound of Formula I is combined in a pharmaceutical combination formulation, or dosing regimen as combination therapy, with a second compound that has anti-hyperproliferative properties or that is useful for treating a hyperproliferative disorder (e.g., cancer).
  • the second compound of the pharmaceutical combination formulation or dosing regimen preferably has complementary activities to the compound of Formula I such that they do not adversely affect each other.
  • Such compounds are suitably present in combination in amounts that are effective for the purpose intended.
  • a composition of this invention comprises a compound of Formula I, or a stereoisomer, geometric isomer, tautomer, solvate, metabolite, or pharmaceutically acceptable salt or prodrug thereof, in combination with a chemotherapeutic agent such as described herein.
  • the combination therapy may be administered as a simultaneous or sequential regimen. When administered sequentially, the combination may be administered in two or more administrations.
  • the combined administration includes coadministration, using separate formulations or a single pharmaceutical formulation, and consecutive administration in either order, wherein preferably there is a time period while both (or all) active agents simultaneously exert their biological activities.
  • Suitable dosages for any of the above coadministered agents are those presently used and may be lowered due to the combined action (synergy) of the newly identified agent and other chemotherapeutic agents or treatments.
  • the combination therapy may provide "synergy” and prove “synergistic", i.e., the effect achieved when the active ingredients used together is greater than the sum of the effects that results from using the compounds separately.
  • a synergistic effect may be attained when the active ingredients are: (1) co-formulated and administered or delivered simultaneously in a combined, unit dosage formulation; (2) delivered by alternation or in parallel as separate formulations; or (3) by some other regimen.
  • a synergistic effect may be attained when the compounds are administered or delivered sequentially, e.g., by different injections in separate syringes.
  • an effective dosage of each active ingredient is administered sequentially, i.e., serially
  • effective dosages of two or more active ingredients are administered together.
  • a compound of Formula I or a stereoisomer, geometric isomer, tautomer, solvate, metabolite, or pharmaceutically acceptable 111-03-PCT - P2338R1 - 02120.004WO1
  • Combination therapies may be combined with other chemotherapeutic, hormonal or antibody agents such as those described herein, as well as combined with surgical therapy and radiotherapy.
  • Combination therapies according to the present invention thus comprise the administration of at least one compound of Formula I, or a stereoisomer, geometric isomer, tautomer, solvate, metabolite, or pharmaceutically acceptable salt or prodrug thereof, and the use of at least one other cancer treatment method.
  • the amounts of the compound(s) of Formula I and the other pharmaceutically active chemotherapeutic agent(s) and the relative timings of administration will be selected in order to achieve the desired combined therapeutic effect.
  • the invention includes metabolites of compounds of Formula I, including compounds produced by a process comprising contacting a compound of this invention with a mammal for a period of time sufficient to yield a metabolic product thereof.
  • Metabolite products typically are identified by preparing a radiolabeled (e.g., 14 C or 3 H) isotope of a compound of the invention, administering it parenterally in a detectable dose (e.g., greater than about 0.5 mg/kg) to an animal such as rat, mouse, guinea pig, monkey, or to man, allowing sufficient time for metabolism to occur (typically about 30 seconds to 30 hours) and isolating its conversion products from the urine, blood or other biological samples.
  • a detectable dose e.g., greater than about 0.5 mg/kg
  • the metabolite structures are determined in conventional fashion, e.g., by MS, LC/MS or NMR analysis. In general, analysis of metabolites is done in the same way as conventional drug metabolism studies well known to those skilled in the art.
  • the metabolite products so long as they are not otherwise found in vivo, are useful in diagnostic assays for therapeutic dosing of the compounds of the invention.
  • kits containing materials useful for the treatment of the diseases and disorders described above.
  • the kit comprises a container comprising a quinoline compound of Formula I, or a stereoisomer, geometric isomer, tautomer, solvate, metabolite, or pharmaceutically acceptable salt or prodrug thereof.
  • the kit may further comprise a label or package insert on or associated with the container.
  • package insert is used to refer to 111-03-PCT- P2338R1 - 02120.004WO1
  • Suitable containers include, for example, bottles, vials, syringes, blister pack, etc.
  • the container may be formed from a variety of materials such as glass or plastic.
  • the container may hold a compound of Formula I or a formulation thereof which is effective for treating the condition and may have a sterile access port (for example, the container may be an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle).
  • At least one active agent in the composition is a compound of Formula I.
  • the label or package insert indicates that the composition is used for treating the condition of choice, such as cancer.
  • the label or package insert may indicate that the patient to be treated is one having a disorder such as a hyperproliferative disorder, neurodegeneration, cardiac hypertrophy, pain, migraine or a neurotraumatic disease or event.
  • the label or package inserts indicates that the composition comprising a compound of Formula I can be used to treat a disorder resulting from abnormal cell growth.
  • the label or package insert may also indicate that the composition can be used to treat other disorders.
  • the article of manufacture may further comprise a second container comprising a pharmaceutically acceptable buffer, such as bacteriostatic water for injection (BWFI), phosphate-buffered saline, Ringer's solution and dextrose solution. It may further include other materials desirable from a commercial and user standpoint, including other buffers, diluents, filters, needles, and syringes.
  • BWFI bacteriostatic water for injection
  • phosphate-buffered saline such as bacteriostatic water for injection (BWFI), phosphate-buffered saline, Ringer's solution and dextrose solution.
  • BWFI bacteriostatic water for injection
  • phosphate-buffered saline such as phosphate-buffered saline, Ringer's solution and dextrose solution.
  • dextrose solution such as bacteriostatic water for injection (BWFI), phosphate-buffered saline, Ringer's solution and dext
  • the kit may further comprise directions for the administration of the compound of
  • kits are suitable for the delivery of solid oral forms of a compound of Formula I, such as tablets or capsules.
  • a kit preferably includes a number of unit dosages.
  • kits can include a card having the dosages oriented in the order of their intended use.
  • An example of such a kit is a "blister pack". Blister packs are well known in the packaging industry and are widely used for packaging pharmaceutical unit dosage forms.
  • a memory aid can be provided, for example in the form of numbers, letters, or other markings or with a calendar insert, designating the days in the treatment schedule in which the dosages can be administered.
  • a kit may comprise (a) a first container with a compound of Formula I contained therein; and optionally (b) a second container with a second pharmaceutical formulation contained therein, wherein the second pharmaceutical formulation comprises a second compound with anti-hyperproliferative activity.
  • the kit may further comprise a third container comprising a pharmaceutically- acceptable buffer, such as bacteriostatic water for injection (BWFI), phosphate-buffered saline, Ringer's solution and dextrose solution. It may further include other materials desirable from a commercial and user standpoint, including other buffers, diluents, filters, needles, and syringes.
  • BWFI bacteriostatic water for injection
  • phosphate-buffered saline such as bacteriostatic water for injection (BWFI), phosphate-buffered saline, Ringer's solution and dextrose solution. It may further include other materials desirable from a commercial and user standpoint, including other buffers, diluents,
  • the kit may comprise a container for containing the separate compositions such as a divided bottle or a divided foil packet, however, the separate compositions may also be contained within a single, undivided container.
  • the kit comprises directions for the administration of the separate components.
  • the kit form is particularly advantageous when the separate components are preferably administered in different dosage forms (e.g., oral and parenteral), are administered at different dosage intervals, or when titration of the individual components of the combination is desired by the prescribing physician.
  • Step A Preparation of 4-chloro-5-methyl-6-(2-methylbenzyl)pyrimidine: 2-
  • Methylbenzylzinc chloride 25 ml of 0.5 M THF solution, 12 mmol was added to a solution of 4,6-dichloro-5-methylpyrimidine (2.0 g, 12 mmol) and bis(triphenylphosphine) palladium(II) chloride (0.4 g. 0.6 mmol) in THF (20 mL). The reaction mixture was heated to reflux for 2 hours, cooled to room temperature, and then poured onto water (10 mL). The reaction mixture was extracted with ethyl acetate, and the organic layer was washed with brine, dried over sodium sulfate, filtered, and concentrated.
  • Step B Preparation of 4-(benzyloxy)-5-methyl-6-(2-methylbenzyl)pyrimidine:
  • Step C Preparation of 5-methyl-6-(2-methylbenzyl)pyrimidin-4-ol: 4-
  • Step D Preparation of 3-(3-fluoro-4-hydroxyphenyl)-5-methyl-6-(2- methylbenzyl)pyrimidin-4(3H)-one: Copper(I) iodide (90 mg, 0.5 mmol) was added to a solution of 5-methyl-6-(2-methylbenzyl)pyrimidin-4-ol (1.0 g, 5.0 mmol), 4-bromo-2-fluorophenol (0.90 g, 5.0 mmol), N,N'-dimethylethylenediamine (80 mg, 0.90 mmol) and potassium phosphate (2.0 g, 9.0 mmol).
  • Step E Preparation of 3-(3-fluoro-4-(6-methoxy-7-(3-morpholino- propoxy)quinolin-4-yloxy)phenyl)-5-methyl-6-(2-methylbenzyl)pyrimidin-4(3H)-one: DMAP (0.75 mg, 0.0062 mmol) was added to a suspension of 3-(3-fluoro-4-hydroxyphenyl)-5-methyl-6- (2-methylbenzyl)pyrimidin-4(3H)-one (20 mg, 0.062 mmol) and 4-chloro-6-methoxy-7-(3- morpholinopropoxy)quinoline (prepared according to WO 01/55116, Example 2, 21 mg, 0.062 mmol).
  • Step A Preparation of 4-benzyl-6-chloropyrimidine: Prepared from 4,6- dichloropyrimidine (2.0 g, 13 mmol) and benzyl zinc chloride (0.5 M solution in THF, 27 mL, 13 mmol) according to the procedure described for Example 1, Step A. The crude product was purified by silica gel flash column chromatography (1:10 Et 2 O/Hexane) to yield the product (1.3 g, 47%) as a yellow liquid.
  • Step B Preparation of 4-benzyl-6-(benzyloxy)pyrimidine: Prepared from 4- benzyl-6-chloropyrimidine (1.1 g, 5.4 mmol) according to the procedure described for Example 1, Step B. The crude was purified by by silica gel flash column chromatography (1:1 Et 2 O/Hexane) to yield the product (1.3 g, 88%) as a colorless liquid.
  • Step C Preparation of 6-benzylpyrimidin-4-ol: Prepared from 4-benzyl-6-
  • Step D Preparation of 6-benzyl-3-(3-fluoro-4-hydroxyphenyl)pyrimidin-4(3H)- one: Prepared from 6-benzylpyrimidin-4-ol (0.50 g, 2.7 mmol) according to the procedure described for Example 1, Step D. The crude was purified by silica gel flash column chromatography (1:1 EtOAc/Hexane) to yield the product (0.50 g, 63%) as a white solid.
  • Step E Preparation of 6-benzyl-3-(3-fluoro-4-(6-methoxy-7-(3- morpholinopropoxy)quinolin-4-yloxy)phenyl)pyrimidin-4(3H)-one: Prepared from 6-benzyl-3- (3-fiuoro-4-hydroxyphenyl)pyrimidin-4(3H)-one (18 mg, 0.059 mmol) according to the procedure described for Example 1, Step E. The crude product was purified by silica gel flash 111-03-PCT- P2338R1 - 02120.004WO1
  • Step A Preparation of 4-benzyl-6-chloro-5-methylpyrimidine: Prepared from benzyl zinc bromide (0.5 M solution in THF, 25 mL, 12 mmol) and 4,6-dichloro-5- methylpyrimidine (2.0 g, 12 mmol) according to the procedure described for Example 1, Step A. The crude was purified by silica gel flash column chromatography (1:5 EtOAc/Hexane) to yield the product (0.86 g, 32%) as a colorless oil.
  • 1 H NMR (CDCl 3 , 400 MHz) ⁇ 8.78 (s, IH), 7.28-7.33 (m, 2H), 7.18-7.26 (m, 3H), 4.19 (s, 2H), 2.35 (s, 3H).
  • Step B Preparation of 4-benzyl-6-(benzyloxy)-5-methylpyrimidine: Prepared from 4-benzyl-6-chloro-5-methylpyrimidine (0.8 g, 4.0 mmol) according to the procedure described for Example 1, Step B. The crude product was purified by silica gel flash column chromatography (1:9 Et 2 O/Hexane) to yield the product (1.0 g, 94%) as a colorless oil.
  • Step C Preparation of 6-benzyl-5-methylpyrimidin-4-ol: Prepared from 4- benzyl-6-(benzyloxy)-5-methylpyrimidine (1.0 g, 3.0 mmol) according to the procedure described for Example 1, Step C, to yield the product (0.50 g, 73%) as a white solid.
  • Step D Preparation of 6-benzyl-3-(3-fluoro-4-hydroxyphenyl)-5- methylpyrimidin-4(3H)-one: Prepared from 6-benzyl-5-methylpyrimidin-4-ol (0.16 g, 0.80 mmol) according to the procedure described for Example 1, Step D. The crude product was purified by silica gel flash column chromatography (1 :1 EtOAc/Hexane) to yield the product (0.10 g, 64%) as a white solid.
  • Step E Preparation of 6-benzyl-3-(3-fluoro-4-(6-methoxy-7-(3- morpholinopropoxy)quinolin-4-yloxy)phenyl)-5-methylpyrimidin-4(3H)-one: Prepared from 6- benzyl-3-(3-fluoro-4-hydroxyphenyl)-5-methylpyrimidin-4(3H)-one (18 mg, 0.06 mmol) according to the procedure described for Example 1, Step E. The crude product was purified by silica gel flash column chromatography (1:10 MeOH/EtOAc) to yield 103 (10 mg, 28%) as a white solid.
  • Step A Preparation of (3-benzylpiperidin-l-yl)(3-fluoro-4- methoxyphenyl)methanone: Triethylamine (2 mL, 0.01 mol) was added into a solution of 3- fluoro-4-methoxybenzoyl chloride (500 mg, 2.65 mmol) and 3-benzylpiperidine hydrochloride (561 mg, 2.65 mmol) in CH 2 Cl 2 (20 mL). The reaction mixture was stirred for 30 minutes at room temperature and then poured into water (10 mL).
  • Step B Preparation of (3-benzylpiperidin-l-yl)(3-fluoro-4- hydroxyphenyl)methanone: Boron tribromide (0.5 ml, 6.57 mmol) was added into a solution of (3-benzylpiperidin-l-yl)(3-fluoro-4-methoxyphenyl)methanone (0.86 g, 2.63 mmol) in CH 2 Cl 2 (2 mL) at O 0 C. The reaction mixture was stirred for 30 minutes at room temperature and then poured into water (10 mL).
  • Step C Preparation of (3-benzylpiperidin-l-yl)(3-fluoro-4-(6-methoxy-7-(3- morpholinopropoxy)quinolin-4-yloxy)phenyl)methanone: Prepared from (3-benzylpiperidin-l- 111-03-PCT- P2338R1 - 02120.004WO1
  • Step A Preparation of (4-benzylpiperidin-l-yl)(3-fluoro-4- methoxyphenyl)methanone: Prepared from 3-fluoro-4-methoxybenzoyl chloride (570 mg, 3.02 mmol) and 4-benzylpiperidine (530 mg, 3.02 mmol) according to the procedure described for Example 4, Step A, to yield the product (920 mg, 93%) as a white solid.
  • Step B Preparation of (4-benzylpiperidin-l-yl)(3-fluoro-4- hydroxyphenyl)methanone: Prepared from (4-benzylpiperidin-l-yl)(3-fluoro-4- methoxyphenyl)methanone (130 mg, 0.40 mmol) according to the procedure described for Example 4, Step B, to yield the product (120 mg, 96%) as a white solid.
  • Step C (4-benzylpiperidin-l-yl)(4-(6,7-dimethoxyquinolin-4-yloxy)-3- fluorophenyl)methanone: Prepared from 4-chloro-6,7-dimethoxyquinoline (for preparation see reference in Example 5) (79 mg, 0.35 mmol) and (4-benzylpiperidin-l-yl)(3-fluoro-4- hydroxyphenyl)methanone (110 g, 0.35 mmol) according to the procedure described for Example 1, Step E. The crude product was purified by silica gel flash column chromatography (1:10 MeOH/EtOAc) to yield 106 (20 mg, 11%) as a white solid.
  • Step A Preparation of 3-benzyl-5-(4-(benzyloxy)-3-fluorophenyl)pyrimidin-
  • Step B Preparation of 3-benzyl-5-(3-fiuoro-4-hydroxyphenyl)pyrimidin-4(3H)- one: Prepared from 3-benzyl-5-(4-(benzyloxy)-3-fluorophenyl)pyrimidin-4(3H)-one (0.3 g, 0.8 mmol) according to the procedure described for Example 1, Step C, to yield the product (0.2 g, 87%) as a white solid.
  • Step C Preparation of 3-benzyl-5-(4-(7-(benzyloxy)-6-methoxyquinolin-4- yloxy)-3-fluorophenyi)pyrimidin-4(3H)-one: Prepared from 7-(benzyloxy)-4-chloro-6- methoxyquinoline (prepared according to WO2005030140, Example 32, 200 mg, 0.67 mmol) and 3-benzyl-5-(3-fluoro-4-hydroxyphenyl)pyrimidin-4(3H)-one (198 mg, 0.67 mmol) according to the procedure described for Example 1, Step E. The crude product was purified by silica gel flash column chromatography (1:10 MeOH/EtOAc) to yield the product (100 mg, 37%) as a white solid. LRMS (ESI pos) m/e 560 (M+l).
  • Step D Preparation of 3-benzyl-5-(3-fluoro-4-(7-hydroxy-6-rnethoxyquinolin-4- yloxy)phenyl)pyrimidin-4(3H)-one: Prepared from 3-benzyl-5-(4-(7-(benzyloxy)-6- methoxyquinolin-4-yloxy)-3-fluorophenyl)pyrimidin-4(3H)-one (90 mg, 0.16 mmol) according to the procedure described for Example 1, Step C, to yield the product (50 mg, 66%) as a white solid.
  • Step E Preparation of 3-benzyl-5-(3-fluoro-4-(6-methoxy-7-(3- morpholinopropoxy)quinolin-4-yloxy)phenyl)pyrimidin-4(3H)-one: Cesium carbonate (6.9 mg, 0.021 mmol) was added to a solution of 3-benzyl-5-(3-fluoro-4-(7-hydroxy-6-methoxyquinolin-4- yloxy)phenyl)pyrimidin-4(3H)-one (10 mg, 0.02 mmol) and 4-(3-chloropropyl)morpholine (3.5 mg, 0.021 mmol).
  • reaction mixture was heated to 5O 0 C for 1 hour, and then poured onto water (1 mL).
  • the reaction mixture was extracted with EtOAc, and the organic layer was washed with brine, dried over sodium sulfate, filtered, and concentrated.
  • the residue was purified by silica gel flash column chromatography (1:10 MeOH/EtOAc) to yield 107 (6 mg, 47%) as a yellow solid.
  • Step A Preparation of 5-bromo-3-(4-chlorobenzyl)pyrimidin-4(3H)-one: Sodium hydride (0.34 g, 8.6 mmol) was added into a solution of 5-bromopyrimidin-4(3H)-one (prepared according to Thomas J Kress (1985) J. Org. Chem. 50:3073-6, 1.5 g, 8.57 mmol) in THF (10 niL) and DMF (6 mL). The reaction was stirred for 10 minutes and 4-chlorobenzyl bromide (1.76 g, 8.57 mmol) was added.
  • Step B Preparation of 5-(4-(benzyloxy)-3-fluorophenyl)-3-(4- chlorobenzyl)pyrimidin-4(3H)-one: Prepared from 5-bromo-3-(4-chlorobenzyl)pyrimidin-4(3H)- one (0.39 g, 1.3 mmol) according to the procedure described for Example 7, Step A. The crude was purified by silica gel flash column chromatography (1: 1 EtOAc/Hexane) to yield the product (0.17 g, 31%) as a white solid.
  • Step C Preparation of 3-(4-chlorobenzyl)-5-(3-fluoro-4- hydroxyphenyl)pyrimidin-4(3H)-one: Prepared from 5-(4-(benzyloxy)-3-fluorophenyl)-3-(4- chlorobenzyl)pyrimidin-4(3H)-one (0.17 g, 0.40 mmol) according to the procedure described for Example 1, Step C, to yield the product (0.1 g, 75%) as a yellow solid.
  • Step D Preparation of 3-(4-chlorobenzyl)-5-(3-fluoro-4-(6-methoxy-7-(3- morpholinopropoxy)quinolin-4-yloxy)phenyl)pyrimidin-4(3H)-one: Prepared from 3-(4- chlorobenzyl)-5-(3-fluoro-4-hydroxyphenyl)pyrimidin-4(3H)-one (27 mg, 0.08 mmol) according to the procedure described for Example 1, Step E. The crude was purified by silica gel flash column chromatography (1:10 MeOH/EtOAc) to yield 113 (10 mg, 36%) as a yellow solid.
  • Step A Preparation of (6-(benzyloxy)pyridin-2-yl)(phenyl)methanol: nBuLi (2.5
  • Step B Preparation of 6-benzylpyridin-2-ol: Palladium on carbon (10%, 1.5 g, 1.4 mmol) was added into a solution of (6-(benzyloxy)pyridin-2-yl)(phenyl)methanol (4 g, 14 mmol) in MeOH (20 mL). The reaction was pressurized with hydrogen using a balloon, stirred for 2 111-03-PCT- P2338R1 - 02120.004WO1
  • Step C Preparation of 6-benzyl-3-bromopyridin-2-ol: Bromine (0.14 mL, 2.7 mmol) was added into a solution of 6-benzylpyridin-2-ol (0.5 g, 2.7 mmol) in CH 2 Cl 2 (5 mL). The reaction was stirred for 20 minutes at room temperature and then poured into 10% aqueous sodium bisulfite solution (10 mL). The reaction mixture was extracted with CH 2 Cl 2 and the organic layer was washed with brine, dried over sodium sulfate, filtered, and concentrated to yield the product (0.59 g, 82%) as a yellow solid. LRMS (ESI pos) m/e 263 (M+l).
  • Step D Preparation of 6-benzyl-3-(4-(benzyloxy)-3-fluorophenyl)pyridin-2(lH)- one: Prepared from 6-benzyl-3-bromopyridin-2-ol (0.63 g, 2.39 mmol) according to the procedure described for Example 7, Step A. The crude product was purified by silica gel flash column chromatography (1:1 EtOAc/Hexane) to yield the product (200 mg, 22%) as a white solid.
  • Step E Preparation of 6-benzyl-3-(3-fluoro-4-hydroxyphenyl)pyridin-2(lH)-one:
  • Step F Preparation of 6-benzyl-3-(4-(6,7-dimethoxyquinolin-4-yloxy)-3- fluorophenyl)pyridin-2(lH)-one: Prepared from 4-chloro-6,7-dimethoxyquinoline (prepared according to reference in Example 5) (85 mg, 0.38 mmol) and 6-benzyl-3-(3-fluoro-4- hydroxyphenyl)pyridin-2(lH)-one (93 mg, 0.31 mmol) according to the procedure described for Example 1, Step E. The crude product was purified by silica gel flash column chromatography (1:10 MeOH/EtOAc) to yield 114 (50 mg, 33%) as a white solid.
  • Example 14 (Example 14, Step E, 30 mg, 0.10 mmol) according to the procedure described for Example 1, Step E.
  • the crude product was purified by silica gel flash column chromatography (1:10 MeOH/EtOAc) to yield 115 (32 mg, 53%) as a white solid.
  • Step A Preparation of (2-(benzyloxy)-5-bromopyridin-4-yl)(phenyl)methanol:
  • Step B Preparation of 4-benzylpyridin-2(lH)-one: Prepared from (2-(benzyloxy)-
  • Step C Preparation of 4-benzyl-l-(3-fluoro-4-hydroxyphenyl)pyridin-2(lH)-one:
  • Step D Preparation of 4-benzyl-l-(3-fluoro-4-(6-methoxy-7-(3- morpholinopropoxy)quinolin-4-yloxy)phenyl)pyridin-2(lH)-one: Prepared from 4-benzyl-l-(3- fluoro-4-hydroxyphenyl)pyridin-2(lH)-one (30 mg, 0.10 mmol) according to the procedure described for Example 1, Step E. The crude product was purified by silica gel flash column chromatography (1:10 MeOH/EtOAc) to yield 116 (41 mg, 68%) as a white solid.
  • Step A Preparation of 5-bromo-3-(2-chlorobenzyl)pyrimidin-4(3H)-one:
  • Step B Preparation ⁇ of 5-(4-(benzyloxy)-3-fluorophenyl)-3-(2- chlorobenzyl)pyrimidin-4(3H)-one: Prepared from 5-bromo-3-(2-chlorobenzyl)pyrimidin-4(3H)- one (0.37 g, 1.2 mmol) according to the procedure described for Example 7, Step A. The crude product was purified by silica gel flash column chromatography (1:1 EtOAc/Hexane) to yield the product (0.30 g, 58%) as a white solid.
  • Step C Preparation of 3-(2-chlorobenzyl)-5-(3-fluoro-4- hydroxyphenyl)pyrimidin-4(3H)-one: Prepared from 5-(4-(benzyloxy)-3-fluorophenyl)-3-(2- chlorobenzyl)pyrimidin-4(3H)-one (0.4 g, 1.0 mmol) according to the procedure described for Example 1, Step C, to yield the product (0.20 g, 64%) as a white solid.
  • Step D Preparation of 3-(2-chlorobenzyl)-5-(4-(6,7-dimethoxyquinolin-4-yloxy)-
  • 3-fluorophenyl)pyrimidin-4(3H)-one Prepared from 4-chloro-6,7-dimethoxyquinoline (prepared according to reference procedure in Example 5) (60 mg, 0.27 mmol) and 3-(2-chlorobenzyl)-5-(3- fluoro-4-hydroxyphenyl)pyrimidin-4(3H)-one (89 mg, 0.27 mmol) according to the procedure described for Example 1, Step E. The crude product was purified by silica gel flash column chromatography (1:10 MeOH/EtOAc) to yield 119 (20 mg, 14%) as a white solid.
  • Step A Preparation of 5-bromo-3-(2-methylbenzyl)pyrimidin-4(3H)-one:
  • Step B Preparation of 5-(4-(benzyloxy)-3-fluorophenyl)-3-(2- methylbenzyl)pyrimidin-4(3H)-one: Prepared from 5-bromo-3-(2-chlorobenzyl)pyrimidin-4(3H)- one (280 mg, 1.0 mmol) according to the procedure described for Example 7, Step A. The crude product was purified by silica gel flash column chromatography (1:1 EtOAc/Hexane) to yield the product (0.31 g, 77%) as a white solid.
  • Step C Preparation of 5-(3-fluoro-4-hydroxyphenyl)-3-(2- methylbenzyl)pyrimidin-4(3H)-one: Prepared frcm 5-(4-(benzyloxy)-3-fluorophenyl)-3-(2- methylbenzyl)pyrimidin-4(3H)-one (0.31 g, 0.77 mmol) according to the procedure described for Example 1, Step C, to yield the product (0.20 g, 84%) as a white solid.
  • LRMS (ESI pos) m/e 331 (M+l).
  • Step D Preparation of 5-(4-(6,7-dimethoxyquinolin-4-yloxy)-3-fluorophenyl)-3-
  • (2-methylbenzyl)pyrimidin-4(3H)-one Prepared from 4-chloro-6,7-dimethoxyquinoline (prepared according to reference procedure in Example 5) (54 mg, 0.24 mmol) and 5-(3-fluoro-4- hydroxyphenyl)-3-(2-methylbenzyl)pyrimidin-4(3H)-one (75 mg, 0.24 mmol) according to the procedure described for Example 1, Step E.
  • the crude product was purified by silica gel flash column chromatography (1:10 MeOH/EtOAc) to yield 120 (30 mg, 25%) as a white solid.
  • Step A Preparation of 5-bromo-3-(3-chlorobenzyl)pyrimidin-4(3H)-one:
  • Step B Preparation of 5-(4-(benzyloxy)-3-fluorophenyl)-3-(3- chlorobenzyl)pyrimidin-4(3H)-one: Prepared from 5-bromo-3-(3-chlorobenzyl)pyrimidin-4(3H)- one (0.41 g, 1.38 mmol) according to the procedure described for Example 7, Step A. The crude product was purified by silica gel flash column chromatography (1:1 EtOAc/Hexane) to yield the product (0.42 g, 72%) as a white solid. 1 H NMR (CDCl 3 , 400 MHz) ⁇ 8.15 (s, IH), 8.04 (s, IH), 111-03-PCT - P2338R1 - 02120.004WO1
  • Step C Preparation of 3-(3-chlorobenzyl)-5-(3-fluoro-4- hydroxyphenyl)pyrimidin-4(3H)-one: Prepared from 5-(4-(benzyloxy)-3-fluorophenyl)-3-(3- chlorobenzyl)pyrimidin-4(3H)-one (0.42 g, 1.0 mmol) according to the procedure described for Example 1, Step C, to yield the product (0.20 g, 61%) as a white solid.
  • LRMS (ESI pos) m/e 331 (M+l).
  • Step D Preparation of 3-(3-chlorobenzyl)-5-(4-(6,7-dimethoxyquinolin-4-yloxy)-
  • 3-fluorophenyl)pyrimidin-4(3H)-one Prepared from 4-chloro-6,7-dimethoxyquinoline (prepared according to reference procedure in Example 5) (64 mg, 0.29 mmol) and 3-(3-chlorobenzyl)-5-(3- fluoro-4-hydroxyphenyl)pyrimidin-4(3H)-one (95 mg, 0.30 mmol) according to the procedure described for Example 1, Step E.
  • the crude product was purified by silica gel flash column chromatography (1:10 MeOH/EtOAc) to yield 121 (20 mg, 13%) as a white solid.
  • Step A Preparation of 5-bromo-3-(2-fluorobenzyl)pyrimidin-4(3H)-one: Prepared from l-(bromomethyl)-2-fluorobenzene (1.58 g, 8.4 mmol) according to the procedure described for Example 13, Step A. The crude product was purified by silica gel flash column chromatography (1:1 EtOAc/Hexane) to yield the product (0.55 g, 23%) as a white solid. LRMS (ESI pos) m/e 283 (M+l).
  • Step B Preparation of 5-(4-(benzyloxy)-3-fluorophenyl)-3-(3- chlorobenzyl)pyrimidin-4(3H)-one: Prepared from 5-bromo-3-(2-fluorobenzyl)pyrimidin-4(3H)- one (0.55 g, 1.9 mmol) according to the procedure described for Example 7, Step A. The crude product was purified by silica gel flash column chromatography (1:1 EtOAc/Hexane) to yield the product (0.35 g, 44%) as a white solid. LRMS (ESI pos) m/e 405 (M+l).
  • Step C Preparation of 5-(3-fluoro-4-hydroxyphenyl)-3-(2- fluorobenzyl)pyrimidin-4(3H)-one: Prepared from 5-(4-(benzyloxy)-3-fluorophenyl)-3-(3- chlorobenzyl)pyrimidin-4(3H)-one (0.35 g, 8.6 mmol) according to the procedure described for Example 1, Step C, to yield the product (0.20 g, 74%) as a white solid.
  • LRMS (ESI pos) m/e 315 (M+l).
  • Step D Preparation of 5-(4-(6,7-dimethoxyquinolin-4-yloxy)-3-fluorophenyl)-3-
  • (2-fluorobenzyl)pyrimidin-4(3H)-one Prepared from 4-chloro-6,7-dimethoxyquinoline (prepared according to reference procedure in Example 5) (100 mg, 0.45 mmol) and 3-(3-chlorobenzyl)-5- (3-fluoro-4-hydroxyphenyl)pyrimidin-4(3H)-one (92 mg, 0.29 mmol) according to the procedure described for Example 1, Step E. The crude product was purified by silica gel flash column chromatography (1:10 MeOH/EtOAc) to yield 123 (41 mg, 28%) as a white solid.
  • Step A Preparation of 5-bromo-3-(4-fluorobenzyl)pyrimidin-4(3H)-one: Prepared from l-(bromomethyl)-4-fluorobenzene (1.58 g, 8.30 mmol) according to the procedure described for Example 13, Step A. The crude product was purified by silica gel flash column chromatography (1:1 EtOAc/Hexane) to yield the product (0.42 g, 17%) as a white solid. LRMS (ESI pos) m/e 283 (M+l).
  • Step B Preparation of 5-(4-(benzyloxy)-3-fiuorophenyl)-3-(4- fluorobenzyl)pyrimidin-4(3H)-one: Prepared from 5-bromo-3-(4-fiuorobenzyl)pyrimidin-4(3H)- one (0.42 g, 1.5 mmol) according to the procedure described for Example 7, Step A. The crude product was purified by silica gel flash column chromatography (1:1 EtOAc/Hexane) to yield the product (0.33 g, 55%) as a white solid. LRMS (ESI pos) m/e 405 (M+l).
  • Step C Preparation of 5-(3-fluoro-4-hydroxyphenyl)-3-(4- fluorobenzyl)pyrimidin-4(3H)-one: Prepared from 5-(4-(benzyloxy)-3-fluorophenyl)-3-(4- fluorobenzyl)pyrimidin-4(3H)-one (0.33 g, 0.82 mmol) according to the procedure described for Example 1, Step C, to yield the product (0.20 g, 78%) as a white solid.
  • LRMS (ESI pos) m/e 315 (M+l).
  • Step D Preparation of 5-(4-(6,7-dimethoxyquinolin-4-yloxy)-3-fluorophenyl)-3-
  • (4-fluorobenzyl)pyrimidin-4(3H)-one Prepared from 4-chloro-6,7-dimethoxyquinoline (prepared according to reference procedure in Example 5) (105 mg, 0.47 mmol) and 5-(3-fluoro-4- hydroxyphenyl)-3-(4-fluorobenzyl)pyrimidin-4(3H)-one (90 mg, 0.29 mmol) according to the procedure described for Example 1, Step E. The crude product was purified by silica gel flash column chromatography (1:10 MeOH/EtOAc) to yield 124 (35 mg, 24 %) as a white solid.
  • Step A Preparation of 5-bromo-3-(4-methylbenzyl)pyrimidin-4(3H)-one:
  • Step B Preparation of 5-(4-(benzyloxy)-3-fluorophenyl)-3-(4- methylbenzyl)pyrimidin-4(3H)-one: Prepared from 5-bromo-3-(4-methylbenzyl)pyrimidin- 4(3H)-one (0.14 g, 0.5 mmol) according to the procedure described for Example 7, Step A. The crude product was purified by silica gel flash column chromatography (1 :1 EtOAc/Hexane) to yield the product (0.15 g, 75%) as a white solid. LRMS (ESI pos) m/e 401 (M+l).
  • Step C Preparation of 5-(3-fluoro-4-hydroxyphenyl)-3-(4- methylbenzyl)pyrimidin-4(3H)-one: Prepared from 5-(4-(benzyloxy)-3-fluorophenyl)-3-(4- methylbenzyl)pyrimidin-4(3H)-one (0.15 g, 0.38 mmol) according to the procedure described for Example 1, Step C, to yield the product (0.10 g, 86%) as a white solid.
  • LRMS (ESI pos) m/e 311 (M+l).
  • Step D 5-(4-(6,7-dimethoxyquinolin-4-yloxy)-3-fluorophenyl)-3-(4- methylbenzyl)pyrimidin-4(3H)-one: Prepared from 4-chloro-6,7-dimethoxyquinoline (prepared according to reference procedure in Example 5) (105 mg, 0.50 mmol) and 5-(3-fluoro-4- hydroxyphenyl)-3-(4-methylbenzyl)pyrimidin-4(3H)-one (90 g, 0.29 mmol) according to the procedure described for Example 1, Step E.
  • Step A Preparation of 5-bromo-3-(3,4-dichlorobenzyl)pyrimidin-4(3H)-one:
  • Step B Preparation of 5-(4-(benzyloxy)-3-fluorophenyl)-3-(3,4- dichlorobenzyl)pyrimidin-4(3H)-one: Prepared from 5-bromo-3-(3,4-dichlorobenzyl)pyrimidin- 4(3H)-one (0.63 g, 0.20 mmol) according to the procedure described for Example 7, Step A. The crude product was purified by silica gel flash column chromatography (1:1 EtOAc/Hexane) to yield the product (0.11 g, 13%) as a white solid. LRMS (ESI pos) m/e 455 (M+l).
  • Step C Preparation of 3-(3,4-dichlorobenzyl)-5-(3-fluoro-4- hydroxyphenyl)pyrimidin-4(3H)-one: Prepared from 5-(4-(benzyloxy)-3-fluorophenyl)-3-(3,4- dichlorobenzyl)pyrimidin-4(3H)-one (0.11 g, 0.24 mmol) according to the procedure described for Example 1, Step C, to yield the product (50 mg, 56%) as a white solid.
  • Step D 3-(3,4-dichlorobenzyl)-5-(4-(6,7-dimethoxyquinolin-4-yloxy)-3- fluorophenyl)pyrimidin-4(3H)-one: Prepared from 4-chloro-6,7-dimethoxyquinoline (prepared according to reference procedure in Example 5) (60 mg, 0.14 mmol) and 3-(3,4-dichlorobenzyl)- 5-(3-fluoro-4-hydroxyphenyl)pyrimidin-4(3H)-one (50 mg, 0.1 mmol) according to the procedure described for Example 1, Step E.
  • Step A Preparation of 5-bromo-3-(4-(trifluoromethyl)benzyl)pyrimidin-4(3H)- one: Prepared from l-(bromomethyl)-4-(trifluoromethyl)benzene (2.0 g, 8.3 mmol) according to the procedure described for Example 13, Step A. The crude product was purified by silica gel flash column chromatography (1:1 EtOAc/Hexane) to yield the product (0.28 g, 9.8%) as a white solid. LRMS (ESI pos) m/e 333 (M+l).
  • Step B Preparation of 5-(4-(benzyloxy)-3-fluorophenyl)-3-(4-
  • Step D Preparation of 5-(4-(6,7-dimethoxyquinolin-4-yloxy)-3-fluorophenyl)-3-
  • (4-(trifluoromethyl)benzyl)pyrimidin-4(3H)-one Prepared from 4-chloro-6,7- dimethoxyquinoline (prepared according to reference procedure in Example 5) (100 mg, 0.45 mmol) and 5-(3-fluoro-4-hydroxyphenyl)-3-(4-(trifluoromethyl)benzyl)pyrimidin-4(3H)-one (100 mg, 0.22 mmol) according to the procedure described for Example 1, Step E. The crude product was purified by silica gel flash column chromatography (1:10 MeOH/EtOAc) to yield 127 (38 mg, 31 %) as a white solid.
  • Step A Preparation of 5-bromo-3-(4-chloro-2-fluorobenzyl)pyrimidin-4(3H)-one:
  • Step B Preparation of 5-(4-(benzyloxy)-3-fluorophenyl)-3-(4-chloro-2- fluorobenzyl)pyrimidin-4(3H)-one: Prepared from 5-bromo-3-(4-chloro-2- fluorobenzyl)pyrimidin-4(3H)-one (0.81 g, 2.5 mmol) according to the procedure described for Example 7, Step A. The crude product was purified by silica gel flash column chromatography (EtOAc) to yield the product (0.74 g, 66%) as a white solid. LRMS (ESI pos) m/e 439 (M+l).
  • Step C Preparation of 3-(4-chloro-2-fluorobenzyl)-5-(3-fluoro-4- hydroxyphenyl)pyrimidin-4(3H)-one: Prepared from 5-(4-(benzyloxy)-3-fluorophenyl)-3-(4- chloro-2-fluorobenzyl)pyrimidin-4(3H)-one (0.74 g, 1.7 mmol) according to the procedure described for Example 1, Step C, to yield the product (0.5 g, 85%) as a white solid.
  • LRMS (ESI pos) m/e 349 (M+l).
  • Step D Preparation of 3-(4-chloro-2-fluorobenzyl)-5-(4-(6,7-dimethoxyquinolin-
  • Step A Preparation of 5-bromo-3-(2-chloro-4-fluorobenzyl)pyrimidin-4(3H)-one:
  • Step B Preparation of 5-(4-(benzyloxy)-3-fluorophenyl)-3-(2-chloro-4- fiuorobenzyl)pyrimidin-4(3H)-one: Prepared from 5-bromo-3-(2-chloro-4- fluorobenzyl)pyrimidin-4(3H)-one (0.66 g, 2.1 mmol) according to the procedure described for Example 7, Step A. The crude product was purified by silica gel flash column chromatography (1:1 EtOAc/Hexane) to yield the product (0.84 g, 92%) as a white solid. LRMS (ESI pos) m/e 439 (M+l).
  • Step C Preparation of 3-(2-chloro-4-fluorobenzyl)-5-(3-fluoro-4- hydroxyphenyl)pyrimidin-4(3H)-one: Prepared from 5-(4-(benzyloxy)-3-fluorophenyl)-3-(2- chloro-4-fluorobenzyl)pyrimidin-4(3H)-one (0.84 g, 1.5 mmol) according to the procedure described for Example 1, Step C, to yield the product (0.5 g, 75%) as a white solid.
  • Step D Preparation of 3-(2-chloro-4-fluorobenzyl)-5-(4-(6,7-dimethoxyquinolin-
  • Example 30 Preparation of 3-(4-chloro-2,6-difluorobenzyl)-5-(4-(6,7- dimethoxyquinolin-4-yloxy)-3-fluorophenyl)pyrimidin-4(3H)-one 130 111-03-PCT - P2338R1 - 02120.0Q4WO1
  • Step A Preparation of 5-bromo-3-(4-chloro-2,6-difluorobenzyl)pyrimidin-4(3H)- one: Prepared from 2-(bromomethyl)-5-chloro-l,3-difluorobenzene (2.0 g, 8.3 mmol) according to the procedure described for Example 13, Step A. The crude product was purified by silica gel flash column chromatography (1:1 EtOAc/Hexane) to yield the product (0.76 g, 26%) as a white solid. LRMS (ESI pos) m/e 335 (M+l).
  • Step B Preparation of 5-(4-(benzyloxy)-3-fluorophenyl)-3-(4-chloro-2,6- difluorobenzyl)pyrimidin-4(3H)-one: Prepared from 5-bromo-3-(4-chloro-2,6- difluorobenzyl)pyrimidin-4(3H)-one (0.76 g, 2.2 mmol) according to the procedure described for Example 7, Step A. The crude product was purified by silica gel flash column chromatography (1:1 EtOAc/Hexane) to yield the product (0.52 g, 50%) as a white solid. LRMS (ESI pos) m/e 457 (M+l).
  • Step C Preparation of 3-(4-chloro-2,6-difluorobenzyl)-5-(3-fluoro-4- hydroxyphenyl)pyrimidin-4(3H)-one: Prepared from 5-(4-(benzyloxy)-3-fluorophenyl)-3-(4- chloro-2,6-difluorobenzyl)pyrimidin-4(3H)-one (0.52 g, 1.14 mmol) according to the procedure described for Example 1, Step C, to yield the product (0.4 g, 97%) as a white solid.
  • LRMS (ESI pos) m/e 367 (M+l).
  • Step D Preparation of 3-(4-chloro-2,6-difluorobenzyl)-5-(4-(6,7- dimethoxyquinolin-4-yloxy)-3-fluorophenyl)pyrimidin-4(3H)-one: Prepared from 4-chloro-6,7- dimethoxyquinoline (prepared according to reference procedure in Example 5) (100 mg, 0.45 mmol) and 3-(4-chloro-2,6-difluorobenzyl)-5-(3-fluoro-4-hydroxyphenyl)pyrimidin-4(3H)-one (164 mg, 0.45 mmol) according to the procedure described for Example 1, Step E.
  • Step A Preparation of 5-bromo-3-(3,4-dimethylbenzyl)pyrimidin-4(3H)-one:
  • Step B Preparation of 5-(4-(benzyloxy)-3-fluorophenyl)-3-(3,4- dimethylbenzyl)pyrimidin-4(3H)-one: Prepared from 5-bromo-3-(3,4-dimethylbenzyl)pyrimidin- 4(3H)-one (0.68 g, 2.3 mmol) according to the procedure described for Example 7, Step A. The crude product was purified by silica gel flash column chromatography (1:1 EtOAc/Hexane) to yield the product (0.17 g, 17%) as a white solid. LRMS (ESI pos) m/e 415 (M+l).
  • Step C Preparation of 3-(3,4-dimethylbenzyl)-5-(3-fluoro-4- hydroxyphenyl)pyrimidin-4(3H)-one: Prepared from 5-(4-(benzyloxy)-3-fluorophenyl)-3-(3,4- dimethylbenzyl)pyrimidin-4(3H)-one (0.17 g, 0.4 mmol) according to the procedure described for Example 1, Step C, to yield the product (0.1 g, 77%) as a white solid.
  • LRMS (ESI pos) m/e 325 (M+l).
  • Step D Preparation of 5-(4-(6,7-dimethoxyquinolin-4-yloxy)-3-fIuorophenyl)-3-
  • (3,4-dimethylbenzyl)pyrimidin-4(3H)-one Prepared from 4-chloro-6,7-dimethoxyquinoline (prepared according to reference procedure in Example 5) (100 mg, 0.45 mmol) and 3-(3,4- dimethylbenzyl)-5-(3-fluoro-4-hydroxyphenyl)pyrimidin-4(3H)-one (145 mg, 0.45 mmol) according to the procedure described for Example 1, Step E. The crude product was purified by silica gel flash column chromatography (1:10 MeOH/EtOAc) to yield 131 (2 mg, 1%) as a white solid.
  • Step A Preparation of 5-bromo-3-(4-fluoro-3-methylbenzyl)pyrimidin-4(3H)- one: Prepared from 4-(bromomethyl)-l-fluoro-2-methylbenzene (0.57 g, 2.8 mmol) according to the procedure described for Example 13, Step A. The crude product was purified by silica gel flash column chromatography (1:1 EtOAc/Hexane) to yield the product (0.46 g, 54%) as a white solid. LRMS (ESI pos) m/e 296 (M+l).
  • Step B Preparation of 5-(4-(benzyloxy)-3-fluorophenyl)-3-(4-fluoro-3- methylbenzyl)pyrimidin-4(3H)-one: Prepared from 5-bromo-3-(4-fluoro-3- methylbenzyl)pyrimidin-4(3H)-one (0.46 g, 1.5 mmol) according to the procedure described for Example 7, Step A. The crude product was purified by silica gel flash column chromatography (1:1 EtOAc/Hexane) to yield the product (86 mg, 13%) as a white solid. LRMS (ESI pos) m/e 419 (M+l).
  • Step C Preparation of 3-(4-fluoro-3-methylbenzyl)-5-(3-fluoro-4- hydroxyphenyl)pyrimidin-4(3H)-one: Prepared 5-(4-(benzyloxy)-3-fluorophenyl)-3-(4-fluoro-3- methylbenzyl)pyrimidin-4(3H)-one (86 mg, 0.2 mmol) according to the procedure described for Example 1, Step C, to yield the product (50 mg, 74%) as a white solid.
  • Step D Preparation of 5-(4-(6,7-dimethoxyquinolin-4-yloxy)-3-fluorophenyl)-3-
  • (4-fluoro-3-methylbenzyl)pyrimidin-4(3H)-one Prepared from 4-chloro-6,7-dimethoxyquinoline (prepared according to reference procedure in Example 5) (40 mg, 0.18 mmol) and 3-(4-fluoro- 3-methylbenzyl)-5-(3-fluoro-4-hydroxyphenyl)pyrimidin-4(3H)-one (59 mg, 0.18 mmol) according to the procedure described for Example 1, Step E. The crude product was purified by silica gel flash column chromatography (1:10 MeOH/EtOAc) to yield 132 (1.6 mg, 2%) as a white solid.
  • Step A Preparation of (5-bromo-2-methoxypyridin-4-yl)(phenyl)methanol:
  • Step B Preparation of (5-bromo-2-methoxypyridin-4-yl)(phenyl)methanone: PCC
  • Step C Preparation of 4-benzoyl-5-bromopyridin-2(lH)-one: (5-bromo-2- methoxypyridin-4-yl)(phenyl)methanone (1.3 g, 4.5 mmol) and pyridine hydrochloride (2 g, 17 mmol) were heated at 150 0 C for 1 hour. CH 2 Cl 2 (30 mL) was added into the hot mixture. The mixture was cooled and the solvent was evaporated. The residue was purified by silica gel flash column chromatography (2:1 EtOAc/Hexane) to yield the product (0.3 g, 24%) as a white solid.
  • Step D Preparation of 4-benzoyl-l-(3-fluoro-4-hydroxyphenyl)pyridin-2(lH)- one: Prepared from 4-benzoyl-5-bromopyridin-2(lH)-one (170 mg, 0.61 mmol) according to the procedure described for Example 1, Step D. The crude product was purified by silica gel flash column chromatography (EtOAc) to yield the product (170 mg, 90%) as a brown solid.
  • EtOAc silica gel flash column chromatography
  • Step E Preparation of 4-benzoyl-l-(4-(6,7-dimethoxyquinolin-4-yloxy)-3- fluorophenyl)pyridin-2(lH)-one: Prepared from 4-chloro-6,7-dimethoxyquinoline (prepared 111-03-PCT - P2338R1 - 02120.004WO1
  • Example 34 60 mg, 0.16 mmol
  • the crude product was purified by silica gel flash column chromatography (EtOAc) to yield 139 (40 mg, 53 %) as a white solid.
  • Step A Preparation of l-(3-fluoro-4-hydroxyphenyl)-4-methylpyridin-2(lH)-one:
  • Step B Preparation of l-(4-(6,7-dimethoxyquinolin-4-yloxy)-3-fluorophenyl)-4- methylpyridin-2(lH)-one: Prepared from l-(3-fluoro-4-hydroxyphenyl)-4-methylpyridin-2(lH)- one (0.1 g, 0.46 mmol) and 4-chloro-6,7-dimethoxyquinoline (prepared according to reference procedure in Example 5) (0.12 g, 0.55 mmol) using the procedure described for Example 1, Step E. The reaction mixture was purified by silica gel flash column chromatography (1:10 MeOH/EtOAc) to yield 140 (50 mg, 27%) as a white solid.
  • Step A Preparation of 5-(4-(benzyloxy)-3-fluorophenyl)pyrimidin-4(3H)-one:
  • Step B Preparation of 5-(3-fluoro-4-hydroxyphenyl)pyrimidin-4(3H)-one:
  • Step C Preparation of 5-(4-(6,7-dimethoxyquinolin-4-yloxy)-3- fluorophenyl)pyrimidin-4(3H)-one: Prepared from 5-(3-fluoro-4-hydroxyphenyl)pyrimidin- 4(3H)-one (40 mg, 0.19 mmol) and 4-chloro-6,7-dimethoxyquinoline (prepared according to reference procedure in Example 5) (43 mg, 0.19 mmol) according to the procedure described for Example 1, Step E, to yield 143 (1 mg, 1%) as white solid.
  • Step A Preparation of 2-amino-N-(3-fluoro-4-methoxyphenyl)benzamide: To a stirred suspension of isatoic anhydride (1.63 g, 10 mmol) in 15 mL dioxane at room temperature 111-03-PCT - P2338R1 - 02120.004WO1
  • Step B Preparation of N-(2-aminobenzyl)-3-fluoro-4-methoxyaniline: To a stirred suspension of lithium aluminum hydride (121 mg, 3.2 mmol) in 2 mL dioxane at reflux under nitrogen was added 2-amino-N-(3-fluoro-4-methoxyphenyl)benzamide (260 mg, 1 mmol) as a solution in 2 mL dioxane. A vigorous reaction was evident. After refluxing overnight the reaction was cooled to room temperature and quenched by sequential treatment with H 2 O (150 uL), 15% NaOH (150 uL) and H 2 O (450 uL).
  • Step D Preparation of 3-(3-fluoro-4-hydroxyphenyl)-3,4-dihydroquinazolin-
  • Step E Preparation of 3-(3-fluoro-4-(6-methoxy-7-(3- morpholinopropoxy)quinolin-4-yloxy)phenyl)-3,4-dihydroquinazolin-2(lH)-one.
  • Step E substituting 3-(3-fiuoro-4-hydroxyphenyl)-3,4- dihydroquinazolin-2( 1 H)-one for 3 -(3 -fluoro-4-hydroxyphenyl)-5 -methyl-6-(2- methylbenzyl)pyrimidin-4(3H)-one to give 145 after silica gel chromatography as a white foam (23 mg, 53% yield).
  • Step A Preparation of 4-(2-fluoro-4-(piperazin-l-yl)phenoxy)-6,7- dimethoxyquinoline: Pd2(dba)3 (20 mg) was added into a suspension of 4-(4-bromo-2- fluorophenoxy)-6,7-dimethoxyquinoline (Example 34, 100 mg, 0.264 mmol), piperazine (228 mg, 2.64 mmol), Xanthphos (100 mg) and potassium phosphate (200 mg) in toluene (10 mL). The reaction mixture was heated to 100 0 C for 10 hours. The reaction mixture was filtered through a pad of silica gel.
  • Step B Preparation of (4-(4-(6,7-dimethoxyquinolin-4-yloxy)-3- fluorophenyl)piperazin-l-yl)(phenyl)methanone: Benzoyl chloride (11 mg, 0.078 mmol) and triethylamine (0.5 mL) were added into a solution of 4-(2-fluoro-4-(piperazin-l-yl)phenoxy)-6,7- dimethoxyquinoline (30 mg, 0.078 mmol) in CH 2 Cl 2 . After a few minutes, water (1 mL) and CH 2 Cl 2 (2 mL) were added.
  • Step A Preparation of 3-benzyl-5-(4-(benzyloxy)-3-fluorophenyl)pyrimidine-
  • Step B 3-benzyl-5-(3-fluoro-4-hydroxyphenyl)pyrimidine-4(3H)-thione:
  • Step C Preparation of 3-benzyl-5-(4-(6,7-dimethoxyquinolin-4-yloxy)-3- fluorophenyl)pyrimidine-4(3H)-thione: Prepared from 4-chloro-6,7-dimethoxyquinoline (prepared according to reference procedure in Example 5) (40 mg, 0.18 mmol) and 3-(4- chlorobenzyl)-5-(3-fluoro-4-hydroxyphenyl)pyrimidine-4(3H)-thione (62 mg, 0.18 mmol) according to the procedure described for Example 1, Step E to yield 148 (40 mg, 45%) as yellow solid.
  • Step A Preparation of 3-benzyl-5-(4-(benzyloxy)-3-fluorophenyl)pyrimidin-
  • the reaction mixture was heated at 100 0 C for 2 hours, cooled and poured into water (10 mL).
  • the reaction mixture was extracted with ethyl acetate, and the organic layer was washed with brine, dried over sodium sulfate, filtered, and concentrated.
  • the crude product was purified by silica gel flash column chromatography (2:1 EtOAc/Hexane) to yield the product (1.4 g, 32%) as a white solid.
  • Step B Preparation of 3-benzyl-5-(3-fluoro-4-hydroxyphenyl)pyrimidin-4(3H)- one: Prepared from 3-benzyl-5-(4-(benzyloxy)-3-fluorophenyl)pyrimidin-4(3H)-one (0.3 g, 0.8 mmol) according to the procedure described for Example 1, Step C, to yield the product (0.2 g, 87%) as a white solid.
  • LRMS (ESI pos) m/e 297 (M+l). 111 -03-PCT - P2338R 1 - 02120.004WO1
  • Step C Preparation of 3-benzyl-5-(4-(7 ⁇ (benzyloxy)-6-methoxyquinolin-4- yloxy)-3-fluorophenyl)pyrimidin-4(3H)-one: Prepared from 7-(benzyloxy)-4-chloro-6- methoxyquinoline (prepared according to WO 2005/030140, Example32, 200 mg, 0.67 mmol) and 3-benzyl-5-(3-fluoro-4-hydroxyphenyl)pyrimidin-4(3H)-one (198 mg, 0.48 mmol) according to the procedure described for Example 1, Step E. The crude product was purified by silica gel flash column chromatography (1:10 MeOH/EtOAc) to yield 149 (100 mg, 27%) as a white solid. LRMS (ESI pos) m/e 560 (M+l).
  • Step A Preparation of 5-bromo-3-(4-tert-butylbenzyl)pyrimidin-4(3H)-one:
  • Step B Preparation of 5-(4-(benzyloxy)-3-fluorophenyl)-3-(4-tert- butylbenzyl)pyrimidin-4(3H)-one: Prepared from 5-bromo-3-(4-tert-butylbenzyl)pyrimidin- 4(3H)-one (0.65 mg, 2.0 mmol) according to the procedure described for Example 7, Step A. The crude product was purified by silica gel flash column chromatography (1:1 EtOAc/Hexane) to yield the product (0.50 g, 56%) as a white solid. LRMS (ESI pos) m/e 443 (M+l). 111-03-PCT - P2338R1 - 02120.004WO1
  • Step C Preparation of 3-(4-tert-butylbenzyl)-5-(3-fluoro-4- hydroxyphenyl)pyrimidin-4(3H)-one: Prepared from 5-(4-(benzyloxy)-3-fluorophenyl)-3-(4-tert- butylbenzyl)pyrimidin-4(3H)-one (0.50 g, 1.1 mmol) according to the procedure described for Example I 5 Step C, to yield the product (0.30 g, 88%) as a white solid.
  • Step D Preparation of 3-(4-tert-butylbenzyl)-5-(4-(6,7-dimethoxyquinolin-4- yloxy)-3-fluorophenyl)pyrimidin-4(3H)-one: Prepared from 4-chloro-6,7-dimethoxyquinoline (prepared according to reference procedure in Example 5) (100 mg, 0.46 mmol) and 3-(4-tert- butylbenzyl)-5-(3-fluoro-4-hydroxyphenyl)pyrimidin-4(3H)-one (130 mg, 0.30 mmol) according to the procedure described for Example 1, Step E.
  • Step A Preparation of 5-bromo-3-(4-chloro-3-(trifluoromethyl)benzyl)pyrimidin-
  • Step B Preparation of 5-(4-(benzyloxy)-3-fiuorophenyl)-3-(4-chloro-3-
  • Step C Preparation of 3-(4-chloro-3-(trifiuoromethyl)benzyl)-5-(3-fluoro-4- hydroxyphenyl)pyrimidin-4(3H)-one: Prepared from 5-(4-(benzyloxy)-3-fluorophenyl)-3-(4- chloro-3-(trifluoromethyl)benzyl)pyrimidin-4(3H)-one (0.50 g, 1.0 mmol) according to the procedure described for Example 1, Step C, to yield the product (0.40 g, 86%) as a white solid.
  • LRMS (ESI pos) m/e 331 (M+l).
  • Step D Preparation of 3-(4-chloro-3-(trifluoromethyl)benzyl)-5-(4-(6,7- dimethoxyquinolin-4-yloxy)-3-fluorophenyl)pyrimidin-4(3H)-one: Prepared from 4-chloro-6,7- dimethoxyquinoline (prepared according to reference procedure in Example 5) (60 mg, 0.27 mmol) and 3-(4-chloro-3-(trifluoromethyl)benzyl)-5-(3-fluoro-4-hydroxyphenyl)pyrimidin- 4(3H)-one(107 mg, 0.27 mmol) according to the procedure described for Example 1, Step E.
  • Step A Preparation of (4-benzylpiperazm-l-yl)(3-fiuoro-4- methoxyphenyl)methanone: Prepared from 1-benzylpiperazine (2.3 mg, 13 mmol) ) according to the procedure described for Example 4, Step A, to yield the product (2.6 g, 65%) as a white solid.
  • Step B Preparation of (4-benzylpiperazin-l-yl)(3-fluoro-4- hydroxyphenyl)methanone: Prepared from (4-benzylpiperazin-l-yl)(3-fluoro-4- methoxyphenyl)methanone (2.0 g, 6.1 mmol) according to the procedure described for Example 4, Step B, to yield the product (1.0 g, 53%) as a white solid.
  • Step C Preparation of 5 (4-benzylpiperazin-l-yl)(4-(6,7-dimethoxyquinolin-4- yloxy)-3-fluorophenyl)methanone: Prepared from (4-benzylpiperazin-l-yl)(3-fluoro-4- hydroxyphenyl)methanone (18 mg, 0.059 mmol) and 4-chloro-6-methoxy-7-(3- morpholinopropoxy)quinoline (20 mg, 0.059 mmol) according to the procedure described for Example 1, Step E, to yield 153 (40 mg, 18% yield) as a white solid.
  • Step A Preparation of (E)-ethyl 3-amino-2-(4-(benzyloxy)phenylcarbamoyl)-4- phenylbut-2-enoate: A suspension of (E)-ethyl 3-amino-4-phenylbut-2-enoate (1 g, 4.9 mmol) and l-((4-isocyanatophenoxy)methyl)benzene (1.1 g, 4.9 mmol) was heated in DMF (20 mL) at 6O 0 C for 72 hours. The reaction mixture was poured into water (10 mL) and removed the solid by filtration.
  • Step B Preparation of ethyl 4-benzyl-l-(4-(benzyloxy)phenyl)-6-oxo-l,6- dihydropyrimidine-5-carboxylate: Acetic anhydride (1 ml, 10.6 mmol) was added into a solution of (E)-ethyl 3-amino-2-((4-(benzyloxy)phenyl)carbamoyl)-4-phenylbut-2-enoate (1 g, 2.32 mmol) and triethyl orthroformate (5 ml, 30.1 mmol). The reaction was heated at HO 0 C for 16 hours, cooled and solvent was evaporated.
  • Step C Preparation of 6-benzyl-3-(4-hydroxyphenyl)pyrimidin-4(3H)-one: Ethyl
  • Step D Preparation of 6-benzyl-3-(4-(6-methoxy-7-(3- morpholinopropoxy)quinolin-4-yloxy)phenyl)pyrimidin-4(3H)-one: Prepared from 4-chloro-6- methoxy-7-(3-morpholinopropoxy)quinoline (34 mg, 0.10 mmol) and 6-benzyl-3-(4- hydroxyphenyl)pyrimidin-4(3H)-one (28 mg, 0.10 mmol) according to the procedure described for Example 1, Step E. The residue was purified by silica gel flash column chromatography column (1:10 MeOH/EtOAc) to yield 154 (20 mg, 34%) as a yellow solid.
  • Step A preparation of (4,6-dichloropyrimidin-5-yl)(phenyl)methanol:
  • Phenylmagnesium bromide (11 ml, 11 mmol) was added into a solution of 4,6-Dichloro-5- pyrimidinecarbaldehyde (2 g, 11 mmol) in THF (30 mL) at -78 0 C. The reaction was allowed to warm to room temperature and was poured into water (10 mL). The reaction mixture was extracted with EtOAc, and the organic layer was washed with brine, dried over sodium sulfate, filtered, and concentrated. The residue was purified by silica gel flash column chromatography (1:10 Et 2 O/Hexane) to yield the product (2.4 g, 83% yield) as a white solid.
  • IH NMR (CDCl 3 , 400 MHz) ⁇ 8.76 (s, IH), 7.20-7.41 (m, 5H), 6.50 (d, IH), 3.00 (d, IH).
  • Step B Preparation of (4-(benzyloxy)-6-chloropyrimidin-5-yl)(phenyl)methanol:
  • Step C Preparation of 5-benzylpyrimidin-4-ol: Prepared from 5-benzylpyrimidin-
  • Step D Preparation of 5-benzyl-3-(3-fluoro-4-hydroxyphenyl)pyrimidin-4(3H)- one: Prepared from 5-benzylpyrimidin-4-ol (0.1 g, 0.54 mmol) according to the procedure described for Example 1, Step D, to yield the product (20 mg, 13 % yield) as a brown solid.
  • LRMS (ESI pos) m/e 279 (M+l).
  • Step E Preparation of 5-benzyl-3-(3-fiuoro-4-(6-methoxy-7-(3- morpholinopropoxy)quinolin-4-yloxy)phenyl)pyrimidin-4(3H)-one: Prepared from 5-benzyl-3- (3-fluoro-4-hydroxyphenyl)pyrimidin-4(3H)-one (18 mg, 0.059 mmol) according to the procedure described for Example 1, Step E, to yield 155 (1 mg, 2.8% yield) as a light brown solid. LRMS (ESI pos) m/e 597 (M+l).
  • Step A Preparation of 2-benzyl-4-methoxypyrimidine: A solution of 2-chloro-4- methoxypyrimidine (0.500 g, 3.46 mmol) and PdCl 2 (PPh 3 ) 2 (0.121 g, 0.173 mmol) in THF (10 mL) was sparged with N 2 . Benzylzinc(II) bromide (8.30 ml, 4.15 mmol; 0.5 M solution in THF) was added and the reaction mixture was stirred at reflux for 1 hour. The reaction mixture was cooled to room temperature and then partitioned between EtOAc and H 2 O. The phases were separated, and the aqueous phase was re-extracted with EtOAc (Ix).
  • Step B Preparation of 2-benzylpyrimidin-4(3H)-one: To a solution of 2-benzyl-
  • Step C Preparation of 2-benzyl-5-bromopyrimidin-4(3H)-one: To a solution of
  • Step E Preparation of 2-benzyl-5-(3-fluoro-4-hydroxyphenyl)pyrimidin-4(3H)- one: A suspension of 2-benzyl-5-(4-(benzyloxy)-3-fluorophenyl)pyrimidin-4(3H)-one (0.284 g, 0.735 mmol) in trifluoroacetic acid (3 mL) was stirred at 40°C for 2 hours and then at room temperature for 16 hours. The reaction mixture was concentrated to dryness and then purified by silica gel flash column chromatography, eluting with 20:1 dichloromethane/MeOH. The desired product (0.177 g, 81%) was obtained as a white solid.
  • Step F Preparation of 2-benzyl-5-(3-fluoro-4-(6-methoxy-7-(3- morpholinopropoxy)quinolin-4-yloxy)phenyl)pyrimidin-4(3H)-one: To a solution of 2-benzyl-5- (3-fluoro-4-hydroxyphenyl)pyrimidin-4(3H)-one (0.027 g; 0.091 mmol) in toluene (500 uL) in a microwave tube was added 4-(3-(4-chloro-6-methoxyquinolin-7-yloxy)propyl)morpholine (prepared according to the reference in Example 1, step E) (0.031 g, 0.091 mmol) and DMAP (0.011 g, 0.091 mmol).
  • the reaction was stirred at 180°C in the microwave for 2 hours.
  • the mixture was solubilized with a small amount of MeOH and purified by silica gel flash column chromatography, eluting with 9:1 EtOAc/MeOH.
  • the desired product eluted along with residual 4-(3-(4-chloro-6-methoxyquinolin-7-yloxy)propyl)morpholine.
  • the mixture was concentrated and repurified by silica gel flash column chromatography, eluting with a step gradient of CH 2 Cl 2 (150 mL), 2.5/97.5 MeOH/CH 2 Cl 2 (200 mL) and 5/95 MeOH/CH 2 Cl 2 (500 mL) to give 156 (0.022 g, 40%) as a tan foamy solid.
  • Step A Preparation of 2-benzyl-5-(4-(benzyloxy)phenyl)pyrimidin-4(3H)-one:
  • Step C Preparation of 2-benzyl-5-(4-(6-methoxy-7-(3- morpholinopropoxy)quinolin-4-yloxy)phenyl)pyrimidin-4(3H)-one: To a stirred suspension of 2- benzyl-5-(4-hydroxyphenyl)pyrimidin-4(3H)-one (0.021 g, 0.076 mmol) in bromobenzene (700 ⁇ L) at room temperature under N 2 was added 4-(3-(4-chloro-6-methoxyquinolin-7- yloxy)propyl)morpholine (prepared according to the referenced procedure in Example 1, step E) (0.028 g, 0.083 mmol) followed by DMAP (0.001 g, 0.008 mmol).
  • Step A Preparation of 5-bromo-2-chloropyrimidin-4(3H)-one: Prepared from 5- bromo-2,4-dichloropyrimidine (10.00 g, 43.88 mmol) according to the procedure described in EP 1506967. The desired product (4.59 g, 50%) was obtained as a pale yellow solid.
  • 1 H-NMR 400 MHz, DMSO-d 6 ) ⁇ 8.36 (s, IH).
  • LRMS ESI neg
  • m/e 207, 209 M-, Br pattern).
  • Step B Preparation of 5-bromo-2-chloro-3-methylpyrimidin-4(3H)-one: To a solution of 5-bromo-2-chloropyrimidin-4(3H)-one (1.00 g, 4.78 mmol) in DME (12 mL)/DMF (3 mL) under N 2 at 0 0 C was added LiH (0.044 g, 5.25 mmol) and the reaction was stirred at room temperature for 15 minutes. Iodomethane (0.589 ml, 9.45 mmol) was then added and the reaction was stirred at room temperature for 30 minutes and then at 60 0 C for 1.5 hours.
  • LiH 0.044 g, 5.25 mmol
  • Step C Preparation of 2-benzyl-5-bromo-3-methylpyrimidin-4(3H)-one: A solution of 5-bromo-2-chloro-3-methylpyrimidin-4(3H)-one (0.100 g, 0.448 mmol) and PdCl 2 (PPh 3 ) 2 (0.016 g, 0.022 mmol) in THF (4 mL) was sparged with N 2 . Benzylzinc(II) bromide (0.904 ml, 0.452 mmol; 0.5 M solution in THF) was added and the reaction mixture was stirred at reflux for 30 minutes. The reaction mixture was cooled to room temperature and then partitioned between EtOAc and H 2 O.
  • Step D Preparation of 2-benzyl-5-(4-(benzyloxy)-3-fluorophenyl)-3- methylpyrimidin-4(3H)-one: Prepared from 2-benzyl-5-bromo-3-methylpyrimidin-4(3H)-one (0.067 g, 0.240 mmol) according to the procedure described in Step D of Example 56. The crude product was purified by silica gel flash column chromatography, eluting with 10:1 dichloromethane/EtOAc. The desired product (0.082 g, 85%) was obtained as an off-white solid.
  • 1 H-NMR 400 MHz, DMSOd 6 ) ⁇ 8.15 (s, IH), 7.66 (dd, IH), 7.51 (m, IH), 7.47 (m, 2H), 7.43-
  • Step E Preparation of 2-benzyl-5-(3-fluoro-4-hydroxyphenyl)-3- methylpyrimidin-4(3H)-one: Prepared from 2-benzyl-5-(4-(benzyloxy)-3-fiuorophenyl)-3- methylpyrimidin-4(3H)-one (0.082 g, 0.20 mmol), according to the procedure described in Step E of Example 56. The desired product (0.064 g, 100%) was obtained as a pale yellow foamy solid.
  • Step F Preparation of 2-benzyl-5-(3-fluoro-4-(6-methoxy-7-(3- morpholinopropoxy)quinolin-4-yloxy)phenyl)-3-methylpyrimidin-4(3H)-one: Prepared from 2- benzyl-5-(3-fluoro-4-hydroxyphenyl)-3-methylpyrimidin-4(3H)-one (0.025 g, 0.081 mmol), 4-(3- (4-chloro-6-methoxyquinolin-7-yloxy)propyl)morpholine (0.030 g, 0.089 mmol) and catalytic DMAP according to the procedure described in Step C of Example 58, to give 159 (0.006 g, 12%) as a yellow solid.
  • Step A Preparation of 4-chloro-6-methoxy-5-methylpyrimidine: To a solution of 4,6-dichloro-5-methylpyrimidine (1.00 g, 6.13 mmol) in MeOH (50 mL) at 0°C was added solid sodium methoxide (0.348 g, 6.44 mmol) in portions. The reaction mixture was allowed to warm to room temperature and stirred at room temperature for 4 hours and then at 50 0 C for 12 hours. Additional sodium methoxide (0.348 g, 6.44 mmol) was added and the reaction mixture was stirred at 50°C for 4 hours.
  • Step B Preparation of 5-methyl-6-(phenylamino)pyrimidin-4(3H)-one:
  • a sealed tube was a mixture of 4-chloro-6-methoxy-5-methylpyrimidine (0.973 g, 6.14 mmol) and aniline (1.679 ml, 18.41 mmol) in n-BuOH (10 mL).
  • the reaction mixture was stirred at reflux for 5 days and then at room temperature for 5 days.
  • the reaction mixture was a purple suspension after cooling.
  • the resulting solid was filtered and washed with Et 2 O, collected and dried under vacuum.
  • the filtrate was concentrated, dried under vacuum and the trituration procedure was repeated with dichloromethane/Et 2 O.
  • Step C Preparation of 3-(4-(benzyloxy)phenyl)-5-methyl-6-
  • the secondary ligand Nl,N2-dimethylethane-l,2-diamine (0.0132 ml, 0.124 mmol) and additional l-(benzyloxy)-4-iodobenzene (0.020 g, 0.065 mmol) were added.
  • the reaction mixture was then stirred at 110 0 C for an additional 16 hours.
  • the reaction mixture was partitioned between EtOAc and saturated aqueous NaCl. The phases were separated, and the aqueous phase was re-extracted with EtOAc (Ix). The combined organic layers were dried (Na 2 SO 4 ), filtered and concentrated.
  • Step D Preparation of 3-(4-hydroxyphenyl)-5-methyl-6-
  • Step E Preparation of 3-(4-(6-methoxy-7-(3-morpholinopropoxy)quinolin-4- yloxy)phenyl)-5-methyl-6-(phenylamino)pyrimidin-4(3H)-one: Prepared from 3-(4- hydroxyphenyl)-5-methyl-6-(phenylamino)pyrimidin-4(3H)-one (0.019 g, 0.063 mmol), 4-(3-(4- chloro-6-methoxyquinolin-7-yloxy)propyl)morpholine (0.023 g, 0.069 mmol) and catalytic DMAP according to the procedure described in Step C of Example 58, to give 160 (0.026 g, 69%) as a pale yellow foamy solid.
  • Step A Preparation of 3-(3-fluoro-4-hydroxyphenyl)-5-methyl-6-
  • the mixture was flushed with N 2 and stirred at 11O 0 C for 16 hours.
  • the reaction mixture was partitioned between EtOAc and saturated aqueous NaCl. The phases were separated, and the aqueous phase was re-extracted with EtOAc (2x). The combined organic layers were dried (Na 2 SO 4 ), filtered and concentrated.
  • the crude product was purified by silica gel flash column chromatography, eluting with 20:1 dichloromethane/MeOH. The desired product (0.069 g, 45%) was obtained as a brown solid.
  • Step B Preparation of 3-(3-fluoro-4-(6-methoxy-7-(3- morpholinopropoxy)quinolin-4-yloxy)phenyl)-5-methyl-6-(phenylamino)pyrimidin-4(3H)-one: Prepared from 3-(3-fluoro-4-hydroxyphenyl)-5-methyl-6-(phenylamino)pyrimidin-4(3H)-one (0.025 g, 0.0803 mmol), 4-(3-(4-chloro-6-methoxyquinolin-7-yloxy)propyl)morpholine (0.0298 g, 0.0883 mmol) and catalytic DMAP according to the procedure described in Step C of Example 58, to give 161 (0.035 g, 71%) as a pale yellow foamy solid.
  • Step A Preparation of 4-methoxy-N-phenylpyrimidin-2-amine:
  • 2-chloro-4-methoxypyrimidine (1.00 g, 6.92 mmol) in 2-propanol (5 mL).
  • Aniline (0.757 ml, 8.30 mmol) and DIEA (1.45 ml, 8.30 mmol) were added and the reaction mixture was heated at 100°C until the reaction was complete by HPLC.
  • the reaction mixture was cooled to room temperature.
  • the resulting thick suspension was filtered, washed with ethanol, collected and dried under vacuum to yield the desired product (0.164 g) as a white solid.
  • Step B Preparation of 2-(phenylamino)pyrimidin-4(3H)-one: To a solution of 4- methoxy-N-phenylpyrimidin-2 -amine (0.632 g, 3.14 mmol) in acetic acid (20 mL) was added HBr (2.132 ml, 18.84 mmol; 48 wt % in H 2 O). The reaction mixture was heated at 90-95°C for 3 hours. The reaction mixture was cooled to room temperature and diluted with H 2 O. The pH of the reaction mixture was adjusted to 5-6 with 6 M aqueous NaOH which resulted in the formation of a solid precipitate.
  • Step C Preparation of 3-methyl-2-(phenylamino)pyrimidin-4(3H)-one.
  • 2-(phenylamino)pyrimidin-4(3H)-one (0.250 g, 1.34 mmol) in DMF (10 mL) was added LiH (0.012 g, 1.47 mmol).
  • iodomethane (0.166 ml, 2.67 mmol) was added.
  • the reaction was stirred at room temperature for 18 hours.
  • the reaction mixture was quenched with H 2 O and then partitioned between EtOAc and 111-03-PCT - P2338R1 - 02120.004WO1
  • Step D Preparation of 5-bromo-3-methyl-2-(phenylamino)pyrimidin-4(3H)-one:
  • Step F Preparation of 5-(3-fluoro-4-hydroxyphenyl)-3-methyl-2-
  • Step G Preparation of 5-(3-fluoro-4-(6-methoxy-7-(3- morpholinopropoxy)quinolin-4-yloxy)phenyl)-3-methyl-2-(phenylamino)pyrimidin-4(3H)-one: Prepared from 5-(3-fluoro-4-hydroxyphenyl)-3-methyl-2-(phenylamino)pyrimidin-4(3H)-one (0.025 g, 0.0803 mmol), 4-(3-(4-chloro-6-methoxyquinolin-7-yloxy)propyl)morpholine (0.0298 g, 0.0883 mmol) and catalytic DMAP according to the procedure described in Step C of Example 58, to give 162 (0.029, 59%) as a pale yellow foamy solid.
  • Step A Preparation of 5-bromo-2-(cyclopropylmethylamino)-3- methylpyrimidin-4(3H)-one: A mixture of 5-bromo-2-chloro-3-rnethylpyrimidin-4(3H)-one (0.100 g, 0.45 mmol; obtained from Example 59, Step B), cyclopropylmethanamine (0.051 ml, 0.58 mmol) and NaHCO 3 (0.150 g, 1.79 mmol) in n-BuOH (3 mL) was stirred at 60 0 C for 1 hour. The reaction mixture was cooled to room temperature and then diluted with EtOAc.
  • Step C Preparation of 2-(cyclopropylmethylamino)-5-(3-fluoro-4- hydroxyphenyl)-3-methylpyrimidin-4(3H)-one: A solution of 5-(4-(benzyloxy)-3-fluorophenyl)- 2-(cyclopropylmethylamino)-3-methylpyrimidin-4(3H)-one (0.128 g, 0.337 mmol) in TFA (2 mL) was stirred at 40°C for 2 hours and 45 minutes. The reaction mixture was concentrated to dryness and then purified by silica gel flash column chromatography, eluting with 20:1 dichloromethane/MeOH.
  • Step A Preparation of 5-bromo-2-(4-fluorophenylamino)-3-methylpyrimidin-
  • Step B Preparation of 5-(4-(benzyloxy)-3-fluorophenyl)-2-(4- fluorophenylamino)-3-methylpyrimidin-4(3H)-one: Prepared from 5-bromo-2-(4- fluorophenylamino)-3-methylpyrimidin-4(3H)-one (0.130 g, 0.436 mmol) according to the procedure described in Step B of Example 63. The desired product (0.139 g, 76%) was obtained as a grey/white solid.
  • Step C Preparation of 5-(3-fluoro-4-hydroxyphenyl)-2-(4-fluorophenylamino)-3- methylpyrimidin-4(3H)-one: Prepared from 5-(4-(benzyloxy)-3-fluorophenyl)-2-(4- fluorophenylamino)-3-methylpyrimidin-4(3H)-one (0.139 g, 0.331 mmol) according to the procedure described in Step C of Example 63. The desired product (0.089 g, 82%) was obtained as a white solid.
  • Step D Preparation of 5-(3-fluoro-4-(6-methoxy-7-(3- mo ⁇ holinopropoxy)quinolin-4-yloxy)phenyl)-2-(4-fluorophenylamino)-3-methylpyrimidin- 4(3H)-one: Prepared from 5-(3-fluoro-4-hydroxyphenyl)-2-(4-fluorophenylamino)-3- methylpyrimidin-4(3H)-one (0.025 g, 0.0759 mmol), 4-(3-(4-chloro-6-methoxyquinolin-7- yloxy)propyl)morpholine (0.0281 g, 0.0835 mmol) and catalytic DMAP according to the procedure described in Step C of Example 58, to give 164 (0.026 g, 54%) as a pale yellow solid.
  • Step A Preparation of 5-bromo-2-chloro-3-ethylpyrimidin-4(3H)-one: Prepared from 5-bromo-2-chloropyrimidin-4(3H)-one (1.00 g, 4.775 mmol; obtained from Example 59, Step A) according to the procedure described in Step B of Example 59, substituting iodoethane in place of iodomethane. The desired product (0.411 g, 36%) was obtained as a yellow crystalline waxy solid.
  • 1 H-NMR 400 MHz, DMSO-d 6 ) ⁇ 8.26 (s, IH), 4.16 (q, 3H), 1.25 (t, 4H).
  • LRMS (ESI pos) m/e 237, 239 (M+, Br pattern).
  • Step B Preparation of 5-bromo-3-ethyl-2-(phenylamino)pyrimidin-4(3H)-one:
  • Step E Preparation of 3-ethyl-5-(3-fluoro-4-(6-methoxy-7-(3- morpholinopropoxy)quinolin-4-yloxy)phenyl)-2-(phenylamino)pyrimidin-4(3H)-one: Prepared from 3-ethyl-5-(3-fluoro-4-hydroxyphenyl)-2-(phenylamino)pyrimidin-4(3H)-one (0.025 g, 0.0768 mmol), 4-(3-(4-chloro-6-methoxyquinolin-7-yloxy)propyl)mo ⁇ holine (0.0285 g, 0.0845 mmol) and catalytic DMAP according to the procedure described in Step C of Example 58, to give 165 (0.016 g, 33%) as a pale yellow solid.

Abstract

Compounds of Formula (I), and stereoisomers, geometric isomers, tautomers, solvates, metabolites, salts and pharmaceutically acceptable prodrugs thereof, are useful for inhibiting receptor tyrosine kinases and for treating hyperproliferative disorders mediated thereby. Methods of using compounds of Formula (I), and stereoisomers, geometric isomers, tautomers, solvates and pharmaceutically acceptable salts thereof, for in vitro, in situ, and in vivo diagnosis, prevention or treatment of such disorders in mammalian cells, or associated pathological conditions are disclosed.

Description

QUINOLINE COMPOUNDS AND METHODS OF USE FIELD OF THE INVENTION
[0001] The invention relates to quinoline compounds having protein tyrosine kinase activity. The quinoline compounds may be useful in the treatment of hyperproliferative disorders, such as cancer, in mammals. The invention also relates to pharmaceutical compositions and formulations, methods of synthesis, and methods of use such as treating hyperproliferative disorders.
BACKGROUND OF THE INVENTION
[0002] Met tyrosine kinase is a high-affinity transmembrane receptor for the hepatocyte growth factor (HGF, Bottaro et al (1991) Science 251:802-804). Met was cloned, named (Cooper et al (1984) 311:29-33) and identified as an oncogene (Park et al (1986) Cell 45:895-904). When deregulated by overexpression or mutations, Met receptor tyrosine kinase leads to tumor growth and invasion (Cristiani et al (2005) Biochem. 44:14110-14119). Stimulation of Met by the ligand HGF, also known as Scatter Factor, initiates numerous physiological processes, including cell proliferation, scattering, morphogenic differentiation, angiogenesis, wound healing, tissue regeneration, and embryological development (Parr et al (2004) Clin. Cancer Res. 10(1, Pt. 1) 202-211; Comoglio et al (2002) J. Clin. Invest. 109:857-862; Maulik et al (2002) Cytokine Growth Factor Reviews 13:41-59; Hecht et al (2004) Cancer Res. 64(17):6109-6118). Receptor c-Met is rapidly internalized via clathrin-coated vesicles and traffics through an early endosomal compartment after hepatocyte growth factor stimulation. c-Met accumulates progressively in perinuclear compartments, which in part include the Golgi (Kermorgant et al (2003) J. of Biol. Chem. 278(31):28921-28929).
[0003] The phenomena of: deregulation or dysregulation of Met and/or HGF; Met overexpression; and Met mutations are implicated in uncontrolled cell proliferation and survival. Such factors play key roles in early-stage tumorigenesis, invasive growth of cancer cells, and metastasis (Danilkovitch-Miagkova et al (2002) J. Clin. Invest. 109(7):863-867; Di Renzo et al (1994) Int. J. Cancer 58:658-662; Matsumoto et al (1994) J. Biol. Chem. 269:31807-31813; Tusolino et al (1998) J. Cell Biol. 142:1145-1156; Jeffers et al (1996) MoI. Cell. Biol. 16:1115- 1125; Wong et al (2004) Exper. Cell Res. 299(l):248-256; Konda et al (2004) J. Urology 171(6), Pt. 1:2166-2170; Heideman et al (2004) J. Gene Med. 6(3):317-327; Ma et al (2003) Cancer Res. 63(19):6272-6281; Maulik et al (2002) Clin. Cancer Res. 8:620-627), making Met an important target for anticancer drug development (Cohen, P. (2002) Nat. Rev. Drug Discovery 1:309-315). Overexpression of Met and HGF is associated with poor prognosis. 111-03-PCT - P2338R1 - 02120.004WO1
2
[0004] Much evidence supports the role of HGF as a regulator of carcinogenesis, cancer invasion and metastasis (for review see: Herynk, M. H., and Radinsky, R. (2000) In Vivo 14:587- 596; Jiang et al (1999) Crit. Rev. Oncol. Hematol. 29:209-248; Longati (2001) Curr. Drug Targets 2:41-55; Maulik et al, (2002) Cytokine Growth Factor Rev. 13:41-59; Parr, C, and Jiang, W. G., (2001) Histol. Histopathol. 16:251-268). Recent data demonstrating the suppression of cancer cell proliferation, survival, and invasion upon inhibition of Met binding to HGF and Met receptor dimerization (Furge et al (2001) Proc. Natl. Acad. Sci. USA 98:10722-10727; Michieli et al (2004) Cancer Cell 6:61-73) confirm the relevance of Met in neoplasia and provide further proof of concept for the development of small-molecule compounds for antineoplastic therapy, e.g. against multiple myeloma (Hov et al. (2004) Clin. Cancer Res. 10(19):6686-6694). Inhibition of Met results in slowing tumor growth in tumor xenograft mouse models. Antibodies specific for c-Met have been expressed to block binding of HGF to c-Met (US 2005/0037431; US 2004/0166544). c-Met is also over-expressed in both non-small cell lung cancer and small cell lung cancer cells, in lung, breast, colon and prostate tumors (Herynk et al (2003) Cancer Res. 63(l l):2990-2996; Maulik et al (2002) Clin. Cancer Res. 8:620-627). Since c-Met appears to play an important role in oncogenesis of a variety of tumors, various inhibition strategies have been employed to therapeutically target this receptor tyrosine kinase. The usefulness of inhibiting the protein-tyrosine kinase c-Met for inhibiting tumor growth and invasion has been shown in many well documented preclinical experiments (Abounader et al (1999) J. Natl. Cancer Inst. 91:1548-1556; Laterra et al (1997) Lab. Invest. 76:565-577; Tomioka, D. (2001) Cancer Res. 61:7518-7524; Wang et al (2001) J. Cell Biology 153:1023-1033).
[0005] Protein kinases (PK) are enzymes that catalyze the phosphorylation of hydroxy groups on tyrosine, serine and threonine residues of proteins by transfer of the terminal (gamma) phosphate from ATP. Through signal transduction pathways, these enzymes modulate cell growth, differentiation and proliferation, i.e., virtually all aspects of cell life in one way or another depend on PK activity. Furthermore, abnormal PK activity has been related to a host of disorders, ranging from relatively non-life threatening diseases such as psoriasis to extremely virulent diseases such as glioblastoma (brain cancer). Protein kinases include two classes; protein tyrosine kinases (PTK) and serine-threonine kinases (STK).
[0006] One of the prime aspects of PTK activity is their involvement with growth factor receptors which are cell-surface proteins. When bound by a growth factor ligand, growth factor receptors are converted to an active form which interacts with proteins on the inner surface of a cell membrane. This leads to phosphorylation on tyrosine residues of the receptor and other proteins and to the formation inside the cell of complexes with a variety of cytoplasmic signaling 111-03-PCT - P2338R1 - 02120.004WO1
3 molecules that, in turn, effect numerous cellular responses such as cell division (proliferation), cell differentiation, cell growth, expression of metabolic effects to the extracellular microenvironment, etc. For a more complete discussion, see Schlessinger and Ullrich, (1992) Neuron 9:303-391, which is incorporated by reference, including any drawings, as if fully set forth herein.
[0007] Growth factor receptors with PTK activity are known as receptor tyrosine kinases
(RTK, Plowman et al (1994) DN&P, 7(6):334-339), which comprise a large family of transmembrane receptors with diverse biological activity. Met is one member of the tyrosine kinase growth factor receptor family, and often referred to as c-Met or human hepatocyte growth factor receptor tyrosine kinase (hHGFR). The expression of c-Met is thought to play a role in primary tumor growth and metastasis (Kim et al. Clin. Cancer Res. (2003) 9(14):5161-5170). [0008] Modulation of the HGF/c-met signaling pathway may be effected by regulating binding of HGF beta chain to c-Met. In particular embodiments, the zymogen-like form of HGF beta mutant was shown to bind Met with 14-fold lower affinity than the wild-type serine protease-like form, suggesting optimal interactions result from conformational changes upon cleavage of the single-chain form (US 2005/0037431). Extensive mutagenesis of the HGF beta region corresponding to the active site and activation domain of serine proteases showed that 17 of the 38 purified two-chain HGF mutants resulted in impaired cell migration or Met phosphorylation but no loss in Met binding. However, reduced biological activities were well correlated with reduced Met binding of corresponding mutants of HGF beta itself in assays eliminating dominant alpha-chain binding contributions.
[0009] Protein-tyrosine kinases (PTK) are critical components of signaling pathways that control cellular proliferation and differentiation. PTK are subdivided into two large families, receptor tyrosine kinases (RTK) and non-receptor tyrosine kinases (NRTK). RTK span the plasma membrane and contain an extra-cellular domain, which binds ligand, and an intracellular portion, which possesses catalytic activity and regulatory sequences. Most RTK, like the hepatocyte growth factor receptor c-Met, possess a single polypeptide chain and are monomeric in the absence of ligand. Ligand binding to the extracellular portion of RTK, dimerizes monomeric receptors, resulting in autophosphorylation of specific tyrosine residues in the cytoplasmic portion (for review see: Blume-Jensen, P., and Hunter, T., Nature (2001) 411:355- 365; Hubbard, S. R., et al, J. Biol. Chem. 273 (1998) 11987-11990; Zwick, E., et al, Trends MoI. Med. (2002) 8:17-23). In general, tyrosine autophosphorylation either stimulates the intrinsic catalytic kinase activity of the receptor or generates recruitment sites for downstream signaling 111-03-PCT- P2338R1 - 02120.004WO1
4 proteins containing phosphotyrosine-recognition domains, such as the Src homology 2 (SH2) domain or the phosphotyrosine-binding (PTB) domain.
[0010] c-Met inhibitors have been reported (US 5,792,783; US 5,834,504; US 5,880,141;
US 6,297,238; US 6,599,902; US 6,790,852; US 2003/0125370; US 2004/0242603; US 2004/0198750; US 2004/0110758; US 2005/0009845; US 2005/0009840; US 2005/0245547; US 2005/0148574; US 2005/0101650; US 2005/0075340; US 2006/0009453; US 2006/0009493; WO 98/007695; WO 2003/000660; WO 2003/087026; WO 2003/097641; WO 2004/076412; WO 2005/004808; WO 2005/121125; WO 2005/030140; WO 2005/070891; WO 2005/080393; WO 2006/014325; WO 2006/021886; WO 2006/021881). PHA-665752 is a small molecule, ATP-competitive, active-site inhibitor of the catalytic activity of c-Met, as well as phenotypes such as cell growth, cell motility, invasion, and morphology of a variety of tumor cells (Ma et al (2005) Clin. Cancer Res. 11:2312-2319; Christensen et al (2003) Cancer Res. 63:7345-7355).
SUMMARY OF THE INVENTION
[0011] In one aspect, the invention relates to quinoline compounds that are inhibitors of receptor tyrosine kinases (RTK), including c-Met. Certain hyperproliferative disorders are characterized by the overactivation of c-Met kinase function, for example by mutations or overexpression of the protein. Accordingly, the compounds of the invention are useful in the treatment of hyperproliferative disorders such as cancer.
[0012] More specifically, one aspect of the invention provides quinoline compounds of
Formula I:
Figure imgf000005_0001
[0013] and stereoisomers, geometric isomers, tautomers, solvates, metabolites, salts, and pharmaceutically acceptable prodrugs thereof, wherein R1, R2, R4, R5 and L are as defined herein. [0014] Another aspect of the invention provides a pharmaceutical composition comprising a quinoline compound of Formula I and a pharmaceutically acceptable carrier. The pharmaceutical composition may further comprise one or more additional therapeutic agents selected from anti-proliferative agents, anti-inflammatory agents, immunomodulatory agents, neurotropic factors, agents for treating cardiovascular disease, agents for treating liver disease, 111-03-PCT - P2338R1 - 02120.004WO1
5 anti-viral agents, agents for treating blood disorders, agents for treating diabetes, and agents for treating immunodeficiency disorders.
[0015] Another aspect of the invention provides methods of inhibiting c-Met kinase activity, comprising contacting a c-Met kinase with an effective inhibitory amount of a quinoline compound of Formula I, or a stereoisomer, geometric isomer, tautomer, solvate, metabolite, or pharmaceutically acceptable salt or prodrug thereof.
[0016] Another aspect of the invention provides methods of preventing or treating a disease or disorder modulated by c-Met kinases, comprising administering to a mammal in need of such treatment an effective amount of a compound of Formula I, or a stereoisomer, geometric isomer, tautomer, solvate, metabolite, or pharmaceutically acceptable salt or prodrug thereof. Examples of such diseases, conditions and disorders include, but are not limited to, hyperproliferative disorders (e.g., cancer, including melanoma and other cancers of the skin), neurodegeneration, cardiac hypertrophy, pain, migraine, neurotraumatic diseases, stroke, diabetes, hepatomegaly, cardiovascular disease, Alzheimer's disease, cystic fibrosis, viral diseases, autoimmune diseases, atherosclerosis, restenosis, psoriasis, allergic disorders, inflammation, neurological disorders, hormone-related diseases, conditions associated with organ transplantation, immunodeficiency disorders, destructive bone disorders, proliferative disorders, infectious diseases, conditions associated with cell death, thrombin-induced platelet aggregation, chronic myelogenous leukemia (CML), liver disease, pathologic immune conditions involving T cell activation, and CNS disorders.
[0017] Another aspect of the invention provides methods of preventing or treating a hyperproliferative disorder, comprising administering to a mammal in need of such treatment an effective amount of a compound of Formula I, or a stereoisomer, geometric isomer, tautomer, solvate, metabolite, or pharmaceutically acceptable salt or prodrug thereof, alone or in combination with one or more additional compounds having anti-hyperproliferative properties. [0018] In a further aspect the present invention provides a method of using a compound of this invention to treat a disease or condition modulated by c-Met in a mammal. [0019] An additional aspect of the invention is the use of a compound of this invention in the preparation of a medicament for the treatment or prevention of a disease or condition modulated by c-Met in a mammal.
[0020] Another aspect of the invention includes kits comprising a compound of Formula
I, or a stereoisomer, geometric isomer, tautomer, solvate, metabolite, or pharmaceutically acceptable salt or prodrug thereof, a container, and optionally a package insert or label indicating a treatment. 111-03-PCT - P2338R1 - 02120.004WO1
6
[0021] Another aspect of the invention includes methods of preparing, methods of separating, and methods of purifying compounds of Formula I.
[0022] Additional advantages and novel features of this invention shall be set forth in part in the description that follows, and in part will become apparent to those skilled in the art upon examination of the following specification or may be learned by the practice of the invention. The advantages of the invention may be realized and attained by means of the instrumentalities, combinations, compositions, and methods particularly pointed out in the appended claims.
DETAILED DESCRIPTION OF EXEMPLARY EMBODIMENTS
[0023] Reference will now be made in detail to certain embodiments of the invention, examples of which are illustrated in the accompanying structures and formulas. While the invention will be described in conjunction with the enumerated embodiments, it will be understood that they are not intended to limit the invention to those embodiments. On the contrary, the invention is intended to cover all alternatives, modifications, and equivalents which may be included within the scope of the present invention as defined by the claims. One skilled in the art will recognize many methods and materials similar or equivalent to those described herein, which could be used in the practice of the present invention. The present invention is in no way limited to the methods and materials described. In the event that one or more of the incorporated literature, patents, and similar materials differs from or contradicts this application, including but not limited to defined terms, term usage, described techniques, or the like, this application controls. DEFINITIONS
[0024] The term "alkyl" as used herein refers to a saturated linear or branched-chain monovalent hydrocarbon radical of one to twelve carbon atoms, wherein the alkyl radical may be optionally substituted independently with one or more substituents described below. Examples of alkyl groups include, but are not limited to, methyl (Me, -CH3), ethyl (Et, -CH2CH3), 1 -propyl (n- Pr, n-propyl, -CH2CH2CH3), 2-propyl (i-Pr, i-propyl, -CH(CH3)2), 1 -butyl (n-Bu, n-butyl, -CH2CH2CH2CH3), 2-methyl-l -propyl (i-Bu, i-butyl, -CH2CH(CH3)2), 2-butyl (s-Bu, s-butyl, -CH(CH3)CH2CH3), 2-methyl-2-propyl (t-Bu, t-butyl, -C(CH3)3), 1-pentyl (n-pentyl, -CH2CH2CH2CH2CH3), 2-pentyl (-CH(CH3)CH2CH2CH3), 3-pentyl (-CH(CH2CH3)2), 2-methyl- 2-butyl (-C(CHs)2CH2CH3), 3-methyl-2-butyl (-CH(CH3)CH (CH3)2), 3-methyl-l -butyl (-CH2CH2CH(CH3)2), 2-methyl-l -butyl (-CH2CH(CH3)CH2CH3), 1-hexyl (-CH2CH2CH2 CH2CH2CH3), 2-hexyl (-CH(CH3)CH2CH2CH2CH3), 3-hexyl (-CH(CH2CH3)(CH2CH2CH3)), 2-methyl-2-pentyl (-C(CH3)2CH2CH2CH3), 3-methyl-2-pentyl (-CH(CH3)CH(CH3)CH2CH3), 111-03-PCT - P2338R1 - 02120.004WO1
7
4-methyl-2-pentyl (-CH(CH3)CH2CH(CH3)2), 3-methyl-3-pentyl (-C(CH3)(CH2CH3)2), 2-methyl- 3-pentyl (-CH(CH2CH3)CH(CH3)2), 2,3-dimethyl-2-butyl (-C(CH3)2CH(CH3)2), 3,3-dimethyl-2- butyl (-CH(CH3)C(CH3)3, 1-heptyl, 1-octyl, and the like.
[0025] The term "alkenyl" refers to linear or branched-chain monovalent hydrocarbon radical of two to twelve carbon atoms with at least one site of unsaturation, i.e., a carbon-carbon, sp2 double bond, wherein the alkenyl radical may be optionally substituted independently with one or more substituents described herein, and includes radicals having "cis" and "trans" orientations, or alternatively, "E" and "Z" orientations. Examples include, but are not limited to, ethylenyl or vinyl (-CH=CH2), allyl (-CH2CH=CH2), and the like.
[0026] The term "alkynyl" refers to a linear or branched monovalent hydrocarbon radical of two to twelve carbon atoms with at least one site of unsaturation, i.e., a carbon-carbon, sp triple bond, wherein the alkynyl radical may be optionally substituted independently with one or more substituents described herein. Examples include, but are not limited to, ethynyl (-C≡CH), propynyl (propargyl, -CH2C≡CH), and the like.
[0027] The terms "carbocycle", "carbocyclyl", "carbocyclic ring" and "cycloalkyl" refer to a monovalent or multivalent non-aromatic, saturated or partially unsaturated ring having 3 to 12 carbon atoms as a monocyclic ring or 7 to 12 carbon atoms as a bicyclic ring. Bicyclic carbocycles having 7 to 12 atoms can be arranged, for example, as a bicyclo [4,5], [5,5], [5,6] or [6,6] system, and bicyclic carbocycles having 9 or 10 ring atoms can be arranged as a bicyclo [5,6] or [6,6] system, or as bridged systems such as bicyclo[2.2.1]heptane, bicyclo[2.2.2]octane and bicyclo[3.2.2]nonane. Examples of monocyclic carbocycles include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, 1-cyclopent-l-enyl, l-cyclopent-2-enyl, l-cyclopent-3-enyl, cyclohexyl, 1-cyclohex-l-enyl, l-cyclohex-2-enyl, l-cyclohex-3-enyl, cyclohexadienyl, cycloheptyl, cyclooctyl, cyclononyl, cyclodecyl, cycloundecyl, cyclododecyl, and the like. [0028] "Aryl" means a monovalent or multivalent aromatic hydrocarbon radical of 6-20 carbon atoms derived by the removal of one or more hydrogen atoms from a carbon atom of a parent aromatic ring system. Some aryl groups are represented in the exemplary structures as "Ar". Aryl includes bicyclic radicals comprising an aromatic ring fused to a saturated, partially unsaturated ring, or aromatic carbocyclic or heterocyclic ring. Typical aryl groups include, but are not limited to, radicals derived from benzene, substituted benzenes, naphthalene, anthracene, biphenyl, indenyl, indanyl, 1,2-dihydronapthalene, 1,2,3,4-tetrahydronapthyl, and the like. [0029] Examples of aryl fused to a heterocyclic ring include, but are not limited to, the structure: 111-03-PCT- P2338R1 - 02120.004WO1
Figure imgf000009_0001
wherein n is 0, 1 or 2. Examples of aryl fused to a carbocylic ring include, but are not limited to, the structures:
Figure imgf000009_0002
wherein R5 is as defined herein..
[0030] The terms "heterocycle," "hetercyclyl" and "heterocyclic ring" are used interchangeably herein and refer to a saturated or a partially unsaturated (i.e., having one or more double and/or triple bonds within the ring) carbocyclic, monovalent or multivalent radical of 3 to 20 carbon atoms, and in which at least one ring atom is a heteroatom selected from nitrogen, oxygen and sulfur, the remaining ring atoms being C, where one or more ring atoms is optionally substituted independently with one or more substituents described below. A heterocycle may be a monocycle having 3 to 7 ring members (2 to 6 carbon atoms and 1 to 3 heteroatoms selected from N, O, P, and S, wherein the S is optionally substituted with one or more oxo to provide the group SO or SO2) or a bicycle having 7 to 10 ring members (4 to 9 carbon atoms and 1 to 3 heteroatoms selected from N, O, P, and S, wherein the S is optionally substituted with one or more oxo to provide the group SO or SO2), for example: a bicyclo [4,5], [5,5], [5,6], or [6,6] system. Heterocycles are described in Paquette, Leo A.; "Principles of Modern Heterocyclic Chemistry" (W.A. Benjamin, New York, 1968), particularly Chapters 1, 3, 4, 6, 7, and 9; "The Chemistry of Heterocyclic Compounds, A series of Monographs" (John Wiley & Sons, New York, 1950 to present), in particular Volumes 13, 14, 16, 19, and 28; and J. Am. Chem. Soc. (1960) 82:5566. The heterocyclyl may be a carbon radical or heteroatom radical. The term "heterocycle" includes heterocycloalkoxy. "Heterocyclyl" also includes radicals where heterocycle radicals are fused with a saturated, partially unsaturated ring, or aromatic carbocyclic or heterocyclic ring. Examples of heterocyclic rings include, but are not limited to, pyrrolidinyl, 111-03-PCT - P2338R1 - 02120.004WO1
9 tetrahydrofliranyl, dihydrofuranyl, tetrahydrothienyl, tetrahydropyranyl, dihydropyranyl, tetrahydrothiopyranyl, piperidino, morpholino, thiomorpholino, thioxanyl, piperazinyl, homopiperazinyl, azetidinyl, oxetanyl, thietanyl, homopiperidinyl, oxepanyl, thiepanyl, oxazepinyl, diazepinyl, thiazepinyl, 2-pyrrolinyl, 3-pyrrolinyl, indolinyl, 2H-pyranyl, 4H-pyranyl, dioxanyl, 1,3-dioxolanyl, pyrazolinyl, dithianyl, dithiolanyl, dihydropyranyl, dihydrothienyl, dihydrofuranyl, pyrazolidinylimidazolinyl, imidazolidinyl, 1,2,3,4-tetrahydroisoquinolinyl, 3- azabicyco[3.1.0]hexanyl, 3-azabicyclo[4.1.0]heptanyl, azabicyclo[2.2.2]hexanyl, 3H-indolyl quinolizinyl and N-pyridyl ureas. Spiro moieties are also included within the scope of this definition. Examples of a heterocyclic group wherein 2 ring carbon atoms are substituted with oxo (=O) moieties are pyrimidindionyl and 1,1-dioxo-thiomorpholinyl. The heterocycle groups herein are optionally substituted independently with one or more substituents described herein. [0031] The term "heteroaryl" refers to a monovalent or multivalent aromatic radical of 5-
, 6-, or 7-membered rings, and includes fused ring systems (at least one of which is aromatic) of 1 to 20 carbon atoms, and containing one or more heteroatoms independently selected from nitrogen, oxygen, and sulfur. Examples of heteroaryl groups are pyridinyl (including, for example, 2-hydroxypyridinyl), imidazolyl, imidazopyridinyl, pyrimidinyl (including, for example, 4-hydroxypyrimidinyl), pyrazolyl, triazolyl, pyrazinyl, tetrazolyl, furyl, thienyl, isoxazolyl, thiazolyl, oxazolyl, isothiazolyl, pyrrolyl, quinolinyl, isoquinolinyl, indolyl, benzimidazolyl, benzofuranyl, cinnolinyl, indazolyl, indolizinyl, phthalazinyl, pyridazinyl, triazinyl, isoindolyl, pteridinyl, purinyl, oxadiazolyl, triazolyl, thiadiazolyl, thiadiazolyl, furazanyl, benzofurazanyl, benzothiophenyl, benzothiazolyl, benzoxazolyl, quinazolinyl, quinoxalinyl, naphthyridinyl, and furopyridinyl. Heteroaryl groups are optionally substituted independently with one or more substituents described herein. [0032] Examples of heteroaryl fused to an aryl ring include, but are not limited to:
Figure imgf000010_0001
wherein R5 and R10 are as defined herein.
[0033] The heterocycle or heteroaryl groups may be C-attached or N-attached where such is possible. By way of example and not limitation, carbon bonded heterocycles or heteroaryls are bonded at position 2, 3, 4, 5, or 6 of a pyridine, position 3, 4, 5, or 6 of a pyridazine, position 2, 4, 5, or 6 of a pyrimidine, position 2, 3, 5, or 6 of a pyrazine, position 2, 3, 4, or 5 of a furan, tetrahydrofuran, thiofuran, thiophene, pyrrole or tetrahydropyrrole, position 2, 4, or 5 of an oxazole, imidazole or thiazole, position 3, 4, or 5 of an isoxazole, pyrazole, or 111-03-PCT- P2338R1 - 02120.004WO1
10 isothiazole, position 2 or 3 of an aziridine, position 2, 3, or 4 of an azetidine, position 2, 3, 4, 5, 6, 7, or 8 of a quinoline or position 1, 3, 4, 5, 6, 7, or 8 of an isoquinoline.
[0034] By way of example and not limitation, nitrogen bonded heterocycles or heteroaryls are bonded at position 1 of an aziridine, azetidine, pyrrole, pyrrolidine, 2-pyrroline, 3- pyrroline, imidazole, imidazolidine, 2-imidazoline, 3-imidazoline, pyrazole, pyrazoline, 2- pyrazoline, 3-pyrazoline, piperidine, piperazine, indole, indoline, lH-indazole, position 2 of a isoindole, or isoindoline, position 4 of a morpholine, and position 9 of a carbazole, or β-carboline. [0035] "Substituted alkyl", "substituted alkenyl", "substituted alkynyl", "substituted aryl", "substituted heteroaryl", "substituted heterocyclyl" and "substituted cycloalkyl" mean alkyl, alkenyl, alkynyl, aryl, heteroaryl, heterocyclyl and cycloalkyl, respectively, in which one or more hydrogen atoms are each independently replaced with a substituent. Typical substituents include, but are not limited to, F, Cl, Br, I, CN, CF3, OR, R, =O, =S, =NR, =N+(O)(R), =N(OR), =N+(O)(OR), =N-NRR', -C(=O)R, -C(O)OR, -C(=O)NRR', -NRR', -N+RR1R", -N(R)C(=0)R, -N(R)C(=0)0R', -N(R)C(=O)NR'R", -SR, -OC(=O)R, -OC(O)OR, -OC(O)NRR', -OS(O)2(OR), -OP(O)(OR)(OR'), -OP(OR)(OR'), -P(O)(OR)(OR'), -P(O)(OR)NR1R", -S(O)R, -S(O)2R, -S(O)2NR, -S(O)(OR), -S(O)2(OR), -SC(O)R, -SC(O)OR, O and -SC(O)NRR'; wherein each R, R1 and R" is independently selected from H, C1-Ci2 alkyl, C2-C8 alkenyl, C2-C8 alkynyl, C6-C20 aryl and C2-C20 heterocyclyl. Substituents may also be combinations of alkyl, alkenyl, alkynyl, carbocycle, aryl, and heteroaryl radicals, such as cyclopropylmethyl, cyclohexylethyl, benzyl, and N-ethylmorpholino, and substituted forms thereof.
[0036] The terms "treat" and "treatment" refer to both therapeutic treatment and prophylactic or preventative measures, wherein the object is to prevent or slow down (lessen) an undesired physiological change or disorder, such as the development or spread of cancer. For purposes of this invention, beneficial or desired clinical results include, but are not limited to, alleviation of symptoms, diminishment of extent of disease, stabilized (i.e., not worsening) state of disease, delay or slowing of disease progression, amelioration or palliation of the disease state, and remission (whether partial or total), whether detectable or undetectable. "Treatment" can also mean prolonging survival as compared to expected survival if not receiving treatment. Those in need of treatment include those already with the condition or disorder as well as those prone to have the condition or disorder or those in which the condition or disorder is to be prevented. [0037] The phrase "therapeutically effective amount" means an amount of a compound of the present invention that (i) treats or prevents the particular disease, condition, or disorder, (ii) attenuates, ameliorates, or eliminates one or more symptoms of the particular disease, condition, 111-03-PCT - P2338R1 - 02120.004WO1
11 or disorder, or (iii) prevents or delays the onset of one or more symptoms of the particular disease, condition, or disorder described herein. In the case of cancer, the therapeutically effective amount of the drug may reduce the number of cancer cells; reduce the tumor size; inhibit (i.e., slow to some extent and preferably stop) cancer cell infiltration into peripheral organs; inhibit (i.e., slow to some extent and preferably stop) tumor metastasis; inhibit, to some extent, tumor growth; and/or relieve to some extent one or more of the symptoms associated with the cancer. To the extent the drug may prevent growth and/or kill existing cancer cells, it may be cytostatic and/or cytotoxic. For cancer therapy, efficacy can be measured, for example, by assessing the time to disease progression (TTP) and/or determining the response rate (RR). [0038] The terms "cancer" and "cancerous" refer to or describe the physiological condition in mammals that is typically characterized by unregulated cell growth. A "tumor" comprises one or more cancerous cells. Examples of cancer include, but are not limited to, carcinoma, lymphoma, blastoma, sarcoma, and leukemia or lymphoid malignancies. More particular examples of such cancers include squamous cell cancer (e.g., epithelial squamous cell cancer), lung cancer including small- cell lung cancer, non-small cell lung cancer ("NSCLC"), adenocarcinoma of the lung and squamous carcinoma of the lung, cancer of the peritoneum, hepatocellular cancer, gastric or stomach cancer including gastrointestinal cancer, pancreatic cancer, glioblastoma, cervical cancer, ovarian cancer, liver cancer, bladder cancer, hepatoma, breast cancer, colon cancer, rectal cancer, colorectal cancer, endometrial or uterine carcinoma, salivary gland carcinoma, kidney or renal cancer, prostate cancer, vulval cancer, thyroid cancer, hepatic carcinoma, anal carcinoma, penile carcinoma, as well as head and neck cancer. [0039] A "chemotherapeutic agent" is a chemical compound useful in the treatment of cancer. Examples of chemotherapeutic agents include Erlotinib (TARCEVA®, Genentech/OSI Pharm.), Bortezomib (VELCADE®, Millennium Pharm.), Fulvestrant (FASLODEX®, AstraZeneca), Sutent (SUl 1248, Pfizer), Letrozole (FEMARA®, Novartis), Imatinib mesylate (GLEEVEC®, Novartis), PTK787/ZK 222584 (Novartis), Oxaliplatin (Eloxatin®, Sanofi), 5-FU (5-fluorouracil), Leucovorin, Rapamycin (Sirolimus, RAPAMUNE®, Wyeth), lapatinib (TYKERB®, GlaxoSmithKline PLC), Lonafarnib (SCH 66336), Sorafenib (BAY43-9006, Bayer Labs), and Gefitinib (IRESSA®, AstraZeneca), AG1478, AG1571 (SU 5271; Sugen), alkylating agents such as thiotepa and CYTOXAN® cyclosphosphamide; alkyl sulfonates such as busulfan, improsulfan and piposulfan; aziridines such as benzodopa, carboquone, meturedopa, and uredopa; ethylenimines and methylamelamines including altretamine, triethylenemelamine, triethylenephosphoramide, triethylenethiophosphoramide and trimethylomelamine; acetogenins (especially bullatacin and bullatacinone); a camptothecin (including the synthetic analog topotecan); bryostatin; callystatin; CC- 1065 (including its adozelesin, carzelesin and bizelesin 111-03-PCT- P2338R1 - 02120.004WO1
12 synthetic analogs); cryptophycins (particularly cryptophycin 1 and cryptophycin 8); dolastatin; duocarmycin (including the synthetic analogs, KW-2189 and CBl-TMl); eleutherobin; pancratistatin; a sarcodictyin; spongistatin; nitrogen mustards such as chlorambucil, chlornaphazine, cholophosphamide, estramustine, ifosfamide, mechlorethamine, mechlorethamine oxide hydrochloride, melphalan, novembichin, phenesterine, prednimustine, trofosfamide, uracil mustard; nitrosureas such as carmustine, chlorozotocin, fotemustine, lomustine, nimustine, and ranimnustine; antibiotics such as the enediyne antibiotics (e.g., calicheamicin, especially calicheamicin gammall and calicheamicin omegall (Angew Chem. Intl. Ed. Engl. (1994) 33:183-186); dynemicin, including dynemicin A; bisphosphonates, such as clodronate; an esperamicin; as well as neocarzinostatin chromophore and related chromoprotein enediyne antibiotic chromophores), aclacinomysins, actinomycin, authramycin, azaserine, bleomycins, cactinomycin, carabicin, carminomycin, carzinophilin, chromomycinis, dactinomycin, daunorubicin, detorubicin, 6-diazo-5-oxo-L-norleucine, ADRIAMYCIN® (doxorubicin), morpholino-doxorubicin, cyanomorpholino-doxorubicin, 2-pyrrolino-doxorubicin and deoxydoxorubicin), epirubicin, esorubicin, idarubicin, marcellomycin, mitomycins such as mitomycin C, mycophenolic acid, nogalamycin, olivomycins, peplomycin, potfϊromycin, puromycin, quelamycin, rodorubicin, streptonigrin, streptozocin, tubercidin, ubenimex, zinostatin, zorubicin; anti-metabolites such as methotrexate and 5-fluorouracil (5-FU); folic acid analogs such as denopterin, methotrexate, pteropterin, trimetrexate; purine analogs such as fludarabine, 6-mercaptopurine, thiamiprine, thioguanine; pyrimidine analogs such as ancitabine, azacitidine, 6-azauridine, carmofur, cytarabine, dideoxyuridine, doxifluridine, enocitabine, floxuridine; androgens such as calusterone, dromostanolone propionate, epitiostanol, mepitiostane, testolactone; anti-adrenals such as aminoglutethimide, mitotane, trilostane; folic acid replenisher such as frolinic acid; aceglatone; aldophosphamide glycoside; aminolevulinic acid; eniluracil; amsacrine; bestrabucil; bisantrene; edatraxate; defofamine; demecolcine; diaziquone; elfornithine; elliptinium acetate; an epothilone; etoglucid; gallium nitrate; hydroxyurea; lentinan; lonidainine; maytansinoids such as maytansine and ansamitocins; mitoguazone; mitoxantrone; mopidanmol; nitraerine; pentostatin; phenamet; pirarubicin; losoxantrone; podophyllinic acid; 2-ethylhydrazide; procarbazine; PSK® polysaccharide complex (JHS Natural Products, Eugene, OR); razoxane; rhizoxin; sizofϊran; spirogermanium; tenuazonic acid; triaziquone; 2,2',2"-trichlorotriethylamine; trichothecenes (especially T-2 toxin, verracurin A, roridin A and anguidine); urethan; vindesine; dacarbazine; mannomustine; mitobronitol; mitolactol; pipobroman; gacytosine; arabinoside ("Ara-C"); cyclophosphamide; thiotepa; taxoids, e.g., TAXOL® (paclitaxel; Bristol-Myers Squibb .Oncology, Princeton, N.J.), ABRAXANE™ (Cremophor-free), albumin-engineered nanoparticle formulations of paclitaxel (American 111-03-PCT - P2338R1 - 02120.004WO1
13
Pharmaceutical Partners, Schaumberg, Illinois), and TAXOTERE® (docetaxel; Rhόne-Poulenc Rorer, Antony, France); chloranbucil; GEMZAR® (gemcitabine); 6-thioguanine; mercaptopurine; methotrexate; platinum analogs such as cisplatin and carboplatin; vinblastine; etoposide (VP- 16); ifosfamide; mitoxantrone; vincristine; NAVELBINE® (vinorelbine); novantrone; teniposide; edatrexate; daunomycin; aminopterin; capecitabine (XEL)DA®, Hoffman LaRoche Inc); ibandronate; CPT-I l; topoisomerase inhibitor RFS 2000; difluoromethylornithine (DMFO); retinoids such as retinoic acid; capecitabine; and pharmaceutically acceptable salts, acids and derivatives of any of the above. [0040] Also included in the definition of "chemotherapeutic agent" are: (i) anti -hormonal agents that act to regulate or inhibit hormone action on tumors such as anti-estrogens and selective estrogen receptor modulators (SERMs), including, for example, tamoxifen (including NOLVADEX®; tamoxifen citrate), raloxifene, droloxifene, 4-hydroxytamoxifen, trioxifene, keoxifene, LYl 17018, onapristone, and FARESTON® (toremifine citrate); (ii) aromatase inhibitors that inhibit the enzyme aromatase, which regulates estrogen production in the adrenal glands, such as, for example, 4(5)-imidazoles, aminoglutethimide, MEGASE® (megestrol acetate), AROMASIN® (exemestane; Pfizer), formestanie, fadrozole, RIVISOR® (vorozole), FEMARA® (letrozole; Novartis), and ARIMIDEX® (anastrozole; AstraZeneca); (iii) anti- androgens such as flutamide, nilutamide, bicalutamide, leuprolide, and goserelin; as well as troxacitabine (a 1,3-dioxolane nucleoside cytosine analog); (iv) protein kinase inhibitors; (v) lipid kinase inhibitors; (vi) antisense oligonucleotides, particularly those which inhibit expression of genes in signaling pathways implicated in aberrant cell proliferation, such as, for example, PKC- alpha, RaIf and H-Ras; (vii) ribozymes such as VEGF expression inhibitors (e.g., ANGIOZYME®) and HER2 expression inhibitors; (viii) vaccines such as gene therapy vaccines, for example, ALLOVECTIN®, LEUVECTIN®, and VAXID®; PROLEUKIN® rIL-2; a topoisomerase 1 inhibitor such as LURTOTECAN®; ABARELIX® rmRH; (ix) anti-angiogenic agents such as bevacizumab (AVASTIN® and LUCENTIS®, Genentech); (x) therapeutic antibodies such as HERCEPTIN®, AVASTIN®, LUCENTIS®; (xi) antibody-drug conjugates such as MYLOTARG®; and (xii) pharmaceutically acceptable salts, acids and derivatives of any of the above.
[0041] The term "prodrug" as used in this application refers to a precursor or derivative form of a compound of the invention that is less cytotoxic to cells compared to the parent compound or drug and is capable of being enzymatically or hydrolytically activated or converted into the more active parent form. See, e.g., Wilman, "Prodrugs in Cancer Chemotherapy" Biochemical Society Transactions, 14, pp. 375-382, 615th Meeting Belfast (1986) and Stella et al, "Prodrugs: A Chemical Approach to Targeted Drug Delivery," Directed Drug Delivery, 111-03-PCT - P2338R1 - 02120.004WO1
14
Borchardt et al, (ed.), pp. 247-267, Humana Press (1985). The prodrugs of this invention include, but are not limited to, phosphate-containing prodrugs, thiophosphate-containing prodrugs, sulfate-containing prodrugs, peptide-containing prodrugs, D-amino acid-modified prodrugs, glycosylated prodrugs, β-lactam-containing prodrugs, optionally substituted phenoxyacetamide- containing prodrugs, optionally substituted phenylacetamide-containing prodrugs, 5- fluorocytosine and other 5-fluorouridine prodrugs which can be converted into the more active cytotoxic free drug. Examples of cytotoxic drugs that can be derivatized into a prodrug form for use in this invention include, but are not limited to, compounds of the invention and chemotherapeutic agents such as described above.
[0042] A "metabolite" is a product produced through metabolism in the body of a specified compound or salt thereof. Metabolites of a compound may be identified using routine techniques known in the art and their activities determined using tests such as those described herein. Such products may result for example from the oxidation, reduction, hydrolysis, amidation, deamidation, esterification, deesterification, enzymatic cleavage, and the like, of the administered compound. Accordingly, the invention includes metabolites of compounds of the invention, including compounds produced by a process comprising contacting a compound of this invention with a mammal for a period of time sufficient to yield a metabolic product thereof. [0043] A "liposome" is a small vesicle composed of various types of lipids, phospholipids and/or surfactant which is useful for delivery of a drug (such as the c-Met inhibitors disclosed herein and, optionally, a chemotherapeutic agent) to a mammal. The components of the liposome are commonly arranged in a bilayer formation, similar to the lipid arrangement of biological membranes.
[0044] The term "package insert" is used to refer to instructions customarily included in commercial packages of therapeutic products, that contain information about the indications, usage, dosage, administration, contraindications and/or warnings concerning the use of such therapeutic products.
[0045] The term "chiral" refers to molecules which have the property of non- superimposability of the mirror image partner, while the term "achiral" refers to molecules which are superimposable on their mirror image partner.
[0046] The term "stereoisomers" refers to compounds which have identical chemical constitution, but differ with regard to the arrangement of the atoms or groups in space. [0047] "Diastereomer" refers to a stereoisomer with two or more centers of chirality and whose molecules are not mirror images of one another. Diastereomers have different physical properties, e.g. melting points, boiling points, spectral properties, and reactivities. Mixtures of 111-03-PCT - P2338R1 - 02120.004WO1
15 diastereomers may separate under high resolution analytical procedures such as electrophoresis and chromatography.
[0048] "Enantiomers" refer to two stereoisomers of a compound which are non- superimposable mirror images of one another.
[0049] Stereochemical definitions and conventions used herein generally follow S. P.
Parker, Ed., McGraw-Hill Dictionary of Chemical Terms (1984) McGraw-Hill Book Company, New York; and Eliel, E. and Wilen, S., "Stereochemistry of Organic Compounds", John Wiley & Sons, Inc., New York, 1994. The compounds of the invention may contain asymmetric or chiral centers, and therefore exist in different stereoisomeric forms. It is intended that all stereoisomeric forms of the compounds of the invention, including but not limited to, diastereomers, enantiomers and atropisomers, as well as mixtures thereof such as racemic mixtures, form part of the present invention. Many organic compounds exist in optically active forms, i.e., they have the ability to rotate the plane of plane-polarized light. In describing an optically active compound, the prefixes D and L, or R and S, are used to denote the absolute configuration of the molecule about its chiral center(s). The prefixes d and 1 or (+) and (-) are employed to designate the sign of rotation of plane-polarized light by the compound, with (-) or 1 meaning that the compound is levorotatory. A compound prefixed with (+) or d is dextrorotatory. For a given chemical structure, these stereoisomers are identical except that they are mirror images of one another. A specific stereoisomer may also be referred to as an enantiomer, and a mixture of such isomers is often called an enantiomeric mixture. A 50:50 mixture of enantiomers is referred to as a racemic mixture or a racemate, which may occur where there has been no stereoselection or stereospecificity in a chemical reaction or process. The terms "racemic mixture" and "racemate" refer to an equimolar mixture of two enantiomeric species, devoid of optical activity. [0050] The term "tautomer" or "tautomeric form" refers to structural isomers of different energies which are interconvertible via a low energy barrier. For example, proton tautomers (also known as prototropic tautomers) include interconversions via migration of a proton, such as keto- enol and imine-enamine isomerizations. Valence tautomers include interconversions by reorganization of some of the bonding electrons.
[0051] A "salt," as used herein, refers to organic or inorganic salts of a compound of the invention. Exemplary salts include, but are not limited, to sulfate, citrate, acetate, oxalate, chloride, bromide, iodide, nitrate, bisulfate, phosphate, acid phosphate, isonicotinate, lactate, salicylate, acid citrate, tartrate, oleate, tannate, pantothenate, bitartrate, ascorbate, succinate, maleate, gentisinate, fumarate, gluconate, glucuronate, saccharate, formate, benzoate, glutamate, methanesulfonate, ethanesulfonate, benzenesulfonate, /?-toluenesulfonate, and pamoate (i.e., 1,1'- 111-03-PCT - P2338R1 - 02120.004WO1
16 methylene-bis-(2-hydroxy-3-naphthoate)) salts. A salt may involve the inclusion of another molecule such as an acetate ion, a succinate ion or other counter ion. The counter ion may be any organic or inorganic moiety that stabilizes the charge on the parent compound. Furthermore, a salt may have more than one charged atom in its structure. Instances where multiple charged atoms are part of the salt can have multiple counter ions. Hence, a salt can have one or more charged atoms and/or one or more counter ion.
[0052] If the compound of the invention is a base, the desired salt may be prepared by any suitable method available in the art, for example, treatment of the free base with an inorganic acid, such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid and the like, or with an organic acid, such as acetic acid, maleic acid, succinic acid, mandelic acid, fumaric acid, malonic acid, pyruvic acid, oxalic acid, glycolic acid, salicylic acid, a pyranosidyl acid, such as glucuronic acid or galacturonic acid, an alpha hydroxy acid, such as citric acid or tartaric acid, an amino acid, such as aspartic acid or glutamic acid, an aromatic acid, such as benzoic acid or cinnamic acid, a sulfonic acid, such as p-toluenesulfonic acid or ethanesulfonic acid, or the like.
[0053] If the compound of the invention is an acid, the desired salt may be prepared by any suitable method, for example, treatment of the free acid with an inorganic or organic base, such as an amine (primary, secondary or tertiary), an alkali metal hydroxide or alkaline earth metal hydroxide, or the like. Illustrative examples of suitable salts include, but are not limited to, organic salts derived from amino acids, such as glycine and arginine, ammonia, primary, secondary, and tertiary amines, and cyclic amines, such as piperidine, morpholine and piperazine, and inorganic salts derived from sodium, calcium, potassium, magnesium, manganese, iron, copper, zinc, aluminum and lithium.
[0054] In certain embodiments, the salt is a pharmaceutically acceptable salt. The phrase
"pharmaceutically acceptable" indicates that the substance or composition must be compatible chemically and/or toxicologically, with the other ingredients comprising a formulation, and/or the mammal being treated therewith.
[0055] The compounds of Formula I also include salts of such compounds which are not necessarily pharmaceutically acceptable salts, and which may be useful as intermediates for preparing and/or purifying compounds of Formula I and/or for separating enantiomers of compounds of Formula I.
[0056] A "solvate" refers to an association or complex of one or more solvent molecules and a compound of the invention. Examples of solvents that form solvates include, but are not 111-03-PCT - P2338R1 - 02120.004WO1
17 limited to, water, isopropanol, ethanol, methanol, DMSO, ethyl acetate, acetic acid, and ethanolamine. The term "hydrate" refers to the complex where the solvent molecule is water. [0057] The term "protecting group" or "Pg" refers to a substituent that is commonly employed to block or protect a particular functionality while reacting other functional groups on the compound. For example, an "amino-protecting group" is a substituent attached to an amino group that blocks or protects the amino functionality in the compound. Suitable amino-protecting groups include acetyl, trifluoroacetyl, t-butoxycarbonyl (BOC), benzyloxycarbonyl (CBZ) and 9- fluorenylmethylenoxycarbonyl (Fmoc). Similarly, a "hydroxy-protecting group" refers to a substituent of a hydroxy group that blocks or protects the hydroxy functionality. Suitable protecting groups include acetyl and silyl. A "carboxy-protecting group" refers to a substituent of the carboxy group that blocks or protects the carboxy functionality. Common carboxy-protecting groups include -CH2CH2SO2Ph, cyanoethyl, 2-(trimethylsilyl)ethyl, 2- (trimethylsilyl)ethoxymethyl, 2-(p-toluenesulfonyl) ethyl, 2-(p-nitrophenylsulfenyl)ethyl, 2- (diphenylphosphino)-ethyl, nitroethyl and the like. For a general description of protecting groups and their use, see T. W. Greene, Protective Groups in Organic Synthesis, John Wiley & Sons, New York, 1991.
[0058] The terms "compound of this invention," and "compounds of the present invention" and "compounds of Formula I" include compounds of Formula I and stereoisomers, geometric isomers, tautomers, solvates, metabolites, salts and pharmaceutically acceptable prodrugs thereof.
[0059] The term "mammal" includes, but is not limited to, humans, dogs, cats, horses, cows, pigs, and sheep, and poultry. C-MET INHIBITOR COMPOUNDS
[0060] The present invention provides quinoline compounds, and pharmaceutical formulations thereof, that are potentially useful in the treatment of diseases, conditions and/or disorders modulated by c-Met. More specifically, the present invention provides compounds of Formula I:
Figure imgf000018_0001
111-03-PCT - P2338R1 - 02120.004WO1
18
[0061] and stereoisomers, geometric isomers, tautomers, solvates, metabolites, salts, and pharmaceutically acceptable prodrugs thereof, wherein:
[0062] R1, R2 and R4 are independently selected from H, F, Cl, Br, I, CN,
-(CR14R15XNR10R11, -C(=Y)R10, -C(=Y)OR10, -CC=Y)NR10R1 \
-C(=O)NR12(CR14R15)tNR10Rn, -NO2, -NR10R11, -NR!0C(=Y)Rπ, -NR10CC=Y)OR1 ', -NR12C(^Y)NR10R11, -NR12SO2NR10R11, -OR10, -OC(^Y)R10, -OC(=Y)OR10, -OC(=Y)NR10Rπ, -OP(= Y)(OR10XOR11), -OP(OR10)(ORn), -P(=Y)(OR10)(ORπ), -SR10, -S(O)R10, -S(O)2R10, -S(O)2NR10R11, -SC(=Y)R10, -SC(=Y)OR10, C1-C12 alkyl, C2-C8 alkenyl, C2-C8 alkynyl, C3-Ci2 carbocyclyl, C2-C2O heterocyclyl, C6-C20 aryl, and Ci-C20 heteroaryl, where said alkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl and heteroaryl are optionally substituted with one or more groups independently selected from F, Cl, Br, I, CN, CF3, -NO2, oxo, -C(=Y)R10, -C(=Y)OR10, -C(=Y)NR10Rn, -(CR14R15)n-NR10Rπ, -NR10C (= Y)R10, -NR10C(=Y)ORπ, -NR12C(=Y)NR10Rn, -NR12SO2R10, -NR10, -OR10, -OC(=Y)R10, -OC(=Y)OR10, -OC(^Y)NR10R11, -OS(O)2(OR10), -OP(=Y)(OR10)(ORπ), -OP(OR10)(ORπ), SR10, -S(O)R10, -S(O)2R10, -S(O)2NR10R11, -S(O)(OR10), -S(O)2(OR10), -SC(=Y)R10, -SC(=Y)OR10, -SC(=Y)NR10Rπ, Cj-Ci2 alkyl, C2-C8 alkenyl, C2-C8 alkynyl, C3-Ci2 carbocyclyl, C2-C20 heterocyclyl, C6-C20 aryl, C]-C2O heteroaryl, -(CR14R15),-NR12C(=O)(CR14R15)NR10Rπ, and -(CR14R15X-NR10R1 ], [0063] with the proviso that at least one of R1 and R2 is not H;
[0064] L is C3-Ci2 carbocyclyl, C2-C20 heterocyclyl, C6-C20 aryl or Cj-C20 heteroaryl, wherein said carbocyclyl, heterocyclyl, aryl and heteroaryl are optionally substituted with one or more groups independently selected from R4 and R10, with the proviso that L is not naphthyl; [0065] R5 is -C(=Y)R13, -C(=Y)NR10R13, -NR10R13, -NR10C(=Y)R13,
-NR10C(=Y)OR13, -NR12SO2R10, -NR12C(=Y1)(CR14R15)C(=Y2)NR10R11, C3-Cj2 carbocyclyl, C2-C20 heterocyclyl, C6-C20 aryl, or Ci-C20 heteroaryl, wherein said carbocyclyl, heterocyclyl, aryl, and heteroaryl are optionally substituted with one or more groups independently selected from oxo, F, Cl, Br, I, SO2RC, CN, 0Ra, (CH2)n-NRaRb, C(=O)NRaRb, C(=O)Ra, CRaC(=O)Rb, NHSO2RC, CF3, C1-C12 alkyl, C2-C8 alkenyl, C2-C8 alkynyl, (CH2)n-(C6-C20 aryl), (CH2)n- cycloalkyl, CH(OH)-aryl, CH(CO2CH3)aryl, and (CH2)n-(Ci-C20 heteroaryl), and wherein any aryl or heteroaryl of the one or more groups is optionally substituted with one or more Rd; [0066] R10, R11 and R12 are independently H, Ci-Cj2 alkyl, C2-C8 alkenyl, C2-C8 alkynyl, C3-Ci2 carbocyclyl, C2-C20 heterocyclyl, C6-C20 aryl, or Ci-C20 heteroaryl, wherein said alkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl, and heteroaryl are optionally 111-03-PCT - P2338R1 - 02120.004WO1
19 substituted with one or more groups independently selected from F, Cl, Br, I, SO2R0, CN, ORa, NRaRb, C(=O)NRaRb, CRaC(=O)Rb, Ci-C12 alkyl, C2-C8 alkenyl, C2-C8 alkynyl, C3-C12 carbocyclyl, C2-C2O heterocyclyl optionally substituted with Ci-C6 alkyl, CH2OH or SO2Me, Ce-C2O aryl, and Cj-C2O heteroaryl optionally substituted with Ci-C6 alkyl, [0067] or R10 and R11 together with the nitrogen to which they are attached optionally form a saturated, partially unsaturated or fully unsaturated C3-C20 heterocyclic ring optionally containing one or more additional ring atoms selected from N, O or S, wherein said heterocyclic ring is optionally substituted with one or more groups independently selected from oxo, (CH2)nORa, NRaRb, CF3, F, Cl, Br, I, SO2R3, C(=O)Ra, NR10C(=Y)Rn, C(=Y)NR10Rn, Ci-Cj2 alkyl, C2-C8 alkenyl, C2-C8 alkynyl, C3-Ci2 carbocyclyl, C2-C2O heterocyclyl, C6-C2O aryl and Ci-C20 heteroaryl;
[0068] R13 is H, Ci-C6 alkyl, -(CR14R15)n-cycloalkyl, -(CR14R15)n-heterocyclyl,
-(CR14R15)n-aryl, -(CR14R15)n-heteroaryl, (CR14R15)n-O-(CRI4R15)m-aryl, (CR14R15)-N(SO2Ra)- (CR14R15)Rn, (CR14R15)n-heterocyclyl-(CR14R15),-aryl, or (CR14R15)-NR10C(O)aryl, where said cycloalkyl, heterocyclyl, aryl, and heteroaryl portions are optionally substituted with one or more groups independently selected from F, Cl, Br, I, oxo, SO2RC, CN, ORa, C(=O)Ra, C(=O)ORa, NRaRb, NRaC(=O)Rb, O-(CH2)-aryl, CJ-CI2 alkyl, C2-C8 alkenyl, C2-C8 alkynyl, C3-Cj2 carbocyclyl, C2-C20 heterocyclyl, CO-C20 aryl, and C]-C20 heteroaryl; [0069] each R14 and R15 is independently H, Cj-C12 alkyl, or (CH2)t-aryl,
[0070] or R14 and R15 together with the atoms to which they are attached form a saturated or partially unsaturated C3-Cj2 carbocyclic ring,
[0071] or R10 and R15 together with the atoms to which they are attached form a saturated or partially unsaturated C2-Cj2 heteorcyclic ring,
[0072] or R14 is null and R10 and R15 together with the atoms to which they are attached form a 5-6 membered heteroaryl ring,
[0073] or R12 and R14 together with the atoms to which they are attached form a saturated or partially unsaturated C2-Cj2 heteorcyclic ring;
[0074] Ra and Rb are independently H, CJ-CJ2 alkyl, C2-C8 alkenyl, C2-C8 alkynyl,
C3-Cj2 carbocyclyl, C2-C20 heterocyclyl, C6-C2O aryl, or Cj-C20 heteroaryl, wherein said alkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl, and heteroaryl are optionally substituted with one or more alkyl groups; 111-03-PCT- P2338R1 - 02120.004WO1
20
[0075] R° is C]-Ci2 alkyl or C6-C20 aryl, wherein said alkyl and aryl are optionally substituted with one or more groups independently selected from F, Cl, Br, I, ORa and C(=O)NRaRb;
[0076] Rd is F, Cl, Br, I, CF3, SO2RC, CN, ORa, NRaRb, C(=O)NRaRb, CRaC(=O)Rb,
Ci-C12 alkyl, C2-Cg alkenyl, C2-Cg alkynyl, C6-C2O aryl, or C1-C20 heteroaryl; [0077] Y, Y1 and Y2 are independently O or S;
[0078] t is 1, 2, 3, 4, 5 or 6; and
[0079] n and m are independently 0, 1, 2, 3, 4, 5 or 6.
[0080] In certain embodiments, one or both of R1 and R2 is -OR10 where R10 is Ci-Ci2 alkyl. For example, in one embodiment one or both of R1 and R2 are methoxy. [0081] In other embodiments, one or both of R1 and R2 is -OR10 where R10 is Ci-Ci2 alkyl substituted with NRaRb or C2-C2O heterocyclyl, wherein said heterocyclyl is optionally substituted with C1-C6 alkyl, CH2OH or SO2Me. [0082] Exemplary embodiments include the structures:
Figure imgf000021_0001
where the wavy line is the attachment site to the quinoline ring.
[0083] In other embodiments, R1 is methoxy and R2 is selected from
Figure imgf000021_0002
[0084] In other embodiments, one or both of R1 and R2 is -OR10 wherein R10 is C]-Ci2 alkyl substituted with Ci-C20 heteroaryl, wherein said heteroaryl is optionally substituted with C]-C6 alkyl. Exemplary embodiments include the structures: 111-03-PCT - P2338R1 - 02120.004WO1
21
Figure imgf000022_0001
where the wavy line is the attachment site to the quinoline ring.
[0085] In other embodiments, one or both of R1 and R2 are independently selected from
C2-C8 alkynyl substituted by -(CR14R15)t-NR12C(=O)(CR14R15)NR10Rπ or -(CR14R15^NR10R1 \ including the exemplary structures:
Figure imgf000022_0002
[0086] In other embodiments, one or both of R1 and R2 are independently selected from optionally substituted aryl or heteroaryl, including the exemplary structures:
Figure imgf000022_0003
111-03-PCT - P2338R1 - 02120.004WO1
Figure imgf000023_0001
[0087] In other embodiments, one or both of R1 and R2 may be independently selected from -C(=O)NR10Rn or -(CR14R15)tNR10Rπ.
[0088] In other embodiments, one or both of R! and R2 is independently alkyl optionally substituted with one or more groups independently selected from OR10, NR10R11, heterocyclyl and heteroaryl. Examples include, but are not limited to, methyl, -CH2OH, -CH2CH2OH,
-CH2CH2CH2OH, and -CH(OH)CH2OH.
[0089] In other embodiments, one or both of R1 and R2 are independently -OR , including the exemplary structure:
Figure imgf000023_0002
[0090] In exemplary embodiments, each R4 is H. [0091] In certain embodiments, L-R5 is (C3-Ci2 carbocyclyO-R5, including the exemplary structures:
Figure imgf000023_0003
where the wavy line indicates the point of attachment to the 4-oxy position of the quinoline ring. [0092] In certain embodiments, L-R5 is (C2-C20 heterocyclyl)-R5 wherein said heterocyclyl is optionally substituted, including the exemplary structures: 111-03-PCT- P2338R1 - 02120.004WO1
23
Figure imgf000024_0001
where the wavy line indicates the point of attachment to the 4-oxy position of the quinoline ring. [0093] In certain embodiments, L-R5 is (C6-C2o aryl)-R5 wherein said aryl is optionally substituted, including the exemplary structures:
Figure imgf000024_0002
where the wavy line indicates the point of attachment to the 4-oxy position of the quinoline ring and each R4 is independent of the other.
[0094] Exemplary embodiments where L-R5 is (C6-C2O aryl)-R5 include the structures:
Figure imgf000024_0003
[0095] Other exemplary embodiments where L-R5 is (C6-C20 aryl)-R5, include the structures: 111-03-PCT- P2338R1 - 02120.004WO1
24
Figure imgf000025_0001
[0096] Other exemplary embodiments where L-R5 is (C6-C2O aryl)-R include the structures:
Figure imgf000025_0002
[0097] Other exemplary embodiments where L-R5 is (C6-C20 aryl)-R5 include the structures:
Figure imgf000025_0003
111-03-PCT - P2338R1 - 02120.004WO1
25
[0098] In certain embodiments, L-R5 is (Ci-C20 heteroaryl)-R5. Exemplary embodiments include the structures:
Figure imgf000026_0001
where the wavy line indicates the point of attachment to the 4-oxy position of the quinoline ring. [0099] Exemplary embodiments where L-R5 is (C]-C2O heteroaryl)-R5 also include the structures:
Figure imgf000026_0002
[00100] In certain embodiments, R5 is -C(=Y)R13. In certain embodiments, R13 is
-(CR14R15)n-cycloalkyl, -(CR14R15)n-aryl, -(CR14R15)n-O-(CR14R15)m-aryl, or -(CR14R15V heterocyclyl-(CR14R15)raryl, wherein said heterocyclyl portion is optionally substituted with SO2R0 or C]-Ci2 alkyl. Exemplary embodiments include the structures: 111-03-PCT - P2338R1 - 02120.004WO1
26
Figure imgf000027_0001
where the wavy line indicates the point of attachment to L.
[00101] In certain embodiments, R5 is -C(=Y)NR10R13. In certain embodiments, R10 is H or Ci-Ci2 alkyl, and R13 is H, Q-C6 alkyl, -(CR14R15)n-cycloalkyl, or -(CR14R15)n-aryl, wherein said alkyl, cycloalkyl, and aryl portions are optionally substituted with F or 0Ra. Exemplary embdodiments of R5 include the structures:
Figure imgf000027_0002
where the wavy line indicates the point of attachment to L.
[00102] In certain embodiments, R5 is -NR10R13. In certain embodiemts, R10 is H or
Ci-Ci2 alkyl, and R13 is -(CR14R15)n-heterocyclyl or -(CR14R15)n-heteroaryl, wherein said heterocyclyl and heteroaryl are optionally substituted with ORa, SO2R0 or O-(CH2)-aryl. Exemplary embdodiments of R5 include the structures:
Figure imgf000027_0003
where the wavy line indicates the point of attachment to L.
[00103] In certain embodiments, R5 is -NR12CC=Y1 )(CR14R15)C(=Y2)NR10Rn, wherein
R15 and R10 optionally together with the atoms to which they are attached form a 5-6 membered 111-03-PCT - P2338R1 - 02120.004WO1
27 heterocyclic ring, and wherein R14 and the adjacent saturated ring carbon together with the atoms to which they are attached optionally form a fused cyclopropyl ring. Exemplary embodiments include the structures:
Figure imgf000028_0001
where the wavy line indicates the point of attachment to L. Exemplary embodiments of R5 include the structures:
Figure imgf000028_0002
[00104] In other embodiments, R5 is -NR12C(=Y1)(CR14R15)C(=Y2)NRR1 ' wherein R4 is null and R10 and R15 together with the nitrogen atom to which they are attached form a heteroaryl ring optionally having an additional ring nitrogen atom. Exemplary embodiments of R5 include the structures: 111-03-PCT - P2338R1 - 02120.004WO1
28
Figure imgf000029_0001
where Y1 and Y2 are independently selected from O and S; and where the wavy line indicates the point of attachment to L. In certain embodiments, R11 is aryl or a Ci-Ci2 alkyl substituted with aryl, wherein said aryl portions are optionally substituted. Particular embodiments include the structures:
Figure imgf000029_0002
wherein the cyclohexyl and phenyl groups are optionally substituted with one or more R groups independently selected from F, Cl, Br, I, SO2RC, CN, ORa, NRaRb, C(=O)NRaRb, CRaC(=O)Rb, Ci-Ci2 alkyl, C2-C8 alkenyl, C2-C8 alkynyl, C6-C20 aryl, and C]-C20 heteroaryl. [00105] In certain embodiments, R5 is -NR12C(=Y1)(CR14R15)C(- Y^NR10R1 \ wherein
R12 and R14 together with the atoms to which they are attached form a 5-6 membered heterocyclic ring. Exemplary embodiments include, but are not limited to 111-03-PCT - P2338R1 - 02120.004WO1
29
Figure imgf000030_0001
[00106] A particular example includes
Figure imgf000030_0002
[00107] In other embodiments, R5 is -NR10C(=Y)R13. In certain embodiments, R13 is C1-
C6 alkyl, (CRHR15)n-O-(CR14R15)m-aryl, (CR14R15)-aryl, (CR14R15)-heteroaryl, (CR14R15)- heterocyclyl, (CR14R15)-N(SO2Ra)(CR14R15)Rπ, or (CR14R15)NR10C(=O)-aryl, wherein said alkyl, aryl, heteroaryl and heterocyclyl portions are optionally substituted. Exemplary embodiments of R5 include the structures:
Figure imgf000030_0003
111-03-PCT- P2338R1 - 02120.004WO1
30
Figure imgf000031_0001
where the wavy line indicates the point of attachment to L.
[00108] In certain embodiments, R5 is -NR10C(=Y)OR13, including the exemplary structures:
Figure imgf000031_0002
where the wavy line indicates the point of attachment to L.
[00109] In certain embodiments, R5 is -NR12SO2R10, including where R10 is alkyl or optionally substituted aryl. Exemplary embodiments include the structures: 111-03-PCT- P2338R1 - 02120.004WO1
31
Figure imgf000032_0001
where the wavy line indicates the point of attachment to L.
[00110] In certain embodiments, R5 is an optionally substituted carbocyclyl, including the exemplary structures:
Figure imgf000032_0002
where the wavy line indicates the point of attachment to L.
[00111] In certain embodiments, R5 is an optionallysubstituted heterocyclyl, including the exemplary structures:
Figure imgf000032_0003
where the wavy line indicates the point of attachment to L.
[00112] In certain embodiments, R5 is an optionally substituted aryl, including the exemplary structures: 111-03-PCT - P2338R1 - 02120.004WO1
32
Figure imgf000033_0001
where the wavy line indicates the point of attachment to L.
[00113] In certain embodiments, R5 is an optionally substituted heteroaryl, including the exemplary structures:
Figure imgf000033_0002
where R^ is H, Ci-C12 alkyl, C3-Ci2 cycloalkyl, C6-C20 aryl, or Ci-C20 heteroaryl, and Ir 211 and
R are independently selected from H or Ci-Ci2 alkyl, wherein said alkyl, cycloalkyl, aryl, heteroaryl are optionally substituted with one or more groups independently selected from F, Cl, Br, I and Ci-Ci2 alkyl; and where the wavy line indicates the point of attachment to L. In certain embodiments, R20 is H. 111-03-PCT - P2338R1 - 02120.004WO1
33
[00114] In certain embodiments, R5 is an optionally substituted heteroaryl, including the exemplary structures:
Figure imgf000034_0001
where the phenyl groups are optionally substituted with one or more Rd groups independently selected from F, Cl, Br, I, CF3, SO2RC, CN, ORa, NRaRb, C(=O)NRaRb, CRaC(=O)Rb, Ci-Cj2 alkyl, C2-C8 alkenyl, C2-C8 alkynyl, C6-C20 aryl, and Ci-C20 heteroaryl; and each Re is independently H, Cj-C4 alkyl or NHC(=O)-aryl wherein said aryl is optionally substituted with halogen.
[00115] In certain embodiments, R5 is an optionally substituted heteroaryl, including the exemplary structures: 111-03-PCT- P2338R1 - 02120.004WO1
34
Figure imgf000035_0001
[00116] The quinoline compounds of the invention may contain asymmetric or chiral centers, and therefore exist in different stereoisomeric forms. It is intended that all stereoisomeric forms of the compounds of the invention, including but not limited to, diastereomers, enantiomers and atropisomers, as well as mixtures thereof such as racemic mixtures, form part of the present invention.
[00117] In addition, the present invention embraces all geometric and positional isomers.
For example, if a quinoline compound of the present invention incorporates a double bond or a fused ring, the cis- and trans-forms, as well as mixtures thereof, are embraced within the scope of the invention. Both the single positional isomers and mixture of positional isomers, e.g., resulting from the N-oxidation of the pyrimidine and pyrazine rings, are also within the scope of the present invention.
[00118] In the structures shown herein, where the stereochemistry of any particular chiral atom is not specified, then all stereoisomers are contemplated and included as the compounds of the invention. Where stereochemistry is specified by a solid wedge or dashed line representing a particular configuration, then that stereoisomer is so specified and defined.
[00119] The compounds of the present invention may exist in unsolvated as well as solvated forms with pharmaceutically acceptable solvents such as water, ethanol, and the like, and it is intended that the invention embrace both solvated and unsolvated forms.
[00120] The compounds of the present invention may also exist in different tautomeric forms, and all such forms are embraced within the scope of the invention. The term "tautomer" or "tautomeric form" refers to structural isomers of different energies which are interconvertible via a low energy barrier. For example, proton tautomers (also known as prototropic tautomers) include interconversions via migration of a proton, such as keto-enol and imine-enamine isomerizations. Valence tautomers include interconversions by reorganization of some of the bonding electrons.
[00121] The present invention also embraces isotopically-labeled compounds of the present invention which are identical to those recited herein, but for the fact that one or more atoms are replaced by an atom having an atomic mass or mass number different from the atomic 111-03-PCT - P2338R1 - 02120.004WO1
35 mass or mass number usually found in nature. All isotopes of any particular atom or element as specified are contemplated within the scope of the compounds of the invention, and their uses. Exemplary isotopes that can be incorporated into compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine, chlorine and iodine, such as 2H, 3H, 11C, 13C, 14C, 13N, 15N, 15O, 17O, 180, 32P, 33P, 35S, 18F, 36Cl, 123I and 125I. Certain isotopically- labeled compounds of the present invention (e.g., those labeled with 3H and 14C) are useful in compound and/or substrate tissue distribution assays. Tritiated (3H) and carbon- 14 (14C) isotopes are useful for their ease of preparation and detectability. Further, substitution with heavier isotopes such as deuterium (i.e., 2H) may afford certain therapeutic advantages resulting from greater metabolic stability (e.g., increased in vivo half-life or reduced dosage requirements) and hence may be preferred in some circumstances. Positron emitting isotopes such as 15O, 13N, 11C and 18F are useful for positron emission tomography (PET) studies to examine substrate receptor occupancy. Isotopically labeled compounds of the present invention can generally be prepared by following procedures analogous to those disclosed in the Schemes and/or in the Examples herein below, by substituting an isotopically labeled reagent for a non-isotopically labeled reagent.
SYNTHESIS OF cMET INHIBITOR COMPOUNDS
[00122] Quinoline compounds of Formula I of the present invention may be synthesized by synthetic routes that include processes analogous to those well-known in the chemical arts, particularly in light of the description contained herein. The starting materials are generally available from commercial sources such as Aldrich Chemicals (Milwaukee, WI) or are readily prepared using methods well known to those skilled in the art (e.g., prepared by methods generally described in Louis F. Fieser and Mary Fieser, Reagents for Organic Synthesis, v. 1-19, Wiley, N.Y. (1967-1999 ed.), or Beilsteins Handbuch der organischen Chemie, 4, Aufl. ed. Springer- Verlag, Berlin, including supplements (also available via the Beilstein online database). [00123] In certain embodiments, compounds of Formula I may be readily prepared using procedures well-known to prepare quinoline compounds; and other heterocycles, which are described in: Comprehensive Heterocyclic Chemistry, Editors Katritzky and Rees, Pergamon Press, 1984; Klemm et al (1970) J. Hetero. Chem. 7(2):373-379; Klemm et al (1974) J. Hetero. Chem. 11(3): 355-361; Klemm et al (1976) J. Hetero. Chem. 13:273-275; Klemm et al (1985) J. Hetero. Chem. 22(5):1395-1396; Bisagni et al (1974) Bull. Soc. Chim. Fr. (3-4, Pt. 2):515-518; Frehel et al (1984) Heterocycles 22(5):1235-1247; WO 93/13664; WO 2004/012671; WO 2005/061476; U.S. Application Publication Nos. 2003/0045540, US 2003/0105089, and 2004/0024210; and U.S. Patent Nos. 5,252,581, 6,232,320, and 6,579,882. 111-03-PCT- P2338R1 - 02120.004WO1
36
[00124] Compounds of Formula I may be prepared singly or as compound libraries comprising at least 2, for example 5 to 1,000 compounds, or 10 to 100 compounds. Libraries of compounds of Formula I may be prepared by a combinatorial 'split and mix' approach or by multiple parallel syntheses using either solution phase or solid phase chemistry, by procedures known to those skilled in the art. Thus according to a further aspect of the invention there is provided a compound library comprising at least 2 compounds, or pharmaceutically acceptable salts thereof.
[00125] For illustrative purposes, Schemes 1-20 show general methods for preparing the compounds of the present invention as well as key intermediates. For a more detailed description of the individual reaction steps, see the Examples section below. Those skilled in the art will appreciate that other synthetic routes may be used to synthesize the inventive compounds. Although specific starting materials and reagents are depicted in the Schemes and discussed below, other starting materials and reagents can be easily substituted to provide a variety of derivatives and/or reaction conditions. In addition, many of the compounds prepared by the methods described below can be further modified in light of this disclosure using conventional chemistry well known to those skilled in the art.
[00126] In preparing compounds of Formulas I, protection of remote functionality (e.g., primary or secondary amine) of intermediates may be necessary. The need for such protection will vary depending on the nature of the remote functionality and the conditions of the preparation methods. Suitable amino-protecting groups (NH-Pg) include acetyl, trifluoroacetyl, t- butoxycarbonyl (BOC), benzyloxycarbonyl (CBz) and 9-fluorenylmethyleneoxycarbonyl (Fmoc). The need for such protection is readily determined by one skilled in the art. For a general description of protecting groups and their use, see T. W. Greene, Protective Groups in Organic Synthesis, John Wiley & Sons, New York, 1991.
111-03-PCT - P2338R1 - Q2120.004WO1
37
Figure imgf000038_0001
1) R11OH coupling
2) Nitro reduction
Figure imgf000038_0002
Scheme 1
[00127] Scheme 1 shows a general method for the synthesis of intermediate compound 4, which is useful for the synthesis of compounds of Formula I. Syntheses of 4-phenoxy-6,7- dialkoxyquinolines have been previously reported in US 2004/0242603; US 2005/0002326; WO 2005/030140; J. Med. Chem. (2005) 48:1359-1366. As shown in Scheme 1, reaction of a 4- hydroxy-6,7-dialkoxyquinoline 1 with a variably substituted p-halonitroarene or heteroarene wherein X is F or Cl and Y is N or CH using an appropriate base (e.g. Cs2CO3, NaH, KOt-Bu, or the like) provides intermediate 2. The protecting group PG can then be removed (in the case where PG = benzyl, HBr or TFA can be used for the deprotection) to give intermediate 3 and a new 7-alkoxy substituent introduced, typically using Mitsunobu conditions or alkylation with an alkyl halide and base. Nitro reduction under hydrogenation conditions or with zinc in acetic acid gave key intermediate 4. Alternatively, exchange of the protecting group PG exchange for R11 could be omitted from the sequence and thus provide compound 4 with the 6,7-alkoxy substituents originally contained in intermediate 1. Compound 4 can then be coupled with an appropriate acid (as prepared according to Schemes 13-17 below) using standard amide bond construction methods to provide compound 5. 111-03-PCT - P2338R1 - 02120.004WO1
38
Figure imgf000039_0001
Scheme 2
[00128] Scheme 2 shows a general synthetic route for the synthesis of compound 9, which is useful for the synthesis of compounds of Formula I. A 4-chloro-6,7-dialkoxyquinoline, such as compound 6, can be prepared by chlorination of the corresponding hydroxyquinoline (compound 1, Scheme 1) typically using POCl3, MeSO2Cl and the like. The protecting group PG can then be removed (in the case where PG = benzyl, HBr, or TFA can be used for the deprotection) and a new 7-alkoxy substituent introduced, typically using Mitsunobu conditions or alkylation with an alkyl halide and base to give compound 7. Compound 7 can then be reacted under basic conditions, typically DMAP in bromobenzene or Cs23 in DMF, with a functionalized phenol 8 (general schemes for synthesis of preferred functionalized phenols are shown below) to give rise to compound 9. Compound 9 can optionally be further manipulated depending on the phenol functionalization. Alternatively, PG exchange for R11 can be omitted from the sequence and thus provide intermediate 9 with the 6,7-alkoxy substituents originally contained in intermediate 6.
Figure imgf000039_0002
10 11 12
Figure imgf000039_0003
13 14
Scheme 3
[00129] Scheme 3 shows a route for the preparation of phenol compound 14.
Commercially available 2-chloro-4-methoxypyrimidine 10 is reacted with the appropriate zinc 111-03-PCT - P2338R1 - 02120.004WO1
39 reagent and palladium catalyst to give 2-substituted 4-methoxypyrimidine 11. Deprotection of the methoxypyrimidine with HBr in acetic acid provides 2-substituted pyrimidinone 12. Bromination in the 5-position gives pyrimidinone intermediate 13. Suzuki coupling of compound 13 to an appropriate boronic acid gives a bicyclic intermediate, which after final deprotection of the phenol gives compound 14 which can be reacted with the appropriate core intermediate 7 as in Scheme 2 to provide compound 9.
Figure imgf000040_0001
Figure imgf000040_0002
20 Scheme 4
[00130] Scheme 4 shows a route for the preparation of the 1 -substituted pyrimidinone intermediate 20 (wherein R10 is independently selected from H, alkyl, aryl and heteroaryl). 5- bromo-2,4-dichloropyrimidine 15 is hydrolyzed with NaOH to give 5-bromo-2-chloropyrimidin- 4(3H)-one 16 as described in EP1506967 Al. Alkylation of compound 16 to provide the 1- substituted pyrimidinone can be accomplished with an alkylation agent (e.g. iodomethane, or the like) mediated by an appropriate base (e.g. sodium alkoxides, lithium or sodium hydride, or the like) providing a mixture of isomers 17 and 18. Isomers 17 and 18 can be separated using purification techniques known to those skilled in the art (e.g. flash chromatography, reverse phase HPLC, or the like). Compound 18 is reacted with the appropriate zinc reagent and palladium catalyst to give 2-substituted intermediate 19. Suzuki coupling of compound 19 to an appropriate boronic acid followed by final deprotection of the phenol gives compound 20, which can be reacted with the appropriate core intermediate 7 as in Scheme 2 to provide compound 9. 111-03-PCT - P2338R1 - 02120.004WO1
40
Figure imgf000041_0001
21 22 23
Figure imgf000041_0002
24 25
Figure imgf000041_0003
26 27
Scheme 5
[00131] Scheme 5 shows a method for preparing phenol intermediate 27 (wherein R10 is independently selected from H, alkyl, aryl and heteroaryl). Nucleophilic substitution of 2-chloro- 4-methoxypyrimidine 21 with a compound of the formula HY-R10, (wherein Y is O, N or S) can be accomplished in an appropriate solvent such as n-butanol, at refluxing temperature to give intermediate 22. Deprotection of the methoxypyrimidine with HBr in acetic acid provides 2- substituted pyrimidinone 23. Alkylation of 23 to provide the 1 -substituted pyrimidinone can be accomplished with an alkylation agent (e.g. iodomethane, or the like) mediated by an appropriate base (e.g. sodium alkoxides, lithium or sodium hydride, or the like) providing a mixture of isomers 24 and 25. Isomers 24 and 25 can be separated using purification techniques known to those skilled in the art (e.g. flash chromatography, reverse phase HPLC, or the like). Bromination in the 5-position with a brominating agent such as Br2 or NBS gives compound 26. Suzuki coupling of compound 26 to an appropriate boronic acid gives a bicyclic intermediate which after final deprotection of the phenol gives compound 27. Compound 27 can be reacted with the appropriate core intermediate 7 as in Scheme 2 to provide compound 9.
Figure imgf000041_0004
111-03-PCT - P2338R1 - 02120.004WO1
41
Scheme 6
[00132] Scheme 6 shows an alternative route to compound 27 (wherein R10 is independently selected from H, alkyl, aryl and heteroaryl). Nucleophilic substitution of compound 28 with a compound of the formula HY-R10 (wherein Y is O, N or S) can be accomplished at elevated temperature with a base such as NaHCO3 in an appropriate solvent such as n-butanol to give intermediate 26. Suzuki coupling of compound 26 to an appropriate boronic acid gives a bicyclic intermediate which after final deprotection of the phenol gives compound 27. Intermediate 27 can then be reacted with the appropriate core intermediate 7 as in Scheme 2 to give compound 9.
Figure imgf000042_0001
Scheme 7
[00133] Scheme 7 shows a route to phenol intermediate 32 (wherein R10 is independently selected from H, alkyl, aryl and heteroaryl). Nucleophilic substitution of compound 29 with NaOMe can be accomplished at elevated temperature in an appropriate solvent such as methanol. Nucleophilic substitution of compound 30 with a compound of the formula HY-R10, (wherein Y is O, N or S), to form the 5-substituted pyrimidinone 31 can be accomplished at elevated temperature with a base such as NaHCO3 in an appropriate solvent such as n-butanol. Under these reaction conditions, deprotection of the methoxypyrimidine to the pyrimidinone can also be achieved. Alternatively, deprotection of the methoxypyrimidine can be accomplished with HBr in acetic acid. Copper (I)-mediated coupling of compound 31 to an appropriate halide provides compound 32. In some instances, the halide used in the coupling reaction contains a standard protecting group. In those cases, the protecting group can be removed by standard conditions known in the art. Compound 32 can then be reacted with the appropriate core intermediate 7 as in Scheme 2 to provide compound 9. 111-03-PCT - P2338R1 - 02120.004WO1
42
Figure imgf000043_0001
Figure imgf000043_0002
Scheme 8
[00134] Scheme 8 shows a route for the preparation of the 1 -substituted pyridone intermediate 38 (wherein R10 is independently selected from H, alkyl, aryl and heteroaryl). Alkylation of 6-chloropyridin-2-ol 33 to provide the 1 -substituted pyridone 35 can be accomplished with an alkylation agent (e.g. iodomethane, or the like) mediated by an appropriate base (e.g. potassium carbonate, sodium alkoxides, lithium or sodium hydride, or the like) providing a mixture of isomers 34 and 35. Isomers 34 and 35 can be separated using purification techniques known to those skilled in the art (e.g. flash chromatography, reverse phase HPLC, or the like). Compound 35 is reacted with the appropriate zinc reagent and palladium catalyst to give 6-substituted compound 36. Bromination of the 3-position with a brominating agent such as Br2 or NBS gives pyridone intermediate 37. Suzuki coupling of compound 37 to an appropriate boronic acid followed by final deprotection of the phenol gives compound 38, which can then be reacted with the appropriate core intermediate 7 as in Scheme 2 to give compound 9.
Figure imgf000043_0003
Deprotection
Figure imgf000043_0004
Figure imgf000043_0005
42 43
Scheme 9
[00135] Scheme 9 shows a method for preparing phenol intermediate 43 (wherein R10 is independently selected from H, alkyl, aryl and heteroaryl). 1 -Substituted pyridone 35, which can 111-03-PCT- P2338R1 - 02120.004WO1
43 be synthesized as shown in Scheme 8, is brominated with a brominating agent such as Br2 or NBS providing a mixture of isomers 39 and 40. Isomers 39 and 40 can be separated using purification techniques known to those skilled in the art (e.g. flash chromatography, reverse phase HPLC, or the like). Suzuki coupling of compound 40 with an appropriate boronic acid gives compound 41. Nucleophilic substitution of compound 41 with a compound of the formula HY- R10, (wherein Y is O, N or S) can be accomplished in an appropriate solvent such as THF, mediated by an appropriate base such as LDA, LiHMDS, NaHMDS, or KHMDS at appropriate temperatures (-78 °C to room temperature) to give compound 42. Final deprotection of the phenol gives compound 43, which can then be reacted with appropriate core intermediate 7 as in Scheme 2 to give compound 9.
Figure imgf000044_0001
49 50
Scheme 10
[00136] Scheme 10 shows a route for the preparation of the 6-acyl pyridin-2(lH)-one phenol compound 48. Base-mediated halogen exchange of the commercially available bromopyridine 44 followed by quenching with an aldehyde gives the secondary alcohol compound 45. Oxidation of the alcohol followed by demethylation gives compound 46. Bromination of compound 46 followed by a Suzuki coupling with an appropriate boronic acid gives a coupling compound 47. Final deprotection of the phenol gives compound 48, which can then be reacted with appropriate core intermediate 7 as in Scheme 2 to give compound 9. Sodium borohydride reduction of this compound gives compound 49, and acetylation of compound 49 gives intermediate 50. Compounds 49 and 50 can also be reacted with appropriate compound 7 as in Scheme 2 to provide compound 9. 111-03-PCT - P2338R1 - 02120.004WO1
44
Figure imgf000045_0001
Scheme 11
[00137] Scheme 11 shows a route for the preparation of 3-benzyl substituted pyrimidin-
4(3H)-one phenol compound 53 which is useful for the synthesis of compounds of Formula I. 5- Bromopyrimidin-4(3H)-one 51 is reacted with a base such as NaH, and an appropriate bromide or chloride of formula R10- Y-X to give the corresponding 3-benzyl-5-bromopyrimidin-4(3H)-one 52. Suzuki coupling of compound 52 with an appropriate boronic acid gives a coupling intermediate, which after final deprotection of the phenol gives compound 53, which can be reacted with appropriate core intermediate 7 as in Scheme 2 to provide compound 9.
Figure imgf000045_0002
Scheme 12
[00138] Scheme 12 shows a route for the preparation of the 5-benzyl-3-(4- hydroxyphenyl)pyrimidin-4(3H)-one phenol intermediate 58 which is useful for the synthesis of compounds of Formula I. Commercially available 4,6-dichloropyrimidine-5-carbaldehyde 54 is reacted with the appropriate substituted phenyl magnesium halide to give the secondary alcohol 55. Monobenzylation gives compound 56, which is subjected to hydrogenation to provide compound 57. Copper (I)-mediated coupling of compound 57 to an appropriate phenol provides the desired compound 58, which can be reacted with appropriate core intermediate 7 as in Scheme 2 to provide compound 9. 111-03-PCT- P2338R1 - 02120.004WO1
45
Figure imgf000046_0001
59 60 61
cyclization
Figure imgf000046_0002
62
Scheme 13
[00139] The pyridazino carboxylic acid compound 62 can be prepared using methods described by McNab H. et al (1982) J. Chem. Soc. Perkin Trans. 1:1845 as depicted in Scheme 13. Substituted hydrazine 59 can be converted to hydrazono acetaldehyde 60 with standard dehydrating conditions such as acetic acid at room temperature. The carbonyl group condensation product 61 is prepared in a suitable organic solvent such as toluene, benzene or dioxane at room temperature using piperidinium acetate as catalyst. Carboxylic acid pyridazinone 62 is prepared from hydrazono ethylidene 61 by cyclization under basic conditions (sodium methoxide in methanol) at 7O0C. When R2 or R3 = CH3 or alkyl, the desired product 62 can be obtained in a one-pot reaction through condensation and cyclization of compound 60. Compound 62 may then be used to acylate aniline intermediate 4 whose preparation is described in Scheme 1 to prepare compounds of Formula I.
1. hydrolysis
2. hydrogenation (X= H, F, alkyl)
Figure imgf000046_0004
Figure imgf000046_0003
63 64
Scheme 14
[00140] Scheme 14 shows a route for the preparation of oxo-4-phenyl-3,4- dihydropyrazine-2 -carboxylic acids. The pyrazine-2-carbonitrile 63 was prepared using methods described by Hoornaert, G., et al (1983) J. Heterocyclic Chem. 20:919 and Hoornaert, G., et al (1990) Tetrahedron 46:5715. The pyrazine-2-carboxylic acid 64 can be prepared by hydrolysis to the carboxylic acid followed by removal of the chloro group under hydrogenolysis conditions to 111-03-PCT - P2338R1 - 02120.004WO1
46 give the desired 3-oxo-4-phenyl-3,4-dihydropyrazine-2-carboxylic acid 64. The acid 64 can then be coupled via standard amide bond forming techniques to an aniline bearing core 4, prepared according to Scheme 1 to provide final compound 5.
Figure imgf000047_0001
65
Scheme 15
[00141] The substituted pyrazino carboxylic acids 67 can be prepared as in Scheme 15.
Methyl 3-oxo-3,4-dihydropyrazine-2-carboxylate 65 can be converted to alkylpyrazino carboxylate 66 by standard basic alkylation conditions with alkyl halides. These conditions include but are not limited to treatment with K2CO3 in acetone or DMF at room or elevated temperature, or NaH in THF at ambient or elevated temperature, followed by addition of the alkyl halide. In certain embodiments, this alkylation is achieved with LiH in DMF at O0C, followed by addition of alkyl chloride or alkyl bromide or alkyl iodide and warming to room temperature. Carboxylic acid 67 can then be prepared using standard saponification conditions such as LiOH or NaOH in standard mixed aqueous/organic solvent systems. The acid 67 can then be coupled via standard amide bond forming techniques to an aniline bearing core 4 constructed according to Scheme 1 to provide final compound 5.
Figure imgf000047_0002
68 69 70
Scheme 16
[00142] Scheme 16 shows a method for preparing pyrrolidin-2-one intermediate 70
(wherein R12 is independently selected from H, alkyl, aryl and heteroaryl). Carbonylation of the N-substituted pyrrolidin-2-one 68 can be completed by treatment with LDA, followed by quenching with methyl carbonochloridate to give ester 69. Hydrolysis of the ester with appropriate base, such as TMSOK, KOH, etc, yields the corresponding acid 70. The acid 70 can then be coupled via standard amide bond forming techniques to an aniline bearing core such as 4 according to Scheme 1 to provide compound 5. 111-03-PCT - P2338R1 - 02120.004WO1
47
cyclization
Figure imgf000048_0001
hydrolysis
Figure imgf000048_0002
Figure imgf000048_0003
73 74
Scheme 17
[00143] Scheme 17 shows a method for the preparation of the fused bicyclic cyclopropane lactam ester 74. An optionally substituted allylic amine 71 is acylated with a malonyl chloride ester under basic conditions to give the allylic amide intermediate 72. Cyclization under conditions which generate the malonyl carbine (preferably manganese III acetate catalyzed) provide the fused cyclopropyllactam compound 73. Deprotection under basic conditions (typically LiOH or NaOH in an aqueous/organic solvent mixture) provides the intermediate acid 74. The acid 74 can then be coupled via standard amide bond forming techniques to aniline bearing cores such as 4 constructed according to Scheme 1 to provide final compound 5.
Figure imgf000048_0004
Scheme 18
[00144] Scheme 18 shows a route for the preparation of (piperidin-l-yl)methanone phenol intermediate 78 which is useful for the synthesis of compounds of Formula I. Substituted and O- protected benzoyl chlorides of type 76 are reacted with an appropriate amine 75 to form the corresponding amide 77, which after final deprotection of the phenol gives compound 78. Compound 78 can then be reacted with the appropriate core intermediate 7 as in Scheme 2 to provide compound 9. 111-03-PCT - P2338R1 - 02120.004WO1
48
HY-R10
Pd Catalyst
Figure imgf000049_0001
Figure imgf000049_0002
Scheme 19
[00145] Scheme 19 shows a method for the preparation of phenolic intermediate 82. 2,5- dibromopyridine 79 is treated with the appropriate boronic acid under Suzuki type reaction conditions to give selective coupling at the pyridine 2-position to provide compound 80. Buchwald type palladium coupling of compound 80 with an appropriate heteroatom bearing an R10 group gives the protected compound 81. Final deprotection of compound 81 gives compound 82 which can be reacted with an appropriate core intermediate 7 as in Scheme 2 to provide compound 9.
Figure imgf000049_0003
Pd Catalyst
83 84
Figure imgf000049_0004
Scheme 20
[00146] Scheme 20 shows a method for the preparation of phenolic intermediate 86. 2,5-
Dibromopyrimidine 83 is treated with the appropriate heteroatom bearing an R10 group with heating in an appropriate solvent such as 1-propanol. Reaction occurs selectively at the 2- position to give the bromopyrimidine intermediate 84. Suzuki coupling to the appropriately substituted boronic acid gives intermediate 85, which after deprotection gives the phenolic 111-03-PCT - P2338R1 - 02120.004WO1
49 compound 86. Compound can be reacted with appropriate core intermediate 7 as in Scheme 2 to provide compound 9. METHODS OF SEPARATION
[00147] In the methods of preparing the compounds of this invention, it may be advantageous to separate reaction products from one another and/or from starting materials. The desired products of each step or series of steps is separated and/or purified (hereinafter separated) to the desired degree of homogeneity by the techniques common in the art. Typically such separations involve multiphase extraction, crystallization from a solvent or solvent mixture, distillation, sublimation, or chromatography. Chromatography can involve any number of methods including, for example: reverse-phase and normal phase; size exclusion; ion exchange; high, medium and low pressure liquid chromatography methods and apparatus; small scale analytical; simulated moving bed (SMB) and preparative thin or thick layer chromatography, as well as techniques of small scale thin layer and flash chromatography.
[00148] Another class of separation methods involves treatment of a mixture with a reagent selected to bind to or render otherwise separable a desired product, unreacted starting material, reaction by product, or the like. Such reagents include adsorbents or absorbents such as activated carbon, molecular sieves, ion exchange media, or the like. Alternatively, the reagents can be acids in the case of a basic material, bases in the case of an acidic material, binding reagents such as antibodies, binding proteins, selective chelators such as crown ethers, liquid/liquid ion extraction reagents (LIX), or the like.
[00149] Selection of appropriate methods of separation depends on the nature of the materials involved. For example, boiling point and molecular weight in distillation and sublimation, presence or absence of polar functional groups in chromatography, stability of materials in acidic and basic media in multiphase extraction, and the like. One skilled in the art will apply techniques most likely to achieve the desired separation.
[00150] Diastereomeric mixtures can be separated into their individual diastereomers on the basis of their physical chemical differences by methods well known to those skilled in the art, such as by chromatography and/or fractional crystallization. Enantiomers can be separated by converting the enantiomeric mixture into a diastereomeric mixture by reaction with an appropriate optically active compound (e.g., chiral auxiliary such as a chiral alcohol or Mosher's acid chloride), separating the diastereomers and converting (e.g., hydrolyzing) the individual diastereoisomers to the corresponding pure enantiomers. Also, some of the compounds of the present invention may be atropisomers (e.g., substituted biaryls) and are considered as part of this invention. Enantiomers can also be separated by use of a chiral FIPLC column. 111-03-PCT- P2338R1 - 02120.004WO1
50
[00151] A single stereoisomer, e.g., an enantiomer, substantially free of its stereoisomer may be obtained by resolution of the racemic mixture using a method such as formation of diastereomers using optically active resolving agents (Eliel, E. and Wilen, S. "Stereochemistry of Organic Compounds," John Wiley & Sons, Inc., New York, 1994; Lochmuller, C. H., (1975) J. Chromatogr., 113(3):283-302). Racemic mixtures of chiral compounds of the invention can be separated and isolated by any suitable method, including: (1) formation of ionic, diastereomeric salts with chiral compounds and separation by fractional crystallization or other methods, (2) formation of diastereomeric compounds with chiral derivatizing reagents, separation of the diastereomers, and conversion to the pure stereoisomers, and (3) separation of the substantially pure or enriched stereoisomers directly under chiral conditions. See: "Drug Stereochemistry, Analytical Methods and Pharmacology," Irving W. Wainer, Ed., Marcel Dekker, Inc., New York (1993).
[00152] Under method (1), diastereomeric salts can be formed by reaction of enantiomerically pure chiral bases such as brucine, quinine, ephedrine, strychnine, α-methyl-β- phenylethylamine (amphetamine), and the like with asymmetric compounds bearing acidic functionality, such as carboxylic acid and sulfonic acid. The diastereomeric salts may be induced to separate by fractional crystallization or ionic chromatography. For separation of the optical isomers of amino compounds, addition of chiral carboxylic or sulfonic acids, such as camphorsulfonic acid, tartaric acid, mandelic acid, or lactic acid can result in formation of the diastereomeric salts.
[00153] Alternatively, by method (2), the substrate to be resolved is reacted with one enantiomer of a chiral compound to form a diastereomeric pair (E. and Wilen, S. "Stereochemistry of Organic Compounds", John Wiley & Sons, Inc., 1994, p. 322). Diastereomeric compounds can be formed by reacting asymmetric compounds with enantiomerically pure chiral derivatizing reagents, such as menthyl derivatives, followed by separation of the diastereomers and hydrolysis to yield the pure or enriched enantiomer. A method of determining optical purity involves making chiral esters, such as a menthyl ester, e.g., (-) menthyl chloroformate in the presence of base, or Mosher ester, oc-methoxy-α- (trifluoromethyl)phenyl acetate (Jacob III (1982) J. Org. Chem 47:4165), of the racemic mixture, and analyzing the 1H NMR spectrum for the presence of the two atropisomeric enantiomers or diastereomers. Stable diastereomers of atropisomeric compounds can be separated and isolated by normal- and reverse-phase chromatography following methods for separation of atropisomeric naphthyl-isoquinolines (WO 96/15111). By method (3), a racemic mixture of two enantiomers can be separated by chromatography using a chiral stationary phase ("Chiral Liquid 111-03-PCT - P2338R1 - 02120.004WO1
51
Chromatography" (1989) W. J. Lough, Ed., Chapman and Hall, New York; Okamoto, J. Chromatogr., (1990) 513:375-378). Enriched or purified enantiomers can be distinguished by methods used to distinguish other chiral molecules with asymmetric carbon atoms, such as optical rotation and circular dichroism.
[00154] Exemplary compounds of this invention include compounds 101-205 as described in Examples 1-105. BIOLOGICAL EVALUATION
[00155] Determination of the activity of c-Met kinase activity of a compound of Formula I is possible by a number of direct and indirect detection methods. One example of an assay used for the determination of c-Met kinase activity is based on an enzyme linked immunosorbant assay (ELISA). The assay includes a compound of Formula I, c-Met (His-tagged recombinant human Met (amino acids 974-end), expressed by baculovirus), and ATP in assay buffer, as described in Example 106.
[00156] In MKN45 cells, the activity of c-Met inhibitors of Formula I was determined by the in vitro fluorescence assay as described in Example 107.
[00157] Exemplary compounds described herein were prepared, characterized, and assayed for their c-Met binding activity and in vitro activity against tumor cells. The range of c- Met binding activities was less than 1 nM to about 10 μM. Certain exemplary compounds of the invention had c-Met binding activity IC50 values less than 10 nM. Certain compounds of the invention had MKN45 cell-based activity IC50 values less than 100 nM. ADMINISTRATION OF COMPOUNDS OF FORMULA I
[00158] The compounds of the invention may be administered by any route appropriate to the condition to be treated. Suitable routes include oral, parenteral (including subcutaneous, intramuscular, intravenous, intraarterial, intradermal, intrathecal and epidural), transdermal, rectal, nasal, topical (including buccal and sublingual), vaginal, intraperitoneal, intrapulmonary and intranasal. For local immunosuppressive treatment, the compounds may be administered by intralesional administration, including perfusing or otherwise contacting the graft with the inhibitor before transplantation. It will be appreciated that the preferred route may vary with for example the condition of the recipient. Where the compound is administered orally, it may be formulated as a pill, capsule, tablet, etc. with a pharmaceutically acceptable carrier or excipient. Where the compound is administered parenterally, it may be formulated with a pharmaceutically acceptable parenteral vehicle and in a unit dosage injectable form, as detailed below. METHODS OF TREATMENT WITH COMPOUNDS OF FORMULA I 111-03-PCT - P2338R1 - 02120.004WO1
52
[00159] Compounds of the present invention are useful for treating diseases, conditions and/or disorders including, but not limited to, those characterized by over expression of receptor tyrosine kinases (RTK), e.g. c-Met kinase. Accordingly, another aspect of this invention includes methods of treating or preventing diseases or conditions that can be treated or prevented by inhibiting receptor tyrosine kinases (RTK), including c-Met. In one embodiment, the method comprises administering to a mammal in need thereof a therapeutically effective amount of a compound of Formula I, or a stereoisomer, geometric isomer, tautomer, solvate, metabolite, or pharmaceutically acceptable salt or prodrug thereof.
[00160] Diseases and conditions treatable according to the methods of this invention include, but are not limited to, cancer, stroke, diabetes, hepatomegaly, cardiovascular disease, Alzheimer's disease, cystic fibrosis, viral disease, autoimmune diseases, atherosclerosis, restenosis, psoriasis, allergic disorders, inflammation, neurological disorders, a hormone-related disease, conditions associated with organ transplantation, immunodeficiency disorders, destructive bone disorders, proliferative disorders, infectious diseases, conditions associated with cell death, thrombin-induced platelet aggregation, chronic myelogenous leukemia (CML), liver disease, pathologic immune conditions involving T cell activation, and CNS disorders in a patient. In one embodiment, a human patient is treated with a compound of Formula I and a pharmaceutically acceptable carrier, adjuvant, or vehicle, wherein said compound of Formula I is present in an amount to detectably inhibit c-Met kinase activity.
[00161] Cancers which can be treated according to the methods of this invention include, but are not limited to, breast, ovary, cervix, prostate, testis, genitourinary tract, esophagus, larynx, glioblastoma, neuroblastoma, stomach, skin, keratoacanthoma, lung, epidermoid carcinoma, large cell carcinoma, non-small cell lung carcinoma (NSCLC), small cell carcinoma, lung adenocarcinoma, bone, colon, adenoma, pancreas, adenocarcinoma, thyroid, follicular carcinoma, undifferentiated carcinoma, papillary carcinoma, seminoma, melanoma, sarcoma, bladder carcinoma, liver carcinoma and biliary passages, kidney carcinoma, myeloid disorders, lymphoid disorders, hairy cells, buccal cavity and pharynx (oral), lip, tongue, mouth, pharynx, small intestine, colon-rectum, large intestine, rectum, brain and central nervous system, Hodgkin's and leukemia.
[00162] Cardiovascular diseases which can be treated according to the methods of this invention include, but are not limited to, restenosis, cardiomegaly, atherosclerosis, myocardial infarction, and congestive heart failure.
[00163] Neurodegenerative disease which can be treated according to the methods of this invention include, but are not limited to, Alzheimer's disease, Parkinson's disease, amyotrophic 111-03-PCT - P2338R1 - 02120.004WO1
53 lateral sclerosis, Huntington's disease, and cerebral ischemia, and neurodegenerative disease caused by traumatic injury, glutamate neurotoxicity and hypoxia.
[00164] Inflammatory diseases which can be treated according to the methods of this invention include, but are not limited to, rheumatoid arthritis, psoriasis, contact dermatitis, and delayed hypersensitivity reactions.
[00165] Another aspect of this invention provides a compound of this invention for use in the treatment of the diseases or conditions described herein in a mammal, for example, a human, suffering from such disease or condition. Also provided is the use of a compound of this invention in the preparation of a medicament for the treatment of the diseases and conditions described herein in a warm-blooded animal, such as a mammal, for example a human, suffering from such disorder.
PHARMACEUTICAL FORMULATIONS
[00166] In order to use a compound of this invention for the therapeutic treatment
(including prophylactic treatment) of mammals including humans, it is normally formulated in accordance with standard pharmaceutical practice as a pharmaceutical composition. According to this aspect of the invention there is provided a pharmaceutical composition comprising a compound of this invention in association with a pharmaceutically acceptable diluent or carrier. [00167] A typical formulation is prepared by mixing a compound of the present invention and a carrier, diluent or excipient. Suitable carriers, diluents and excipients are well known to those skilled in the art and include materials such as carbohydrates, waxes, water soluble and/or swellable polymers, hydrophilic or hydrophobic materials, gelatin, oils, solvents, water and the like. The particular carrier, diluent or excipient used will depend upon the means and purpose for which the compound of the present invention is being applied. Solvents are generally selected based on solvents recognized by persons skilled in the art as safe (GRAS) to be administered to a mammal. In general, safe solvents are non-toxic aqueous solvents such as water and other nontoxic solvents that are soluble or miscible in water. Suitable aqueous solvents include water, ethanol, propylene glycol, polyethylene glycols (e.g., PEG 400, PEG 300), etc. and mixtures thereof. The formulations may also include one or more buffers, stabilizing agents, surfactants, wetting agents, lubricating agents, emulsifiers, suspending agents, preservatives, antioxidants, opaquing agents, glidants, processing aids, colorants, sweeteners, perfuming agents, flavoring agents and other known additives to provide an elegant presentation of the drug (i.e., a compound of the present invention or pharmaceutical composition thereof) or aid in the manufacturing of the pharmaceutical product (i.e., medicament). 111-03-PCT - P2338R1 - 02120.004WO1
54
[00168] The formulations may be prepared using conventional dissolution and mixing procedures. For example, the bulk drug substance (i.e., compound of the present invention or stabilized form of the compound (e.g., complex with a cyclodextrin derivative or other known complexation agent) is dissolved in a suitable solvent in the presence of one or more of the excipients described above. The compound of the present invention is typically formulated into pharmaceutical dosage forms to provide an easily controllable dosage of the drug and to enable patient compliance with the prescribed regimen.
[00169] The pharmaceutical composition (or formulation) for application may be packaged in a variety of ways depending upon the method used for administering the drug. Generally, an article for distribution includes a container having deposited therein the pharmaceutical formulation in an appropriate form. Suitable containers are well known to those skilled in the art and include materials such as bottles (plastic and glass), sachets, ampoules, plastic bags, metal cylinders, and the like. The container may also include a tamper-proof assemblage to prevent indiscreet access to the contents of the package. In addition, the container has deposited thereon a label that describes the contents of the container. The label may also include appropriate warnings.
[00170] Pharmaceutical formulations of the compounds of the present invention may be prepared for various routes and types of administration. For example, a compound of Formula I having the desired degree of purity may optionally be mixed with pharmaceutically acceptable diluents, carriers, excipients or stabilizers (Remington's Pharmaceutical Sciences (1980) 16th edition, Osol, A. Ed.), in the form of a lyophilized formulation, milled powder, or an aqueous solution. Formulation may be conducted by mixing at ambient temperature at the appropriate pH, and at the desired degree of purity, with physiologically acceptable carriers, i.e., carriers that are non-toxic to recipients at the dosages and concentrations employed. The pH of the formulation depends mainly on the particular use and the concentration of compound, but may range from about 3 to about 8. Formulation in an acetate buffer at pH 5 is a suitable embodiment. [00171] The compound of this invention for use herein is preferably sterile. In particular, formulations to be used for in vivo administration must be sterile. Such sterilization is readily accomplished by filtration through sterile filtration membranes.
[00172] The compound ordinarily can be stored as a solid composition, a lyophilized formulation or as an aqueous solution.
[00173] The pharmaceutical compositions of the invention will be formulated, dosed and administered in a fashion, i.e., amounts, concentrations, schedules, course, vehicles and route of administration, consistent with good medical practice. Factors for consideration in this context 111-03-PCT- P2338R1 - 02120.004WOl
55 include the particular disorder being treated, the particular mammal being treated, the clinical condition of the individual patient, the cause of the disorder, the site of delivery of the agent, the method of administration, the scheduling of administration, and other factors known to medical practitioners. The "therapeutically effective amount" of the compound to be administered will be governed by such considerations, and is the minimum amount necessary to prevent, ameliorate, or treat the coagulation factor mediated disorder. Such amount is preferably below the amount that is toxic to the host or renders the host significantly more susceptible to bleeding. [00174] As a general proposition, the initial pharmaceutically effective amount of the inhibitor administered parenterally per dose will be in the range of about 0.01-100 mg/kg, namely about 0.1 to 20 mg/kg of patient body weight per day, with the typical initial range of compound used being 0.3 to 15 mg/kg/day.
[00175] Acceptable diluents, carriers, excipients and stabilizers are nontoxic to recipients at the dosages and concentrations employed, and include buffers such as phosphate, citrate and other organic acids; antioxidants including ascorbic acid and methionine; preservatives (such as octadecyldimethylbenzyl ammonium chloride; hexamethonium chloride; benzalkonium chloride, benzethonium chloride; phenol, butyl or benzyl alcohol; alkyl parabens such as methyl or propyl paraben; catechol; resorcinol; cyclohexanol; 3-pentanol; and m-cresol); low molecular weight (less than about 10 residues) polypeptides; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, histidine, arginine, or lysine; monosaccharides, disaccharides and other carbohydrates including glucose, mannose, or dextrins; chelating agents such as EDTA; sugars such as sucrose, mannitol, trehalose or sorbitol; salt-forming counter-ions such as sodium; metal complexes (e.g., Zn-protein complexes); and/or non-ionic surfactants such as TWEEN®, PLURONICS® or polyethylene glycol (PEG). The active pharmaceutical ingredients may also be entrapped in microcapsules prepared, for example, by coacervation techniques or by interfacial polymerization, for example, hydroxymethylcellulose or gelatin-microcapsules and poly- (methylmethacylate) microcapsules, respectively, in colloidal drug delivery systems (for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules) or in macroemulsions. Such techniques are disclosed in Remington's Pharmaceutical Sciences 16th edition, Osol, A. Ed. (1980).
[00176] Sustained-release preparations of compounds of Formulas I may be prepared.
Suitable examples of sustained-release preparations include semipermeable matrices of solid hydrophobic polymers containing a compound of Formula I, which matrices are in the form of shaped articles, e.g., films, or microcapsules. Examples of sustained-release matrices include 111-03-PCT - P2338R1 - 02120.004WO1
56 polyesters, hydrogels (for example, poly(2-hydroxyethyl-methacrylate), or poly(vinyl alcohol)), polylactides (U.S. Patent No. 3,773,919), copolymers of L-glutamic acid and gamma-ethyl-L- glutamate, non-degradable ethylene-vinyl acetate, degradable lactic acid-glycolic acid copolymers such as the LUPRON DEPOT® (injectable microspheres composed of lactic acid- glycolic acid copolymer and leuprolide acetate) and poly-D-(-)-3-hydroxybutyric acid. [00177] The formulations include those suitable for the administration routes detailed herein. The formulations may conveniently be presented in unit dosage form and may be prepared by any of the methods well known in the art of pharmacy. Techniques and formulations generally are found in Remington's Pharmaceutical Sciences (Mack Publishing Co., Easton, PA). Such methods include the step of bringing into association the active ingredient with the carrier which constitutes one or more accessory ingredients. In general the formulations are prepared by uniformly and intimately bringing into association the active ingredient with liquid carriers or finely divided solid carriers or both, and then, if necessary, shaping the product. [00178] Formulations of a compound of Formula I suitable for oral administration may be prepared as discrete units such as pills, capsules, cachets or tablets each containing a predetermined amount of a compound of Formula I.
[00179] Compressed tablets may be prepared by compressing in a suitable machine the active ingredient in a free-flowing form such as a powder or granules, optionally mixed with a binder, lubricant, inert diluent, preservative, surface active or dispersing agent. Molded tablets may be made by molding in a suitable machine a mixture of the powdered active ingredient moistened with an inert liquid diluent. The tablets may optionally be coated or scored and optionally are formulated so as to provide slow or controlled release of the active ingredient therefrom.
[00180] Tablets, troches, lozenges, aqueous or oil suspensions, dispersible powders or granules, emulsions, hard or soft capsules, e.g., gelatin capsules, syrups or elixirs may be prepared for oral use. Formulations of compounds of Formula I intended for oral use may be prepared according to any method known to the art for the manufacture of pharmaceutical compositions and such compositions may contain one or more agents including sweetening agents, flavoring agents, coloring agents and preserving agents, in order to provide a palatable preparation. Tablets containing the active ingredient in admixture with non-toxic pharmaceutically acceptable excipient which are suitable for manufacture of tablets are acceptable. These excipients may be, for example, inert diluents, such as calcium or sodium carbonate, lactose, calcium or sodium phosphate; granulating and disintegrating agents, such as maize starch, or alginic acid; binding agents, such as starch, gelatin or acacia; and lubricating 111-03-PCT - P2338R1 - 02120.004WO1
57 agents, such as magnesium stearate, stearic acid or talc. Tablets may be uncoated or may be coated by known techniques including microencapsulation to delay disintegration and adsorption in the gastrointestinal tract and thereby provide a sustained action over a longer period. For example, a time delay material such as glyceryl monostearate or glyceryl distearate alone or with a wax may be employed.
[00181] For treatment of the eye or other external tissues, e.g., mouth and skin, the formulations are preferably applied as a topical ointment or cream containing the active ingredient(s) in an amount of, for example, 0.075 to 20% w/w. When formulated in an ointment, the active ingredients may be employed with either a paraffinic or a water-miscible ointment base. Alternatively, the active ingredients may be formulated in a cream with an oil-in-water cream base.
[00182] If desired, the aqueous phase of the cream base may include a polyhydric alcohol, i.e., an alcohol having two or more hydroxyl groups such as propylene glycol, butane 1,3-diol, mannitol, sorbitol, glycerol and polyethylene glycol (including PEG 400) and mixtures thereof. The topical formulations may desirably include a compound which enhances absorption or penetration of the active ingredient through the skin or other affected areas. Examples of such dermal penetration enhancers include dimethyl sulfoxide and related analogs. [00183] The oily phase of the emulsions of this invention may be constituted from known ingredients in a known manner. While the phase may comprise merely an emulsifier, it desirably comprises a mixture of at least one emulsifier with a fat or an oil or with both a fat and an oil. Preferably, a hydrophilic emulsifier is included together with a lipophilic emulsifier which acts as a stabilizer. It is also preferred to include both an oil and a fat. Together, the emulsifier(s) with or without stabilizer(s) make up the so-called emulsifying wax, and the wax together with the oil and fat make up the so-called emulsifying ointment base which forms the oily dispersed phase of the cream formulations. Emulsifiers and emulsion stabilizers suitable for use in the formulation of the invention include TWEEN® 60, Span® 80, cetostearyl alcohol, benzyl alcohol, myristyl alcohol, glyceryl mono-stearate and sodium lauryl sulfate.
[00184] Aqueous suspensions of Formula I compounds contain the active materials in admixture with excipients suitable for the manufacture of aqueous suspensions. Such excipients include a suspending agent, such as sodium carboxymethylcellulose, croscarmellose, povidone, methylcellulose, hydroxypropyl methylcellulose, sodium alginate, polyvinylpyrrolidone, gum tragacanth and gum acacia, and dispersing or wetting agents such as a naturally occurring phosphatide (e.g., lecithin), a condensation product of an alkylene oxide with a fatty acid (e.g., polyoxyethylene stearate), a condensation product of ethylene oxide with a long chain aliphatic 111-03-PCT - P2338R1 - 02120.004WO1
58 alcohol (e.g., heptadecaethyleneoxycetanol), a condensation product of ethylene oxide with a partial ester derived from a fatty acid and a hexitol anhydride (e.g., polyoxyethylene sorbitan monooleate). The aqueous suspension may also contain one or more preservatives such as ethyl or n-propyl p-hydroxybenzoate, one or more coloring agents, one or more flavoring agents and one or more sweetening agents, such as sucrose or saccharin.
[00185] The pharmaceutical compositions of compounds of Formula I may be in the form of a sterile injectable preparation, such as a sterile injectable aqueous or oleaginous suspension. This suspension may be formulated according to the known art using those suitable dispersing or wetting agents and suspending agents which have been mentioned above. The sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally acceptable diluent or solvent, such as a solution in 1,3-butanediol or prepared as a lyophilized powder. Among the acceptable vehicles and solvents that may be employed are water, Ringer's solution and isotonic sodium chloride solution. In addition, sterile fixed oils may conventionally be employed as a solvent or suspending medium. For this purpose any bland fixed oil may be employed including synthetic mono- or diglycerides. In addition, fatty acids such as oleic acid may likewise be used in the preparation of injectables.
[00186] The amount of active ingredient that may be combined with the carrier material to produce a single dosage form will vary depending upon the host treated and the particular mode of administration. For example, a time-release formulation intended for oral administration to humans may contain approximately 1 to 1000 mg of active material compounded with an appropriate and convenient amount of carrier material which may vary from about 5 to about 95% of the total compositions (weight:weight). The pharmaceutical composition can be prepared to provide easily measurable amounts for administration. For example, an aqueous solution intended for intravenous infusion may contain from about 3 to 500 μg of the active ingredient per milliliter of solution in order that infusion of a suitable volume at a rate of about 30 mL/hr can occur.
[00187] Formulations suitable for parenteral administration include aqueous and nonaqueous sterile injection solutions which may contain anti-oxidants, buffers, bacteriostats and solutes which render the formulation isotonic with the blood of the intended recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents.
[00188] Formulations suitable for topical administration to the eye also include eye drops wherein the active ingredient is dissolved or suspended in a suitable carrier, especially an aqueous solvent for the active ingredient. The active ingredient is preferably present in such formulations 111-03-PCT - P2338R1 - 02120.004WO1
59 in a concentration of about 0.5 to 20% w/w, for example about 0.5 to 10% w/w, for example about 1.5% w/w.
[00189] Formulations suitable for topical administration in the mouth include lozenges comprising the active ingredient in a flavored basis, usually sucrose and acacia or tragacanth; pastilles comprising the active ingredient in an inert basis such as gelatin and glycerin, or sucrose and acacia; and mouthwashes comprising the active ingredient in a suitable liquid carrier. [00190] Formulations for rectal administration may be presented as a suppository with a suitable base comprising for example cocoa butter or a salicylate.
[00191] Formulations suitable for intrapulmonary or nasal administration have a particle size for example in the range of 0.1 to 500 microns (including particle sizes in a range between 0.1 and 500 microns in increments microns such as 0.5, 1, 30 microns, 35 microns, etc.), which is administered by rapid inhalation through the nasal passage or by inhalation through the mouth so as to reach the alveolar sacs. Suitable formulations include aqueous or oily solutions of the active ingredient. Formulations suitable for aerosol or dry powder administration may be prepared according to conventional methods and may be delivered with other therapeutic agents such as compounds heretofore used in the treatment or prophylaxis disorders as described below. [00192] Formulations suitable for vaginal administration may be presented as pessaries, tampons, creams, gels, pastes, foams or spray formulations containing in addition to the active ingredient such carriers as are known in the art to be appropriate.
[00193] The formulations may be packaged in unit-dose or multi-dose containers, for example sealed ampoules and vials, and may be stored in a fϊeeze-dried (lyophilized) condition requiring only the addition of the sterile liquid carrier, for example water, for injection immediately prior to use. Extemporaneous injection solutions and suspensions are prepared from sterile powders, granules and tablets of the kind previously described. Preferred unit dosage formulations are those containing a daily dose or unit daily sub-dose, as herein above recited, or an appropriate fraction thereof, of the active ingredient.
[00194] The invention further provides veterinary compositions comprising at least one active ingredient as above defined together with a veterinary carrier therefore. Veterinary carriers are materials useful for the purpose of administering the composition and may be solid, liquid or gaseous materials which are otherwise inert or acceptable in the veterinary art and are compatible with the active ingredient. These veterinary compositions may be administered parenterally, orally or by any other desired route. COMBINATION THERAPY 111-03-PCT- P2338R1 - 02120.Q04WO1
60
[00195] The compounds of Formula I may be employed alone or in combination with other therapeutic agents for the treatment of a disease or disorder described herein, such as a hyperproliferative disorder (e.g., cancer). In certain embodiments, a compound of Formula I is combined in a pharmaceutical combination formulation, or dosing regimen as combination therapy, with a second compound that has anti-hyperproliferative properties or that is useful for treating a hyperproliferative disorder (e.g., cancer). The second compound of the pharmaceutical combination formulation or dosing regimen preferably has complementary activities to the compound of Formula I such that they do not adversely affect each other. Such compounds are suitably present in combination in amounts that are effective for the purpose intended. In one embodiment, a composition of this invention comprises a compound of Formula I, or a stereoisomer, geometric isomer, tautomer, solvate, metabolite, or pharmaceutically acceptable salt or prodrug thereof, in combination with a chemotherapeutic agent such as described herein. [00196] The combination therapy may be administered as a simultaneous or sequential regimen. When administered sequentially, the combination may be administered in two or more administrations. The combined administration includes coadministration, using separate formulations or a single pharmaceutical formulation, and consecutive administration in either order, wherein preferably there is a time period while both (or all) active agents simultaneously exert their biological activities.
[00197] Suitable dosages for any of the above coadministered agents are those presently used and may be lowered due to the combined action (synergy) of the newly identified agent and other chemotherapeutic agents or treatments.
[00198] The combination therapy may provide "synergy" and prove "synergistic", i.e., the effect achieved when the active ingredients used together is greater than the sum of the effects that results from using the compounds separately. A synergistic effect may be attained when the active ingredients are: (1) co-formulated and administered or delivered simultaneously in a combined, unit dosage formulation; (2) delivered by alternation or in parallel as separate formulations; or (3) by some other regimen. When delivered in alternation therapy, a synergistic effect may be attained when the compounds are administered or delivered sequentially, e.g., by different injections in separate syringes. In general, during alternation therapy, an effective dosage of each active ingredient is administered sequentially, i.e., serially, whereas in combination therapy, effective dosages of two or more active ingredients are administered together.
[00199] In a particular embodiment of anti-cancer therapy, a compound of Formula I, or a stereoisomer, geometric isomer, tautomer, solvate, metabolite, or pharmaceutically acceptable 111-03-PCT - P2338R1 - 02120.004WO1
61 salt or prodrug thereof, may be combined with other chemotherapeutic, hormonal or antibody agents such as those described herein, as well as combined with surgical therapy and radiotherapy. Combination therapies according to the present invention thus comprise the administration of at least one compound of Formula I, or a stereoisomer, geometric isomer, tautomer, solvate, metabolite, or pharmaceutically acceptable salt or prodrug thereof, and the use of at least one other cancer treatment method. The amounts of the compound(s) of Formula I and the other pharmaceutically active chemotherapeutic agent(s) and the relative timings of administration will be selected in order to achieve the desired combined therapeutic effect. METABOLITES OF COMPOUNDS OF FORMULA I
[00200] Also falling within the scope of this invention are the in vivo metabolic products of quinoline compounds of Formula I described herein. Such products may result for example from the oxidation, reduction, hydrolysis, amidation, deamidation, esterification, deesterification, enzymatic cleavage, and the like, of the administered compound. Accordingly, the invention includes metabolites of compounds of Formula I, including compounds produced by a process comprising contacting a compound of this invention with a mammal for a period of time sufficient to yield a metabolic product thereof.
[00201] Metabolite products typically are identified by preparing a radiolabeled (e.g., 14C or 3H) isotope of a compound of the invention, administering it parenterally in a detectable dose (e.g., greater than about 0.5 mg/kg) to an animal such as rat, mouse, guinea pig, monkey, or to man, allowing sufficient time for metabolism to occur (typically about 30 seconds to 30 hours) and isolating its conversion products from the urine, blood or other biological samples. These products are easily isolated since they are labeled (others are isolated by the use of antibodies capable of binding epitopes surviving in the metabolite). The metabolite structures are determined in conventional fashion, e.g., by MS, LC/MS or NMR analysis. In general, analysis of metabolites is done in the same way as conventional drug metabolism studies well known to those skilled in the art. The metabolite products, so long as they are not otherwise found in vivo, are useful in diagnostic assays for therapeutic dosing of the compounds of the invention. ARTICLES OF MANUFACTURE
[00202] In another embodiment of the invention, an article of manufacture, or "kit", containing materials useful for the treatment of the diseases and disorders described above is provided. In one embodiment, the kit comprises a container comprising a quinoline compound of Formula I, or a stereoisomer, geometric isomer, tautomer, solvate, metabolite, or pharmaceutically acceptable salt or prodrug thereof. The kit may further comprise a label or package insert on or associated with the container. The term "package insert" is used to refer to 111-03-PCT- P2338R1 - 02120.004WO1
62 instructions customarily included in commercial packages of therapeutic products, that contain information about the indications, usage, dosage, administration, contraindications and/or warnings concerning the use of such therapeutic products. Suitable containers include, for example, bottles, vials, syringes, blister pack, etc. The container may be formed from a variety of materials such as glass or plastic. The container may hold a compound of Formula I or a formulation thereof which is effective for treating the condition and may have a sterile access port (for example, the container may be an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle). At least one active agent in the composition is a compound of Formula I. The label or package insert indicates that the composition is used for treating the condition of choice, such as cancer. In addition, the label or package insert may indicate that the patient to be treated is one having a disorder such as a hyperproliferative disorder, neurodegeneration, cardiac hypertrophy, pain, migraine or a neurotraumatic disease or event. In one embodiment, the label or package inserts indicates that the composition comprising a compound of Formula I can be used to treat a disorder resulting from abnormal cell growth. The label or package insert may also indicate that the composition can be used to treat other disorders. Alternatively, or additionally, the article of manufacture may further comprise a second container comprising a pharmaceutically acceptable buffer, such as bacteriostatic water for injection (BWFI), phosphate-buffered saline, Ringer's solution and dextrose solution. It may further include other materials desirable from a commercial and user standpoint, including other buffers, diluents, filters, needles, and syringes.
[00203] The kit may further comprise directions for the administration of the compound of
Formula I and, if present, the second pharmaceutical formulation. For example, if the kit comprises a first composition comprising a compound of Formula I and a second pharmaceutical formulation, the kit may further comprise directions for the simultaneous, sequential or separate administration of the first and second pharmaceutical compositions to a patient in need thereof. [00204] In another embodiment, the kits are suitable for the delivery of solid oral forms of a compound of Formula I, such as tablets or capsules. Such a kit preferably includes a number of unit dosages. Such kits can include a card having the dosages oriented in the order of their intended use. An example of such a kit is a "blister pack". Blister packs are well known in the packaging industry and are widely used for packaging pharmaceutical unit dosage forms. If desired, a memory aid can be provided, for example in the form of numbers, letters, or other markings or with a calendar insert, designating the days in the treatment schedule in which the dosages can be administered. 111-03-PCT- P2338R1 - 02120.004WO1
63
[00205] According to one embodiment, a kit may comprise (a) a first container with a compound of Formula I contained therein; and optionally (b) a second container with a second pharmaceutical formulation contained therein, wherein the second pharmaceutical formulation comprises a second compound with anti-hyperproliferative activity. Alternatively, or additionally, the kit may further comprise a third container comprising a pharmaceutically- acceptable buffer, such as bacteriostatic water for injection (BWFI), phosphate-buffered saline, Ringer's solution and dextrose solution. It may further include other materials desirable from a commercial and user standpoint, including other buffers, diluents, filters, needles, and syringes. [00206] In certain other embodiments wherein the kit comprises a composition of Formula
I and a second therapeutic agent, the kit may comprise a container for containing the separate compositions such as a divided bottle or a divided foil packet, however, the separate compositions may also be contained within a single, undivided container. Typically, the kit comprises directions for the administration of the separate components. The kit form is particularly advantageous when the separate components are preferably administered in different dosage forms (e.g., oral and parenteral), are administered at different dosage intervals, or when titration of the individual components of the combination is desired by the prescribing physician.
EXAMPLES
[00207] In order to illustrate the invention, the following examples are included.
However, it is to be understood that these examples do not limit the invention and are only meant to suggest a method of practicing the invention. Persons skilled in the art will recognize that the chemical reactions described may be readily adapted to prepare a number of other c-Met inhibitors of the invention, and alternative methods for preparing the compounds of this invention are deemed to be within the scope of this invention. For example, the synthesis of non- exemplified compounds according to the invention may be successfully performed by modifications apparent to those skilled in the art, e.g., by appropriately protecting interfering groups, by utilizing other suitable reagents known in the art other than those described, and/or by making routine modifications of reaction conditions. Alternatively, other reactions disclosed herein or known in the art will be recognized as having applicability for preparing other compounds of the invention.
[00208] In the examples described below, unless otherwise indicated all temperatures are set forth in degrees Celsius. Reagents were purchased from commercial suppliers such as Aldrich Chemical Company, Lancaster, TCI or Maybridge, and were used without further purification unless otherwise indicated. 111-03-PCT - P2338R1 - 02120.004WO1
64
[00209] The reactions set forth below were done generally under a positive pressure of nitrogen or argon or with a drying tube (unless otherwise stated) in anhydrous solvents, and the reaction flasks were typically fitted with rubber septa for the introduction of substrates and reagents via syringe. Glassware was oven dried and/or heat dried.
[00210] Column chromatography was conducted on a Biotage system (Manufacturer:
Dyax Corporation) having a silica gel column or on a silica SEP PAK® cartridge (Waters). 1H NMR spectra were recorded on a Varian instrument operating at 400 MHz. 1H NMR spectra were obtained as CDCI3, dβ-DMSO, CH3OD or dδ-acetone solutions (reported in ppm), using chloroform as the reference standard (7.25 ppm). When peak multiplicities are reported, the following abbreviations are used: s (singlet), d (doublet), t (triplet), m (multiplet), br (broadened), dd (doublet of doublets), dt (doublet of triplets). Coupling constants, when given, are reported in Hertz (Hz).
[00211] Example 1 Preparation of 3-(3-fluoro-4-(6-methoxy-7-(3-morpholino- propoxy)quinolin-4-yloxy)phenyl)-5-methyl-6-(2-methylbenzyl)pyrimidin-4(3H)-one 101
Figure imgf000065_0001
[00212] Step A: Preparation of 4-chloro-5-methyl-6-(2-methylbenzyl)pyrimidine: 2-
Methylbenzylzinc chloride (25 ml of 0.5 M THF solution, 12 mmol) was added to a solution of 4,6-dichloro-5-methylpyrimidine (2.0 g, 12 mmol) and bis(triphenylphosphine) palladium(II) chloride (0.4 g. 0.6 mmol) in THF (20 mL). The reaction mixture was heated to reflux for 2 hours, cooled to room temperature, and then poured onto water (10 mL). The reaction mixture was extracted with ethyl acetate, and the organic layer was washed with brine, dried over sodium sulfate, filtered, and concentrated. The residue was purified by silica gel flash column chromatography (1:10 Et2O/Hexane) to yield the product (1.0 g, 35%) as a white solid. 1H NMR (CDCl3, 400 MHz) δ 8.75 (s, IH), 7.09-7.22 (m, 4H), 6.84 (d, J = 7.81 Hz, IH), 4.15 (s, IH), 2.38 (s, 3H), 2.32 (s, 3H).
[00213] Step B: Preparation of 4-(benzyloxy)-5-methyl-6-(2-methylbenzyl)pyrimidine:
Potassium hydroxide (0.48 g, 8.6 mmol) was added to a solution of 4-chloro-5-methyl-6-(2- methylbenzyl)pyrimidine (1.0 g, 4.3 mmol), 18-crown-6 (0.11 g, 0.43 mmol) and benzyl alcohol (0.45 ml, 4.3 mmol) in toluene (20 mL). The reaction mixture was heated to reflux for 2 hours, 111-03-PCT- P2338R1 - 02120.004WO1
65 cooled to room temperature, and then poured into water (10 mL). The reaction mixture was extracted with ethyl acetate, and the organic layer was washed with brine, dried over sodium sulfate, filtered, and concentrated. The residue was purified by silica gel flash column chromatography (1:1 Et2O/Hexane) to yield the product (1.5 g, 92%) as a white solid. 1H NMR (CDCl3, 400 MHz) δ 8.60 (s, IH), 7.44-7.48 (m, 2H), 7.30-7.42 (m, 3H), 7.06-7.20 (m, 3H), 6.88 (d, J = 7.42 Hz, IH), 5.45 (s, 2H), 2.33 (s, 3H), 2.15 (s, 3H).
[00214] Step C: Preparation of 5-methyl-6-(2-methylbenzyl)pyrimidin-4-ol: 4-
(benzyloxy)-5-methyl-6-(2-methylbenzyl)pyrimidine (1.5 g, 4.9 mmol) was dissolved in trifluoroacetic acid (10 mL). The reaction mixture was heated at 6O0C for 4 hours, cooled to room temperature, and then solvent was evaporated to yield the product (1.5 g, 98%) as white solid. 1H NMR (DMSOd6, 400 MHz) δ 8.12 (s, IH), 7.30-7.40 (m, IH), 7.10-7.15 (m, IH), 7.00-7.10 (m, IH), 6.90 (d, J = 7.42 Hz, IH), 3.82 (s, 2H), 2.45 (s, 3H), 2.22 (s, 3H). LRMS (ESI pos) m/e 215 (M+l).
[00215] Step D: Preparation of 3-(3-fluoro-4-hydroxyphenyl)-5-methyl-6-(2- methylbenzyl)pyrimidin-4(3H)-one: Copper(I) iodide (90 mg, 0.5 mmol) was added to a solution of 5-methyl-6-(2-methylbenzyl)pyrimidin-4-ol (1.0 g, 5.0 mmol), 4-bromo-2-fluorophenol (0.90 g, 5.0 mmol), N,N'-dimethylethylenediamine (80 mg, 0.90 mmol) and potassium phosphate (2.0 g, 9.0 mmol). The reaction mixture was heated to reflux for 12 hours, cooled to room temperature, and then filtered through a pad of celite. The filtrate was concentrated and the residue was purified by silica gel flash column chromatography (2:1 EtOAc/hexanes) to yield the product (0.6 g, 40%) as a white solid. 1H NMR (CDCl3, 400 MHz) δ 7.98 (s, IH), 7.15-7.24 (m, 3H), 7.03-7.08 (m, 2H), 6.88-6.94 (m, 2H), 3.98 (2, 2H), 2.37 (s, 3H), 2.21 (s, 3H). LRMS (ESI pos) m/e 325 (M+l).
[00216] Step E: Preparation of 3-(3-fluoro-4-(6-methoxy-7-(3-morpholino- propoxy)quinolin-4-yloxy)phenyl)-5-methyl-6-(2-methylbenzyl)pyrimidin-4(3H)-one: DMAP (0.75 mg, 0.0062 mmol) was added to a suspension of 3-(3-fluoro-4-hydroxyphenyl)-5-methyl-6- (2-methylbenzyl)pyrimidin-4(3H)-one (20 mg, 0.062 mmol) and 4-chloro-6-methoxy-7-(3- morpholinopropoxy)quinoline (prepared according to WO 01/55116, Example 2, 21 mg, 0.062 mmol). The reaction mixture was heated at 15O0C for 12 hours, cooled to room temperature and purified directly by silica gel flash column chromatography (1:10 MeOH/EtOAc) to yield 101 (10 mg, 26%) as a light brown solid. 1H NMR (CDCl3, 400 MHz) δ 8.54 (s, IH), 8.05 (s, IH), 7.52 (s, IH), 7.46 (s, IH), 7.34-7.42 (m, 2H), 7.12-7.24 (m, 4H), 7.05-7.12 (m, IH), 6.50-6.56 (m, IH), 4.24-4.32 (m,2H), 4.04 (s, 3H), 3.94-4.02 (m, 2H), 3.68-3.80 (m, 4H), 2.56-2.62 (m,2H), 2.44-2.52 (m, 4H), 2.38 (s, 3H), 2.22 (s, 3H), 2.10-2.18 (m,2H). LRMS (ESI pos) m/e 625 (M+l). 111-03-PCT - P2338R1 - 02120.004WO1
66
[00217] Example 2 Preparation of 6-benzyl-3-(3-fluoro-4-(6-methoxy-7-(3- moφholinopropoxy)quinolin-4-yloxy)phenyl)pyrimidin-4(3H)-one 102
Figure imgf000067_0001
[00218] Step A: Preparation of 4-benzyl-6-chloropyrimidine: Prepared from 4,6- dichloropyrimidine (2.0 g, 13 mmol) and benzyl zinc chloride (0.5 M solution in THF, 27 mL, 13 mmol) according to the procedure described for Example 1, Step A. The crude product was purified by silica gel flash column chromatography (1:10 Et2O/Hexane) to yield the product (1.3 g, 47%) as a yellow liquid. 1H NMR (CDCl3, 400 MHz) δ 8.86 (s, IH), 7.33-7.38 (m, 2H), 7.28- 7.32 (m, IH), 7.24-7.28 (m, 2H), 7.13 (d, J = 0.78 Hz, IH), 4.11 (s, 2H).
[00219] Step B: Preparation of 4-benzyl-6-(benzyloxy)pyrimidine: Prepared from 4- benzyl-6-chloropyrimidine (1.1 g, 5.4 mmol) according to the procedure described for Example 1, Step B. The crude was purified by by silica gel flash column chromatography (1:1 Et2O/Hexane) to yield the product (1.3 g, 88%) as a colorless liquid. 1H NMR (CDCl3, 400 MHz) δ 8.76 (s, IH), 7.36-7.46 (m, 3H), 7.29-7.35 (m, 3H), 7.24-7.27 (m, 4H), 6.52 (s, IH), 5.40 (s, 2H), 4.20 (s, 2H).
[00220] Step C: Preparation of 6-benzylpyrimidin-4-ol: Prepared from 4-benzyl-6-
(benzyloxy)pyrimidine was (1.0 g, 3.6 mmol) according to the procedure described for Example 1, Step C, to yield the product (0.63 g, 94%) as a white solid. 1H NMR (CDCl3, 400 MHz) δ 8.06 (s, IH), 7.30-7.36 (m, 2H), 7.23-7.29 (m, 3H), 6.24 (s, IH), 3.90 (s, 3H).
[00221] Step D: Preparation of 6-benzyl-3-(3-fluoro-4-hydroxyphenyl)pyrimidin-4(3H)- one: Prepared from 6-benzylpyrimidin-4-ol (0.50 g, 2.7 mmol) according to the procedure described for Example 1, Step D. The crude was purified by silica gel flash column chromatography (1:1 EtOAc/Hexane) to yield the product (0.50 g, 63%) as a white solid. 1H NMR (DMSO-d6, 400 MHz) δ 10.30 (s, IH), 8.32 (s, IH), 7.30-7.38 (m, 4H), 7.20-7.30 (m, IH), 7.00-7.10 (m, 2H), 6.32 (s, IH), 5.76 (s, IH), 3.83 (s, 2H). LRMS (ESI pos) m/e 297 (M+ 1). [00222] Step E: Preparation of 6-benzyl-3-(3-fluoro-4-(6-methoxy-7-(3- morpholinopropoxy)quinolin-4-yloxy)phenyl)pyrimidin-4(3H)-one: Prepared from 6-benzyl-3- (3-fiuoro-4-hydroxyphenyl)pyrimidin-4(3H)-one (18 mg, 0.059 mmol) according to the procedure described for Example 1, Step E. The crude product was purified by silica gel flash 111-03-PCT- P2338R1 - 02120.004WO1
67 column chromatography (1 :10 MeOH/EtOAc) to yield 102 (10 mg, 28%) as a white solid. 1H NMR (CDCl3, 400 MHz) δ 8.54 (d, J = 5.01 Hz, IH), 8.14 (s, IH), 7.52 (s, IH), 7.46 (s, IH), 7.34-7.42 (m, 4H), 7.28-7.34 (m, 3H), 7.20-7.24 (m, IH), 6.54 (d, J = 5.47 Hz, IH), 4.26 (m, 2H), 4.04 (s, 3H), 3.90-3.94 (m,2H), 3.70-3.76 (m, 4H), 2.54-2.60 (m, 2H), 2.44-2.52 (m, 4H), 2.10- 2.18 (m, 2H). LRMS (ESI pos) m/e 597 (M+l).
[00223] Example 3 Preparation of 6-benzyl-3-(3-fluoro-4-(6-methoxy-7-(3- morpholinopropoxy)quinolin-4-yloxy)phenyl)-5-methylpyrimidin-4(3H)-one 103
Figure imgf000068_0001
[00224] Step A: Preparation of 4-benzyl-6-chloro-5-methylpyrimidine: Prepared from benzyl zinc bromide (0.5 M solution in THF, 25 mL, 12 mmol) and 4,6-dichloro-5- methylpyrimidine (2.0 g, 12 mmol) according to the procedure described for Example 1, Step A. The crude was purified by silica gel flash column chromatography (1:5 EtOAc/Hexane) to yield the product (0.86 g, 32%) as a colorless oil. 1H NMR (CDCl3, 400 MHz) δ 8.78 (s, IH), 7.28-7.33 (m, 2H), 7.18-7.26 (m, 3H), 4.19 (s, 2H), 2.35 (s, 3H).
[00225] Step B: Preparation of 4-benzyl-6-(benzyloxy)-5-methylpyrimidine: Prepared from 4-benzyl-6-chloro-5-methylpyrimidine (0.8 g, 4.0 mmol) according to the procedure described for Example 1, Step B. The crude product was purified by silica gel flash column chromatography (1:9 Et2O/Hexane) to yield the product (1.0 g, 94%) as a colorless oil. IH NMR (CDCl3, 400 MHz) δ 8.62 (s, IH), 7.42-7.45 (m, 2H), 7.32-7.40 (m, 3H), 7.27-7.30 (m, 2H), 7.18- 7.24 (m, 3H), 5.42 (s, 2H), 4.20(s. 2H), 2.20 (s, 3H).
[00226] Step C: Preparation of 6-benzyl-5-methylpyrimidin-4-ol: Prepared from 4- benzyl-6-(benzyloxy)-5-methylpyrimidine (1.0 g, 3.0 mmol) according to the procedure described for Example 1, Step C, to yield the product (0.50 g, 73%) as a white solid. 1H NMR (DMSO-d6, 400 MHz) δ 8.20 (s, IH), 7.18-7.33 (m, 5H), 3.91 (s, 2H), 1.99 (s, 3H). [00227] Step D: Preparation of 6-benzyl-3-(3-fluoro-4-hydroxyphenyl)-5- methylpyrimidin-4(3H)-one: Prepared from 6-benzyl-5-methylpyrimidin-4-ol (0.16 g, 0.80 mmol) according to the procedure described for Example 1, Step D. The crude product was purified by silica gel flash column chromatography (1 :1 EtOAc/Hexane) to yield the product (0.10 g, 64%) as a white solid. 1H NMR (DMSOd6, 400 MHz) δ 8.21 (s, IH), 7.34-7.38 (m, 111-03-PCT - P2338R1 - 02120.004WO1
68
IH), 7.28-7.32 (m, 4H), 7.20-7.24 (m, IH), 7.00-7.10 (m, 2H), 3.94 (s, 2H), 2.08 (s, 3H). LRMS (ESI pos) m/e 311 (M+ 1).
[00228] Step E: Preparation of 6-benzyl-3-(3-fluoro-4-(6-methoxy-7-(3- morpholinopropoxy)quinolin-4-yloxy)phenyl)-5-methylpyrimidin-4(3H)-one: Prepared from 6- benzyl-3-(3-fluoro-4-hydroxyphenyl)-5-methylpyrimidin-4(3H)-one (18 mg, 0.06 mmol) according to the procedure described for Example 1, Step E. The crude productwas purified by silica gel flash column chromatography (1:10 MeOH/EtOAc) to yield 103 (10 mg, 28%) as a white solid. 1H NMR (CDCl3, 400 MHz) δ 8.52 (s, IH), 8.06 (s, IH), 7.52 (s, IH), 7.46 (s, IH), 7.20-7.40 (m, 8H), 6.52 (s, IH), 4.24-4.32 (m, 2H), 4.04 (s, 3H), 3.98-4.02 (m, 2H), 3.70-3.78 (m, 4H), 2.54-2.62 (m, 2H), 2.42-2.54 (m, 4H), 2.24 (s, 3H), 2.10-2.18 (m, 2H). LRMS (ESI pos) m/e 611 (M+l).
[00229] Example 4 Preparation of (3-benzylpiperidin-l-yl)(3-fluoro-4-(6-methoxy-7-
(3-morpholinopropoxy)quinolin-4-yloxy)phenyl)methanone 104
Figure imgf000069_0001
[00230] Step A: Preparation of (3-benzylpiperidin-l-yl)(3-fluoro-4- methoxyphenyl)methanone: Triethylamine (2 mL, 0.01 mol) was added into a solution of 3- fluoro-4-methoxybenzoyl chloride (500 mg, 2.65 mmol) and 3-benzylpiperidine hydrochloride (561 mg, 2.65 mmol) in CH2Cl2 (20 mL). The reaction mixture was stirred for 30 minutes at room temperature and then poured into water (10 mL). The reaction mixture was extracted with CH2Cl2, and the organic layer was washed with brine, dried over sodium sulfate, filtered, and concentrated to yield the product (0.80 g, 92%) as a white solid. LRMS (ESI pos) m/e 328 (M+l). [00231] Step B: Preparation of (3-benzylpiperidin-l-yl)(3-fluoro-4- hydroxyphenyl)methanone: Boron tribromide (0.5 ml, 6.57 mmol) was added into a solution of (3-benzylpiperidin-l-yl)(3-fluoro-4-methoxyphenyl)methanone (0.86 g, 2.63 mmol) in CH2Cl2 (2 mL) at O0C. The reaction mixture was stirred for 30 minutes at room temperature and then poured into water (10 mL). The reaction mixture was extracted with CH2Cl2, and the organic layer was washed with brine, dried over sodium sulfate, filtered, and concentrated to yield the product (0.74 g, 90% yield) as a white solid. LRMS (ESI pos) m/e 314 (M+l).
[00232] Step C: Preparation of (3-benzylpiperidin-l-yl)(3-fluoro-4-(6-methoxy-7-(3- morpholinopropoxy)quinolin-4-yloxy)phenyl)methanone: Prepared from (3-benzylpiperidin-l- 111-03-PCT- P2338R1 - 02120.004WO1
69 yl)(3-fluoro-4-hydroxyphenyl)methanone (30 mg, 0.10 mmol) according to the procedure described for Example 1, Step E. The crude product was purified by silica gel flash column chromatography (1:10 MeOH/EtOAc) to yield 104 (10 mg, 28%) as a white solid. 1H NMR (CDCl3, 400 MHz) δ 8.50 (s, IH), 7.50 (s, IH), 7.40 (s, IH), 7.00-7.30 (m, 9H), 4.50-4.60 (m, 2H), 4.20-4.30 (m, 4H), 4.00 (s, 3H), 3.70-3.8- (m, 6H), 2.70-2.80 (m, 2H), 2.60-2.70 (m, 4H), 2.40-2.60 (m, 4H), 2.10-2.20 (m, 2H), 1.80-1.90 (m, IH). LRMS (ESI pos) m/e 614 (M+l). [00233] Example 5 Preparation of (3-benzylpiperidm-l-yl)(4-(6,7-dimethoxyquinolin-
4-yloxy)-3-fluorophenyl)methanone 105
Figure imgf000070_0001
[00234] Prepared from 4-chloro-6,7-dimethoxyquinoline (prepared according to Kazuo
Kubo (2005) Journal of Medicinal Chemistry 48:1359-1366, 70 mg, 0.31 mmol) and (3- benzylpiperidin-l-yl)(3-fluoro-4-hydroxyphenyl)methanone (Example 4, Step B, 98 mg, 0.31 mmol) according to the procedure described for Example 1, Step E. The crude product was purified by silica gel flash column chromatography (1:10 MeOH/EtOAc) to yield 105 (40 mg, 26%) as a white solid. 1H NMR (CDCl3, 400 MHz) δ 8.52-8.56 (m, IH), 7.55 (s, IH), 7.45 (s, IH), 7.00-7.30 (m, 8H), 6.20-6.50 (m, IH), 4.40-4.70 (m, IH), 4.06 (s, 6H), 3.50-3.80 (m, IH, 2.40-3.20 (m, 4H), 1.60-2.00 (m, 2H), 1.40-1.60 (m, IH), 1.20-1.40 (m, 2H). LRMS (ESI pos) m/e 501 (M+l).
[00235] Example 6 Preparation of (4-benzylpiperidin-l-yl)(4-(6,7-dimethoxy- quinolin-4-yloxy)-3-fluorophenyl)methanone 106
Figure imgf000070_0002
[00236] Step A: Preparation of (4-benzylpiperidin-l-yl)(3-fluoro-4- methoxyphenyl)methanone: Prepared from 3-fluoro-4-methoxybenzoyl chloride (570 mg, 3.02 mmol) and 4-benzylpiperidine (530 mg, 3.02 mmol) according to the procedure described for Example 4, Step A, to yield the product (920 mg, 93%) as a white solid. 1H NMR (CDCl3, 400 MHz) δ 7.25-7.30 (m, 2H), 7.12-7.23 (m, 5H), 6.93-6.98 (m, IH), 3.91 (s, 3H), 2.60-2.90 (m, 111-03-PCT - P2338R1 - 02120.004WO1
70
IHO, 2.50-2.60 (m, 3H), 1.58-1.82 (m, 4H), 1.10-1.30 (m, 2H), 1.00-1.10 (m, IH). LRMS (ESI pos) m/e 328 (M+l).
[00237] Step B: Preparation of (4-benzylpiperidin-l-yl)(3-fluoro-4- hydroxyphenyl)methanone: Prepared from (4-benzylpiperidin-l-yl)(3-fluoro-4- methoxyphenyl)methanone (130 mg, 0.40 mmol) according to the procedure described for Example 4, Step B, to yield the product (120 mg, 96%) as a white solid. LRMS (ESI pos) m/e 314 (M+1).
[00238] Step C: (4-benzylpiperidin-l-yl)(4-(6,7-dimethoxyquinolin-4-yloxy)-3- fluorophenyl)methanone: Prepared from 4-chloro-6,7-dimethoxyquinoline (for preparation see reference in Example 5) (79 mg, 0.35 mmol) and (4-benzylpiperidin-l-yl)(3-fluoro-4- hydroxyphenyl)methanone (110 g, 0.35 mmol) according to the procedure described for Example 1, Step E. The crude product was purified by silica gel flash column chromatography (1:10 MeOH/EtOAc) to yield 106 (20 mg, 11%) as a white solid. 1H NMR (CDCl3, 400 MHz) δ 8.50- 8.55 (m, IH), 7.55 (s, IH), 7.44 (s, IH), 7.10-7.40 (m, 8H), 6.40-6.50 (m, IH), 4.60-4.80 (m, IH), 4.06 (s, 6H), 3.70-3.82 (m, IH), 2.40-3.20 (m, 4H), 1.60-1.90 (m, 3H), 1.20-1.30 (m, IH). LRMS (ESI pos) m/e 501 (M+l).
[00239] Example 7 Preparation of 3-benzyl-5-(3-fluoro-4-(6-methoxy-7-(3-(piperidin-
1 -yl)propoxy)quinolin-4-yloxy)phenyl)pyrimidin-4(3H)-one 107
Figure imgf000071_0001
[00240] Step A: Preparation of 3-benzyl-5-(4-(benzyloxy)-3-fluorophenyl)pyrimidin-
4(3H)-one: Tetrakis(triphenylphosphine)palladium(0) (0.65 g, 0.57 mmol) was added into a suspension of 3-benzyl-5-bromopyrimidin-4(3H)-one (prepared according to Gurnos Jones Journal of the Chemical Society, Perkin Transactions 1: Organic and Bio-Organic Chemistry (1972-1999) 1983, 11:2645-8, 3.0 g, 11 mmol), 4-benzyloxy-3-fluorobenzeneboronic acid (3.3 g, 14 mmol) and lithium chloride (2.4 g, 57 mmol) in dioxane (100 mL) and 2M aqueous sodium carbonate solution (50 mL). The reaction mixture was heated at 1000C for 2 hours, cooled and poured into water (10 mL). The reaction mixture was extracted with ethyl acetate, and the organic layer was washed with brine, dried over sodium sulfate, filtered, and concentrated. The residue was purified by silica gel flash column chromatography (2:1 EtOAc/Hexane) to yield the 111-03-PCT- P2338R1 - 02120.004WO1
71 product (1.4 g, 32%) as a white solid. 1H NMR (CDCl3, 400 MHκ) δ 8.15 (s, IH), 8.01 (s, IH), 7.53 (dd, J = 12.5, 2.34 Hz, IH), 7.43-7.47 (m, 2H), 7.30-7.42 (m, 9H), 7.00-7.05 (m, IH), 5.18 (s, 2H), 5.17 (s, 2H). LRMS (ESI pos) m/e 387 (M+l).
[00241] Step B: Preparation of 3-benzyl-5-(3-fiuoro-4-hydroxyphenyl)pyrimidin-4(3H)- one: Prepared from 3-benzyl-5-(4-(benzyloxy)-3-fluorophenyl)pyrimidin-4(3H)-one (0.3 g, 0.8 mmol) according to the procedure described for Example 1, Step C, to yield the product (0.2 g, 87%) as a white solid. LRMS (ESI pos) m/e 297 (M+l).
[00242] Step C: Preparation of 3-benzyl-5-(4-(7-(benzyloxy)-6-methoxyquinolin-4- yloxy)-3-fluorophenyi)pyrimidin-4(3H)-one: Prepared from 7-(benzyloxy)-4-chloro-6- methoxyquinoline (prepared according to WO2005030140, Example 32, 200 mg, 0.67 mmol) and 3-benzyl-5-(3-fluoro-4-hydroxyphenyl)pyrimidin-4(3H)-one (198 mg, 0.67 mmol) according to the procedure described for Example 1, Step E. The crude product was purified by silica gel flash column chromatography (1:10 MeOH/EtOAc) to yield the product (100 mg, 37%) as a white solid. LRMS (ESI pos) m/e 560 (M+l).
[00243] Step D: Preparation of 3-benzyl-5-(3-fluoro-4-(7-hydroxy-6-rnethoxyquinolin-4- yloxy)phenyl)pyrimidin-4(3H)-one: Prepared from 3-benzyl-5-(4-(7-(benzyloxy)-6- methoxyquinolin-4-yloxy)-3-fluorophenyl)pyrimidin-4(3H)-one (90 mg, 0.16 mmol) according to the procedure described for Example 1, Step C, to yield the product (50 mg, 66%) as a white solid. LRMS (ESI pos) m/e 470 (M+l).
[00244] Step E: Preparation of 3-benzyl-5-(3-fluoro-4-(6-methoxy-7-(3- morpholinopropoxy)quinolin-4-yloxy)phenyl)pyrimidin-4(3H)-one: Cesium carbonate (6.9 mg, 0.021 mmol) was added to a solution of 3-benzyl-5-(3-fluoro-4-(7-hydroxy-6-methoxyquinolin-4- yloxy)phenyl)pyrimidin-4(3H)-one (10 mg, 0.02 mmol) and 4-(3-chloropropyl)morpholine (3.5 mg, 0.021 mmol). The reaction mixture was heated to 5O0C for 1 hour, and then poured onto water (1 mL). The reaction mixture was extracted with EtOAc, and the organic layer was washed with brine, dried over sodium sulfate, filtered, and concentrated. The residue was purified by silica gel flash column chromatography (1:10 MeOH/EtOAc) to yield 107 (6 mg, 47%) as a yellow solid. 1H NMR (CDCb 400 MHz) δ 8.46 (m, IH), 8.23 (s, IH), 8.13(s, IH), 7.70-7.76 (m, IH), 7.50-7.60 (m, 2H), 7.30-7.48 (m, 2H), 7.20-7.30 (m, 3H), 6.44-6.50 (m, IH), 5.30 (s, 2H), 5.20 (s, 2H), 4.24-4.34 (m, 2H), 4.04 (s, 3H), 3.66-3.80 (m, 4H), 2.44-2.64 (m, 4H), 2.10-2.20 (m, 2H). LRMS (ESI pos) m/e 597 (M+l).
[00245] Example 8 Preparation of 5-(4-(7-(2-(lH-imidazol-l-yl)ethoxy)-6- methoxyquinolin-4-yloxy)-3-fluorophenyl)-3-benzylpyrimidin-4(3H)-one 108 111-03-PCT - P2338R1 - 02120.004WO1
72
Figure imgf000073_0001
[00246] Prepared from 3-benzyl-5-(3-fluoro-4-(7-hydroxy-6-methoxyquinolin-4- yloxy)phenyl)pyrimidin-4(3H)-one (Example 7, Step D, 10 mg, 0.02 mmol) and l-(2- chloroethyl)-lH-imidazole hydrochloride (10 mg, 0.06 mmol) according to the procedure described for Example 7, Step E. The crude product was purified by silica gel flash column chromatography (1:10 MeOH/EtOAc) to yield 108 (5 mg, 52%) as a white solid. LRMS (ESI pos) m/e 564 (M+l).
[00247] Example 9 Preparation of 3-benzyl-5-(3-fluoro-4-(6-methoxy-7-(3-(4- methylpiperazin-l-yl)propoxy)quinolin-4-yloxy)phenyl)pyrimidin-4(3H)-one 109
Figure imgf000073_0002
[00248] Prepared from 3-benzyl-5-(3-fluoro-4-(7-hydroxy-6-methoxyquinolin-4- yloxy)phenyl)pyrimidin-4(3H)-one (Example 7, Step D, 22 mg, 0.05 mmol) and l-(3- chloropropyl)-4-methylpiperazine hydrochloride (45 mg, 0.21 mmol) according to the procedure described for Example 7, Step E. The crude product was purified by silica gel flash column chromatography (1 :10 MeOH/EtOAc) to yield 109 (20 mg, 70%) as a yellow solid. LRMS (ESI pos) m/e 610 (M+l).
[00249] Example 10 Preparation of 3-benzyl-5-(3-fluoro-4-(6-methoxy-7-(3-(4- methylpiperazin- 1 -yl)propoxy)quinolin-4-yloxy)phenyl)pyrimidin-4(3H)-one Hydrogen Chloride 110
Figure imgf000073_0003
111 -03-PCT - P2338Rl - 02120.004WO 1
73
[00250] HCl (2.0 M in ether, 1 mL) was added in a solution of 3-benzyl-5-(3-fluoro-4-(6- methoxy-7-(3-(4-methylpiperazin-l-yl)propoxy)quinolin-4-yloxy)phenyl)pyrimidin-4(3H)-one (12 mg, 0.02 mmol) in ether (1 mL). The reaction mixture was stirred for 20 minutes and the solvent was evaporated to yield 110 (12 mg, 81%) as a white solid. 1H NMR (DMSOd6, 400 MHz) δ 11.50-12.50 (s, br, 4H), 8.80-9.00 (m, 2H), 8.40 (s, IH), 7.90-8.10 (m, IH), 7.60-7.80 (m, 2H), 7.20-7.60 (m, 5H), 6.90-7.10 (m, IH), 5.20-5.25 (m, 2H), 4.20-5.00 (m, 4H), 4.00 (s, 3H), 3.60-4.00 (m, 4H), 3.20-3.60 (m, 6H), 300 (m, 3H). LRMS (ESI pos) m/e 610 (M+l). [00251] Example 11 Preparation of 3-benzyl-5-(3-fluoro-4-(6-methoxy-7-(3-(piperidin-
1 -yl)propoxy)quinolin-4-yloxy)phenyl)pyrimidin-4(3H)-one 111
Figure imgf000074_0001
[00252] Prepared from l-(3-chloropropyl)piperidine hydrochloride (46 mg, 0.05 mmol) and 3-benzyl-5-(3-fluoro-4-(7-hydroxy-6-methoxyquinolin-4-yloxy)phenyl)pyrimidin-4(3H)-one (Example 7, Step D, 22 mg, 0.05 mmol) according to the procedure described for Example 7, Step E. The crude product was purified by silica gel flash column chromatography (1:10 MeOH/EtOAc) to yield 111 (10 mg, 36%) as a yellow solid. 1H NMR (DMSO-d6, 400 MHz) δ 8.80 (s, IH), 8.47-8.52 (m, IH), 8.36 (s, IH), 7.90-7.92 (m, IH), 7.70-7.75 (m, IH), 7.45-7.55 (m, 2H), 7.30-7.42 (m, 6H), 6.48-6.52 (m, IH), 5.22 (s, 2H), 4.15-4.30 (m, 2H), 3.94 (s, 3H), 3.35- 3.40 (m, 2H), 2.62-2.70 (m, 2H), 2.30-2.32 (m, 2H), 1.45-1.60 (m, 4H), 1.30-1.42 (m, 2H). LRMS (ESI pos) m/e 595 (M+l).
[00253] Example 12 Preparation of 3-benzyl-5-(3-fluoro-4-(6-methoxy-7-(3-(piperidin- l-yl)propoxy)quinolin-4-yloxy)phenyl)pyrimidin-4(3H)-one Hydrogen Chloride 112
Figure imgf000074_0002
111-03-PCT - P2338R1 - 02120.004WO1
74
[00254] Prepared from 3-benzyl-5-(3-fluoro-4-(6-methoxy-7-(3-(piperidin-l- yl)propoxy)quinolin-4-yloxy)phenyl)pyrimidin-4(3H)-one (Example 11, 9 mg, 0.02 mmol) according to the procedure described for Example 10, to yield 112 (1.2 mg, 90%) as a white solid. 1H NMR (DMSOd6, 400 MHz) δ 9.20 (s, IH), 8.80 (s, IH), 8.40 (s, IH), 7.90-8.00 (m, IH), 7.75-7.80 (m, IH), 7.65 (s, IH), 7.50-7.60 (m, IH), 7.35-7.40 (m, 5H), 7.30-7.35 (m, 2H), 6.70 (s, IH), 5.40 (s, 2H), 4.50-4.60 (m, 2H), 4.00 (s, 3H), 3.20-3.30 (m, 2H), 2.80-3.00 (m, 2H), 2.20- 2.40 (m, 4H), 1.80-1.90 (m, 2H), 1.60-1.70 (m, 2H), 1.30-1.50 (m, 2H). LRMS (ESI pos) m/e 595 (M+l).
[00255] Example 13 Preparation of 3-(4-chlorobenzyl)-5-(3-fluoro-4-(6-methoxy-7-(3- moφholinopropoxy)quinolin-4-yloxy)phenyl)pyrimidin-4(3H)-one 113
Figure imgf000075_0001
[00256] Step A: Preparation of 5-bromo-3-(4-chlorobenzyl)pyrimidin-4(3H)-one: Sodium hydride (0.34 g, 8.6 mmol) was added into a solution of 5-bromopyrimidin-4(3H)-one (prepared according to Thomas J Kress (1985) J. Org. Chem. 50:3073-6, 1.5 g, 8.57 mmol) in THF (10 niL) and DMF (6 mL). The reaction was stirred for 10 minutes and 4-chlorobenzyl bromide (1.76 g, 8.57 mmol) was added. The reaction was stirred for 30 minutes, poured into water (10 mL), and diluted with ethyl acetate. The reaction mixture was extracted with ethyl acetate, and the organic layer was washed with brine, dried over sodium sulfate, filtered, and concentrated. The residue was purified by silica gel flash column chromatography (2:1 EtOAc/Hexane) to yield the product (0.39 g, 15 %) as a white solid. 1H NMR (CDCl3, 400 MHz) δ 8.20 (s, IH), 8.10 (s, IH), 7.29- 7.38 (m, 4H), 5.10 (s, 2H). LRMS (ESI pos) m/e 300 (M+l).
[00257] Step B: Preparation of 5-(4-(benzyloxy)-3-fluorophenyl)-3-(4- chlorobenzyl)pyrimidin-4(3H)-one: Prepared from 5-bromo-3-(4-chlorobenzyl)pyrimidin-4(3H)- one (0.39 g, 1.3 mmol) according to the procedure described for Example 7, Step A. The crude was purified by silica gel flash column chromatography (1: 1 EtOAc/Hexane) to yield the product (0.17 g, 31%) as a white solid. 1H NMR (CDCl3, 400 MHz) δ 8.15 (s, IH), 8.02 (s, IH), 7.50- 7.54 (m, IH), 7.42-7.46 (m, 2H), 7.35-7.42 (m, 2HO, 7.30-7.35 (m, 5H), 7.00-7.05 (m, IHO, 5.18 (s, 2H), 5.12 (s, 2H)XRMS (ESI pos) m/e 421 (M+l). 111-03-PCT- P2338R1 - 02120.004WO1
75
[00258] Step C: Preparation of 3-(4-chlorobenzyl)-5-(3-fluoro-4- hydroxyphenyl)pyrimidin-4(3H)-one: Prepared from 5-(4-(benzyloxy)-3-fluorophenyl)-3-(4- chlorobenzyl)pyrimidin-4(3H)-one (0.17 g, 0.40 mmol) according to the procedure described for Example 1, Step C, to yield the product (0.1 g, 75%) as a yellow solid. LRMS (ESI pos) m/e 331 (M+l).
[00259] Step D: Preparation of 3-(4-chlorobenzyl)-5-(3-fluoro-4-(6-methoxy-7-(3- morpholinopropoxy)quinolin-4-yloxy)phenyl)pyrimidin-4(3H)-one: Prepared from 3-(4- chlorobenzyl)-5-(3-fluoro-4-hydroxyphenyl)pyrimidin-4(3H)-one (27 mg, 0.08 mmol) according to the procedure described for Example 1, Step E. The crude was purified by silica gel flash column chromatography (1:10 MeOH/EtOAc) to yield 113 (10 mg, 36%) as a yellow solid. 1H NMR (CDCl3, 400 MHz) δ 8.40-8.60 (m, IH), 8.20-8.40 (m, 2H), 7.70-7.80 (m, IH), 7.50-7.60 (m, 2H), 7.40-7.50 (m, 6H), 6.40-6.60 (m, IH), 5.10-5.30 (m, 2H), 4.20-4.40 (m, 2H), 4.00 (s, 3H), 3.60-3.80 (m, 4H), 2.30-2.70 (m, 4H), 2.00-2.30 (m, 2H), 1.50-1.80 (m, 2H). LRMS (ESI pos) m/e 631 (M+l).
[00260] Example 14 Preparation of 6-benzyl-3-(4-(6,7-dimethoxyquinolin-4-yloxy)-3- fluorophenyl)pyridin-2( 1 H)-one 114
Figure imgf000076_0001
[00261] Step A: Preparation of (6-(benzyloxy)pyridin-2-yl)(phenyl)methanol: nBuLi (2.5
M in hexanes, 18.2 ml, 45.4 mmol) was added into a solution of 2-(benzyloxy)-6-bromopyridine (10 g, 37.9 mmol) in THF (200 mL) at -780C for 30 minutes. Benzaldehyde (4.59 ml, 45.4 mmol) was added, the reaction was stirred for 20 minutes at that temperature and poured onto water (10 mL). The reaction mixture was extracted with EtOAc, and the organic layer was washed with brine, dried over sodium sulfate, filtered, and concentrated. The residue was purified by silica gel flash column chromatography (1:2 EtOAc/Hexane) to yield the product (9.9 g, 90%) as a colorless oil. 1H NMR (CDCl3, 400 MHz) δ 7.50-7.55 (m, IH), 7.44-7.47 (m, 2H), 7.35-7.41 (m, 2H), 7.30-7.35 (m, 5H), 6.68-6.72 (m, 2H), 5.66 (d, J = 4.69 Hz, 2H), 5.44 (s, 2H), 4.70 (d, J - 5.08 Hz, IH).
[00262] Step B: Preparation of 6-benzylpyridin-2-ol: Palladium on carbon (10%, 1.5 g, 1.4 mmol) was added into a solution of (6-(benzyloxy)pyridin-2-yl)(phenyl)methanol (4 g, 14 mmol) in MeOH (20 mL). The reaction was pressurized with hydrogen using a balloon, stirred for 2 111-03-PCT- P2338R1 - 02120.004WO1
76 hours and filtered through a pad of celite. The filtrate was concentrated to yield the product (2 g, 79% yield) as a white solid. 1H NMR (DMS0-d6, 400 MHz) δ 7.28-7.35 (m, 5H), 7.18-7.26 (m, IH), 6.10-6.16 (m, IH), 5.92-5.98 (m, IH), 3.80 (s, 2H).
[00263] Step C: Preparation of 6-benzyl-3-bromopyridin-2-ol: Bromine (0.14 mL, 2.7 mmol) was added into a solution of 6-benzylpyridin-2-ol (0.5 g, 2.7 mmol) in CH2Cl2 (5 mL). The reaction was stirred for 20 minutes at room temperature and then poured into 10% aqueous sodium bisulfite solution (10 mL). The reaction mixture was extracted with CH2Cl2 and the organic layer was washed with brine, dried over sodium sulfate, filtered, and concentrated to yield the product (0.59 g, 82%) as a yellow solid. LRMS (ESI pos) m/e 263 (M+l). [00264] Step D: Preparation of 6-benzyl-3-(4-(benzyloxy)-3-fluorophenyl)pyridin-2(lH)- one: Prepared from 6-benzyl-3-bromopyridin-2-ol (0.63 g, 2.39 mmol) according to the procedure described for Example 7, Step A. The crude product was purified by silica gel flash column chromatography (1:1 EtOAc/Hexane) to yield the product (200 mg, 22%) as a white solid. 1H NMR (DMSOd6, 400 MHz) δ 12.00 (s, IH), 7.68-7.74 (m, IH), 7.62 (d, J = 7.42 Hz, IH), 7.42-7.52 (m, 3H), 7.38-7.44 (m, 2H), 7.30-7.38 (m, 5H), 7.20-7.28 (m, 2HO, 6.04-6.10 (m, IH), 5.20 (s, 2H), 3.80 (s, 2H). LRMS (ESI pos) m/e 386 (M+l).
[00265] Step E: Preparation of 6-benzyl-3-(3-fluoro-4-hydroxyphenyl)pyridin-2(lH)-one:
Prepared from 6-benzyl-3-(4-(benzyloxy)-3-fluorophenyl)pyridin-2(lH)-one (150 mg, 0.39 mmol) according to the procedure described for Example 14, Step B. The crude product was purified by silica gel flash column chromatography (1 :1 EtOAc/Hexane) to yield the product (100 mg, 87%) as a white solid. 1H NMR (DMSOd6, 400 MHz) δ 11.90 (s, IH), 10.00 (s, IH), 7.60- 7.66 (m, IH), 7.54-7.58 (m, IH), 7.32-7.40 (m, 5H), 7.22-7.30 (m, IH), 6.92-6.96 (m, IHO, 6.02- 6.08 (m, IH), 3.80 (s, 2H). LRMS (ESI pos) m/e 296 (M+l).
[00266] Step F: Preparation of 6-benzyl-3-(4-(6,7-dimethoxyquinolin-4-yloxy)-3- fluorophenyl)pyridin-2(lH)-one: Prepared from 4-chloro-6,7-dimethoxyquinoline (prepared according to reference in Example 5) (85 mg, 0.38 mmol) and 6-benzyl-3-(3-fluoro-4- hydroxyphenyl)pyridin-2(lH)-one (93 mg, 0.31 mmol) according to the procedure described for Example 1, Step E. The crude product was purified by silica gel flash column chromatography (1:10 MeOH/EtOAc) to yield 114 (50 mg, 33%) as a white solid. 1H NMR (DMSOd6, 400 MHz) δ 10.40 (s, IH), 8.45-8.48 (m, IHO, 7.70-7.76 (m, IH), 7.52-7.80 (m, 2H), 7.40 (s, IH), 7.28-7.36 (m, 2H), 7.20-7.36 (m, 5H), 6.44-6.50 (m, IHO, 6.14-6.20 (m, IHO, 4.03 (s, 3H), 4.02 (s, 3H), 3.91 (s, 2H). ). LRMS (ESI pos) m/e 483 (M+l).
[00267] Example 15 Preparation of 6-benzyl-3-(3-fluoro-4-(6-methoxy-7-(3- morpholinopropoxy)quinolin-4-yloxy)phenyl)pyridin-2( 1 H)-one 115 111-03-PCT - P2338R1 - 02120.004WO1
77
Figure imgf000078_0001
[00268] Prepared from 3-benzyl-5-(3-fluoro-4-hydroxyphenyl)pyrimidin-4(3H)-one
(Example 14, Step E, 30 mg, 0.10 mmol) according to the procedure described for Example 1, Step E. The crude product was purified by silica gel flash column chromatography (1:10 MeOH/EtOAc) to yield 115 (32 mg, 53%) as a white solid. 1H NMR (DMSOd6, 400 MHz) δ 12.00 (s, IH), 8.40-8.45 (m, IH), 8.02-8.08 (m, IH), 7.88-7.96 (m, IH), 7.66-7.76 (m, IH), 7.46- 7.50 (m, IH), 7.26-7.40 (m, 4H), 7.18-7.26 (m, IH), 7.02-7.14 (m, IH), 6.52-6.58 (m, IHO, 6.40- 6.46 (m, IH), 6.04-6.12 (m, IH), 4.10-4.20 (m, 2H), 3.90 (s, 3H0, 3.78-3.84 (m, 2H), 3.50-3.58 (m, 4H), 2.90 (s, 2H), 2.20-2.35 (m, 4H), 1.85-2.00 (m, 2H). LRMS (ESI pos) m/e 596 (M+l). [00269] Example 16 Preparation of 4-benzyl-l-(3-fluoro-4-(6-methoxy-7-(3- morpholinopropoxy)quinolin-4-yloxy)phenyl)pyridin-2(lH)-one 116
Figure imgf000078_0002
[00270] Step A: Preparation of (2-(benzyloxy)-5-bromopyridin-4-yl)(phenyl)methanol:
Prepared from 2-(benzyloxy)-5-bromopyridine (1 g, 3.8 mmol) according to the procedure described for Example 14, Step A. The crude product was purified by silica gel flash column chromatography (1:9 Et2O/Hexane) to yield the product (1.4 g, 29%) as a colorless oil. 1H NMR (CDCl3, 400 MHz) δ 8.19 (s, IH), 7.43-7.47 (m, 2H), 7.30-7.40 (m, 8H), 7.18 (s, IH), 5.99 (d, J = 3.90Hz, IH), 5.31-5.40 (m, 2H).
[00271] Step B: Preparation of 4-benzylpyridin-2(lH)-one: Prepared from (2-(benzyloxy)-
5-bromopyridin-4-yl)(phenyl)methanol (0.40 g, 1.1 mmol) according to the procedure described for Example 14, Step B, to yield the product (0.20 g, 100%) as a white solid. 1H NMR (CDCl3, 400 MHz) δ 7.80 (s, IH), 7.26-7.36 (m, 4H), 7.10-7.20 (m, 2H), 6.80 (s, IHO, 6.60 (s, IH), 3.93 (s, 2H).
[00272] Step C: Preparation of 4-benzyl-l-(3-fluoro-4-hydroxyphenyl)pyridin-2(lH)-one:
Prepared from 4-benzylpyridin-2(lH)-one (0.20 g, 1.1 mmol) according to the procedure 111-03-PCT - P2338R1 - 02120.004WO1
78 described for Example 1, Step D. The crude product was purified by silica gel flash column chromatography (EtOAc) to yield the product (0.29 g, 91%) as a white solid. 1H NMR (DMSO- d6, 400 MHz) δ 10.17 (s, IH), 7.50 (d, J = 7.0 H), 7.28-7.38 (m, 4H), 7.20-7.38 (m, 2H), 6.96- 7.04 (m, 2H), 6.29-6.30 (m, IH), 6.12-6.16 (m, IH), 3.80 (s, 2H). LRMS (ESI pos) m/e 296 (M+l).
[00273] Step D: Preparation of 4-benzyl-l-(3-fluoro-4-(6-methoxy-7-(3- morpholinopropoxy)quinolin-4-yloxy)phenyl)pyridin-2(lH)-one: Prepared from 4-benzyl-l-(3- fluoro-4-hydroxyphenyl)pyridin-2(lH)-one (30 mg, 0.10 mmol) according to the procedure described for Example 1, Step E. The crude product was purified by silica gel flash column chromatography (1:10 MeOH/EtOAc) to yield 116 (41 mg, 68%) as a white solid. 1H NMR (DMSO-d6, 400 MHz) δ 8.45-8.50 (m, IH), 7.62-7.68 (m, IHO, 7.56-7.60 (m, IH), 7.48-7.54 (m, IH), 7.46-7.48 (m, IH), 7.36-7.38 (m,lH), 7.28-7.39 (m, 5H), 7.18-7.24 (m, IH), 6.46-6.50 (m, IH), 6.30 (s, IH), 6.16-6.20 (m, IH), 4.12-4.20 (m, 2H), 3.90(s, IH), 3.80 (s, IH), 3.50-3.58 (M, 4H)), 2.40 (s, 2H), 2.30-2.38 (m, 4H), 1.85-2.00 (m, 2H). LRMS (ESI pos) m/e 596(M+1). [00274] Example 17 Preparation of 4-benzyl-l-(4-(6,7-dimethoxyquinolin-4-yloxy)-3- fluorophenyl)pyridin-2(lH)-one 117
Figure imgf000079_0001
[00275] Prepared from 4-chloro-6,7-dimethoxyquinoline (prepared according to reference procedure in Example 5) (0.13 g, 0.57 mmol) and 4-benzyl-l-(3-fluoro-4-hydroxyphenyl)pyridin- 2(lH)-one (Example 16, Step C, 0.14 g, 0.34 mmol) according to the procedure described for Example 1, Step E. The crude product was purified by silica gel flash column chromatography (1:10 MeOH/EtOAc) to yield 117 (82 mg, 50%) as a white solid. 1H NMR (DMSO-d6, 400 MHz) δ 8.50 (d, IH), 7.67-7.72 (m, IHO, 7.62-7.64 (m, IH), 7.54-7.60 (m, IHO, 7.52 (s, IHO, 7.42 (s, IH), 7.32-7.40 (m, 5H0, 7.24-7.30 (m, IH), 6.52-6.56 (m, IHO, 6.36-6.38 (m, IHO, 6.22- 6.26 (m,lH), 3.96 (s, 3H), 3.95 (s, 3H), 3.84 (s, 2H). LRMS (ESI pos) m/e 483 (M+l). [00276] Example 18 Preparation of 3-benzyl-5-(4-(6,7-dimethoxyquinolin-4-yloxy)-3- fluorophenyl)pyrimidin-4(3H)-one 118 111-03-PCT - P2338R1 - 02120.004WO1
79
Figure imgf000080_0001
[00277] Prepared from 4-chloro-6,7-dimethoxyquinoline (prepared according to reference procedure in Example 5) (91 mg, 0.41 mmol) and 3-benzyl-5-(3-fiuoro-4- hydroxyphenyl)pyrimidin-4(3H)-one (Example 7, Step D, 100 mg, 0.34 mmol) according to the procedure described for Example 1, Step E. The crude product was purified by silica gel flash column chromatography (1:10 MeOH/EtOAc) to yield 118 (50 mg, 61%) as a white solid. 1H NMR (DMSOd6, 400 MHz) δ 8.80 (s, IH), 8.50 (d, J = 5.08 Hz, IH), 8.36 (s, IH), 7.88-7.94 (m, IH), 7.71-7.78 (m, IH), 7.46-7.56 (m, 2H), 7.30-7.42 (m, 5H), 6.48-6.54 (m, IH), 5.22 (s, 2H), 3.95 (s, 6H). LRMS (ESI pos) m/e 484 (M+l).
[00278] Example 19 Preparation of 3-(2-chlorobenzyl)-5-(4-(6,7-dimethoxyquinolin-4- yloxy)-3-fluorophenyl)pyrimidin-4(3H)-one 119
Figure imgf000080_0002
[00279] Step A: Preparation of 5-bromo-3-(2-chlorobenzyl)pyrimidin-4(3H)-one:
Prepared from l-(bromomethyl)-2-chlorobenzene (1.0 g, 5.7 mmol) according to the procedure described for Example 13, Step A. The crude product was purified by silica gel flash column chromatography (1:1 EtOAc/Hexane) to yield the product (0.37 g, 22%) as a white solid. 1H NMR (CDCl3, 400 MHz) δ 8.24 (s, IH), 8.20 (s, IH), 7.50-7.54 (m, IHO, 7.42-7.44 (m, IH), 7.28-7.35 (m, 2H), 5.25 (s, IH). LRMS (ESI pos) m/e 299 (M+l).
[00280] Step B: Preparation ■ of 5-(4-(benzyloxy)-3-fluorophenyl)-3-(2- chlorobenzyl)pyrimidin-4(3H)-one: Prepared from 5-bromo-3-(2-chlorobenzyl)pyrimidin-4(3H)- one (0.37 g, 1.2 mmol) according to the procedure described for Example 7, Step A. The crude product was purified by silica gel flash column chromatography (1:1 EtOAc/Hexane) to yield the product (0.30 g, 58%) as a white solid. 1H NMR (CDCl3, 400 MHz) δ 8.25 (s, IH), 8.02 (s, IH), 7.26-7.55 (m, HH), 7.00-7.05 (m, IH), 5.28 (s, IH), 5.18 (s, IH). LRMS (ESI pos) m/e 421 (M+l). 111-03-PCT - P2338R1 - 02120.004WO1
80
[00281] Step C: Preparation of 3-(2-chlorobenzyl)-5-(3-fluoro-4- hydroxyphenyl)pyrimidin-4(3H)-one: Prepared from 5-(4-(benzyloxy)-3-fluorophenyl)-3-(2- chlorobenzyl)pyrimidin-4(3H)-one (0.4 g, 1.0 mmol) according to the procedure described for Example 1, Step C, to yield the product (0.20 g, 64%) as a white solid. LRMS (ESI pos) m/e 331 (M+l).
[00282] Step D: Preparation of 3-(2-chlorobenzyl)-5-(4-(6,7-dimethoxyquinolin-4-yloxy)-
3-fluorophenyl)pyrimidin-4(3H)-one: Prepared from 4-chloro-6,7-dimethoxyquinoline (prepared according to reference procedure in Example 5) (60 mg, 0.27 mmol) and 3-(2-chlorobenzyl)-5-(3- fluoro-4-hydroxyphenyl)pyrimidin-4(3H)-one (89 mg, 0.27 mmol) according to the procedure described for Example 1, Step E. The crude product was purified by silica gel flash column chromatography (1:10 MeOH/EtOAc) to yield 119 (20 mg, 14%) as a white solid. 1H NMR (DMSO-d6, 400 MHz) δ 8.70 (s, IH), 8.47-8.51 (m, IH), 8.41 (s, IH), 7.90-7.95 (m, IH), 7.73- 7.26 (m, IH), 7.50-7.55 (m, 3H), 7.41-7.44 (m, IH), 7.34-7.40 (m, 2H), 7.14-7.16 (m, IH), 6.49- 6.52 (m, IH), 5.75 (s, 2H), 3.95 (s, 6H). LRMS (ESI pos) m/e 518 (M+l). [00283] Example 20 Preparation of 5-(4-(6,7-dimethoxyquinolin-4-yloxy)-3- fluorophenyl)-3-(2-methylbenzyl)pyrimidin-4(3H)-one 120
Figure imgf000081_0001
[00284] Step A: Preparation of 5-bromo-3-(2-methylbenzyl)pyrimidin-4(3H)-one:
Prepared from 5-bromo-3-(2-methylbenzyl)pyrimidin-4(3H)-one (1.0 g, 5.7 mmol) according to the procedure described for Example 13, Step A. The crude product was purified by silica gel flash column chromatography (1 :1 EtOAc/Hexane) to yield the product (0.28 g, 18%) as a white solid. 1H NMR (CDCl3, 400 MHz) δ 8.21 (s, IH), 7.90 (s, IH), 7.21-7.30 (m, 3H), 7.12-7.15 (m, IH), 5.16 (s, 2H), 2.32 (s, 3H). LRMS (ESI pos) m/e 281 (M+l).
[00285] Step B: Preparation of 5-(4-(benzyloxy)-3-fluorophenyl)-3-(2- methylbenzyl)pyrimidin-4(3H)-one: Prepared from 5-bromo-3-(2-chlorobenzyl)pyrimidin-4(3H)- one (280 mg, 1.0 mmol) according to the procedure described for Example 7, Step A. The crude product was purified by silica gel flash column chromatography (1:1 EtOAc/Hexane) to yield the product (0.31 g, 77%) as a white solid. 1H NMR (CDCl3, 400 MHz) δ 8.04 (s, IH), 7.95 (s, IH), 7.53-7.58 (m, IH), 7.43-7.47 (m, 2H), 7.35-7.44 (m, 2H), 7.20-7.30 (m, 6H), 7.01-7.06 (m, IH), 5.19 (s, 2H), 5.18 (s, 2H), 2.35 (s, 3H). LRMS (ESI pos) m/e 421 (M+l). 111-03-PCT - P2338R1 - 02120.004WO1
81
[00286] Step C: Preparation of 5-(3-fluoro-4-hydroxyphenyl)-3-(2- methylbenzyl)pyrimidin-4(3H)-one: Prepared frcm 5-(4-(benzyloxy)-3-fluorophenyl)-3-(2- methylbenzyl)pyrimidin-4(3H)-one (0.31 g, 0.77 mmol) according to the procedure described for Example 1, Step C, to yield the product (0.20 g, 84%) as a white solid. LRMS (ESI pos) m/e 331 (M+l).
[00287] Step D: Preparation of 5-(4-(6,7-dimethoxyquinolin-4-yloxy)-3-fluorophenyl)-3-
(2-methylbenzyl)pyrimidin-4(3H)-one: Prepared from 4-chloro-6,7-dimethoxyquinoline (prepared according to reference procedure in Example 5) (54 mg, 0.24 mmol) and 5-(3-fluoro-4- hydroxyphenyl)-3-(2-methylbenzyl)pyrimidin-4(3H)-one (75 mg, 0.24 mmol) according to the procedure described for Example 1, Step E. The crude product was purified by silica gel flash column chromatography (1:10 MeOH/EtOAc) to yield 120 (30 mg, 25%) as a white solid. 1H NMR (DMSO-d6, 400 MHz) δ 8.64 (s, IH), 8.50 (d, J - 5.08 Hz, IH), 8.40 (s, IH), 7.90-7.94 (m,, IH), 7.74-7.78 (m, IH), 7.48-7.56 (m, 2H), 7.42 (s, IH), 7.14-7.26 (m, 3H), 6.94-6.98 (m, IH), 6.50-6.54 (m, IHO, 5.20 (s, 2H), 3.96 (s, 3H), 3.95 (s, 3H), 2.40 (s, 3H). LRMS (ESI pos) m/e 498 (M+l).
[00288] Example 21 Preparation of 3-(3-chlorobenzyl)-5-(4-(6,7-dimethoxyquinolin-4- yloxy)-3-fluorophenyl)pyrimidin-4(3H)-one 121
Figure imgf000082_0001
[00289] Step A: Preparation of 5-bromo-3-(3-chlorobenzyl)pyrimidin-4(3H)-one:
Prepared from l-(bromomethyl)-3-chlorobenzene (1.5 g, 8.6 mmol) according to the procedure described for Example 13, Step A. The crude product was purified by silica gel flash column chromatography (1:1 EtOAc/Hexane) to yield the product (0.41 g, 16%) as a white solid. 1H NMR (CDCl3, 400 MHz) δ 8.21 (s, IH), 8.12 (s, IH), 7.28-7.35 (m, 3H), 7.23-7.25 (m, IH), 5.11 (s, 2H). LRMS (ESI pos) m/e 301 (M+l).
[00290] Step B: Preparation of 5-(4-(benzyloxy)-3-fluorophenyl)-3-(3- chlorobenzyl)pyrimidin-4(3H)-one: Prepared from 5-bromo-3-(3-chlorobenzyl)pyrimidin-4(3H)- one (0.41 g, 1.38 mmol) according to the procedure described for Example 7, Step A. The crude product was purified by silica gel flash column chromatography (1:1 EtOAc/Hexane) to yield the product (0.42 g, 72%) as a white solid. 1H NMR (CDCl3, 400 MHz) δ 8.15 (s, IH), 8.04 (s, IH), 111-03-PCT - P2338R1 - 02120.004WO1
82
7.50-7.55 (m, IH), 7.43-7.47 (m, 2H), 7.30-7.42 (m, 7H), 7.28-7.30 (m, IH), 7.00-7.06 (m, IH), 5.18 (s, 2H), 5.12 (s, 2H). LRMS (ESI pos) m/e 421 (M+l).
[00291] Step C: Preparation of 3-(3-chlorobenzyl)-5-(3-fluoro-4- hydroxyphenyl)pyrimidin-4(3H)-one: Prepared from 5-(4-(benzyloxy)-3-fluorophenyl)-3-(3- chlorobenzyl)pyrimidin-4(3H)-one (0.42 g, 1.0 mmol) according to the procedure described for Example 1, Step C, to yield the product (0.20 g, 61%) as a white solid. LRMS (ESI pos) m/e 331 (M+l).
[00292] Step D: Preparation of 3-(3-chlorobenzyl)-5-(4-(6,7-dimethoxyquinolin-4-yloxy)-
3-fluorophenyl)pyrimidin-4(3H)-one: Prepared from 4-chloro-6,7-dimethoxyquinoline (prepared according to reference procedure in Example 5) (64 mg, 0.29 mmol) and 3-(3-chlorobenzyl)-5-(3- fluoro-4-hydroxyphenyl)pyrimidin-4(3H)-one (95 mg, 0.30 mmol) according to the procedure described for Example 1, Step E. The crude product was purified by silica gel flash column chromatography (1:10 MeOH/EtOAc) to yield 121 (20 mg, 13%) as a white solid. 1H NMR (DMSO-d6, 400 MHz) δ 8.80 (s, IH), 8.50 (d, J - 5.08 Hz, IH), 8.36 (s, IH), 7.88-7.94 (m, IH), 7.72-7. ,76 (m, IH), 7.48-7.54 (m, 3H), 7.36-7.44 (m, 4H), 6.50-6.52 (m, IH), 5.20 (s, 3H), 3.96 (s, 3H), 3.95 (s, 3H). LRMS (ESI pos) m/e 518 (M+l).
[00293] Example 22 Preparation of 3-(4-chlorobenzyl)-5-(4-(6,7-dimethoxyquinolin-4- yloxy)-3-fluorophenyl)pyrimidin-4(3H)-one 122
Figure imgf000083_0001
[00294] Prepared from 4-chloro-6,7-dimethoxyquinoline (prepared according to reference procedure in Example 5) (60 mg, 0.27 mmol) and 3-(4-chlorobenzyl)-5-(3-fluoro-4- hydroxyphenyl)pyrimidin-4(3H)-one (Example, Step C, 89 mg, 0.20 mmol) according to the procedure described for Example 1, Step E. The crude product was purified by silica gel flash column chromatography (1:10 MeOH/EtOAc) to yield 122 (37 mg, 36%) as a white solid. 1H NMR (DMSOd6, 400 MHz) d 8.80 (s, IH), 8.50 (d, J = 5.5 Hz, IH), 8.36 (s, IH), 7.88-7.94 (m, IH), 7.70-7.76 (m, IH), 7.48-7.54 (m, 2H), 7.40-7.46 (m, 5H), 6.48-6.52 (m, IH), 5.20 (s, 2H), 3.96 (s, 3H), 3.95 (s, 3H). LRMS (ESI pos) m/e 518 (M+l).
[00295] Example 23 Preparation of 5-(4-(6,7-dimethoxyquinolin-4-yloxy)-3- fluorophenyl)-3-(2-fluorobenzyl)pyrimidin-4(3H)-one 123 111-03-PCT- P2338R1 - 02120.004WO1
83
Figure imgf000084_0001
[00296] Step A: Preparation of 5-bromo-3-(2-fluorobenzyl)pyrimidin-4(3H)-one: Prepared from l-(bromomethyl)-2-fluorobenzene (1.58 g, 8.4 mmol) according to the procedure described for Example 13, Step A. The crude product was purified by silica gel flash column chromatography (1:1 EtOAc/Hexane) to yield the product (0.55 g, 23%) as a white solid. LRMS (ESI pos) m/e 283 (M+l).
[00297] Step B: Preparation of 5-(4-(benzyloxy)-3-fluorophenyl)-3-(3- chlorobenzyl)pyrimidin-4(3H)-one: Prepared from 5-bromo-3-(2-fluorobenzyl)pyrimidin-4(3H)- one (0.55 g, 1.9 mmol) according to the procedure described for Example 7, Step A. The crude product was purified by silica gel flash column chromatography (1:1 EtOAc/Hexane) to yield the product (0.35 g, 44%) as a white solid. LRMS (ESI pos) m/e 405 (M+l).
[00298] Step C: Preparation of 5-(3-fluoro-4-hydroxyphenyl)-3-(2- fluorobenzyl)pyrimidin-4(3H)-one: Prepared from 5-(4-(benzyloxy)-3-fluorophenyl)-3-(3- chlorobenzyl)pyrimidin-4(3H)-one (0.35 g, 8.6 mmol) according to the procedure described for Example 1, Step C, to yield the product (0.20 g, 74%) as a white solid. LRMS (ESI pos) m/e 315 (M+l).
[00299] Step D: Preparation of 5-(4-(6,7-dimethoxyquinolin-4-yloxy)-3-fluorophenyl)-3-
(2-fluorobenzyl)pyrimidin-4(3H)-one: Prepared from 4-chloro-6,7-dimethoxyquinoline (prepared according to reference procedure in Example 5) (100 mg, 0.45 mmol) and 3-(3-chlorobenzyl)-5- (3-fluoro-4-hydroxyphenyl)pyrimidin-4(3H)-one (92 mg, 0.29 mmol) according to the procedure described for Example 1, Step E. The crude product was purified by silica gel flash column chromatography (1:10 MeOH/EtOAc) to yield 123 (41 mg, 28%) as a white solid. 1H NMR (DMSO-d6, 400 MHz) δ 8.73 (s, IH), 8.50 (d, J= 5.1 Hz., IH), 8.38 (s, IH), 7.88-7.94 (m, IH), 7.70-7.76 (m, IH), 7.46-7.56 (m, 2H), 7.32-7.44 (m, 3H), 7.18-7.30 (m, 2H), 6.48-6.54 (m, IH), 5.26 (s, 2H), 3.95 (s, 3H), 3.94 (s, 3H). LRMS (ESI pos) m/e 502 (M+l).
[00300] Example 24 Preparation of 5-(4-(6,7-dimethoxyquinolin-4-yloxy)-3- fluorophenyl)-3-(4-fluorobenzyl)pyrimidin-4(3H)-one 124 111-03-PCT- P2338R1 - 02120.004WO1
Figure imgf000085_0001
[00301] Step A: Preparation of 5-bromo-3-(4-fluorobenzyl)pyrimidin-4(3H)-one: Prepared from l-(bromomethyl)-4-fluorobenzene (1.58 g, 8.30 mmol) according to the procedure described for Example 13, Step A. The crude product was purified by silica gel flash column chromatography (1:1 EtOAc/Hexane) to yield the product (0.42 g, 17%) as a white solid. LRMS (ESI pos) m/e 283 (M+l).
[00302] Step B: Preparation of 5-(4-(benzyloxy)-3-fiuorophenyl)-3-(4- fluorobenzyl)pyrimidin-4(3H)-one: Prepared from 5-bromo-3-(4-fiuorobenzyl)pyrimidin-4(3H)- one (0.42 g, 1.5 mmol) according to the procedure described for Example 7, Step A. The crude product was purified by silica gel flash column chromatography (1:1 EtOAc/Hexane) to yield the product (0.33 g, 55%) as a white solid. LRMS (ESI pos) m/e 405 (M+l).
[00303] Step C: Preparation of 5-(3-fluoro-4-hydroxyphenyl)-3-(4- fluorobenzyl)pyrimidin-4(3H)-one: Prepared from 5-(4-(benzyloxy)-3-fluorophenyl)-3-(4- fluorobenzyl)pyrimidin-4(3H)-one (0.33 g, 0.82 mmol) according to the procedure described for Example 1, Step C, to yield the product (0.20 g, 78%) as a white solid. LRMS (ESI pos) m/e 315 (M+l).
[00304] Step D: Preparation of 5-(4-(6,7-dimethoxyquinolin-4-yloxy)-3-fluorophenyl)-3-
(4-fluorobenzyl)pyrimidin-4(3H)-one: Prepared from 4-chloro-6,7-dimethoxyquinoline (prepared according to reference procedure in Example 5) (105 mg, 0.47 mmol) and 5-(3-fluoro-4- hydroxyphenyl)-3-(4-fluorobenzyl)pyrimidin-4(3H)-one (90 mg, 0.29 mmol) according to the procedure described for Example 1, Step E. The crude product was purified by silica gel flash column chromatography (1:10 MeOH/EtOAc) to yield 124 (35 mg, 24 %) as a white solid. 1H NMR (DMSOd6, 400 MHz) δ 8.80 (s, IH), 8.50 (d, J= 5.47 Hz, IH), 8.35 (s, IH), 7.88-7.94 (m, IH), 7.71-7.76 (m, IH), 7.46-7.54 (m, 4H), 7.42 (s, IH), 7.18-7.24 (m, 2H), 6.50 (d, J = 4.3 Hz, IH), 5.20 (s, 2H), 3.96 (s, 3H), 3.95 (s, 3H). LRMS (ESI pos) m/e 502 (M+l). [00305] Example 25 Preparation of 5-(4-(6,7-dimethoxyquinolin-4-yloxy)-3- fluorophenyl)-3-(4-methylbenzyl)pyrimidin-4(3H)-one 125 111-03-PCT - P2338R1 - 02120.004WO1
85
Figure imgf000086_0001
[00306] Step A: Preparation of 5-bromo-3-(4-methylbenzyl)pyrimidin-4(3H)-one:
Prepared from l-(bromomethyl)-4-fluorobenzene (1.55 g, 8.3 mmol) according to the procedure described for Example 13, Step A. The crude product was purified by silica gel flash column chromatography (1:1 EtOAc/Hexane) to yield the product (0.14 g, 6%) as a white solid. LRMS (ESI pos) m/e 281 (M+l).
[00307] Step B: Preparation of 5-(4-(benzyloxy)-3-fluorophenyl)-3-(4- methylbenzyl)pyrimidin-4(3H)-one: Prepared from 5-bromo-3-(4-methylbenzyl)pyrimidin- 4(3H)-one (0.14 g, 0.5 mmol) according to the procedure described for Example 7, Step A. The crude product was purified by silica gel flash column chromatography (1 :1 EtOAc/Hexane) to yield the product (0.15 g, 75%) as a white solid. LRMS (ESI pos) m/e 401 (M+l). [00308] Step C: Preparation of 5-(3-fluoro-4-hydroxyphenyl)-3-(4- methylbenzyl)pyrimidin-4(3H)-one: Prepared from 5-(4-(benzyloxy)-3-fluorophenyl)-3-(4- methylbenzyl)pyrimidin-4(3H)-one (0.15 g, 0.38 mmol) according to the procedure described for Example 1, Step C, to yield the product (0.10 g, 86%) as a white solid. LRMS (ESI pos) m/e 311 (M+l).
[00309] Step D: 5-(4-(6,7-dimethoxyquinolin-4-yloxy)-3-fluorophenyl)-3-(4- methylbenzyl)pyrimidin-4(3H)-one: Prepared from 4-chloro-6,7-dimethoxyquinoline (prepared according to reference procedure in Example 5) (105 mg, 0.50 mmol) and 5-(3-fluoro-4- hydroxyphenyl)-3-(4-methylbenzyl)pyrimidin-4(3H)-one (90 g, 0.29 mmol) according to the procedure described for Example 1, Step E. The crude product was purified by silica gel flash column chromatography (1:10 MeOH/EtOAc) to yield 125 (44 mg, 30 %) as a white solid. 1H NMR (DMSOd6, 400 MHz) δ 8.78 (s, IH), 8.50 (d, J = 5.1 Hz, IH), 8.34 (s, IH), 7.88-7.94 (m, IH), 7.71-7.76 (m, IH), 7.53 (s, IH), 7.48-7.54 (m, IH), 7.42 (s, IH), 7.28-7.34 (m, 2H), 7.16- 7.20 (m, 2H), 6.50-6.52 (m, IH), 5.17 (s, 2H), 3.96 (s, 3H), 3.95 (s, 3H). LRMS (ESI pos) m/e 498 (M+l).
[00310] Example 26 Preparation of 3-(3,4-dichlorobenzyl)-5-(4-(6,7- dimethoxyquinolin-4-yloxy)-3-fluorophenyl)pyrimidin-4(3H)-one 126 111-03-PCT- P2338R1 - 02120.004WO1
86
Figure imgf000087_0001
[00311] Step A: Preparation of 5-bromo-3-(3,4-dichlorobenzyl)pyrimidin-4(3H)-one:
Prepared from 4-(bromom ethyl)- 1,2-dichlorobenzene (2.0 g, 8.4 mmol) according to the procedure described for Example 13, Step A. The crude product was purified by silica gel flash column chromatography (1:1 EtOAc/Hexane) to yield the product (0.63 g, 22%) as a white solid. LRMS (ESI pos) m/e 335 (M+l).
[00312] Step B: Preparation of 5-(4-(benzyloxy)-3-fluorophenyl)-3-(3,4- dichlorobenzyl)pyrimidin-4(3H)-one: Prepared from 5-bromo-3-(3,4-dichlorobenzyl)pyrimidin- 4(3H)-one (0.63 g, 0.20 mmol) according to the procedure described for Example 7, Step A. The crude product was purified by silica gel flash column chromatography (1:1 EtOAc/Hexane) to yield the product (0.11 g, 13%) as a white solid. LRMS (ESI pos) m/e 455 (M+l). [00313] Step C: Preparation of 3-(3,4-dichlorobenzyl)-5-(3-fluoro-4- hydroxyphenyl)pyrimidin-4(3H)-one: Prepared from 5-(4-(benzyloxy)-3-fluorophenyl)-3-(3,4- dichlorobenzyl)pyrimidin-4(3H)-one (0.11 g, 0.24 mmol) according to the procedure described for Example 1, Step C, to yield the product (50 mg, 56%) as a white solid. LRMS (ESI pos) m/e 365 (M+l).
[00314] Step D: 3-(3,4-dichlorobenzyl)-5-(4-(6,7-dimethoxyquinolin-4-yloxy)-3- fluorophenyl)pyrimidin-4(3H)-one: Prepared from 4-chloro-6,7-dimethoxyquinoline (prepared according to reference procedure in Example 5) (60 mg, 0.14 mmol) and 3-(3,4-dichlorobenzyl)- 5-(3-fluoro-4-hydroxyphenyl)pyrimidin-4(3H)-one (50 mg, 0.1 mmol) according to the procedure described for Example 1, Step E. The crude product was purified by silica gel flash column chromatography (1:10 MeOH/EtOAc) to yield 126 (10 mg, 13 %) as a white solid. 1H NMR (DMSOd6, 400 MHz) δ 8.78-8.82 (m, IH), 8.48-8.52 (m, IH), 8.34-8.40 (m, IH), 7.86- 7.94 (m, IH), 7.70-7.76 (m, 2H), 7.62-7.68(m,lH), 7.46-7.56 (m, 2H), 7.38-7.46 (m, 2H), 6.48- 6.54 (m,lH), 5.20 (s, 2H), 3.96 (s, 3H), 3.95 (s, 3H). LRMS (ESI pos) m/e 536 (M+l). [00315] Example 27 Preparation of 5-(4-(6,7-dimethoxyquinolin-4-yloxy)-3- fluorophenyl)-3-(4-(trifluoromethyl)benzyl)pyrimidin-4(3H)-one 127 111-03-PCT- P2338R] - 02120.004WO1
Figure imgf000088_0001
[00316] Step A: Preparation of 5-bromo-3-(4-(trifluoromethyl)benzyl)pyrimidin-4(3H)- one: Prepared from l-(bromomethyl)-4-(trifluoromethyl)benzene (2.0 g, 8.3 mmol) according to the procedure described for Example 13, Step A. The crude product was purified by silica gel flash column chromatography (1:1 EtOAc/Hexane) to yield the product (0.28 g, 9.8%) as a white solid. LRMS (ESI pos) m/e 333 (M+l).
[00317] Step B: Preparation of 5-(4-(benzyloxy)-3-fluorophenyl)-3-(4-
(trifluoromethyl)benzyl)pyrimidin-4(3H)-one: Prepared from 5-bromo-3-(4-
(trifluoromethyl)benzyl)pyrimidin-4(3H)-one (0.28 g, 0.25 mmol) according to the procedure described for Example 7, Step A. The crude product was purified by silica gel flash column chromatography (1 :1 EtOAc/Hexane) to yield the product (0.34 g, 90%) as a white solid. LRMS (ESI pos) m/e 455 (M+l).
[00318] Step C: Preparation of 5-(3-fluoro-4-hydroxyphenyl)-3-(4-
(trifluoromethyl)benzyl)pyrimidin-4(3H)-one: Prepared from in 5-(4-(benzyloxy)-3- fluorophenyl)-3-(4-(trifluoromethyl)benzyl)pyrimidin-4(3H)-one (0.34 g, g, 0.75 mmol) according to the procedure described for Example 1, Step C, to yield the product (0.2 g, 72%) as a white solid. LRMS (ESI pos) m/e 365 (M+l).
[00319] Step D: Preparation of 5-(4-(6,7-dimethoxyquinolin-4-yloxy)-3-fluorophenyl)-3-
(4-(trifluoromethyl)benzyl)pyrimidin-4(3H)-one: Prepared from 4-chloro-6,7- dimethoxyquinoline (prepared according to reference procedure in Example 5) (100 mg, 0.45 mmol) and 5-(3-fluoro-4-hydroxyphenyl)-3-(4-(trifluoromethyl)benzyl)pyrimidin-4(3H)-one (100 mg, 0.22 mmol) according to the procedure described for Example 1, Step E. The crude product was purified by silica gel flash column chromatography (1:10 MeOH/EtOAc) to yield 127 (38 mg, 31 %) as a white solid. 1H NMR (DMSO-d6, 400 MHz) δ 8.82 (s, IH), 8.50 (d, J = 5.5 Hz, IH), 8.40 (s, IH), 7.89-7.94 (m, IH), 7.72-7.78 (m, 3H), 7.59-7.64 (m, 2H), 7.48-7.54 (m, 2H), 7.42 (s, IH), 6.48-6.52 (m, IH), 5.30 (s, 2H), 3.96 (s,. 3H), 3.95 (s, 3H). LRMS (ESI pos) m/e 552 (M+l).
[00320] Example 28 Preparation of 3-(4-chloro-2-fluorobenzyl)-5-(4-(6,7- dimethoxyquinolin-4-yloxy)-3-fluorophenyl)pyrimidin-4(3H)-one 128 111-03-PCT - P2338R1 - 02120 004WO1
88
Figure imgf000089_0001
[00321] Step A: Preparation of 5-bromo-3-(4-chloro-2-fluorobenzyl)pyrimidin-4(3H)-one:
Prepared from l-(bromomethyl)-4-chloro-2-fluorobenzene (1.87 g, 8.3 mmol) according to the procedure described for Example 13, Step A. The crude product was purified by silica gel flash column chromatography (1:1 EtOAc/Hexane) to yield the product (0.81 g, 30%) as a white solid. LRMS (ESI pos) m/e 319 (M+l).
[00322] Step B: Preparation of 5-(4-(benzyloxy)-3-fluorophenyl)-3-(4-chloro-2- fluorobenzyl)pyrimidin-4(3H)-one: Prepared from 5-bromo-3-(4-chloro-2- fluorobenzyl)pyrimidin-4(3H)-one (0.81 g, 2.5 mmol) according to the procedure described for Example 7, Step A. The crude product was purified by silica gel flash column chromatography (EtOAc) to yield the product (0.74 g, 66%) as a white solid. LRMS (ESI pos) m/e 439 (M+l). [00323] Step C: Preparation of 3-(4-chloro-2-fluorobenzyl)-5-(3-fluoro-4- hydroxyphenyl)pyrimidin-4(3H)-one: Prepared from 5-(4-(benzyloxy)-3-fluorophenyl)-3-(4- chloro-2-fluorobenzyl)pyrimidin-4(3H)-one (0.74 g, 1.7 mmol) according to the procedure described for Example 1, Step C, to yield the product (0.5 g, 85%) as a white solid. LRMS (ESI pos) m/e 349 (M+l).
[00324] Step D: Preparation of 3-(4-chloro-2-fluorobenzyl)-5-(4-(6,7-dimethoxyquinolin-
4-yloxy)-3-fluorophenyl)pyrimidin-4(3H)-one: Prepared from 4-chloro-6,7-dimethoxyquinoline (prepared according to reference procedure in Example 5) (50 mg, 0.22 mmol) and in 3-(4- chloro-2-fluorobenzyl)-5-(3-fluoro-4-hydroxyphenyl)pyrimidin-4(3H)-one (78 mg, 0.22 mmol) according to the procedure described for Example 1, Step E. The crude product was purified by silica gel flash column chromatography (1:10 MeOH/EtOAc) to yield 128 (8.3 mg, 7%) as a white solid. 1H NMR (DMSOd6, 400 MHz) δ 8.68 (s, IH), 8.44 (s, IH), 8.33 (s, IH), 7.80-8.00 (m, 2H), 7.60-7.70 (m, IH), 7.40-7.50 (m, 3H), 7.30-7.40(m, 2H), 7.20-7.30 (m, IH), 6.40-6.50 (m, IH), 5.19 (s, 2H), 3.90 (s, 6H). LRMS (ESI pos) m/e 536 (M+l).
[00325] Example 29 Preparation of 3-(2-chloro-4-fluorobenzyl)-5-(4-(6,7- dimethoxyquinolin-4-yloxy)-3-fluorophenyl)pyrimidin-4(3H)-one 129 111-03-PCT - P2338R1 - 02120.004WO1
89
Figure imgf000090_0001
[00326] Step A: Preparation of 5-bromo-3-(2-chloro-4-fluorobenzyl)pyrimidin-4(3H)-one:
Prepared from l-(bromomethyl)-2-chloro-4-fluorobenzene (1.87 g, 8.4 mmol) according to the procedure described for Example 13, Step A. The crude product was purified by silica gel flash column chromatography (1:1 EtOAc/Hexane) to yield the product (0.66 g, 24%) as a white solid. LRMS (ESI pos) m/e 319 (M+l).
[00327] Step B: Preparation of 5-(4-(benzyloxy)-3-fluorophenyl)-3-(2-chloro-4- fiuorobenzyl)pyrimidin-4(3H)-one: Prepared from 5-bromo-3-(2-chloro-4- fluorobenzyl)pyrimidin-4(3H)-one (0.66 g, 2.1 mmol) according to the procedure described for Example 7, Step A. The crude product was purified by silica gel flash column chromatography (1:1 EtOAc/Hexane) to yield the product (0.84 g, 92%) as a white solid. LRMS (ESI pos) m/e 439 (M+l).
[00328] Step C: Preparation of 3-(2-chloro-4-fluorobenzyl)-5-(3-fluoro-4- hydroxyphenyl)pyrimidin-4(3H)-one: Prepared from 5-(4-(benzyloxy)-3-fluorophenyl)-3-(2- chloro-4-fluorobenzyl)pyrimidin-4(3H)-one (0.84 g, 1.5 mmol) according to the procedure described for Example 1, Step C, to yield the product (0.5 g, 75%) as a white solid. LRMS (ESI pos) m/e 349 (M+l).
[00329] Step D: Preparation of 3-(2-chloro-4-fluorobenzyl)-5-(4-(6,7-dimethoxyquinolin-
4-yloxy)-3-fluorophenyl)pyrimidin-4(3H)-one: Prepared from 4-chloro-6,7-dimethoxyquinoline (prepared according to reference procedure in Example 5) (80 mg, 0.36 mmol) and 3-(2-chloro-4- fluorobenzyl)-5-(3-fluoro-4-hydroxyphenyl)pyrimidin-4(3H)-one (125 mg, 0.36 mmol) according to the procedure described for Example 1, Step E. The crude product was purified by silica gel flash column chromatography (1:10 MeOH/EtOAc) to yield 129 (4.2 mg, 2.2%) as a white solid. 1H NMR (DMSO-d6, 400 MHz) δ 8.64 (s, IH), 8.45 (s, IH), 8.35 (s, IH), 7.80-7.90 (m, IH). 7.60-7.70 (m, IH), 7.40-7.60 (m, 3H), 7.30-7.40 (m, IH), 7.10-7.30 (m, 2H), 6.40-6.50 (m, IH), 5.20 (s, 2H), 3.90 (s, 6H). LRMS (ESI pos) m/e 536 (M+l).
[00330] Example 30 Preparation of 3-(4-chloro-2,6-difluorobenzyl)-5-(4-(6,7- dimethoxyquinolin-4-yloxy)-3-fluorophenyl)pyrimidin-4(3H)-one 130 111-03-PCT - P2338R1 - 02120.0Q4WO1
90
Figure imgf000091_0001
[00331] Step A: Preparation of 5-bromo-3-(4-chloro-2,6-difluorobenzyl)pyrimidin-4(3H)- one: Prepared from 2-(bromomethyl)-5-chloro-l,3-difluorobenzene (2.0 g, 8.3 mmol) according to the procedure described for Example 13, Step A. The crude product was purified by silica gel flash column chromatography (1:1 EtOAc/Hexane) to yield the product (0.76 g, 26%) as a white solid. LRMS (ESI pos) m/e 335 (M+l).
[00332] Step B: Preparation of 5-(4-(benzyloxy)-3-fluorophenyl)-3-(4-chloro-2,6- difluorobenzyl)pyrimidin-4(3H)-one: Prepared from 5-bromo-3-(4-chloro-2,6- difluorobenzyl)pyrimidin-4(3H)-one (0.76 g, 2.2 mmol) according to the procedure described for Example 7, Step A. The crude product was purified by silica gel flash column chromatography (1:1 EtOAc/Hexane) to yield the product (0.52 g, 50%) as a white solid. LRMS (ESI pos) m/e 457 (M+l).
[00333] Step C: Preparation of 3-(4-chloro-2,6-difluorobenzyl)-5-(3-fluoro-4- hydroxyphenyl)pyrimidin-4(3H)-one: Prepared from 5-(4-(benzyloxy)-3-fluorophenyl)-3-(4- chloro-2,6-difluorobenzyl)pyrimidin-4(3H)-one (0.52 g, 1.14 mmol) according to the procedure described for Example 1, Step C, to yield the product (0.4 g, 97%) as a white solid. LRMS (ESI pos) m/e 367 (M+l).
[00334] Step D: Preparation of 3-(4-chloro-2,6-difluorobenzyl)-5-(4-(6,7- dimethoxyquinolin-4-yloxy)-3-fluorophenyl)pyrimidin-4(3H)-one: Prepared from 4-chloro-6,7- dimethoxyquinoline (prepared according to reference procedure in Example 5) (100 mg, 0.45 mmol) and 3-(4-chloro-2,6-difluorobenzyl)-5-(3-fluoro-4-hydroxyphenyl)pyrimidin-4(3H)-one (164 mg, 0.45 mmol) according to the procedure described for Example 1, Step E. The crude product was purified by silica gel flash column chromatography (1:10 MeOH/EtOAc) to yield 130 (1.4 mg, 1%) as a white solid. 1H NMR (DMSOd6, 400 MHz) δ 8.76(s, IH), 8.49 (s, IH), 8.35 (s, IH), 7.82-7.90 (m, IH), 7.66-7.74 (m, IH), 7.45-7.55 (m, 2H), 7.35-7.40 (m, 3H), 6.44- 6.52 (m, IH), 5.24 (s, 2H), 3.95 (s, 6H). LRMS (ESI pos) m/e 554 (M+l). [00335] Example 31 Preparation of 5-(4-(6,7-dimethoxyquinolin-4-yloxy)-3- fluorophenyl)-3-(3,4-dimethylbenzyl)pyrimidin-4(3H)-one 131 111-03-PCT - P2338R1 - 02120.004WO1
91
Figure imgf000092_0001
[00336] Step A: Preparation of 5-bromo-3-(3,4-dimethylbenzyl)pyrimidin-4(3H)-one:
Prepared from 4-(chloromethyl)-l,2-dimethylbenzene (1.3 g, 8.4 mmol) according to the procedure described for Example 13, Step A. The crude product was purified by silica gel flash column chromatography (1:1 EtOAc/Hexane) to yield the product (0.68 g, 27%) as a white solid. LRMS (ESI pos) m/e 295 (M+l).
[00337] Step B: Preparation of 5-(4-(benzyloxy)-3-fluorophenyl)-3-(3,4- dimethylbenzyl)pyrimidin-4(3H)-one: Prepared from 5-bromo-3-(3,4-dimethylbenzyl)pyrimidin- 4(3H)-one (0.68 g, 2.3 mmol) according to the procedure described for Example 7, Step A. The crude product was purified by silica gel flash column chromatography (1:1 EtOAc/Hexane) to yield the product (0.17 g, 17%) as a white solid. LRMS (ESI pos) m/e 415 (M+l). [00338] Step C: Preparation of 3-(3,4-dimethylbenzyl)-5-(3-fluoro-4- hydroxyphenyl)pyrimidin-4(3H)-one: Prepared from 5-(4-(benzyloxy)-3-fluorophenyl)-3-(3,4- dimethylbenzyl)pyrimidin-4(3H)-one (0.17 g, 0.4 mmol) according to the procedure described for Example 1, Step C, to yield the product (0.1 g, 77%) as a white solid. LRMS (ESI pos) m/e 325 (M+l).
[00339] Step D: Preparation of 5-(4-(6,7-dimethoxyquinolin-4-yloxy)-3-fIuorophenyl)-3-
(3,4-dimethylbenzyl)pyrimidin-4(3H)-one: Prepared from 4-chloro-6,7-dimethoxyquinoline (prepared according to reference procedure in Example 5) (100 mg, 0.45 mmol) and 3-(3,4- dimethylbenzyl)-5-(3-fluoro-4-hydroxyphenyl)pyrimidin-4(3H)-one (145 mg, 0.45 mmol) according to the procedure described for Example 1, Step E. The crude product was purified by silica gel flash column chromatography (1:10 MeOH/EtOAc) to yield 131 (2 mg, 1%) as a white solid. 1H NMR (DMSO-d6, 400 MHz) δ 8.56 (s, IH), 8.51 (s, IH), 8.40 (s, IH), 7.90-8.00 (m, IH), 7.70-7.80 (m, 2H), 7.48-7.60 (m, 2H), 7.38-7.46 (m, IH), 7.02-7.20 (m, 2H), 6.76-6.84 (m, IH), 6.48-6.56 (m, IH), 5.20 (s, 2H), 3.96 (s, 3H), 3.95 (s, 3H), 2.50 (s, 6H). LRMS (ESI pos) m/e 512 (M+l).
[00340] Example 32 Preparation of 5-(4-(6,7-dimethoxyquinolin-4-yloxy)-3- fluorophenyl)-3-(4-fluoro-3-methylbenzyl)pyrimidin-4(3H)-one 132 111-03-PCT- P2338R1 - 02120.004WO1
92
Figure imgf000093_0001
[00341] Step A: Preparation of 5-bromo-3-(4-fluoro-3-methylbenzyl)pyrimidin-4(3H)- one: Prepared from 4-(bromomethyl)-l-fluoro-2-methylbenzene (0.57 g, 2.8 mmol) according to the procedure described for Example 13, Step A. The crude product was purified by silica gel flash column chromatography (1:1 EtOAc/Hexane) to yield the product (0.46 g, 54%) as a white solid. LRMS (ESI pos) m/e 296 (M+l).
[00342] Step B: Preparation of 5-(4-(benzyloxy)-3-fluorophenyl)-3-(4-fluoro-3- methylbenzyl)pyrimidin-4(3H)-one: Prepared from 5-bromo-3-(4-fluoro-3- methylbenzyl)pyrimidin-4(3H)-one (0.46 g, 1.5 mmol) according to the procedure described for Example 7, Step A. The crude product was purified by silica gel flash column chromatography (1:1 EtOAc/Hexane) to yield the product (86 mg, 13%) as a white solid. LRMS (ESI pos) m/e 419 (M+l).
[00343] Step C: Preparation of 3-(4-fluoro-3-methylbenzyl)-5-(3-fluoro-4- hydroxyphenyl)pyrimidin-4(3H)-one: Prepared 5-(4-(benzyloxy)-3-fluorophenyl)-3-(4-fluoro-3- methylbenzyl)pyrimidin-4(3H)-one (86 mg, 0.2 mmol) according to the procedure described for Example 1, Step C, to yield the product (50 mg, 74%) as a white solid. LRMS (ESI pos) m/e 329 (M+l).
[00344] Step D: Preparation of 5-(4-(6,7-dimethoxyquinolin-4-yloxy)-3-fluorophenyl)-3-
(4-fluoro-3-methylbenzyl)pyrimidin-4(3H)-one: Prepared from 4-chloro-6,7-dimethoxyquinoline (prepared according to reference procedure in Example 5) (40 mg, 0.18 mmol) and 3-(4-fluoro- 3-methylbenzyl)-5-(3-fluoro-4-hydroxyphenyl)pyrimidin-4(3H)-one (59 mg, 0.18 mmol) according to the procedure described for Example 1, Step E. The crude product was purified by silica gel flash column chromatography (1:10 MeOH/EtOAc) to yield 132 (1.6 mg, 2%) as a white solid. 1H NMR (DMSO-d6, 400 MHz) δ 8.79 (s, IH), 8.50 (d, J = 5.5 Hz, IH), 8.35 (s, IH), 7.89-7.95 (m, IH), 7.72-7.75 (m, IH), 7.46-7.60 (m, 2H), 7.45 (s, IH), 7.25-7.40 (m, 2H), 7.10- 7.16 (m, IH), 6.48-6.54 (m, IH), 5.16 (s, 2H), 5.96 (s, 3H), 5.95 (s, 3H), 2.22 (s, 3H). LRMS (ESI pos) m/e 516 (M+l).
[00345] Example 33 Preparation of 4-benzoyl-l-(4-(6,7-dimethoxyquinolin-4-yloxy)-
3-fluorophenyl)pyridin-2(lH)-one l33 111-03-PCT - P2338R1 - 02120.004WO1
93
Figure imgf000094_0001
[00346] Step A: Preparation of (5-bromo-2-methoxypyridin-4-yl)(phenyl)methanol:
Prepared from 5-bromo-2-methoxypyridine (10 g, 51 mmol) according to the procedure described for Example 12, Step A. The crude product was purified by silica gel flash column chromatography (1:1 EtOAc/Hexane) to yield the product (8.9 g, 60%) as a colorless liquid. 1H NMR (CDCl3, 400 MHz) δ 8.18 (s, IH), 7.30-7.40 (m, 5H), 7.12 (s, IH), 5.99 (d, J = 3.9 Hz, IH), 3.93 (s, 3H), 2.34 (d, J = 3.9 Hz, IH).
[00347] Step B: Preparation of (5-bromo-2-methoxypyridin-4-yl)(phenyl)methanone: PCC
(2.93 g, 13.6 mmol) and 4A molecular sieves (2 g) were added into a solution of (5-bromo-2- methoxypyridin-4-yl)(phenyl)methanol (2.0 g, 6.80 mmol) in CH2Cl2 (50 mL). The reaction was stirred for 1 hour, was then filtered with a pad of silica gel. The filtrate was concentrated the filtrate to yield the product (1.9 g, 99%) as a yellow liquid. 1H NMR (CDCl3, 400 MHz) δ 8.34 (s, IH), 7.80-7.84 (m, 2H), 7.61-7.66 (m, IH), 7.46-7.52 (m, 2H). 6.72 (s, IH), 3.97 (s, 3H). [00348] Step C: Preparation of 4-benzoyl-5-bromopyridin-2(lH)-one: (5-bromo-2- methoxypyridin-4-yl)(phenyl)methanone (1.3 g, 4.5 mmol) and pyridine hydrochloride (2 g, 17 mmol) were heated at 150 0C for 1 hour. CH2Cl2 (30 mL) was added into the hot mixture. The mixture was cooled and the solvent was evaporated. The residue was purified by silica gel flash column chromatography (2:1 EtOAc/Hexane) to yield the product (0.3 g, 24%) as a white solid. 1H NMR (DMSOd6, 400 MHz) δ 12.00 (s, IH), 7.91 (s, IH), 7.82-7.86 (m, 2H), 7.72-7.78 (m, IH), 7.56-7.64 (m, 2H), 6.53 (s, IH).
[00349] Step D: Preparation of 4-benzoyl-l-(3-fluoro-4-hydroxyphenyl)pyridin-2(lH)- one: Prepared from 4-benzoyl-5-bromopyridin-2(lH)-one (170 mg, 0.61 mmol) according to the procedure described for Example 1, Step D. The crude product was purified by silica gel flash column chromatography (EtOAc) to yield the product (170 mg, 90%) as a brown solid. 1H NMR (DMSO-d6, 400 MHz) δ 10.28 (s, IH), 7.85-7.90 (m, 2H), 7.79 (d, J = 7.0 Hz, IH), 7.72-7.77 (m, IH), 7.59-7.65 (m, 2H), 7.35-7.40 (m, IH), 7.04-7.14 (m, 2H), 6.60 (d, J = 2.0 Hz, IH), 6.46 (dd, J = 7.0, 2.0 Hz, IH). LRMS (ESI pos) m/e 310 (M+l).
[00350] Step E: Preparation of 4-benzoyl-l-(4-(6,7-dimethoxyquinolin-4-yloxy)-3- fluorophenyl)pyridin-2(lH)-one: Prepared from 4-chloro-6,7-dimethoxyquinoline (prepared 111-03-PCT - P2338R1 - 02120.004WO1
94 according to reference procedure in Example 5) (123 mg, 0.55 mmol) and 4-benzoyl-l-(3-fluoro- 4-hydroxyphenyl)pyridin-2(lH)-one (170 mg, 0.55 mmol) according to the procedure described for Example 1, Step E. The crude product was purified by silica gel flash column chromatography (1:10 MeOH/EtOAc) to yield 133 (151 mg, 55%) as a white solid. 1H NMR (DMSO-d6, 400 MHz) δ 8.55 (d, J = 5.5 Hz, IH), 7.88-7.94 (m, 3H), 7.74-7.84 (m, 2H), 7.60- 7.66 (m, 3H), 7.55 (s, IH), 7.48-7.52 (m, IHO, 7.45 (s, IH), 6.66-6.68 (m, IH), 6.54-6.62 (m, 2H). LRMS (ESI pos) m/e 497 (M+l).
[00351] Example 34 Preparation of N-(4-(7-(3-morpholinopropoxy)-6- methoxyquinolin-4-yloxy)-3-fluorophenyl)-2-(pyridin-2-yl)acetamide 134
Figure imgf000095_0001
[00352] A mixture of 3-fluoro-4-(6-methoxy-7-(3-morpholinopropoxy)quinolin-4- yloxy)aniline (prepared in example 72, steps C-F) (10.0 mg, 0.0234 mmol), 2-(pyridin-2-yl)acetic acid (16.0 mg, 0.117 mmol), N1-((ethylimino)methylene)-N3,N3-dimethylpropane-l,3-diamine hydrochloride (22.4 mg, 0.117 mmol), lH-benzo[d][l,2,3]triazol-l-ol (15.8 mg, 0.117 mmol) and N-ethyl-N-isopropylpropan-2-amine (0.0204 ml, 0.117 mmol) in THF (10 mL) was stirred at room temperature for 2 days. Water (10 mL) was added and the aqueous was extracted with CH2Cl2 (3 x 50 mL). The organic layers were combined and dried over Na2SO4. Concentration and purification by silica gel chromatography afforded 134 (1.3 mg, 10.2%). 1H NMR (400 MHz, CDCl3) δ 10.37 (s, IH, NH), 8.65 (d, J = 4.8 Hz, IH), 8.46 (d, J = 5.2 Hz, IH), 7.72-7.79 (m, 2H), 7.57 (s, IH), 7.43 (s, IH), 7.30-7.34 (m, 3H), 7.21 (t, J = 8.6 Hz, IH), 6.37 (d, J = 5.6 Hz, IH), 4.27 (t, J = 6.8 Hz, 2H), 4.04 (s, 3H), 3.91 (s, 2H), 3.73 (t, J = 4.6 Hz, 4H), 2.58 (t, J = 7.2 Hz, 2H), 2.44-2.53 (m, 4H), 2.10-2.17 (m, 2H). LRMS (APCI neg) m/z 545 (M-I). [00353] Example 35 Preparation of 4-(2-fluoro-4-(6-methoxypyridin-3-yl)phenoxy)-
6,7-dimethoxyquinoline 135
Figure imgf000095_0002
111-03-PCT - P2338R1 - 02120.004WO1
95
[00354] Prepared from 2-methoxy-5-pyridine boronic acid (24 mg, 0.16 mmol) and 4-(4- bromo-2-fluorophenoxy)-6,7-dimethoxyquinoline (Example 34, 60 mg, 0.16 mmol) according to the procedure described for Example 7, Step A. The crude product was purified by silica gel flash column chromatography (EtOAc) to yield 135 (30 mg, 47%) as a white solid. 1H NMR (DMSO-d6, 400 MHz) δ 8.60 (s,l HO, 8.51 (s, IH), 8.10-8.18 (m, Ih), 7.80-7.90 (m, IH), 7.60- 7.70 (m, IH), 7.50-7.60 (m, IH), 7.40 (s, IH), 6.90-7.00 (m, IH), 6.50-6.60 (m,lH), 3.96 (s, 6H), 3.92 (s, 3H). LRMS (ESI pos) m/e 407 (M+l).
[00355] Example 36 Preparation of 4-(3-fluoro-4'-phenoxybiphenyl-4-yloxy)-6,7- dimethoxyquinoline 136
Figure imgf000096_0001
[00356] Prepared from 4-phenoxyphenyl boronic acid (109 mg, 0.51 mmol) and 4-(4- bromo-2-fluorophenoxy)-6,7-dimethoxyquinoline (Example 34, 64 mg, 0.17 mmol) according to the procedure described for Example 7, Step A. The crude product was purified by silica gel flash column chromatography (EtOAc) to yield 136 (40 mg, 51 %) as a white solid. 1H NMR (DMSO-d6, 400 MHz) δ 8.51 (d, J = 5.1 Hz, IH), 7.78-7.86 (m, 3H), 7.62-7.68 (m, IH), 7.50- 7.58 (m, 2H), 7.40-7.46 (m, 3H), 7.16-7.22 (m, IH), 7.06-7.14 (m, 4H), 6.54 (d, J = 5.1 HZ, IH), 3.88 (s, 6H). LRMS (ESI pos) m/e 468 (M+l).
[00357] Example 37 Preparation of N-(4!-(6,7-dimethoxyquinolin-4-yloxy)-3'- fluorobiphenyl-4-yl)methanesulfonamide 137
Figure imgf000096_0002
[00358] Prepared from 4-(methylsulfonylamino)phenyl boronic acid (109 mg, 0.51 mmol) and 4-(4-bromo-2-fluorophenoxy)-6,7-dimethoxyquinoline (Example 34, 64 mg, 0.17 mmol) according to the procedure described for Example 7, Step A. The crude product was purified by silica gel flash column chromatography (EtOAc) to yield 137 (55 mg, 69 %) as a white solid. 1H NMR (DMSO-d6, 400 MHz) δ 9.93 (s, IH), 8.51 (s, IH), 7.56-7.82 (m, 3H), 7.50-7.65 (m, 3H), 111-03-PCT - P2338R1 - 02120.004WO1
96
7.43 (s, IH), 7.24-7.38 (m, 2H), 6.50-6.60 (m, IH), 3.96 (m, 6H), 3.05 (s, 3H).LRMS (ESI pos) m/e 469 (M+l).
[00359] Example 38 Preparation of N-cyclopropyl-4'-(6,7-dimethoxyquinolin-4- yloxy)-3'-fluorobiphenyl-4-carboxamide 138
Figure imgf000097_0001
[00360] Prepared from 4-(cyclopropylcarbamoyl)phenyl boronic acid (81 mg, 0.40 mmol) and 4-(4-bromo-2-fluorophenoxy)-6,7-dimethoxyquinoline (Example 34, 50 mg, 0.13 mmol) according to the procedure described for Example 7, Step A. The crude product was purified by silica gel flash column chromatography (EtOAc) to yield 138 (30 mg, 49 %) as a white solid. 1H NMR (DMSOd6, 400 MHz) δ 8.50-8.54 (m, 2H), 7.92-7.96 (m, 3H), 7.85-7.89 (m, IH), 7.72- 7.76 (m, IH), 7.55-7.60 (m, 2H), 7.43 (s, IH), 6.55-6.58 (m, IH), 2.84-2.92 (m, IH), 0.69-0.74 (m, 2H), 0.58-0.64 (m, 2H). LRMS (ESI pos) m/e 459 (M+l).
[00361] Example 39 Preparation of 4-(3-fluoro-4'-(tetrahydro-2H-pyran-2- yloxy)biphenyl-4-yloxy)-6,7-dimethoxyquinoline 139
Figure imgf000097_0002
[00362] Prepared from 4-(tetrahydro-2H-pyran-2-yloxy)phenyl boronic acid (106 mg,
0.48 mmol) and 4-(4-bromo-2-fluorophenoxy)-6,7-dimethoxyquinoline (Example 34, 60 mg, 0.16 mmol) according to the procedure described for Example 7, Step A. The crude product was purified by silica gel flash column chromatography (EtOAc) to yield 139 (40 mg, 53 %) as a white solid. 1H NMR (DMSO-d6, 400 MHz) δ 8.51 (s, IH), 8.50 (s, IH), 7.78-7.82 (m, IH), 7.68- 7.72 (m, 2H), 7.58-7.62 (m, IH), 7.55 (s, IH), 7.48-7.52 (m, IH), 7.43 (s, IH), 7.10-7.15 (m, 2H), 5.52-5.56 (m, IH), 3.75-3.82 (m, IH), 1.70-1.94 (m, 3H), 1.50-1.70 (m, 3H). LRMS (ESI pos) m/e 476 (M+l).
[00363] Example 40 Preparation of l-(4-(6,7-dimethoxyquinolin-4-yloxy)-3- fluorophenyl)-4-methylpyridin-2( 1 H)-one 140 111-03-PCT - P2338R1 - 02120.004WO1
97
Figure imgf000098_0001
[00364] Step A: Preparation of l-(3-fluoro-4-hydroxyphenyl)-4-methylpyridin-2(lH)-one:
Prepared from 2-hydroxy-4-methylpyridine (2.37 g, 21.7 mmol) according to the procedure described for Example 1, Step D. The crude reaction mixture was filtered with a pad of celite and the filtrate was evaporated to yield the product as a brown solid (1.0 g, 21%). 1H NMR (DMSO- d6, 400 MHz) δ 10.17 (s, IH), 7.48 (d, J = 7.0 Hz, IH), 7.20-7.27 (m, IH), 6.95-7.05 (m, 2H), 6.25 (s, IH), 6.14 (dd, J = 7.0, 1.6 Hz, IH), 2.16 (s, 3H). LRMS (ESI pos) m/e 220 (M+l). [00365] Step B: Preparation of l-(4-(6,7-dimethoxyquinolin-4-yloxy)-3-fluorophenyl)-4- methylpyridin-2(lH)-one: Prepared from l-(3-fluoro-4-hydroxyphenyl)-4-methylpyridin-2(lH)- one (0.1 g, 0.46 mmol) and 4-chloro-6,7-dimethoxyquinoline (prepared according to reference procedure in Example 5) (0.12 g, 0.55 mmol) using the procedure described for Example 1, Step E. The reaction mixture was purified by silica gel flash column chromatography (1:10 MeOH/EtOAc) to yield 140 (50 mg, 27%) as a white solid. 1H NMR (CDCl3, 400 MHz) δ 8.52 (s, IH), 7.70 (dd, IH), 7.62 (d, IH), 7.56-7.60 (m, IH), 7.54 (s, IH), 7.44 (s, IH), 6.24 (dd, IH), 3.96 (s, 3H), 3.95 (s, 3H), 2.20 (s, IH). LRMS (ESI pos) m/e 407 (M+l).
[00366] Example 41 Preparation of l-(4-(6,7-dimethoxyquinolin-4-yloxy)-3- fluorophenyl)-4-(hydroxy(phenyl)methyl)pyridin-2(lH)-one 141
Figure imgf000098_0002
[00367] Sodium borohydride (11 mg, 0.30 mmol) was added into a solution of 4-benzoyl- l-(4-(6,7-dimethoxyquinolin-4-yloxy)-3-fluorophenyl)pyridin-2(lH)-one (Example 33, 30 mg, 0.060 mmol) in MeOH (2 mL) at O0C. The reaction was warmed up to room temperature and was stirred for 20 minutes. The reaction was diluted with ethyl acetate (10 mL), washed with sodium bicarbonate and brine, dried with Na2SO4, filter and concentrated. The residue was purified by silica gel flash column chromatography (1:10 MeOH/EtOAc) to yield 141 (30 mg, 95%) as a yellow solid. 1H NMR (DMSO-d6, 400 MHz) δ 8.53 (d, IH), 7.70 (dd, IH), 7.64 (d, IH), 7.54- 111-03-PCT - P2338R1 - 02120.004WO1
98
7.58 (m, IH), 7.52 (s, IH), 7.44-7.48 (m, IH), 7.43 (s, IH), 7.34-7.42 (m, 3H), 7.25-7.30 (m, IH), 6.60 (s, IH), 6.54 (d, IH), 6.28 (d, IH), 6.12 (s, IH), 5.75 (s, IH), 3.96 (s, 3H), 3.95 (s, 3H). LRMS (ESI pos) m/e 499 (M+l).
[00368] Example 42 Preparation of (l-(4-(6,7-dimethoxyquinolin-4-yloxy)-3- fluorophenyl)-2-oxo- 1 ,2-dihydropyridin-4-yl)(phenyl)methyl acetate 142
Figure imgf000099_0001
[00369] Triethylamine (0.1 mL, 0.7 mmol) and acetyl chloride (2.7 mg, 0.034 mmol) was into a solution of l-(4-(6,7-dimethoxyquinolin-4-yloxy)-3-fluorophenyl)-4- (hydroxy(phenyl)methyl)pyridin-2(lH)-one (Example, 41, 17 mg, 0.034 mmol) in CH2Cl2 (2 mL) and was stirred for 10 minutes. Water (1 mL) and ethyl acetate (2 mL) was added. The reaction mixture was extracted with ethyl acetate, and the organic layer was washed with brine, dried over sodium sulfate, filtered, and concentrated. The residue was purified by silica gel flash column chromatography (1:10 MeOH/EtOAc) to yield 142 (10 mg, 43% yield) as a yellow solid. LRMS (ESI pos) m/e 541 (M+l).
[00370] Example 43 Preparation of 5-(4-(6,7-dimethoxyquinolin-4-yloxy)-3- fluorophenyl)pyrimidin-4(3H)-one 143
Figure imgf000099_0002
[00371] Step A: Preparation of 5-(4-(benzyloxy)-3-fluorophenyl)pyrimidin-4(3H)-one:
Prepared from 5-bromopyrimidin-4(3H)-one (300 mg, 1.71 mmol) and 4-(benzyloxy)-3- fluorophenylboronic acid (844 mg, 3.43 mmol) according to the procedure described for Example 7, Step A. The product was crashed out from the solution while the crude was concentrating. The solid was collected to yield the product (305 mg, 60%) as white solid. LRMS (ESI pos) m/e 297 (M+l).
[00372] Step B: Preparation of 5-(3-fluoro-4-hydroxyphenyl)pyrimidin-4(3H)-one:
Prepared from 5-(4-(benzyloxy)-3-fluorophenyl)pyrimidin-4(3H)-one (300 mg, 1.01 mmol) in 111-03-PCT - P2338R1 - 02120.004WO1
99
MeOH (2 niL) and acetic acid (2 niL) according to the procedure described for Example 14, Step B, to yield the product (140 mg, 67%) as white solid. LRMS (ESI pos) m/e 207 (M+l). [00373] Step C: Preparation of 5-(4-(6,7-dimethoxyquinolin-4-yloxy)-3- fluorophenyl)pyrimidin-4(3H)-one: Prepared from 5-(3-fluoro-4-hydroxyphenyl)pyrimidin- 4(3H)-one (40 mg, 0.19 mmol) and 4-chloro-6,7-dimethoxyquinoline (prepared according to reference procedure in Example 5) (43 mg, 0.19 mmol) according to the procedure described for Example 1, Step E, to yield 143 (1 mg, 1%) as white solid. 1H NMR (DMSOd6, 400 MHz) δ 12.93 (s, IH), 8.84-8.54 (m, IH), 8.34 (s, IH), 8.26 (s, IH), 7.96 (d, IH), 7.76 (d, IH), 7.48-7.58 (m, 2H), 7.42 (s, IH), 6.50-6.54 (m, IH), 3.96 (s, 6H). LRMS (ESI pos) m/e 394 (M+l). [00374] Example 44 Preparation of 4-(4'-((lH-pyrazol-l-yl)methyl)-3-fluorobiphenyl-
4-yloxy)-6,7-dimethoxyquinoline 144
Figure imgf000100_0001
[00375] Prepared from 4-(4-bromo-2-fluorophenoxy)-6,7-dimethoxyquinoline (Example
35, 60 mg, 0.16mmol)and lH-pyrazole-l-benzyl-4-boronic acid (96 mg, 0.48 mmol) according to the procedure described for Example 7, Step A. The crude product was purified by silica gel flash column chromatography (EtOAc) to yield 144 (40 mg, 55 %) as a white solid. 1H NMR (DMSO-d6, 400 MHz) δ 8.46-8.56 (m, IH), 8.40 (s, IH), 8.02 (s, IH), 7.50 (d, IH), 7.50-7.65 (m, 2H), 7.24-7.48 (m, 7H), 6.46-6.52 (m, IH), 5.36 (s, 2H), 3.96 (s, 6H). LRMS (ESI pos) m/e 456 (M+l).
[00376] Example 45 Preparation of 3-(4-(7-(3-morpholinopropoxy)-6- methoxyquinolin-4-yloxy)-3-fluorophenyl)-3,4-dihydroquinazolin-2(lH)-one 145
Figure imgf000100_0002
[00377] Step A: Preparation of 2-amino-N-(3-fluoro-4-methoxyphenyl)benzamide: To a stirred suspension of isatoic anhydride (1.63 g, 10 mmol) in 15 mL dioxane at room temperature 111-03-PCT - P2338R1 - 02120.004WO1
100 under nitrogen was added powdered sodium hydroxide (40 mg, 1 mmol) followed by 3-fluoro-4- methoxyaniline (1.41 g, 10 mmol). The mixture was immersed in a room temperature oil bath and slowly heated to reflux. Carbon dioxide gas evolution was evident. After stirring at reflux for 2 hours, the reaction was cooled to room temperature and inorganics were filtered off with dioxane. The filtrate was concentrated to dryness to a brown solid. The crude product was dissolved in a minimum of hot 95% EtOH and with cooling, crystals formed. The crystals were filtered off and rinsed with a minimum of ice cold 95% EtOH to give a tan solid (1.0 g, 39%). 1H -NMR (400 MHz, CDCl3) δ 7.66 (br s, IH), 7.50 (dd, IH), 7.44 (dd, IH), 7.26 (m, IH), 7.17 (m, IH), 6.95 (m, IH), 6.71 (m, 2H), 5.50 (br s, 2H), 3.89 (s, 3H).
[00378] Step B: Preparation of N-(2-aminobenzyl)-3-fluoro-4-methoxyaniline: To a stirred suspension of lithium aluminum hydride (121 mg, 3.2 mmol) in 2 mL dioxane at reflux under nitrogen was added 2-amino-N-(3-fluoro-4-methoxyphenyl)benzamide (260 mg, 1 mmol) as a solution in 2 mL dioxane. A vigorous reaction was evident. After refluxing overnight the reaction was cooled to room temperature and quenched by sequential treatment with H2O (150 uL), 15% NaOH (150 uL) and H2O (450 uL). After stirring for several minutes, the heterogeneous mixture was filtered through GF/F filter paper with dioxane and concentrated to a brown residue (246 mg, 100%). 1H -NMR (400 MHz, CDCl3) δ 7.14 (m, 2H), 6.86 (m, IH), 6.74 (m, 2H), 6.60 (dd, IH), 6.42 (dd, IH), 4.15 (d, 2H), 4.12 (br s, 2H), 3.83 (s, 3H), 3.54 (br s, IH). [00379] Step C: Preparation of 3-(3-fluoro-4-methoxyphenyl)-3,4-dihydroquinazolin-
2(lH)-one: To a stirred suspension of crude N-(2-aminobenzyl)-3-fluoro-4-methoxyaniline (246 mg, 1 mmol) in 10 mL toluene at 0°C under a drying tube was added phosgene solution (20% in toluene, 683 uL, 1.30 mmol). Immediately a bright orange color appeared. The cooling bath was removed and the reaction allowed to warm to room temperature over 30 minutes. The solution was then warmed to reflux. After 1 hour, the reaction was concentrated to dryness and the residue dissolved in a minimum of hot 95% EtOH. A precipitate formed which was isolated by filtration with 95% EtOH and dried to give a tan solid (65 mg, 24%). 1H -NMR (400 MHz, CDCl3) δ 7.23 (m, IH), 7.14 (m, IH), 7.08 (m, 2H), 7.01 (m, 2H), 6.81 (d, IH), 4.80 (s, 2H), 3.91 (s, 3H).
[00380] Step D: Preparation of 3-(3-fluoro-4-hydroxyphenyl)-3,4-dihydroquinazolin-
2(lH)-one: To a stirred solution of 3-(3-fluoro-4-methoxyphenyl)-3,4-dihydroquinazolin-2(lH)- one (60 mg, 0.22 mmol) in 2.2 mL dichloromethane at 00C under a drying tube was added boron tribromide (104 uL, 1.1 mmol) neat by syringe. The solution turned yellow. After 5 minutes, the reaction was quenched by pouring into saturated NaHCO3 (30 mL) with stirring. 9/1 Dichloromethane/methanol (30 mL) was added and the mixture stirred rapidly. The layers were 111-03-PCT - P2338R1 - 02120.004WO1
101 separated and the organics were dried (MgSO4), filtered, and concentrated to a white solid (40 mg, 70%). LRMS (APCI pos) m/e 259 (M+l). 1H-NMR (400 MHz, CDCl3) 57.40 (m, IH), 7.22 (m, IH), 7.07 (m, 2H), 6.98 (m, 2H), 6.83 (m, IH), 4.78 (s, 2H).
[00381] Step E: Preparation of 3-(3-fluoro-4-(6-methoxy-7-(3- morpholinopropoxy)quinolin-4-yloxy)phenyl)-3,4-dihydroquinazolin-2(lH)-one. Prepared according to the procedure of Example 1, step E substituting 3-(3-fiuoro-4-hydroxyphenyl)-3,4- dihydroquinazolin-2( 1 H)-one for 3 -(3 -fluoro-4-hydroxyphenyl)-5 -methyl-6-(2- methylbenzyl)pyrimidin-4(3H)-one to give 145 after silica gel chromatography as a white foam (23 mg, 53% yield). 1H-NMR (400 MHz, CDCl3) δ 8.51 (d, IH), 7.57 (s, IH), 7.45 (s, IH), 7.39 (m, IH), 7.32-7.24 (m, 5H), 7.14 (d, IH), 7.04 (m, IH), 6.80 (d, IH), 6.49 (d, IH), 4.89 (s, 2H), 4.28 (m, 2H), 4.05 (s, 3H), 3.73 (m, 4H), 2.58 (m, 2H), 2.49 (m, 4H), 2.14 (m, 2H). LRMS (apcipos) m/e 559 (M+l).
[00382] Example 46 Preparation of (4-(4-(6,7-dimethoxyquinolin-4-yloxy)-3- fluorophenyl)piperazin-l-yl)(phenyl)methanone 146
Figure imgf000102_0001
[00383] Step A: Preparation of 4-(2-fluoro-4-(piperazin-l-yl)phenoxy)-6,7- dimethoxyquinoline: Pd2(dba)3 (20 mg) was added into a suspension of 4-(4-bromo-2- fluorophenoxy)-6,7-dimethoxyquinoline (Example 34, 100 mg, 0.264 mmol), piperazine (228 mg, 2.64 mmol), Xanthphos (100 mg) and potassium phosphate (200 mg) in toluene (10 mL). The reaction mixture was heated to 1000C for 10 hours. The reaction mixture was filtered through a pad of silica gel. The filtrate was concentrated and the residue was purified by silica gel flash column chromatography (1:1 MeOH/EtOAc) to yield the product (41 mg, 41% yield) as a brown solid. LRMS (ESI pos) m/e 384 (M+l).
[00384] Step B: Preparation of (4-(4-(6,7-dimethoxyquinolin-4-yloxy)-3- fluorophenyl)piperazin-l-yl)(phenyl)methanone: Benzoyl chloride (11 mg, 0.078 mmol) and triethylamine (0.5 mL) were added into a solution of 4-(2-fluoro-4-(piperazin-l-yl)phenoxy)-6,7- dimethoxyquinoline (30 mg, 0.078 mmol) in CH2Cl2. After a few minutes, water (1 mL) and CH2Cl2 (2 mL) were added. The reaction mixture was extracted with ethyl acetate, and the organic layer was washed with brine, dried over sodium sulfate, filtered, and concentrated. The 111-03-PCT - P2338R1 - 02120.004WO1
102 residue was purified by silica gel flash column chromatography (1:10 MeOH/EtOAc) to yield 146 (5 mg, 13% yield) as a brown solid. 1H NMR LRMS (ESI pos) m/e 488 (M+l). [00385] Example 47 Preparation of 4-(2-fluoro-4-(4-(phenylsulfonyl)piperazin-l- yl)phenoxy)-6,7-dimethoxyquinoline 147
Figure imgf000103_0001
[00386] Prepared from benzenesulfonyl chloride (14 mg, 0.078 mmol) and 4-(2-fluoro-4-
(piperazin-l-yl)phenoxy)-6,7-dimethoxyquinoline (Example 46, step A, 30 mg, 0.078 mmol) according to the procedure described for Example 46, Step B. The crude product was purified by silica gel flash column chromatography (1:10 MeOH/EtOAc) to yield 147 (3.2 mg, 7.8% yield) as white solid. 1H NMR (DMSO-d6, 400 MHz) δ 8.42 (d, IH), 7.74-7.84 (m, 2H), 7.65-7.74 (m, 2H), 7.50 (s, IH), 7.39 (s, IH), 7.25-7.32 (m, IH), 7.00-7.08 (m, IH), 6.80-6.88 (m, IH), 6.35 (d, IH), 5.75 (s, IH), 3.95 (s, 6H), 3.00-3.08 (m, 4H). LRMS (ESI pos) m/e 524 (M+l). [00387] Example 48 Preparation of 3-benzyl-5-(4-(6,7-dimethoxyquinolin-4-yloxy)-3- fluorophenyl)pyrimidine-4(3H)-thione 148
Figure imgf000103_0002
[00388] Step A: Preparation of 3-benzyl-5-(4-(benzyloxy)-3-fluorophenyl)pyrimidine-
4(3H)-thione: Lawesson 's reagent (262 mg, 0.647 mmol) was added into a suspension of 3- benzyl-5-(4-(benzyloxy)-3-fluorophenyl)pyrimidin-4(3H)-one (Example 7, Step A, 100 mg, 0.26 mmol) in toluene. The reaction was heated at 12O0C for 12 hours. LCMS indicated the reaction was complete. The reaction mixture was concentrated and the crude product was purified by silica gel flash column chromatography (EtOAc) to yield the product (70 mg, 67%) as a brown solid. LRMS (ESI pos) m/e 403 (M+l).
[00389] Step B: 3-benzyl-5-(3-fluoro-4-hydroxyphenyl)pyrimidine-4(3H)-thione:
Prepared from 3-benzyl-5-(4-(benzyloxy)-3-fluorophenyl)pyrimidine-4(3H)-thione (70 mg, 0.17 mmol) according to the procedure described for Example 1, Step C to yield the product (50 mg, 92%) as brown solid. LRMS (ESI pos) m/e 313 (M+l). 111-03-PCT - P2338R1 - 02120.004WO1
103
[00390] Step C: Preparation of 3-benzyl-5-(4-(6,7-dimethoxyquinolin-4-yloxy)-3- fluorophenyl)pyrimidine-4(3H)-thione: Prepared from 4-chloro-6,7-dimethoxyquinoline (prepared according to reference procedure in Example 5) (40 mg, 0.18 mmol) and 3-(4- chlorobenzyl)-5-(3-fluoro-4-hydroxyphenyl)pyrimidine-4(3H)-thione (62 mg, 0.18 mmol) according to the procedure described for Example 1, Step E to yield 148 (40 mg, 45%) as yellow solid. 1H NMR (DMSO-d6, 400 MHz) δ 9.08 (s, IH), 8.54 (d, IH), 8.11 (s, IH), 7.70 (d, IH), 7.54 (s, IH), 7.47-7.50 (m, 2H), 7.43 (s, IH), 7.35-7.40 (m, 4H), 7.26-7.33 (m, IH), 6.53 (d, IH), 5.76 (s, 2H), 3.96 (s, 3H), 3.96 (s, 3H). LRMS (ESI pos) m/e 500 (M+l).
[00391] Example 49 Preparation of 3-benzyl-5-(4-(7-(benzyloxy)-6-methoxyquinolin-
4-yloxy)-3-fluorophenyl)pyrimidin-4(3H)-one 149
Figure imgf000104_0001
[00392] Step A: Preparation of 3-benzyl-5-(4-(benzyloxy)-3-fluorophenyl)pyrimidin-
4(3H)-one: Tetrakis(triphenylphosphine)palladium(0) (0.65 g, 0.57 mmol) was added into a suspension of 3-benzyl-5-bromopyrimidin-4(3H)-one (prepared according to Gurnos Jones described in Journal of the Chemical Society, Perkin Transactions 1: Organic and Bio-Organic Chemistry (1972-1999) 1983, 11:2645-8, 3.0 g, 11 mmol), 4-benzyloxy-3-fluorobenzeneboronic acid (3.3 g, 14 mmol) and lithium chloride (2.4 g, 57 mmol) in dioxane (100 mL) and 2M aqueous sodium carbonate solution (50 mL). The reaction mixture was heated at 1000C for 2 hours, cooled and poured into water (10 mL). The reaction mixture was extracted with ethyl acetate, and the organic layer was washed with brine, dried over sodium sulfate, filtered, and concentrated. The crude product was purified by silica gel flash column chromatography (2:1 EtOAc/Hexane) to yield the product (1.4 g, 32%) as a white solid. 1H NMR (CDCl3, 400 MHz) δ 8.15 (s, IH), 8.01 (s, IH), 7.53 (dd, J = 12.5, 2.34 Hz, IH), 7.43-7.47 (m, 2H), 7.30-7.42 (m, 9H), 7.00-7.05 (m, IH), 5.18 (s, 2H), 5.17 (s, 2H). LRMS (ESI pos) m/e 387 (M+l). [00393] Step B: Preparation of 3-benzyl-5-(3-fluoro-4-hydroxyphenyl)pyrimidin-4(3H)- one: Prepared from 3-benzyl-5-(4-(benzyloxy)-3-fluorophenyl)pyrimidin-4(3H)-one (0.3 g, 0.8 mmol) according to the procedure described for Example 1, Step C, to yield the product (0.2 g, 87%) as a white solid. LRMS (ESI pos) m/e 297 (M+l). 111 -03-PCT - P2338R 1 - 02120.004WO1
104
[00394] Step C: Preparation of 3-benzyl-5-(4-(7~(benzyloxy)-6-methoxyquinolin-4- yloxy)-3-fluorophenyl)pyrimidin-4(3H)-one: Prepared from 7-(benzyloxy)-4-chloro-6- methoxyquinoline (prepared according to WO 2005/030140, Example32, 200 mg, 0.67 mmol) and 3-benzyl-5-(3-fluoro-4-hydroxyphenyl)pyrimidin-4(3H)-one (198 mg, 0.48 mmol) according to the procedure described for Example 1, Step E. The crude product was purified by silica gel flash column chromatography (1:10 MeOH/EtOAc) to yield 149 (100 mg, 27%) as a white solid. LRMS (ESI pos) m/e 560 (M+l).
[00395] Example 50 Preparation of 3-benzyl-5-(3-fluoro-4-(7-hydroxy-6- methoxyquinolin-4-yloxy)phenyl)pyrimidin-4(3H)-one 150
Figure imgf000105_0001
[00396] Prepared from 3-benzyl-5-(4-(7-(benzyloxy)-6-methoxyquinolin-4-yloxy)-3- fluorophenyl)pyrimidin-4(3H)-one (90 mg, 0.16 mmol) according to the procedure described for Example 1, Step C, to yield 150 (50 mg, 66%) as a white solid. LRMS (ESI pos) m/e 470 (M+l). [00397] Example 51 Preparation of 3-(4-tert-butylbenzyl)-5-(4-(6,7- dimethoxyquinolin-4-yloxy)-3-fluorophenyl)pyrimidin-4(3H)-one 151
Figure imgf000105_0002
[00398] Step A: Preparation of 5-bromo-3-(4-tert-butylbenzyl)pyrimidin-4(3H)-one:
Prepared from l-(bromomethyl)-4-tert-butylbenzene (1.9 g, 8.4 mmol) according to the procedure described for Example 13, Step A. The crude product was purified by silica gel flash column chromatography (1:1 EtOAc/Hexane) to yield the product (0.65 g, 24%) as a white solid. LRMS (ESI pos) m/e 323 (M+l).
[00399] Step B: Preparation of 5-(4-(benzyloxy)-3-fluorophenyl)-3-(4-tert- butylbenzyl)pyrimidin-4(3H)-one: Prepared from 5-bromo-3-(4-tert-butylbenzyl)pyrimidin- 4(3H)-one (0.65 mg, 2.0 mmol) according to the procedure described for Example 7, Step A. The crude product was purified by silica gel flash column chromatography (1:1 EtOAc/Hexane) to yield the product (0.50 g, 56%) as a white solid. LRMS (ESI pos) m/e 443 (M+l). 111-03-PCT - P2338R1 - 02120.004WO1
105
[00400] Step C: Preparation of 3-(4-tert-butylbenzyl)-5-(3-fluoro-4- hydroxyphenyl)pyrimidin-4(3H)-one: Prepared from 5-(4-(benzyloxy)-3-fluorophenyl)-3-(4-tert- butylbenzyl)pyrimidin-4(3H)-one (0.50 g, 1.1 mmol) according to the procedure described for Example I5 Step C, to yield the product (0.30 g, 88%) as a white solid. LRJMS (ESI pos) m/e 353 (M+l).
[00401] Step D: Preparation of 3-(4-tert-butylbenzyl)-5-(4-(6,7-dimethoxyquinolin-4- yloxy)-3-fluorophenyl)pyrimidin-4(3H)-one: Prepared from 4-chloro-6,7-dimethoxyquinoline (prepared according to reference procedure in Example 5) (100 mg, 0.46 mmol) and 3-(4-tert- butylbenzyl)-5-(3-fluoro-4-hydroxyphenyl)pyrimidin-4(3H)-one (130 mg, 0.30 mmol) according to the procedure described for Example 1, Step E. The crude product was purified by silica gel flash column chromatography (1:10 MeOH/EtOAc) to yield 151 (48 mg, 30%) as a white solid. 1H NMR (DMSO-d6, 400 MHz) δ 8.79 (s, IH), 8.50 (d, IH), 8.35 (s, IH), 7.92 (dd, IH), 7.72- 7.76 (m, IH), 7.54 (s, IH), 7.48-7.52 (m, IH), 7.42 (s, IH), 7.36-7.40 (m, 2H), 7.30-7.34 (m, 2H), 6.52 (d, IH), 5.18 (s, 2H), 3.96 (s, 3H), 3.95 (s, 3H). LRMS (ESI pos) m/e 540 (M+l). [00402] Example 52 Preparation of 3-(4-chloro-3-(trifluoromethyl)benzyl)-5-(4-(6,7- dimethoxyquinolin-4-yloxy)-3-fluorophenyl)pyrimidin-4(3H)-one 152
Figure imgf000106_0001
[00403] Step A: Preparation of 5-bromo-3-(4-chloro-3-(trifluoromethyl)benzyl)pyrimidin-
4(3H)-one: Prepared from 4-(bromomethyl)-l-chloro-2-(trifluoromethyl)benzene (0.76 g, 2.8 mmol) according to the procedure described for Example 13, Step A. The crude product was purified by silica gel flash column chromatography (1:1 EtOAc/Hexane) to yield the product (0.41 g, 39%) as a white solid. LRMS (ESI pos) m/e 369 (M+l).
[00404] Step B: Preparation of 5-(4-(benzyloxy)-3-fiuorophenyl)-3-(4-chloro-3-
(trifluoromethyl)benzyl)pyrimidin-4(3H)-one: Prepared from 5-bromo-3-(4-chloro-3- fluorobenzyl)pyrimidin-4(3H)-one (0.41 g, 1.1 mmol) according to the procedure described for Example 7, Step A. The crude product was purified by silica gel flash column chromatography (1:1 EtOAc/Hexane) to yield the product (0.50 g, 92%) as a white solid. LRMS (ESI pos) m/e 489 (M+l). 111-03-PCT- P2338R1 - 02120.004WO1
106
[00405] Step C: Preparation of 3-(4-chloro-3-(trifiuoromethyl)benzyl)-5-(3-fluoro-4- hydroxyphenyl)pyrimidin-4(3H)-one: Prepared from 5-(4-(benzyloxy)-3-fluorophenyl)-3-(4- chloro-3-(trifluoromethyl)benzyl)pyrimidin-4(3H)-one (0.50 g, 1.0 mmol) according to the procedure described for Example 1, Step C, to yield the product (0.40 g, 86%) as a white solid. LRMS (ESI pos) m/e 331 (M+l).
[00406] Step D: Preparation of 3-(4-chloro-3-(trifluoromethyl)benzyl)-5-(4-(6,7- dimethoxyquinolin-4-yloxy)-3-fluorophenyl)pyrimidin-4(3H)-one: Prepared from 4-chloro-6,7- dimethoxyquinoline (prepared according to reference procedure in Example 5) (60 mg, 0.27 mmol) and 3-(4-chloro-3-(trifluoromethyl)benzyl)-5-(3-fluoro-4-hydroxyphenyl)pyrimidin- 4(3H)-one(107 mg, 0.27 mmol) according to the procedure described for Example 1, Step E. The crude product was purified by silica gel flash column chromatography (1:10 MeOH/EtOAc) to yield 152 (12 mg, 7%) as a white solid. 1H NMR (DMSOd6, 400 MHz) δ 8.80 (s, IH), 8.40 (s, IH), 8.30 (s, IH), 7.92 (s, IH), 7.80-7.90 (m, IH), 7.60-7.75 (m, 3H), 7.40-7.50 (m, 2H), 7.30- 7.40 (m, IH), 6.40-6.50 (m, Ih), 5.20 (s,2H), 3.96 (s, 6H). LRMS (ESI pos) m/e 586 (M+l). [00407] Example 53 Preparation of (4-benzylpiperazin-l-yl)(4-(6,7- dimethoxyquinolin-4-yloxy)-3-fluorophenyl)methanone 153
Figure imgf000107_0001
[00408] Step A: Preparation of (4-benzylpiperazm-l-yl)(3-fiuoro-4- methoxyphenyl)methanone: Prepared from 1-benzylpiperazine (2.3 mg, 13 mmol) ) according to the procedure described for Example 4, Step A, to yield the product (2.6 g, 65%) as a white solid. LRMS (ESI pos) m/e 329 (M+l).
[00409] Step B: Preparation of (4-benzylpiperazin-l-yl)(3-fluoro-4- hydroxyphenyl)methanone: Prepared from (4-benzylpiperazin-l-yl)(3-fluoro-4- methoxyphenyl)methanone (2.0 g, 6.1 mmol) according to the procedure described for Example 4, Step B, to yield the product (1.0 g, 53%) as a white solid. LRMS (ESI pos) m/e 315 (M+l). [00410] Step C: Preparation of 5 (4-benzylpiperazin-l-yl)(4-(6,7-dimethoxyquinolin-4- yloxy)-3-fluorophenyl)methanone: Prepared from (4-benzylpiperazin-l-yl)(3-fluoro-4- hydroxyphenyl)methanone (18 mg, 0.059 mmol) and 4-chloro-6-methoxy-7-(3- morpholinopropoxy)quinoline (20 mg, 0.059 mmol) according to the procedure described for Example 1, Step E, to yield 153 (40 mg, 18% yield) as a white solid. 1H NMR (DMSOd6, 400 111-03-PCT - P2338R1 - 02120.Q04WO1
107
MHz) δ 8.50 (d, IH), 7.48-7.56 (m, 3H), 7.42 (s, IH), 7.24-7.36 (m, 6H), 6.58 (d, IH), 3.96 (s, 3H), 3.95 (s, 3H), 3.60-3.70 (m, 2H), 3.30-3.50 (m, 2H), 2.35-2.45 (m, 4H). LRMS (ESI pos) m/e 502 (M+l).
[00411] Example 54 Preparation of 6-benzyl-3-(4-(6-methoxy-7-(3- moφholinopropoxy)quinolin-4-yloxy)phenyl)pyrimidin-4(3H)-one 154
Figure imgf000108_0001
[00412] Step A: Preparation of (E)-ethyl 3-amino-2-(4-(benzyloxy)phenylcarbamoyl)-4- phenylbut-2-enoate: A suspension of (E)-ethyl 3-amino-4-phenylbut-2-enoate (1 g, 4.9 mmol) and l-((4-isocyanatophenoxy)methyl)benzene (1.1 g, 4.9 mmol) was heated in DMF (20 mL) at 6O0C for 72 hours. The reaction mixture was poured into water (10 mL) and removed the solid by filtration. The filtrate was evaporated and the residue was purified by silica gel flash column chromatography (1 :4 EtOAc/hexane) to yield the product (1.8 g, 86%) as a yellow solid.1H NMR (CDCl3, 400 MHz) δ 10.90 (s, IH), 7.20-7.42 (m, 14H), 6.90-6.95 (m, 2H), 5.04 (s, 2H), 4.24 (q, 2H), 4.15 (s, 2H), 1.29 (t, 3H).
[00413] Step B: Preparation of ethyl 4-benzyl-l-(4-(benzyloxy)phenyl)-6-oxo-l,6- dihydropyrimidine-5-carboxylate: Acetic anhydride (1 ml, 10.6 mmol) was added into a solution of (E)-ethyl 3-amino-2-((4-(benzyloxy)phenyl)carbamoyl)-4-phenylbut-2-enoate (1 g, 2.32 mmol) and triethyl orthroformate (5 ml, 30.1 mmol). The reaction was heated at HO0C for 16 hours, cooled and solvent was evaporated. The residue was purified by silica gel flash column chromatography (1 :2 EtOAc/hexane) to yield the product (0.9 g, 88%) as a yellow solid.1H NMR (CDCl3, 400 MHz) δ 8.10 (s, IH), 7.30-7.42 (m, 10H), 7.20-7.28 (m, 2H), 3.96 (s, 2H), 1.36 (q, 3H).
[00414] Step C: Preparation of 6-benzyl-3-(4-hydroxyphenyl)pyrimidin-4(3H)-one: Ethyl
4-benzyl-l-(4-(benzyloxy)phenyl)-6-oxo-l,6-dihydropyrimidine-5-carboxylate (0.87 g, 1.98 mmol) was added into concentrated HCl (20 mL) and acetic acid (20 mL) and stirred for 3 hours. The reaction mixture was extracted with CH2Cl2, and the organic layer was washed with brine, dried over sodium sulfate, filtered, and concentrated. The residue was purified by silica gel flash column chromatography (1:10 Et2O/Hexane) to yield the product (0.2 g, 36% yield) as a yellow 111-03-PCT- P2338R1 - 02120.004WO1
108 solid. 1H NMR (CDCl3, 400 MHz) δ 8.10 (s, IH), 7.25-7.40 (m, 5H), 7.04-7.14 (m, 2H), 6.79- 6.85 (m, 2H), 6.33 (s, IH), 3.91 (s, 2H). LRMS (ESI pos) m/e 279 (M+l). [00415] Step D: Preparation of 6-benzyl-3-(4-(6-methoxy-7-(3- morpholinopropoxy)quinolin-4-yloxy)phenyl)pyrimidin-4(3H)-one: Prepared from 4-chloro-6- methoxy-7-(3-morpholinopropoxy)quinoline (34 mg, 0.10 mmol) and 6-benzyl-3-(4- hydroxyphenyl)pyrimidin-4(3H)-one (28 mg, 0.10 mmol) according to the procedure described for Example 1, Step E. The residue was purified by silica gel flash column chromatography column (1:10 MeOH/EtOAc) to yield 154 (20 mg, 34%) as a yellow solid. 1H NMR (CDCl3, 400 MHz) δ 8.55 (d, IH), 8.14 (s, IH), 7.46 (d, IH), 7.40-7.45 (m, IH), 7.35-7.46 (m, IH), 7.24-7.34 (m, 8H), 6.63 (d, 2H), 6.35 (s, IH), 4.25-4.36 (m, 2H), 4.01 (s, 3H), 3.92 (s, 2H), 3.70-3.75 (m, 4H), 2.55-2.66 (m, 2H), 2.45-2.50 (m, 4H), 2.10-2.15 (m, 2H). LRMS (ESI pos) m/e 579 (M+l). [00416] Example 55 Preparation of 5-benzyl-3-(3-fluoro-4-(6-methoxy-7-(3- morpholinopropoxy)quinolin-4-yloxy)phenyl)pyrimidin-4(3H)-one 155
Figure imgf000109_0001
[00417] Step A: preparation of (4,6-dichloropyrimidin-5-yl)(phenyl)methanol:
Phenylmagnesium bromide (11 ml, 11 mmol) was added into a solution of 4,6-Dichloro-5- pyrimidinecarbaldehyde (2 g, 11 mmol) in THF (30 mL) at -780C. The reaction was allowed to warm to room temperature and was poured into water (10 mL). The reaction mixture was extracted with EtOAc, and the organic layer was washed with brine, dried over sodium sulfate, filtered, and concentrated. The residue was purified by silica gel flash column chromatography (1:10 Et2O/Hexane) to yield the product (2.4 g, 83% yield) as a white solid. IH NMR (CDCl3, 400 MHz) δ 8.76 (s, IH), 7.20-7.41 (m, 5H), 6.50 (d, IH), 3.00 (d, IH).
[00418] Step B: Preparation of (4-(benzyloxy)-6-chloropyrimidin-5-yl)(phenyl)methanol:
KOH (0.44 g, 7.8 mmol) was added into a solution of (4,6-dichloropyrimidin-5- yl)(phenyl)methanol (1.0 g, 3.9 mmol), benzyl alcohol (0.41 ml, 3.9 mmol) and 18-crown-6 (0.21 g, 0.78 mmol) in toluene (50 mL). The solution was heated to reflux for 2 hours. The reaction was allowed to warm to room temperature and was poured into water (10 mL). The reaction mixture was extracted with EtOAc, and the organic layer was washed with brine, dried over sodium sulfate, filtered, and concentrated. The residue was purified by silica gel flash 111-03-PCT- P2338R1 - 02120.004WO1
109 column chromatography (1:10 Et2O/Hexane) to yield the product (0.5 g, 39% yield) as a white solid. LRMS (ESI pos) m/e 327 (M+l).
[00419] Step C: Preparation of 5-benzylpyrimidin-4-ol: Prepared from 5-benzylpyrimidin-
4-ol (0.5 g, 1.5 mmol) according to the procedure described for Example 14, Step B, to yield the product (0.17 g, 60%) as a white solid. LRMS (ESI pos) m/e 187 (M+l).
[00420] Step D: Preparation of 5-benzyl-3-(3-fluoro-4-hydroxyphenyl)pyrimidin-4(3H)- one: Prepared from 5-benzylpyrimidin-4-ol (0.1 g, 0.54 mmol) according to the procedure described for Example 1, Step D, to yield the product (20 mg, 13 % yield) as a brown solid. LRMS (ESI pos) m/e 279 (M+l).
[00421] Step E: Preparation of 5-benzyl-3-(3-fiuoro-4-(6-methoxy-7-(3- morpholinopropoxy)quinolin-4-yloxy)phenyl)pyrimidin-4(3H)-one: Prepared from 5-benzyl-3- (3-fluoro-4-hydroxyphenyl)pyrimidin-4(3H)-one (18 mg, 0.059 mmol) according to the procedure described for Example 1, Step E, to yield 155 (1 mg, 2.8% yield) as a light brown solid. LRMS (ESI pos) m/e 597 (M+l).
[00422] Example 56 Preparation of 2-benzyl-5-(3-fluoro-4-(6-methoxy-7-(3- morpholinopropoxy)quinolin-4-yloxy)phenyl)pyrimidin-4(3H)-one 156
Figure imgf000110_0001
[00423] Step A: Preparation of 2-benzyl-4-methoxypyrimidine: A solution of 2-chloro-4- methoxypyrimidine (0.500 g, 3.46 mmol) and PdCl2(PPh3)2 (0.121 g, 0.173 mmol) in THF (10 mL) was sparged with N2. Benzylzinc(II) bromide (8.30 ml, 4.15 mmol; 0.5 M solution in THF) was added and the reaction mixture was stirred at reflux for 1 hour. The reaction mixture was cooled to room temperature and then partitioned between EtOAc and H2O. The phases were separated, and the aqueous phase was re-extracted with EtOAc (Ix). The combined organic layers were dried (Na2SO4), filtered and concentrated to yield a crude dark brown oil. The crude product was purified by silica gel flash column chromatography, eluting with 20:1 dichloromethane/EtOAc. The desired product (0.676 g, 98%) was obtained as a dark yellow oil. 1H-NMR (400 MHz, CDCl3) δ 8.34 (d, IH), 7.41-7.25 (m, 4H), 7.21 (m, IH), 6.53 (d, IH), 4.16 (s, 2H), 3.95 (s, 3H). LRMS (ESI pos) m/e 201 (M+l). 111-O3-PCT - P2338R1 - 02120.004WO1
110
[00424] Step B: Preparation of 2-benzylpyrimidin-4(3H)-one: To a solution of 2-benzyl-
4-methoxypyrimidine (0.675 g, 3.37 mmol) in AcOH (15 mL) was added HBr (2.28 ml, 20.2 mmol; 48 wt% in H2O). The reaction mixture was stirred at 95°C for 2 hours. The reaction mixture was cooled to room temperature and diluted with H2O. The pH of the reaction mixture was adjusted to 5-6 with 6 M aqueous NaOH and then partitioned between EtOAc and H2O. The phases were separated, and the aqueous phase was re-extracted with EtOAc (Ix). The combined organic layers were dried (Na2SO4), filtered and concentrated to yield a crude yellow solid. Purification of the crude product was achieved by trituration with dichloromethane and diethyl ether. The resulting solid was filtered, washed with diethyl ether, collected and dried under vacuum to yield the desired product (0.531 g, 85%) as an off-white solid. 1H-NMR (400 MHz, DMSO-d6) δ 7.79 (d, IH), 7.31 (m, 4H), 7.23 (m, IH), 6.10 (d, IH), 3.83 (s, 2H). LRMS (ESI pos) m/e 187 (M+ 1).
[00425] Step C: Preparation of 2-benzyl-5-bromopyrimidin-4(3H)-one: To a solution of
2-benzylpyrimidin-4(3H)-one (0.531 g, 2.85 mmol) in CHCl3 (15 mL) and methanol (3 mL) was added bromine (0.146 ml, 2.85 mmol). The reaction mixture was stirred at room temperature for 3 hours and then quenched with 10% sodium bisulfite solution. The reaction mixture was partitioned between EtOAc and H2O. The phases were separated, and the aqueous phase was re- extracted with EtOAc (Ix). The combined organic layers were dried (Na2SO4), filtered and concentrated to yield a crude yellow solid. Purification of the crude product was achieved by trituration with dichloromethane. The resulting solid was filtered, washed with dichloromethane, collected and dried under vacuum to yield the desired product (0.302 g, 40%) as an off-white solid. 1H-NMR (400 MHz, DMSO-d6) δ 13.23 (br s, IH), 8.25 (s, IH), 7.35-7.28 (m, 4H), 7.27- 7.22 (m, IH), 3.87 (s, 2H). LRMS (ESI pos) m/e 265, 267 (M+, Br pattern). [00426] Step D: Preparation of 2-benzyl-5-(4-(benzyloxy)-3-fluorophenyl)pyrimidin-
4(3H)-one: A solution of 2-benzyl-5-bromopyrimidin-4(3H)-one (0.300 g, 1.13 mmol), 4- (benzyloxy)-3-fluorophenylboronic acid (0.334 g, 1.36 mmol), Pd(PPh3)4 (0.065 g, 0.057 mmol) and lithium chloride (0.240 g, 5.66 mmol) in dioxane (3 mL) and 2 M aqueous Na2CO3 (0.3 mL) was stirred at 1000C for 18 hours. After cooling, the reaction mixture was partitioned between EtOAc and H2O. The phases were separated, and the aqueous phase was re-extracted with EtOAc (3x). The combined organic layers were dried (Na2SO4), filtered and concentrated to yield a crude dark yellow solid. Purification of the crude product was achieved by trituration with dichloromethane. The resulting solid was filtered, washed with dichloromethane, collected and dried under vacuum. The filtrate was concentrated and the trituration procedure was repeated (2x) with EtOAc. The solids were combined to yield the desired product (0.284 g, 65%) as a pale 111-03-PCT- P2338R1 - 02120.004WO1
111 yellow solid. 1H-NMR (400 MHz, DMSOd6) δ 12.93 (br s, IH), 8.13 (s, IH), 7.66 (dd, IH), 7.53-7.22 (s, 12H), 5.21 (s, 2H), 3.90 (s, 2H). LRMS (APCI pos) m/e 387 (M+l). [00427] Step E: Preparation of 2-benzyl-5-(3-fluoro-4-hydroxyphenyl)pyrimidin-4(3H)- one: A suspension of 2-benzyl-5-(4-(benzyloxy)-3-fluorophenyl)pyrimidin-4(3H)-one (0.284 g, 0.735 mmol) in trifluoroacetic acid (3 mL) was stirred at 40°C for 2 hours and then at room temperature for 16 hours. The reaction mixture was concentrated to dryness and then purified by silica gel flash column chromatography, eluting with 20:1 dichloromethane/MeOH. The desired product (0.177 g, 81%) was obtained as a white solid. 1H-NMR (400 MHz, DMSOd6) δ 12.89 (br s, IH), 9.98 (s, IH)5 8.08 (s, IH), 7.58 (dd, IH), 7.39-7.30 (m, 5H), 7.28-7.22 (m, IH), 6.96 (dd, IH), 3.89 (s, 2H). LRMS (ESI pos) m/e 297 (M+l).
[00428] Step F: Preparation of 2-benzyl-5-(3-fluoro-4-(6-methoxy-7-(3- morpholinopropoxy)quinolin-4-yloxy)phenyl)pyrimidin-4(3H)-one: To a solution of 2-benzyl-5- (3-fluoro-4-hydroxyphenyl)pyrimidin-4(3H)-one (0.027 g; 0.091 mmol) in toluene (500 uL) in a microwave tube was added 4-(3-(4-chloro-6-methoxyquinolin-7-yloxy)propyl)morpholine (prepared according to the reference in Example 1, step E) (0.031 g, 0.091 mmol) and DMAP (0.011 g, 0.091 mmol). The reaction was stirred at 180°C in the microwave for 2 hours. The mixture was solubilized with a small amount of MeOH and purified by silica gel flash column chromatography, eluting with 9:1 EtOAc/MeOH. The desired product eluted along with residual 4-(3-(4-chloro-6-methoxyquinolin-7-yloxy)propyl)morpholine. The mixture was concentrated and repurified by silica gel flash column chromatography, eluting with a step gradient of CH2Cl2 (150 mL), 2.5/97.5 MeOH/CH2Cl2 (200 mL) and 5/95 MeOH/CH2Cl2 (500 mL) to give 156 (0.022 g, 40%) as a tan foamy solid. 1H-NMR (400 MHz, CDCl3) δ 8.50 (d, IH), 8.22 (s, IH), 7.77 (dd, IH), 7.57 (s, IH), 7.55 (d, IH), 7.45 (s, IH), 7.42-7.28 (m, 6H), 6.50 (m, IH), 4.28 (t, 2H), 4.08 (s, 2H), 4.04 (s, 3H), 3.73 (t, 4H), 2.58 (t, 2H), 2.49 (m, 4H), 2.14 (m, 2H). LRMS (APCI pos) m/e 597 (M+l).
[00429] Example 57 Preparation of 2-benzyl-5-(4-(6,7-dimethoxyquinolin-4-yloxy)-3- fluorophenyl)pyrimidin-4(3H)-one 157
Figure imgf000112_0001
[00430] Prepared according to the method of Example 56, step F, substituting 4-chloro-
6,7-dimethoxyquinoline (reference for preparation given in Example 5)(6.8 mg, 0.03 mmol) for 111-03-PCT - P2338R1 - 02120.004WO1
112
4-(3-(4-chloro-6-methoxyquinolin-7-yloxy)propyl)morpholine to provide 157 (3.3 mg, 34%). 1H-NMR (400 MHz, CDCl3) δ 8.52 (d, IH), 8.22 (s, IH), 7.74 (m, IH), 7.59 (s, IH), 7.54 m, IH), 7.44 (s, IH), 7.40-7.30 (m, 6H), 6.51 (d, IH), 4.08 (s, 2H), 4.06 (s, 3H), 4.05 (s, 3H). LRMS (ESI pos) m/e 484 (M+l).
[00431] Example 58 Preparation of 2-benzyl-5-(4-(6-methoxy-7-(3- morpholinopropoxy)quinolin-4-yloxy)phenyl)pyrimidin-4(3H)-one 158
Figure imgf000113_0001
[00432] Step A: Preparation of 2-benzyl-5-(4-(benzyloxy)phenyl)pyrimidin-4(3H)-one:
Prepared from 2-benzyl-5-bromopyrimidin-4(3H)-one (0.100 g, 0.377 mmol; obtained from Example 56, Step C) according to the procedure described in Step D of Example 56, substituting 4-(benzyloxy)phenylboronic acid in place of 4-(benzyloxy)-3-fluorophenylboronic acid. The desired product (0.116 g, 84%) was obtained as a white solid. 1H-NMR (400 MHz, DMSOd6) δ 12.84 (br s, IH), 8.04 (s, IH), 7.64 (m, 2H), 7.45 (m, 2H), 7.42-7.29 (m, 7H), 7.25 (m, IH), 7.03 (m, 2H), 5.13 (s, 2H), 3.89 (s, 2H). LRMS (APCI pos) m/e 369 (M+l). [00433] Step B: Preparation of 2-benzyl-5-(4-hydroxyphenyl)pyrimidin-4(3H)-one:
Prepared from 2-benzyl-5-(4-(benzyloxy)phenyl)pyrimidin-4(3H)-one (0.116 g, 0.315 mmol) according to the procedure described in Step E of Example 56. The desired product (0.042 g, 48%) was obtained as a white solid. 1H-NMR (400 MHz, DMSO-d6) δ 9.54 (br s, IH), 7.99 (s, IH), 7.52 (m, 2H), 7.37-7.30 (m, 4H), 7.25 (m, IH), 6.77 (m, 2H), 3.89 (s, 2H). LRMS (ESI pos) m/e 279 (M+l).
[00434] Step C: Preparation of 2-benzyl-5-(4-(6-methoxy-7-(3- morpholinopropoxy)quinolin-4-yloxy)phenyl)pyrimidin-4(3H)-one: To a stirred suspension of 2- benzyl-5-(4-hydroxyphenyl)pyrimidin-4(3H)-one (0.021 g, 0.076 mmol) in bromobenzene (700 μL) at room temperature under N2 was added 4-(3-(4-chloro-6-methoxyquinolin-7- yloxy)propyl)morpholine (prepared according to the referenced procedure in Example 1, step E) (0.028 g, 0.083 mmol) followed by DMAP (0.001 g, 0.008 mmol). The reaction mixture was stirred at 1500C for 12 hours and then stirred at room temperature for an additional 9 hours. The reaction mixture was directly purified by silica gel flash column chromatography, eluting with 10:1 dichloromethane/MeOH, to give 158 (0.023 g, 53%) as a pale yellow foamy solid. 1H-NMR 111-03-PCT - P2338R1 - 02120.004WO1
113
(400 MHz, DMSO-d6) δ 12.99 (br s, IH), 8.49 (d, IH), 8.18 (s, IH), 7.86 (m, 2H), 7.50 (s, IH), 7.43-7.22 (m, 8H), 6.54 (d, IH), 4.21 (t, 2H), 3.93 (s, 5H), 3.59 (m, 4H), 2.47 (m, 2H), 2.39 (m, 4H), 1.98 (m, 2H). LRMS (ESI pos) m/e 579 (M+l).
[00435] Example 59 Preparation of 2-benzyl-5-(3-fluoro-4-(6-methoxy-7-(3- morpholinopropoxy)quinolin-4-yloxy)phenyl)-3-methylpyrimidin-4(3H)-one 159
Figure imgf000114_0001
[00436] Step A: Preparation of 5-bromo-2-chloropyrimidin-4(3H)-one: Prepared from 5- bromo-2,4-dichloropyrimidine (10.00 g, 43.88 mmol) according to the procedure described in EP 1506967. The desired product (4.59 g, 50%) was obtained as a pale yellow solid. 1H-NMR (400 MHz, DMSO-d6) δ 8.36 (s, IH). LRMS (ESI neg) m/e 207, 209 (M-, Br pattern). [00437] Step B: Preparation of 5-bromo-2-chloro-3-methylpyrimidin-4(3H)-one: To a solution of 5-bromo-2-chloropyrimidin-4(3H)-one (1.00 g, 4.78 mmol) in DME (12 mL)/DMF (3 mL) under N2 at 00C was added LiH (0.044 g, 5.25 mmol) and the reaction was stirred at room temperature for 15 minutes. Iodomethane (0.589 ml, 9.45 mmol) was then added and the reaction was stirred at room temperature for 30 minutes and then at 600C for 1.5 hours. The reaction mixture was quenched with H2O and then partitioned between EtOAc and saturated aqueous NaCl. The phases were separated, and the aqueous phase was re-extracted with EtOAc (Ix). The combined organic layers were dried (Na2SO4), filtered and concentrated to yield a crude yellow oil. The crude product was purified by silica gel flash column chromatography, eluting with 25:1 dichloromethane/EtOAc. The desired product (0.764 g, 72%) was obtained as a yellow crystalline solid. 1H-NMR (400 MHz, DMSO-d6) δ 8.26 (s, IH), 3.59 (s, 3H). LRMS (ESI pos) m/e 223, 225 (M+, Br pattern).
[00438] Step C: Preparation of 2-benzyl-5-bromo-3-methylpyrimidin-4(3H)-one: A solution of 5-bromo-2-chloro-3-methylpyrimidin-4(3H)-one (0.100 g, 0.448 mmol) and PdCl2(PPh3)2 (0.016 g, 0.022 mmol) in THF (4 mL) was sparged with N2. Benzylzinc(II) bromide (0.904 ml, 0.452 mmol; 0.5 M solution in THF) was added and the reaction mixture was stirred at reflux for 30 minutes. The reaction mixture was cooled to room temperature and then partitioned between EtOAc and H2O. The phases were separated, and the aqueous phase was re- extracted with EtOAc (Ix). The combined organic layers were dried (Na2SO4), filtered and 111-03-PCT - P2338R1 - 02120.004WO1
114 concentrated to yield a crude yellow gum. The crude product was purified by silica gel flash column chromatography, eluting with 20:1 dichloromethane/EtOAc. The desired product (0.067 g, 54%) was obtained as a colorless gum. 1H-NMR (400 MHz, CDCl3) δ 8.21 (s, IH), 7.38-7.27 (m ,3H), 7.19 (m, 2H), 4.14 (s, 2H), 3.50 (s, 3H). LRMS (ESI pos) m/e 279, 281 (M+, Br pattern).
[00439] Step D. Preparation of 2-benzyl-5-(4-(benzyloxy)-3-fluorophenyl)-3- methylpyrimidin-4(3H)-one: Prepared from 2-benzyl-5-bromo-3-methylpyrimidin-4(3H)-one (0.067 g, 0.240 mmol) according to the procedure described in Step D of Example 56. The crude product was purified by silica gel flash column chromatography, eluting with 10:1 dichloromethane/EtOAc. The desired product (0.082 g, 85%) was obtained as an off-white solid. 1H-NMR (400 MHz, DMSOd6) δ 8.15 (s, IH), 7.66 (dd, IH), 7.51 (m, IH), 7.47 (m, 2H), 7.43-
7.38 (m, 2H), 7.38-7.32 (m, 3H), 7.30-7.25 (m, 4H), 5.22 (s, 2H), 4.24 (s, 2H), 3.48 (s, 3H). LRMS (APCI pos) m/e 401 (M+l).
[00440] Step E. Preparation of 2-benzyl-5-(3-fluoro-4-hydroxyphenyl)-3- methylpyrimidin-4(3H)-one: Prepared from 2-benzyl-5-(4-(benzyloxy)-3-fiuorophenyl)-3- methylpyrimidin-4(3H)-one (0.082 g, 0.20 mmol), according to the procedure described in Step E of Example 56. The desired product (0.064 g, 100%) was obtained as a pale yellow foamy solid. 1H-NMR (400 MHz, DMSOd6) δ 10.01 (br s, IH), 8.10 (s, IH), 7.58 (dd, IH), 7.40-7.32 (m, 3H), 7.30-7.23 (m, 3H), 6.97 (dd, IH), 4.24 (s, 2H), 3.47 (s, 3H). LRMS (ESI pos) m/e 311 (M+l).
[00441] Step F. Preparation of 2-benzyl-5-(3-fluoro-4-(6-methoxy-7-(3- morpholinopropoxy)quinolin-4-yloxy)phenyl)-3-methylpyrimidin-4(3H)-one: Prepared from 2- benzyl-5-(3-fluoro-4-hydroxyphenyl)-3-methylpyrimidin-4(3H)-one (0.025 g, 0.081 mmol), 4-(3- (4-chloro-6-methoxyquinolin-7-yloxy)propyl)morpholine (0.030 g, 0.089 mmol) and catalytic DMAP according to the procedure described in Step C of Example 58, to give 159 (0.006 g, 12%) as a yellow solid. 1H-NMR (400 MHz, DMSOd6) δ 8.50 (d, IH), 8.29 (s, IH), 7.92 (dd, IH), 7.74 (m, IH), 7.53 (s, IH), 7.50 (t, IH), 7.42 (s, IH), 7.39-7.34 (m, 2H), 7.32-7.26 (m, 3H), 6.52 (dd, IH), 4.28 (s, 2H), 4.21 (t, 2H), 3.95 (s, 3H), 3.59 (m, 4H), 3.52 (s, 3H), 2.47 (m ,2H),
2.39 (m, 4H), 1.99 (m, 2H). LRMS (APCI pos) m/e 611 (M+l).
[00442] Example 60 Preparation of 3-(4-(6-methoxy-7-(3- moφholinopropoxy)quinolin-4-yloxy)phenyl)-5-methyl-6-(phenylamino)pyrimidin-4(3H)-one
160 111-03-PCT - P2338R1 - 02120.004WO1
115
Figure imgf000116_0001
[00443] Step A: Preparation of 4-chloro-6-methoxy-5-methylpyrimidine: To a solution of 4,6-dichloro-5-methylpyrimidine (1.00 g, 6.13 mmol) in MeOH (50 mL) at 0°C was added solid sodium methoxide (0.348 g, 6.44 mmol) in portions. The reaction mixture was allowed to warm to room temperature and stirred at room temperature for 4 hours and then at 500C for 12 hours. Additional sodium methoxide (0.348 g, 6.44 mmol) was added and the reaction mixture was stirred at 50°C for 4 hours. Additional sodium methoxide (0.348 g, 6.44 mmol; 3 eq total) was added and the reaction mixture was stirred at 5O0C for 20 minutes, when HPLC showed the reaction was complete. The reaction mixture was concentrated and then partitioned between EtOAc and saturated aqueous NH4Cl. The phases were separated, and the aqueous phase was re- extracted with EtOAc (Ix). The combined organic layers were dried (Na2SO4), filtered and concentrated to yield the desired product (0.829 g, 85%) as a colorless oil that was used without further purification. 1H-NMR (400 MHz, CDCl3) δ 8.42 (s, IH), 4.02 (s, 3H). LRMS (ESI pos) m/e l59 (M+l).
[00444] Step B: Preparation of 5-methyl-6-(phenylamino)pyrimidin-4(3H)-one: In a sealed tube was a mixture of 4-chloro-6-methoxy-5-methylpyrimidine (0.973 g, 6.14 mmol) and aniline (1.679 ml, 18.41 mmol) in n-BuOH (10 mL). The reaction mixture was stirred at reflux for 5 days and then at room temperature for 5 days. The reaction mixture was a purple suspension after cooling. The resulting solid was filtered and washed with Et2O, collected and dried under vacuum. The filtrate was concentrated, dried under vacuum and the trituration procedure was repeated with dichloromethane/Et2O. The solids were combined to yield the desired product (0.611 g, 49%) as a lavender solid. 1H-NMR (400 MHz, DMSO-d6) δ 11.88 (br s, IH), 8.07 (s, IH), 7.85 (s, IH), 7.40 (m, 2H), 7.25 (m, 2H), 6.97 (m, IH), 1.91 (s, 3H). LRMS (ESI pos) m/e 202 (M+l).
[00445] Step C: Preparation of 3-(4-(benzyloxy)phenyl)-5-methyl-6-
(phenylamino)pyrimidin-4(3H)-one: To a mixture of 5-methyl-6-(phenylamino)pyrimidin- 4(3H)-one (0.025 g, 0.124 mmol) and l-(benzyloxy)-4-iodobenzene (0.046 g, 0.149 mmol) in dioxane (500 μL) and DMF (~ 4 drops) was added copper (I) iodide (0.005 g, 0.024 mmol), (lS,2S)-cyclohexane-l,2-diamine (0.006 ml, 0.050 mmol), and K3PO4 (0.053 g, 0.248 mmol). 111-03-PCT- P2338R1 - 02120.004WO1
116
The mixture was flushed with N2 and stirred at 110°C for 5 hours, when LC-MS showed some formation of desired product as well as formation of (lS,2S)-Nl-(4- (benzyloxy)phenyl)cyclohexane-l,2-diamine from coupling of ligand to l-(benzyloxy)-4- iodobenzene. Because the reaction had stalled from ligand and l-(benzyloxy)-4-iodobenzene being depleted, the secondary ligand Nl,N2-dimethylethane-l,2-diamine (0.0132 ml, 0.124 mmol) and additional l-(benzyloxy)-4-iodobenzene (0.020 g, 0.065 mmol) were added. The reaction mixture was then stirred at 1100C for an additional 16 hours. The reaction mixture was partitioned between EtOAc and saturated aqueous NaCl. The phases were separated, and the aqueous phase was re-extracted with EtOAc (Ix). The combined organic layers were dried (Na2SO4), filtered and concentrated. The crude product was purified by silica gel flash column chromatography, eluting with 10:1 dichloromethane/EtOAc. The desired product (0.030 g, 63%) was obtained as a white foamy solid. 1H-NMR (400 MHz, DMSO-d6) δ 8.25 (s, IH), 8.11 (s, IH), 7.49-7.38 (m 6H), 7.37-7.25 (m, 5H), 7.11 (m, 2H), 7.01 (m, IH), 5.17 (s, 2H), 1.98 (s, 3H). LRMS (ESI pos) m/e 384 (M+l).
[00446] Step D: Preparation of 3-(4-hydroxyphenyl)-5-methyl-6-
(phenylamino)pyrimidin-4(3H)-one: Prepared from 3-(4-(benzyloxy)phenyl)-5-methyl-6- (phenylamino)pyrimidin-4(3H)-one (0.029 g, 0.074 mmol) according to the procedure described in Step E of Example 56. The desired product (0.020 g, 89%) was obtained as a white solid. 1H- NMR (400 MHz, DMSO-d6) δ 9.73 (s, IH), 8.23 (br s, IH), 8.08 (s, IH), 7.43 (m, 2H), 7.28 (m, 2H), 7.18 (m, 2H), 7.00 (m, IH)5 6.84 (m, 2H), 1.98 (s, 3H). LRMS (ESI pos) m/e 294 (M+l). [00447] Step E: Preparation of 3-(4-(6-methoxy-7-(3-morpholinopropoxy)quinolin-4- yloxy)phenyl)-5-methyl-6-(phenylamino)pyrimidin-4(3H)-one: Prepared from 3-(4- hydroxyphenyl)-5-methyl-6-(phenylamino)pyrimidin-4(3H)-one (0.019 g, 0.063 mmol), 4-(3-(4- chloro-6-methoxyquinolin-7-yloxy)propyl)morpholine (0.023 g, 0.069 mmol) and catalytic DMAP according to the procedure described in Step C of Example 58, to give 160 (0.026 g, 69%) as a pale yellow foamy solid. 1H-NMR (400 MHz, DMSO-d6) δ 8.54 (d, IH), 8.32 (br s, IH), 8.21 (s, IH), 7.56 (m, 2H), 7.49 (s, IH), 7.47-7.36 (m, 5H), 7.29 (m, 2H), 7.02 (m, IH), 6.62 (d, IH), 4.21 (t, 2H), 3.93 (s, 3H), 3.59 (t, 4H), 2.48 (m, 2H), 2.39 (m, 4H), 2.02 (s, 3H), 1.98 (m, 2h). LRMS (APCI pos) m/e 594 (M+l).
[00448] Example 61 Preparation of 3-(3-fluoro-4-(6-methoxy-7-(3- moφholinopropoxy)quinolin-4-yloxy)phenyl)-5-methyl-6-(phenylamino)pyrimidin-4(3H)-one 161 111-03-PCT- P2338R1 - 02120.004WO1
117
Figure imgf000118_0001
[00449] Step A: Preparation of 3-(3-fluoro-4-hydroxyphenyl)-5-methyl-6-
(phenylamino)pyrimidin-4(3H)-one: To a mixture of 5-methyl-6-(phenylamino)pyrimidin-4(3H)- one (0.100 g, 0.497 mmol; obtained from Example XX, Step B) and 4-bromo-2-fluorophenol (0.0765 ml, 0.745 mmol) in dioxane (2 mL) and DMF (-12 drops) was added copper(I) iodide (0.019 g, 0.099 mmol), Nl,N2-dimethylethane-l,2-diamine (0.0214 ml, 0.199 mmol) and K3PO4 (0.211 g, 0.994 mmol). The mixture was flushed with N2 and stirred at 11O0C for 16 hours. The reaction mixture was partitioned between EtOAc and saturated aqueous NaCl. The phases were separated, and the aqueous phase was re-extracted with EtOAc (2x). The combined organic layers were dried (Na2SO4), filtered and concentrated. The crude product was purified by silica gel flash column chromatography, eluting with 20:1 dichloromethane/MeOH. The desired product (0.069 g, 45%) was obtained as a brown solid. 1H-NMR (400 MHz, DMSO-d6) δ 10.22 (s, IH), 8.26 (s, IH), 8.11 (s, IH), 7.42 (m, 2H), 7.33-7.25 (m, 3H), 7.07-6.97 (m, 3H), 1.98 (s, 3H). LRMS (ESI pos) m/e 312 (M+l).
[00450] Step B: Preparation of 3-(3-fluoro-4-(6-methoxy-7-(3- morpholinopropoxy)quinolin-4-yloxy)phenyl)-5-methyl-6-(phenylamino)pyrimidin-4(3H)-one: Prepared from 3-(3-fluoro-4-hydroxyphenyl)-5-methyl-6-(phenylamino)pyrimidin-4(3H)-one (0.025 g, 0.0803 mmol), 4-(3-(4-chloro-6-methoxyquinolin-7-yloxy)propyl)morpholine (0.0298 g, 0.0883 mmol) and catalytic DMAP according to the procedure described in Step C of Example 58, to give 161 (0.035 g, 71%) as a pale yellow foamy solid. 1H-NMR (400 MHz, DMSO-d6) δ 8.54 (d, IH), 8.36 (br s, IH), 8.24 (s, IH), 7.74 (dd, IH), 7.59 (t, IH), 7.54 (s, IH), 7.46-7.42 (m, 4H), 7.30 (m, 2H), 7.04 (m, IH), 6.55 (dd, IH), 4.22 (t, 2H), 3.96 (s, 3H), 3.59 (t, 4H), 2.47 (m, 2H), 2.39 (m, 4H), 2.02 (s, 3H), 1.99 (m, 2H) . LRMS (ESI pos) m/e 612 (M+l). [00451] Example 62 Preparation of 5-(3-fluoro-4-(6-methoxy-7-(3- morpholinopropoxy)quinolin-4-yloxy)phenyl)-3-methyl-2-(phenylamino)pyrimidin-4(3H)-one 162 111-03-PCT - P2338R1 - 02120.004WO1
118
Figure imgf000119_0001
[00452] Step A: Preparation of 4-methoxy-N-phenylpyrimidin-2-amine: In a sealed tube was 2-chloro-4-methoxypyrimidine (1.00 g, 6.92 mmol) in 2-propanol (5 mL). Aniline (0.757 ml, 8.30 mmol) and DIEA (1.45 ml, 8.30 mmol) were added and the reaction mixture was heated at 100°C until the reaction was complete by HPLC. The reaction mixture was cooled to room temperature. The resulting thick suspension was filtered, washed with ethanol, collected and dried under vacuum to yield the desired product (0.164 g) as a white solid. The filtrate was concentrated and then partitioned between EtOAc and saturated aqueous NaCl. The phases were separated, and the aqueous phase was re-extracted with EtOAc (Ix). The combined organic layers were dried (Na2SO4), filtered and concentrated to yield a yellow solid. The crude product was purified by silica gel flash column chromatography, eluting with 25:1 dichloromethane/EtOAc. The desired product (0.548 g) was obtained as a white solid which was combined with the filtered product to yield 0.712 g (51%) total desired product. 1H-NMR (400 MHz, DMSO-d6) δ 9.51 (s, IH), 8.20 (d, IH), 7.77 (d, 2H), 7.27 (t, 2H), 6.94 (t, IH), 6.28 (d, IH), 3.91 (s, 3H). LRMS (ESI pos) m/e 202 (M+l).
[00453] Step B: Preparation of 2-(phenylamino)pyrimidin-4(3H)-one: To a solution of 4- methoxy-N-phenylpyrimidin-2 -amine (0.632 g, 3.14 mmol) in acetic acid (20 mL) was added HBr (2.132 ml, 18.84 mmol; 48 wt % in H2O). The reaction mixture was heated at 90-95°C for 3 hours. The reaction mixture was cooled to room temperature and diluted with H2O. The pH of the reaction mixture was adjusted to 5-6 with 6 M aqueous NaOH which resulted in the formation of a solid precipitate. The solid was filtered, washed with H2O, collected and dried under vacuum to yield the desired product (0.553 g, 94%) as a white solid. 1H-NMR (400 MHz, DMSO-d6) δ 10.74 (br s, IH), 8.81 (br s, IH), 7.76 (s, IH), 7.60 (d, 2H), 7.31 (t, 2H), 7.02 (t, IH), 5.81 (s, IH). LRMS (ESI pos) m/e 188 (M+l).
[00454] Step C: Preparation of 3-methyl-2-(phenylamino)pyrimidin-4(3H)-one. To a solution of 2-(phenylamino)pyrimidin-4(3H)-one (0.250 g, 1.34 mmol) in DMF (10 mL) was added LiH (0.012 g, 1.47 mmol). The reaction mixture was stirred for 25 minutes and then iodomethane (0.166 ml, 2.67 mmol) was added. The reaction was stirred at room temperature for 18 hours. The reaction mixture was quenched with H2O and then partitioned between EtOAc and 111-03-PCT - P2338R1 - 02120.004WO1
119 saturated aqueous NaCl. The phases were separated, and the aqueous phase was re-extracted with EtOAc (Ix). The combined organic layers were dried (Na2SO4), filtered and concentrated to yield a crude yellow oil. The crude product was purified by silica gel flash column chromatography, eluting with 30:1 dichloromethane/methanol. The desired product (0.166 g, 62%) was obtained as a white crystalline solid. 1H-NMR (400 MHz, CDCl3) δ 7.68 (d, IH), 7.46 (m, 2H), 7.39 (t, 2H), 7.19 (t, IH), 6.48 (s, IH), 6.01 (d, IH), 3.58 (s, 3H). LRMS (ESI pos) m/e 202 (M+l).
[00455] Step D: Preparation of 5-bromo-3-methyl-2-(phenylamino)pyrimidin-4(3H)-one:
To a solution of 3-methyl-2-(phenylamino)pyrimidin-4(3H)-one (0.104 g, 0.517 mmol) in CHCl3 (5 mL)/MeOH (1 niL) at 00C was added bromine (0.027 ml, 0.517 mmol). The reaction mixture was stirred for 30 minutes at room temperature and then quenched with 10% aqueous sodium bisulfite solution. The reaction mixture was partitioned between EtOAc and H2O. The phases were separated, and the aqueous phase was re-extracted with EtOAc (Ix). The combined organic layers were dried (Na2SO4), filtered and concentrated to yield the desired product (0.145 g; 100%) as a white solid that was used without further purification. 1H-NMR (400 MHz, DMSO- d6) δ 8.95 (s, IH), 7.94 (s, IH), 7.47 (m, 2H), 7.34 (t, 2H), 7.14 (t, IH), 3.53 (s, 3H). LRMS (ESI pos) m/e 280, 282 (M+l, Br pattern).
[00456] Step E. Preparation of 5-(4-(benzyloxy)-3-fluorophenyl)-3-methyl-2-
(phenylamino)pyrimidin-4(3H)-one: A suspension of 5-bromo-3-methyl-2-
(phenylamino)pyrimidin-4(3H)-one (0.145 g, 0.518 mmol), 4-(benzyloxy)-3-fluorophenylboronic acid (0.153 g, 0.621 mmol), Pd(PPh3)4 (0.030 g, 0.026 mmol) and lithium chloride (0.110 g, 2.59 mmol) in dioxane (1.5 mL) and 2 M aqueous Na2CO3 (1.5 mL) was stirred at 100°C for 20 minutes. The reaction mixture was cooled to room temperature and then partitioned between EtOAc and H2O. The phases were separated, and the aqueous phase was re-extracted with EtOAc (Ix). The combined organic layers were dried (Na2SO4), filtered and concentrated to yield a crude black solid. The crude product was purified by silica gel flash column chromatography, eluting with 10:1 dichloromethane/EtOAc. The desired product (0.133 g, 64%) was obtained as a grey waxy solid. 1H-NMR (400 MHz, DMSO-d6) δ 8.90 (br s, IH), 7.93 (s, IH), 7.59 (dd, IH), 7.55-7.31 (m, 10H), 7.22 (t, IH), 7.14 (t, IH), 5.20 (s, 2H), 3.55 (s, 3H). LRMS (ESI pos) m/e 402 (M+l).
[00457] Step F: Preparation of 5-(3-fluoro-4-hydroxyphenyl)-3-methyl-2-
(phenylamino)pyrimidin-4(3H)-one: A solution of 5-(4-(benzyloxy)-3-fluorophenyl)-3-methyl- 2-(phenylamino)pyrimidin-4(3H)-one (0.133 g, 0.331 mmol) in TFA (1.5 mL) was stirred at 400C for 3.5 hours The reaction mixture was concentrated to dryness and then purified by silica 111-03-PCT - P2338R1 - 02120.004WO1
120 gel flash column chromatography, eluting with 20:1 dichloromethane/MeOH. The desired product (0.103 g, 100%) was obtained as a foamy white solid. 1H-NMR (400 MHz, DMSOd6) δ 9.81 (br s, IH), 8.96 (br s, IH), 7.86 (s, IH), 7.56-7.45 (m, 3H), 7.37 (t, 2H), 7.27 (m, IH), 7.15 (t, IH), 6.92 (t, IH), 3.54 (s, 3H). LRMS (APCI pos) m/e 312 (M+l).
[00458] Step G. Preparation of 5-(3-fluoro-4-(6-methoxy-7-(3- morpholinopropoxy)quinolin-4-yloxy)phenyl)-3-methyl-2-(phenylamino)pyrimidin-4(3H)-one: Prepared from 5-(3-fluoro-4-hydroxyphenyl)-3-methyl-2-(phenylamino)pyrimidin-4(3H)-one (0.025 g, 0.0803 mmol), 4-(3-(4-chloro-6-methoxyquinolin-7-yloxy)propyl)morpholine (0.0298 g, 0.0883 mmol) and catalytic DMAP according to the procedure described in Step C of Example 58, to give 162 (0.029, 59%) as a pale yellow foamy solid. 1H-NMR (400 MHz, DMSOd6) 9.02 (br s, IH), 8.49 (d, IH). 8.10 (s, IH), 7.88 (dd, IH), 7.68 (m, IH), 7.57-7.51 (m, 3H), 7.47-7.35 (m, 4H), 7.16 (t, IH), 6.49 (dd, IH), 4.21 (t, 2H), 3.95 (s, 3H), 3.62-3.56 (m, 7H), 2.47 (m, 2H), 2.39 (m, 4H), 1.99 (m, 2H). LRMS (APCI pos) m/e 612 (M+l).
[00459] Example 63 Preparation of 2-(cyclopropylmethylamino)-5-(3-fluoro-4-(6- methoxy-7-(3-morpholinopropoxy)quinolin-4-yloxy)phenyl)-3-methylpyrimidin-4(3H)-one 163
Figure imgf000121_0001
[00460] Step A: Preparation of 5-bromo-2-(cyclopropylmethylamino)-3- methylpyrimidin-4(3H)-one: A mixture of 5-bromo-2-chloro-3-rnethylpyrimidin-4(3H)-one (0.100 g, 0.45 mmol; obtained from Example 59, Step B), cyclopropylmethanamine (0.051 ml, 0.58 mmol) and NaHCO3 (0.150 g, 1.79 mmol) in n-BuOH (3 mL) was stirred at 600C for 1 hour. The reaction mixture was cooled to room temperature and then diluted with EtOAc. The EtOAc layer was washed with H2O and saturated aqueous NaCl. The aqueous phase was re-extracted with EtOAc (Ix). The combined EtOAc layers were dried (Na2SO4), filtered and concentrated to yield the desired product (0.114 g, 98%) as a pale yellow solid that was used without further purification. 1H-NMR (400 MHz, DMSO-d6) δ 7.93 (s, IH), 7.46 (t, IH), 3.33 (s, 3H), 3.19 (t, 2H), 1.12 (m, IH), 0.43 (m, 2H), 0.24 (m, 2H). LRMS (ESI pos) m/e 258, 260 (M+, Br pattern). [00461] Step B: Preparation of 5-(4-(benzyloxy)-3-fluorophenyl)-2-
(cyclopropylmethylamino)-3-methylpyrimidin-4(3H)-one: A suspension of 5-bromo-2- (cyclopropylmethylamino)-3-methylpyrimidin-4(3H)-one (0.112 g, 0.434 mmol), 4-(benzyloxy)- 111-03-PCT- P2338R1 - 02120.004WO1
121
3-fluorophenylboronic acid (0.128 g, 0.521 mmol), Pd(PPh3)4 (0.025 g, 0.022 mmol) and lithium chloride (0.092 g, 2.17 mmol) in dioxane (1.5 mL) and 2 M aqueous Na2CO3 (1.5 mL) was stirred at 100°C for 30 minutes. The reaction mixture was cooled to room temperature and then partitioned between EtOAc and H2O. The phases were separated, and the aqueous phase was re- extracted with EtOAc (Ix). The combined organic layers were dried (Na2SO4), filtered and concentrated to yield a crude black solid. The crude product was purified by silica gel flash column chromatography, eluting with 10:1 dichloromethane/EtOAc. The desired product (0.128 g, 78%) was obtained as a foamy off-white solid. 1H-NMR (400 MHz, DMSO-d6) δ 7.93 (s, IH), 7.57 (dd, IH), 7.49-7.31 (m, 7H), 7.19 (t, IH), 5.19 (s, 2H), 3.35 (s, 3H), 3.24 (t, 2H), 1.16 (m, IH), 0.44 (m, 2H), 0.25 (m, IH). LRMS (APCI pos) m/e 380 (M+l).
[00462] Step C. Preparation of 2-(cyclopropylmethylamino)-5-(3-fluoro-4- hydroxyphenyl)-3-methylpyrimidin-4(3H)-one: A solution of 5-(4-(benzyloxy)-3-fluorophenyl)- 2-(cyclopropylmethylamino)-3-methylpyrimidin-4(3H)-one (0.128 g, 0.337 mmol) in TFA (2 mL) was stirred at 40°C for 2 hours and 45 minutes. The reaction mixture was concentrated to dryness and then purified by silica gel flash column chromatography, eluting with 20:1 dichloromethane/MeOH. The desired product (0.080 g, 82%) was obtained as a colorless glassy solid. 1H-NMR (400 MHz, DMSO-d6) δ 9.71 (s, IH), 7.87 (s, IH), 7.46 (dd, IH), 7.35 (t, IH), 7.24 (dd, IH), 6.90 (dd, IH), 3.34 (s, 3H), 3.24 (t, 2H), 1.16 (m, IH), 0.44 (m, 2H), 0.26 (m, 2H). LRMS (ESI pos) m/e 290 (M+l).
[00463] Step D. Preparation of 2-(cyclopropylmethylamino)-5-(3-fluoro-4-(6-methoxy-7-
(3-morpholinopropoxy)quinolin-4-yloxy)phenyl)-3-methylpyrimidin-4(3H)-one: Prepared from 2-(cyclopropylmethylamino)-5-(3-fluoro-4-hydroxyphenyl)-3-methylpyrimidin-4(3H)-one (0.025 g, 0.0864 mmol), 4-(3-(4-chloro-6-methoxyquinolin-7-yloxy)propyl)morpholine (0.0320 g, 0.0951 mmol) and catalytic DMAP according to the procedure described in Step C of Example 58, to give 163 (0.030, 60%) as a pale yellow solid. 1H-NMR (400 MHz, DMSO-d6) δ 8.48 (d, IH), 8.11 (s, IH), 7.85 (dd, IH), 7.66 (m, IH), 7.56-7.50 (m, 2H), 7.44-7.39 (m, 2H), 6.48 (dd, IH), 4.21 (t, 2H), 3.96 (s, 3H), 3.59 (t, 4H), 3.38 (s, 3H), 3.28 (m, 2H), 2.47 (m, 2H), 2.39 (m, 4H), 1.98 (m, 2H), 1.18 (m, IH), 0.46 (m, 2H), 0.28 (m, 2H). LRMS (APCI pos) m/e 590 (M+l). [00464] Example 64 Preparation of 5-(3-fluoro-4-(6-methoxy-7-(3- moφholinopropoxy)quinolin-4-yloxy)phenyl)-2-(4-fluorophenylamino)-3-methylpyrimidin- 4(3H)-one 164 111-03-PCT - P2338R1 - 02120.004WO1
122
Figure imgf000123_0001
[00465] Step A: Preparation of 5-bromo-2-(4-fluorophenylamino)-3-methylpyrimidin-
4(3H)-one: Prepared from 5-bromo-2-chloro-3-methylpyrimidin-4(3H)-one (0.100 g, 0.45 mmol; obtained from Example 59, Step B), according to the procedure described in Step A of Example 63, substituting 4-fluorobenzenamine in place of cyclopropylmethanamine. The desired product (0.132, 99%) was obtained as a pale yellow solid. 1H-NMR (400 MHz, DMSOd6) δ 8.97 (br s , IH), 7.93 (s, IH),
7.47 (m, 2H), 7.19 (m, 2H), 3.51 (s, 3H). LRMS (ESI pos) m/e 298, 300 (M+, Br pattern). [00466] Step B: Preparation of 5-(4-(benzyloxy)-3-fluorophenyl)-2-(4- fluorophenylamino)-3-methylpyrimidin-4(3H)-one: Prepared from 5-bromo-2-(4- fluorophenylamino)-3-methylpyrimidin-4(3H)-one (0.130 g, 0.436 mmol) according to the procedure described in Step B of Example 63. The desired product (0.139 g, 76%) was obtained as a grey/white solid. 1H-NMR (400 MHz, DMSO-d6) δ 8.93 (br s, IH), 7.92 (s, IH), 7.58 (dd, IH), 7.52 (m, 2H), 7.49-7.44 (m, 2H), 7.44-7.39 (m, 3H), 7.37-7.31 (m, IH), 7.24-7.16 (m, 3H), 5.19 (s, 2H), 3.53 (s, 3H). LRMS (ESI pos) m/e 420 (M+l).
[00467] Step C. Preparation of 5-(3-fluoro-4-hydroxyphenyl)-2-(4-fluorophenylamino)-3- methylpyrimidin-4(3H)-one: Prepared from 5-(4-(benzyloxy)-3-fluorophenyl)-2-(4- fluorophenylamino)-3-methylpyrimidin-4(3H)-one (0.139 g, 0.331 mmol) according to the procedure described in Step C of Example 63. The desired product (0.089 g, 82%) was obtained as a white solid. 1H-NMR (400 MHz, DMSO-d6) δ 9.78 (s, IH), 8.89 (s, IH), 7.86 (s, IH), 7.56- 7.46 (m, 3H), 7.27 (m, IH), 7.19 (m, 2H), 6.92 (dd, IH), 3.53 (s, 3H). LRMS (ESI pos) m/e 330 (M+l).
[00468] Step D. Preparation of 5-(3-fluoro-4-(6-methoxy-7-(3- moφholinopropoxy)quinolin-4-yloxy)phenyl)-2-(4-fluorophenylamino)-3-methylpyrimidin- 4(3H)-one: Prepared from 5-(3-fluoro-4-hydroxyphenyl)-2-(4-fluorophenylamino)-3- methylpyrimidin-4(3H)-one (0.025 g, 0.0759 mmol), 4-(3-(4-chloro-6-methoxyquinolin-7- yloxy)propyl)morpholine (0.0281 g, 0.0835 mmol) and catalytic DMAP according to the procedure described in Step C of Example 58, to give 164 (0.026 g, 54%) as a pale yellow solid. 1H-NMR (400 MHz, DMSO-d6) δ 9.05 (br s, IH), 8.49 (d, IH), 8.09 (s, IH), 7.87 (dd, IH), 7.67 111-03-PCT- P2338R1 - 02120.004WO1
123
(m, IH), 7.58-7.50 (m, 3H), 7.44 (t, IH), 7.41 (s, IH), 7.22 (m, 2H), 6.49 (dd, IH), 4.21 (t, 2H), 3.95 (s, 3H), 3.59 (t, 4H), 3.57 (s, 3H), 2.47 (m, 2H), 2.39 (m, 4H), 1.98 (m, 2H). LRMS (APCI pos) m/e 630 (M+l).
[00469] Example 65 Preparation of 3-ethyl-5-(3-fluoro-4-(6-methoxy-7-(3- morpholinopropoxy)quinolin-4-yloxy)phenyl)-2-(phenylamino)pyrimidin-4(3H)-one 165
Figure imgf000124_0001
[00470] Step A: Preparation of 5-bromo-2-chloro-3-ethylpyrimidin-4(3H)-one: Prepared from 5-bromo-2-chloropyrimidin-4(3H)-one (1.00 g, 4.775 mmol; obtained from Example 59, Step A) according to the procedure described in Step B of Example 59, substituting iodoethane in place of iodomethane. The desired product (0.411 g, 36%) was obtained as a yellow crystalline waxy solid. 1H-NMR (400 MHz, DMSO-d6) δ 8.26 (s, IH), 4.16 (q, 3H), 1.25 (t, 4H). LRMS (ESI pos) m/e 237, 239 (M+, Br pattern).
[00471] Step B: Preparation of 5-bromo-3-ethyl-2-(phenylamino)pyrimidin-4(3H)-one:
Prepared from 5-bromo-2-chloro-3-ethylpyrimidin-4(3H)-one (0.075 g, 0.316 mmol) according to the procedure described in Step A of Example 63, substituting aniline in place of cyclopropylmethanamine. The desired product (0.091 g, 98%) was obtained as a yellow solid. 1H-NMR (400 MHz, DMSOd6) δ 9.00 (br s, IH), 7.91 (s, IH), 7.46-7.41 (m, 2H), 7.39-7.32 (m, 2H), 7.16 (m, IH), 4.19 (q, 2H), 1.23 (t, 3H). LRMS (APCI pos) m/e 294, 296 (M+, Br pattern). [00472] Step C: Preparation of 5-(4-(benzyloxy)-3-fluorophenyl)-3-ethyl-2-
(phenylamino)pyrimidin-4(3H)-one: Prepared from 5-bromo-3-ethyl-2-(phenylamino)pyrimidin- 4(3H)-one (0.086 g, 0.292 mmol) according to the procedure described in Step B of Example 63. The desired product (0.073 g, 60%) was obtained as a white foamy solid. 1H-NMR (400 MHz, DMSO-d6) δ 8.94 (br s, IH), 7.90 (s, IH), 7.59 (dd, IH), 7.52-7.31 (m, 9H), 7.21 (t, IH), 7.16 (t, IH), 5.20 (s, 2H), 4.22 (q, 2H), 1.25 (t, 3H). LRMS (APCI pos) m/e 416 (M+l). [00473] Step D: Preparation of 3-ethyl-5-(3-fluoro-4-hydroxyphenyl)-2-
(phenylamino)pyrimidin-4(3H)-one: Prepared from 5-(4-(benzyloxy)-3-fluorophenyl)-3-ethyl-2- (phenylamino)pyrimidin-4(3H)-one (0.072 g, 0.17 mmol) according to the procedure described in Step C of Example 63. The desired product (0.056 g, 100%) was obtained as a white solid. 1H- NMR (400 MHz, DMSOd6) δ 9.80 (br s, IH), 8.98 (br s, IH), 7.82 (s, IH), 7.54-7.42 (m, 3H), 111-O3-PCT - P2338R1 - 02120.004WO1
124
7.37 (m, 2H), 7.27 (dd, IH), 7.16 (t, IH), 6.92 (dd, IH), 4.22 (q, 2H), 1.25 (t, 3H). LRMS (APCI pos) m/e 326 (M+l).
[00474] Step E: Preparation of 3-ethyl-5-(3-fluoro-4-(6-methoxy-7-(3- morpholinopropoxy)quinolin-4-yloxy)phenyl)-2-(phenylamino)pyrimidin-4(3H)-one: Prepared from 3-ethyl-5-(3-fluoro-4-hydroxyphenyl)-2-(phenylamino)pyrimidin-4(3H)-one (0.025 g, 0.0768 mmol), 4-(3-(4-chloro-6-methoxyquinolin-7-yloxy)propyl)moφholine (0.0285 g, 0.0845 mmol) and catalytic DMAP according to the procedure described in Step C of Example 58, to give 165 (0.016 g, 33%) as a pale yellow solid. 1H-NMR (400 MHz, DMSOd6) δ 9.07 (br s, IH), 8.49 (d, IH), 8.07 (s, IH), 7.88 (dd, IH), 7.68 (m, IH), 7.55-7.35 (m, 7H), 7.19 (m, IH), 6.49 (dd, IH), 4.30-4.18 (m, 4H), 3.95 (s, 3H), 3.60 (t, 4H), 2.49 (m, 2H), 2.40 (m, 4H), 1.99 (m, 2H), 1.29 (t, 3H). LRMS (APCI pos) m/e 626 (M+l).
[00475] Example 66 Preparation of 5-(3-fluoro-4-(6-methoxy-7-(3- morpholinopropoxy)quinolin-4-yloxy)phenyl)-3-methyl-2-phenoxypyrimidin-4(3H)-one 166
Figure imgf000125_0001
[00476] Step A: Preparation of 5-bromo-3-methyl-2-phenoxypyrimidin-4(3H)-one:
Prepared from 5-brorno-2-chloro-3-methylpyrimidin-4(3H)-one (0.075 g, 0.336 mmol; obtained from Example 59, Step B), according to the procedure described in Step A of Example 63, substituting phenol in place of cyclopropylmethanamine. The desired product (0.058, 62%) was obtained as a white solid. 1H-NMR (400 MHz, DMSO-d6) δ 8.02 (s, IH), 7.49-7.43 (m, 2H), 7.34-7.27 (m, 3H), 3.54 (s, 3H). LRMS (ESI pos) m/e 281, 283 (M+, Br pattern). [00477] Step B: Preparation of 5-(4-(benzyloxy)-3-fluorophenyl)-3-methyl-2- phenoxypyrimidin-4(3H)-one: Prepared from 5-bromo-3-methyl-2-phenoxypyrimidin-4(3H)-one (0.056 g, 0.199 mmol) according to the procedure described in Step B of Example 63. The desired product (0.079 g, 99%) was obtained as a white/grey solid. 1H-NMR (400 MHz, DMSO- d6) δ 7.90 (s, IH), 7.57 (dd, IH), 7.51-7.29 (m, HH), 7.26 (t, IH), 5.21 (s, 2H), 3.57 (s, 3H). LRMS (ESI pos) m/e 403 (M+l).
[00478] Step C: Preparation of 5-(3-fluoro-4-hydroxyphenyl)-3-methyl-2- phenoxypyrimidin-4(3H)-one: Prepared from 5-(4-(benzyloxy)-3-fluorophenyl)-3-methyl-2- phenoxypyrimidin-4(3H)-one (0.078 g, 0.19 mmol) according to the procedure described in Step 111-03-PCT- P2338R1 - 02120.004WO1
125
C of Example 63. The desired product (0.065g, 82%) was obtained as a pale yellow foamy solid. 1H-NMR (400 MHz, DMSOd6) δ 9.94 (br s, IH), 7.85 (s, IH), 7.52-7.44 (m, 3H), 7.35-7.26 (m, 4H), 6.95 (dd, IH), 3.56 (s, 3H). LRMS (APCI pos) m/e 313 (M+l).
[00479] Step D: Preparation of 5-(3-fluoro-4-(6-methoxy-7-(3- morpholinopropoxy)quinolin-4-yloxy)phenyl)-3-methyl-2-phenoxypyrimidin-4(3H)-one: Prepared from 5-(3-fluoro-4-hydroxyphenyl)-3-methyl-2-phenoxypyrimidin-4(3H)-one (0.029 g, 0.086 mmol), 4-(3-(4-chloro-6-methoxyquinolin-7-yloxy)propyl)morpholine (0.0247 g, 0.0733 mmol) and catalytic DMAP according to the procedure described in Step C of Example 58, to give 166 (0.03Og, 66%) as a pale yellow foamy solid. 1H-NMR (400 MHz, DMSO-d6) δ 8.49 (d, IH), 8.07 (s, IH), 7.84 (dd, IH), 7.67 (m, IH), 7.54-7.46 (m, 4H), 7.41 (s, IH), 7.37-7.31 (m, 3H), 6.51 (dd, IH), 4.21 (t, 2H), 3.95 (s, 3H), 3.60 (s, 3H), 3.59 (m, 4H), 2.47 (m, 2H), 2.39 (m, 4H), 1.98 (m, 2H). LRMS (APCI pos) m/e 613 (M+l).
[00480] Example 67 Preparation of 5-(3-fluoro-4-(6-methoxy-7-(3- morpholinopropoxy)quinolin-4-yloxy)phenyl)-3-methyl-2-(methyl(phenyl)amino)pyrimidin- 4(3H)-one 167
Figure imgf000126_0001
[00481] Step A: Preparation of 5-bromo-3-methyl-2-(methyl(phenyl)amino)pyrimidin-
4(3H)-one: Prepared from 5-bromo-2-chloro-3-methylpyrimidin-4(3H)-one (0.100 g, 0.448 mmol; obtained from Example 59, Step B), according to the procedure described in Step A of Example 63, substituting N-methylaniline in place of cyclopropylmethanamine. The desired product (0.085, 65%) was obtained as a white solid. 1H-NMR (400 MHz, DMSOd6) δ 8.23 (s, IH), 7.42-7.36 (m, 2H), 7.20 (m, IH), 7.15-7.10 (m, 2H), 3.32 (s, 3H), 2.92 (s, 3H). LRMS (ESI pos) m/e 294, 296 (M+, Br pattern).
[00482] Step B: 5-(4-(benzyloxy)-3-fluorophenyl)-3-methyl-2-
(methyl(phenyl)amino)pyrimidin-4(3H)-one: Prepared from 5-bromo-3-methyl-2-
(methyl(phenyl)amino)pyrimidin-4(3H)-one (0.083 g, 0.282 mmol) according to the procedure described in Step B of Example 63. The desired product (0.109 g, 93%) was obtained as a pale yellow solid. 1H-NMR (400 MHz, DMSOd6) δ 8.17 (s, IH), 7.66 (dd, IH), 7.53-7.32 (m, 8H), 111-03-PCT - P2338R1 - 02120.004WO1
126
7.27 (t, IH), 7.19 (m, IH), 7.12-7.08 (m, 2H), 5.22 (s, 2H), 3.36 (s, 3H), 2.96 (s, 3H). LRMS (APCI pos) m/e 416 (M+l).
[00483] Step C: Preparation of 5-(3-fluoro-4-hydroxyphenyl)-3-methyl-2-
(methyl(phenyl)amino)pyrimidin-4(3H)-one: Prepared from 5-(4-(benzyloxy)-3-fluorophenyl)-3- methyl-2-(methyl(phenyl)amino)pyrimidin-4(3H)-one (0.109 g, 0.262 mmol) according to the procedure described in Step C of Example 63. The desired product (0.082g, 71%) was obtained as a pale yellow foamy solid. 1H-NMR (400 MHz, DMSO-d6) δ 9.92 (br s, IH), 8.12 (s, IH), 7.58 (dd, IH), 7.44-7.36 (m, 3H), 7.18 (m, IH), 7.09 (m, 2H), 6.97 (dd, IH), 3.35 (s, 3H), 2.97 (s, 3H). LRMS (ESI pos) m/e 326 (M+l).
[00484] Step D: Preparation of 5-(3-fluoro-4-(6-methoxy-7-(3- morpholinopropoxy)quinolin-4-yloxy)phenyl)-3-methyl-2-(methyl(phenyl)amino)pyrimidin- 4(3H)-one: Prepared from 5-(3-fluoro-4-hydroxyphenyl)-3-methyl-2-
(methyl(phenyl)amino)pyrimidin-4(3H)-one (0.025 g, 0.078 mmol), 4-(3-(4-chloro-6- methoxyquinolin-7-yloxy)propyl)morpholine (0.025 g, 0.074 mmol) and catalytic DMAP according to the procedure described in Step C of Example 58, to give 167 (0.021, 45%) as a pale yellow foamy solid. 1H-NMR (400 MHz, DMSOd6) 8.50 (d, IH). 8.33 (s, IH), 7.94 (dd, IH), 7.76 (m, IH), 7.55 (s, IH), 7.50 (t, IH), 7.46-7.40 (m, 3H), 7.22 (m, IH), 7.18-7.33 (m, 2H), 6.51 (dd, IH), 4.21 (t, 2H), 3.96 (s, 3H), 3.59 (t, 4H), 3.40 (s, 3H), 2.98 (s, 3H), 2.48 (m, 2H), 2.40 (m, 4H), 1.99 (m, 2H). LRMS (APCI pos) m/e 626 (M+l).
[00485] Example 68 Preparation of 6-benzyl-3-(3-fluoro-4-(6-methoxy-7-(3- morpholinopropoxy)quinolin-4-yloxy)phenyl)-l-methylpyridin-2(lH)-one 168
Figure imgf000127_0001
[00486] Step A: Preparation of 6-chloro-l-methylpyridin-2(lH)-one: To a solution of 6- chloropyridin-2-ol (10.00 g, 77.19 mmol) in acetone (350 mL) was added K2CO3 (37.34 g, 270.2 mmol) and iodomethane (17.37 ml, 270.2 mmol). The reaction mixture was stirred at room temperature for 1 hour and then at reflux for 16 hours. The reaction mixture was cooled to room temperature and the K2CO3 was filtered off and washed with acetone. The filtrate was then concentrated and the residue was partitioned between H2O and CH2Cl2. The phases were separated, and the aqueous phase was re-extracted with CH2Cl2 (Ix). The combined CH2Cl2 layers were dried (Na2SO4), filtered and concentrated to yield a yellow oil. The crude product 111-03-PCT - P2338R1 - 02120.004WO1
127 was purified by silica gel flash column chromatography, eluting with 10:1 CH2Cl2/EtOAc. The desired product (7.38 g, 67%) was obtained as a white solid. 1H-NMR (400 MHz, CDCl3) δ 7.23 (dd, IH), 6.50 (dd, IH), 6.30 (m, IH), 3.69 (s, 3H). LRMS (ESI pos) m/e 144 (M+l). [00487] Step B: Preparation of 6-benzyl-l-methylpyridin-2(lH)-one: A mixture of 6- chloro-l-methylpyridin-2(lH)-one (0.200 g, 1.39 mmol) and PdCl2(PPh3)2 (0.049 g, 0.070 mmol) in THF (8 niL) was sparged with N2. Benzylzinc (II) bromide (3.06 ml, 1.53 mmol; 0.5 M solution in THF) was added and the reaction mixture was stirred at reflux for 1 hour and then at room temperature for 16 hours. The reaction mixture partitioned between H2O and EtOAc. The phases were separated, and the aqueous phase was re-extracted with EtOAc (Ix). The combined EtOAc layers were dried (Na2SO4), filtered and concentrated to yield a yellow oil. The crude product was purified by silica gel flash column chromatography, eluting with 20:1 CH2Cl2MeOH. The desired product (0.144 g, 52%) was obtained as a yellow oil that crystallized to a waxy solid under vacuum. 1H-NMR (400 MHz, CDCl3) δ 7.37-7.31 (m, 3H), 7.30-7.23 (m, IH), 7.16-7.11 (m, 2H), 6.53 (dd, IH), 5.99 (m, IH), 3.98 (s, 2H), 3.43 (s, 3H). LRMS (APCI pos) m/e 200 (M+l).
[00488] Step C: Preparation of 6-benzyl-3-bromo-l-methylpyridin-2(lH)-one: To a solution of 6-benzyl-l-methylpyridin-2(lH)-one (0.144 g, 0.723 mmol) in CHCl3 (5 mL) was added Br2 (0.037 ml, 0.72 mmol). The reaction mixture was stirred at room temperature for 2 hours and then quenched with 10% sodium bisulfite solution. The reaction mixture was partitioned between EtOAc and H2O. The phases were separated, and the aqueous phase was re- extracted with EtOAc (Ix). The combined organic layers were dried (Na2SO4), filtered and concentrated to yield a yellow oil. The crude product was purified by silica gel flash column chromatography, eluting with 20:1 CH2Cl2/Et0Ac. The desired product (0.087 g, 43%) was obtained as a yellow gum. 1H-NMR (400 MHz, CDCl3) δ 7.38-7.31 (m, 3H), 7.31-7.28 (m, IH), 7.14-7.10 (m, 2H), 5.91 (d, IH), 3.96 (s, 2H), 3.50 (s, 3H). LRMS (APCI pos) m/e 278, 280 (M+, Br pattern).
[00489] Step D: Preparation of 6-benzyl-3-(4-(benzyloxy)-3-fluorophenyl)-l- methylpyridin-2(lH)-one: Prepared from 6-benzyl-3-bromo-l-methylpyridin-2(lH)-one (0.087 g, 0.313 mmol) according to the procedure described in Step B of Example 63. The desired product (0.071 g, 57%) was obtained as yellow gum that crystallized to a waxy solid under vacuum. 1H-NMR (400 MHz, DMSO-d6) δ 7.55 (dd, IH), 7.48-7.27 (m, 10H), 7.17 (m, 2H), 7.00 (t, IH), 6.08 (d, IH), 5.17 (s, 2H), 4.01 (s, 2H), 3.49 (s, 3H). LRMS (ESI pos) m/e 400 (M+l). 111-O3-PCT - P2338R1 - 02120.004WO1
128
[00490] Step E: 6-benzyl-3-(3-fluoro-4-hydroxyphenyl)-l-methylpyridin-2(lH)-one:
Prepared from 6-benzyl-3-(4-(benzyloxy)-3-fluorophenyl)-l-methylpyridin-2(lH)-one (0.071 g, 0.18 mmol) according to the procedure described in Step C of Example 63. The desired product (0.047g, 85%) was obtained as a pale yellow foamy solid. 1H-NMR (400 MHz, DMSOd6) δ 9.87 (s, IH), 7.60 (dd, IH), 7.56 (d, IH), 7.40-7.33 (m, 3H), 7.28 (m, IH), 7.26-7.21 (m, 2H), 6.93 (dd, IH), 6.08 (d, IH), 4.12 (s, 2H), 3.42 (s, 3H). LRMS (ESI pos) m/e 310 (M+l). [00491] Step F: Preparation of 6-benzyl-3-(3-fluoro-4-(6-methoxy-7-(3- moφholinopropoxy)quinolin-4-yloxy)phenyl)-l-methylpyridin-2(lH)-one: Prepared from 6- benzyl-3-(3-fluoro-4-hydroxyphenyl)-l-methylpyridin-2(lH)-one (0.027 g, 0.087 mmol), 4-(3-(4- chloro-6-rnethoxyquinolin-7-yloxy)propyl)morpholine (0.032 g, 0.096 mmol) and catalytic DMAP according to the procedure described in Step C of Example 58, to give 168 (0.037 g, 70%) as a pale yellow foamy solid. 1H-NMR (400 MHz, DMSOd6) δ 8.49 (d, IH), 7.95 (dd, IH), 7.77 (d, IH), 7.72 (m, IH), 7.54 (s, IH), 7.46 (t, IH), 7.43-7.36 (m, 3H), 7.33-7.24 (m, 3H), 6.50 (dd, IH), 6.16 (d, IH), 4.21 (t, 2H), 4.17 (s, 2H), 3.96 (s, 3H), 3.59 (t, 4H), 3.47 (s, 3H), 2.47 (m, 2H), 2.39 (m, 4H), 1.98 (m, 2H). LRMS (ESI pos) m/e 610 (M+l).
[00492] Example 69 Preparation of 3-(3-fluoro-4-(6-methoxy-7-(3- morpholinopropoxy)quinolin-4-yloxy)phenyl)- 1 -methyl-6-(phenylamino)pyridin-2( 1 H)-one 169
Figure imgf000129_0001
[00493] Step A: Preparation of 3-bromo-6-chloro-l-methylpyridin-2(lH)-one: To a solution of 6-chloro-l-methylpyridin-2(lH)-one (0.500 g, 3.48 mmol; obtained from Example 68, Step A) in DMF (15 mL) was added N-bromosuccinimide (0.620 g, 3.48 mmol). The reaction was stirred at room temperature for 2 hours and then quenched with 10% sodium bisulfite solution. The reaction mixture was partitioned between EtOAc and H2O. The phases were separated, and the aqueous phase was re-extracted with EtOAc (Ix). The combined organic layers were dried (Na2SO4), filtered and concentrated to yield a yellow oil. The crude product was purified by silica gel flash column chromatography, eluting with 20:1 CH2Cl2ZEtOAc. The desired product (0.424 g, 55%) was obtained as a white crystalline solid. 1H-NMR (400 MHz, DMSO-d6) δ 7.90 (d, IH), 6.48 (d, IH), 3.63 (s, 3H). LRMS (ESI pos) m/e 222, 224 (M+, Br pattern). Also isolated was 5-bromo-6-chloro-l-methylpyridin-2(lH)-one (0.233 g, 30%) as a 111-03-PCT - P2338R1 - 02120.004WO1
129 white crystalline solid. 1H-NMR (400 MHz, DMSOd6) δ 7.68 (d, IH), 6.42 (d, IH), 3.61 (s, 3H).
[00494] Step B: Preparation of 3-(4-(benzyloxy)-3-fluorophenyl)-6-chloro-l- methylpyridin-2(lH)-one: Prepared from 3-bromo-6-chloro-l-methylpyridin-2(lH)-one (0.050 g, 0.225 mmol) according to the procedure described in Step B of Example 63. The desired product (0.059 g, 76%) was obtained as a pale yellow waxy crystalline solid. 1H-NMR (400 MHz, DMSO-d6) δ 7.68-7.62 (m, 2H), 7.50-7.45 (m, 3H), 7.44-7.32 (m, 3H), 7.26 (t, IH), 6.60 (d, IH), 5.22 (s, 2H), 3.64 (s, 3H). LRMS (ESI pos) m/e 344 (M+l).
[00495] Step C: Preparation of 3-(4-(benzyloxy)-3-fluorophenyl)-l-methyl-6-
(phenylamino)pyridin-2(lH)-one: To a solution of aniline (0.018 ml, 0.203 mmol) in THF (1 mL) at -78 0C is added LiHMDS (0.203 ml, 0.203 mmol; 1 M soln in hexanes) dropwise. The reaction mixture is stirred for 30 minutes at -780C after addition is complete. 3-(4-(benzyloxy)-3- fluorophenyl)-6-chloro-l-methylpyridin-2(lH)-one (0.058 g, 0.169 mmol) is then added dropwise as a solution in THF (1 mL). The reaction mixture is stirred at -780C and slowly warmed to room temperature and stirred for 16 hours. The reaction mixture is quenched with H2O and then partitioned between EtOAc and H2O. The phases are separated, and the aqueous phase is re- extracted with EtOAc (Ix). The combined organic layers are dried (Na2SO4), filtered and concentrated to yield the crude product. The crude product is purified by silica gel flash column chromatography, eluting with 20:1 CH2Cl2ZEtOAc to obtain the desired product. [00496] Step D: Preparation of 3-(3-fluoro-4-hydroxyphenyl)-l-methyl-6-
(phenylamino)pyridin-2(lH)-one: The title compound is prepared from 3-(4-(benzyloxy)-3- fluorophenyl)-l-methyl-6-(phenylamino)pyridin-2(lH)-one according to the procedure described in Step C of Example 63.
[00497] Step E: Preparation of 3-(3-fluoro-4-(6-methoxy-7-(3- morpholinopropoxy)quinolin-4-yloxy)phenyl)- 1 -methyl-6-(phenylamino)pyridin-2( 1 H)-one : Compound 169 is prepared from 3-(3-fluoro-4-hydroxyphenyl)-l-methyl-6- (phenylamino)pyridin-2(lH)-one, 4-(3-(4-chloro-6-methoxyquinolin-7-yloxy)propyl)morpholine and catalytic DMAP according to the procedure described in Step C of Example 58. [00498] Example 70 Preparation of l-(4-(6,7-dimethoxyquinolin-4-yloxy)-3- fluorophenyl)-N-(4-fluorophenyl)-2-oxopyrrolidine-3-carboxamide 170 111-03-PCT - P2338Rl - 02120.004WO1
Figure imgf000131_0001
[00499] Step A: Preparation of ethyl l-(4-(6,7-dimethoxyquinolin-4-yloxy)-3- fluorophenyl)-2-oxopyrrolidine-3-carboxylate: A mixture of 4-(4-bromo-2-fluorophenoxy)-6,7- dimethoxyquinoline (0.172 g, 0.19 mmol, Example 34), ethyl 2-oxopyrrolidine-3-carboxylate (0.025 g, 0.16 mmol), (lR,2R)-cyclohexane-l,2-diamine (0.011 g, 0.60 mmol), CuI (0.009 g, 0.30 mmol), and K3PO4 (0.068 g, 0.32 mmol) was placed in a sealed vial with dioxane (4 mL). The reaction mixture was then flushed with nitrogen, capped and placed in an oil bath at HO0C, and stirred for 20 hours. After the reaction was cooled to room temperture, the mixture was filtered through a pad of celite with EtOAc. After evaporation of the solvent, the crude was purified by silica gel flash column chromatography (1.5% MeOH in CH2Cl2) to afford 7.7 mg (11%) of the desired product. LRMS (ESI pos) m/e 455.2 (M+ 1).
[00500] Step B: Preparation of l-(4-(6,7-dimethoxyquinolin-4-yloxy)-3-fluorophenyl)-2- oxopyrrolidine-3-carboxylic acid: LiOH (0.034 mL, 0.034 mmol, 1.0 M in H2O) was added to a solution of ethyl l-(4-(6,7-dimethoxyquinolin-4-yloxy)-3-fluorophenyl)-2-oxopyrrolidine-3- carboxylate in 2 mL (4:1 ratio of THF:MeOH) at room temperature and stirred for 1 hour. The reaction mixture was acidified to pH 1 with aq. 1 N HCl solution and treated with water (5 mL), extracted with EtOAc, washed with brine, dried over MgSO4, and concentrated to afford 5.0 mg (69%) of the desired product.
[00501] Step C: Preparation of l-(4-(6,7-dimethoxyquinolin-4-yloxy)-3-fluorophenyl)-N-
(4-fluorophenyl)-2-oxopyrrolidine-3-carboxamide: EDCI (6.7 mg, 0.035 mmol) was added to a mixture of 1 -(4-(6,7-dimethoxyquinolin-4-yloxy)-3-fluorophenyl)-2-oxopyrrolidine-3-carboxylic acid (5.0 mg, 0.012 mmol) and HOBt (4.8 mg, 0.035 mmol) in DMF (2 mL) and was stirred at room temperature for 30 min. 4-Fluoroaniline (2.6 mg, 0.023 mmol) was then added followed by Et3N (0.005 mL, 0.035 mmol). After stirring 3 days, the reaction mixture was diluted with EtOAc and washed with saturated aqueous NH4Cl, saturated aqueous NaHCO3, and brine. The organic layer was dried over MgSO4 and concentrated under reduced pressure to give the crude material that was purified by silica gel flash column chromatography (1% MeOH in CH2Cl2) to afford 0.9 mg (15%) of 170. 1H-NMR (400 MHz, CD3OD) δ 8.44 (d, IH), 7.93 (dd, IH), 7.63 (s, IH), 7.62 (m, 2H), 7.55 (d, IH), 7.42 (t, IH), 7.37 (s, IH), 7.08 (t, IH), 6.51 (d, IH), 4.45 (m, 111-03-PCT- P2338R1 - 02120.004WO1
131
IH), 4.02 (s, 6H), 3.99 (m, IH), 3.81 (m, IH), 2.59 (m, IH), 2.51 (m, IH); 19F NMR (376 MHz, CD3OD) δ -120.6, -129.8. LRMS (ESI pos) m/e 520 (M+l).
[00502] Example 71 Preparation of N-(l-(4-(6,7-dimethoxyquinolin-4-yloxy)-3- fluorophenyl)-2-oxo- 1 ,2-dihydropyridin-3-yl)-4-fluorobenzamide 171
Figure imgf000132_0001
[00503] Step A: Preparation of 4-fluoro-N-(2-oxo-l,2-dihydropyridin-3-yl)benzamide:
EDCI (0.52 g, 2.70 mmol) was added to a mixture of 4-fluorobenzoic acid (0.25 g, 1.80 mmol) and HOBt (0.37 g, 2.70 mmol) in DMF (5 mL) and was stirred at room temperature for 30 minutes. 3-Aminopyridin-2(lH)-one (0.10 g, 0.91 mmol) was added followed by Et3N (0.38 mL, 2.70 mmol). After stirring 17 hours, the reaction mixture was diluted with EtOAc and washed with saturated aqueous NH4Cl, saturated aqueous NaHCO3, and brine. The organic layer was dried over MgSO4 and concentrated under reduced pressure to give the crude material that was purified by silica gel flash column chromatography (1% MeOH in CH2Cl2) to afford 0.11 g (52%) of the desired product. 1H-NMR (400 MHz, CDCl3) δ 11.62 (br. s, IH), 9.03 (br. s, IH), 8.63 (dd, IH), 7.96 (m, 2H), 7.19 (t, 2H), 7.11 (dd, IH), 6.41 (t, IH); 19F NMR (376 MHz, CD3OD) δ -107.4. LRMS (ESI pos) m/e 233 (M+l).
[00504] Step B: Preparation of N-(l-(4-(6,7-dimethoxyquinolin-4-yloxy)-3- fluorophenyl)-2-oxo-l,2-dihydropyridin-3-yl)-4-fluorobenzamide: A mixture of 4-(4-bromo-2- fluorophenoxy)-6,7-dimethoxyquinoline (60 mg, 0.158 mmol, Example 34), 4-fluoro-N-(2-oxo- l,2-dihydropyridin-3-yl)benzamide (35 mg, 0.151 mmol), (lR,2R)-cyclohexane-l,2-diamine (6.9 mg, 0.060 mmol), CuI (5.7 mg, 0.030 mmol), and K3PO4 (64 mg, 0.30 mmol) was placed in a sealed vial with dioxane (3 mL). The reaction mixture was then flushed with nitrogen, capped and placed in an oil bath at HO0C, and stirred for 17 hours. After the reaction was cooled to room temperture, the mixture was filtered through a pad of celite with EtOAc. After evaporation of the solvent, the crude was purified by silica gel flash column chromatography (1% MeOH in CH2Cl2 and then 100% Et2O to 3:1 Et2O:EtOAc) to afford 14.2 mg (18%) of 171. 1H-NMR (400 MHz, CD3OD) δ 9.21 (br. s, IH), 8.64 (dd, IH), 8.55 (d, IH), 7.95 (m, 2H), 7.58 (s, IH), 7.50 (s, IH), 7.46 (dd, IH), 7.43 (t, IH), 7.33 (d, IH), 7.18 (m, 3H), 6.56 (d, IH), 6.47 (t, IH), 4.08 (s, 3H), 4.07 (s, 3H); 19F NMR (376 MHz, CD3OD) δ -107.1, -125.8. LRMS (ESI pos) m/e 530 (M+l). 111-03-PCT- P2338R1 - 02120004WO1
132
[00505] Example 72 Preparation of N-(3-fluoro-4-(6-methoxy-7-(3- morpholinopropoxy)quinolin-4-yloxy)phenyl)-l-(4-fluorophenyl)-2-oxopiperidine-3- carboxamide 172
Figure imgf000133_0001
[00506] Step A: Preparation of ethyl l-(4-fluorophenyl)-2-oxopiperidine-3-carboxylate:
A mixture of l-fluoro-4-iodobenzene (1.95 g, 8.76 mmol), ethyl 2-oxopiperidine-3-carboxylate (1.0 g, 5.84 mmol), (lR,2R)-cyclohexane-l,2-diamine (0.27 g, 2.34 mmol), CuI (0.22 g, 1.17 mmol), and K3PO4 (2.48 g, 11.68 mmol) was placed in a sealed vial with dioxane (20 mL). The reaction mixture was then flushed with nitrogen, capped and placed in an oil bath at HO0C, and stirred for 15 hours. After the reaction was cooled to room temperture, the mixture was filtered through a pad of celite with EtOAc. After evaporation of the solvent, the crude was purified by silica gel flash column chromatography (2:l=CH2Cl2:Et20) to afford 1.067 g (69%) of the desired product. 1H-NMR (400 MHz, CDCl3) δ 7.24 (m, 2H), 7.13 (m, 2H), 4.24 (m, 2H), 3.67 (m, 2H), 3.57 (t, IH), 2.27 (m, IH), 2.21 (m, IH), 2.10 (m, IH), 1.95 (m, IH), 1.31 (t, 3H); 19F NMR (376 MHz, CDCl3) δ -115.4. LRMS (ESI pos) m/e 266 (M+l).
[00507] Step B: Preparation of l-(4-fluorophenyl)-2-oxopiperidine-3-carboxylic acid:
LiOH (0.87 mL, 0.87 mmol, 1.0 M in H2O) was added to a solution of ethyl l-(4-fluorophenyl)- 2-oxopiperidine-3-carboxylate in a mixture of THF (3 mL) and MeOH (1 mL) at room temperature for 1 hour. The reaction mixture was acidified to pH 1 with aq. 1 N HCl solution (0.9 mL) and then concentrated to afford the desired product salt. 1H-NMR (400 MHz, CD3OD) δ 7.31 (m, 2H), 7.14 (m, 2H), 3.69 (m, 2H), 3.54 (t, IH), 2.24 (m, 2H), 2.09 (m, IH), 1.99 (m, IH); 19F NMR (376 MHz, CD3OD) δ -117.3. LRMS (ESI neg) m/e 236 (M-I). [00508] Step C: Preparation of 7-(benzyloxy)-4-(2-fluoro-4-nitrophenoxy)-6- methoxyquinoline: To a stirred solution of 7-(benzyloxy)-6-methoxyquinolin-4-ol (prepared according to the method of WO 2005/030140) (2.81 g, 10 mmol) in 30 mL of 1:1 CH3CN:DMF at room temperature under nitrogen was added cesium carbonate (6.52 g, 20 mmol). After 30 minutes, l,2-difluoro-4-nitrobenzene (1.22 mL, 11 mmol) was added. After 3 hours, the reaction was partially concentrated by rotovap and then diluted to 60 mL with EtOAc and washed 4 x 50 mL with a brine/H2O mix. The organics were dried (MgSO4), filtered and concentrated to a 111-03-PCT - P2338R1 - 02120.004WO1
133 residue that was purified by silica gel flash column chromatography (2:3 EtOAc/hexanes). Product containing fractions were pooled and concentrated to a brown solid (1.56 g, 37%). 1H NMR (400 MHz, CDCl3) δ 8.56 (d, IH), 8.19 (dd, IH), 8.13 (m, IH), 7.51 (m, 3H), 7.46 (s, IH), 7.40 (m, 2H), 7.33 (m, 2H), 6.54 (d, IH), 5.34 (s, 2H), 4.04 (s, 3H).
[00509] Step D: Preparation of 4-(2-fluoro-4-nitrophenoxy)-6-methoxyquinolin-7-ol: A solution of 7-(benzyloxy)-4-(2-fluoro-4-nitrophenoxy)-6-methoxyquinoline (1.56 g, 3.71 mmol) was stirred in 11 mL of 33% wt HBr in acetic acid at room temperature under a drying tube. After 4 hours the reaction was diluted with 100 mL Et2O and filtered. The isolated tan solid was washed with Et2O and then dried under high vacuum to give 1.45 g (89%) of the HBr salt. This material was stirred as a suspension in 100 mL 4:1 CH2Cl2:Me0H. 100 mL of H2O was added and then solid NaHCO3 added until pH=7. More MeOH was added until the mixture was a two phase solution. The organics were isolated and the aqueous phase extracted 2 x 50 mL with CH2Cl2. The combined organics were dried (MgSO4), filtered and concentrated to a yellow solid (1.08 g, 88%). HBr Salt 1H NMR (400 MHz, CDC13/CD3OD) δ 8.60 (d, IH), 8.30 (m, 2H), 7.80 (s, IH), 7.64 (m, 2H), 7.33 (s, IH), 6.76 (dd, IH), 4.13 (s, 3H).
[00510] Step E: Preparation of 4-(3-(4-(2-fluoro-4-nitrophenoxy)-6-methoxyquinolin-7- yloxy)propyl)morpholine: To a stirred suspension of 4-(2-fluoro-4-nitrophenoxy)-6- methoxyquinolin-7-ol: A solution of 7-(benzyloxy)-4-(2-fluoro-4-nitrophenoxy)-6- methoxyquinoline (610 mg, 1.85 mmol) in 9.2 mL CH2Cl2 at room temperature under nitrogen was added 3-morpholinopropan-l-ol (307 uL, 2.22 mmol) followed by triphenylphosphine (775 mg, 2.96 mmol) and finally DEAD (465 uL, 2.96 mmol). After stirring overnight, the reaction was concentrated to a residue by rotovap and purified directly by silica gel flash column chromatography (9/1 EtOAc/MeOH). Product containing fractions were pooled and concentrated to a yellow solid (650 mg, 77%). 1H NMR (400 MHz, CDCl3) δ 8.58 (d, IH), 8.19 (dd, IH), 8.14 (m, IH), 7.48
(s, IH), 7.43 (s, IH), 7.33 (dd, IH), 6.55 (d, IH), 4.29 (dd, 2H), 4.01 (s, 3H), 3.73 (m, 4H), 2.58 (dd, 2H), 2.49 (br m, 4H), 2.14 (m, 2H).
[00511] Step F: Preparation of 3-fluoro-4-(6-methoxy-7-(3-morpholinopropoxy)quinolin-
4-yloxy)aniline: A solution of 4-(3-(4-(2-fluoro-4-nitrophenoxy)-6-methoxyquinolin-7- yloxy)propyl)morpholine (620 mg,1.36 mmol) was formed in 75 mL of 95% EtOH and 75 mL EtOAc in a 250 mL Parr Bottle. Pearlman's catalyst (20 wt %, 95 mg, 0.14 g/atom palladium) was added and the reaction put through a vacuum/purge cycle three times with hydrogen gas and then held under 50 psi hydrogen and shaken overnight. The reaction was filtered through GF/F filter paper with 95% EtOH and concentrated to a yellow foam (560 mg, 96%). 1H NMR (400 111-03-PCT - P2338R1 - 02120.004WO1
134
MHz, CDCl3) δ 8.47 (d, IH), 7.58 (s, IH), 7.43 (s, IH), 7.04 (m, IH), 6.57 (m, IH), 6.51 (m, IH),
6.40 (m, IH), 4.27 (m, 2H), 4.04 (s, 3H), 3.73 (m, 4H), 2.58 (m, 2H), 2.49 (br m, 4H), 2.13 (m,
2H).
[00512] Step G: Preparation of N-(3-fluoro-4-(6-methoxy-7-(3- morpholinopropoxy)quinolin-4-yloxy)phenyl)-l-(4-fluorophenyl)-2-oxopiperidine-3- carboxamide: EDCI (29.6 mg, 0.154 mmol) was added to a mixture of l-(4-fluorophenyl)-2- oxopiperidine-3-carboxylic acid (15.3 mg, 0.064 mmol) and HOBt (20.9 mg, 0.154 mmol) in
DMF (2 mL) was stirred at room temperature for 30 minutes. 3-Fluoro-4-(6-methoxy-7-(3- morpholinopropoxy)quinolin-4-yloxy)aniline (11 mg, 0.026 mmol) was added followed by Et3N
(0.022 mL, 0.154 mmol). After stirring 17 hours, the reaction mixture was diluted with EtOAc and washed with saturated aq. NH4Cl, saturated aq. NaHCO3, and brine. The organic layer was dried over MgSO4 and concentrated under reduced pressure to give the crude material that was purified by silica gel flash column chromatography (7% MeOH in CH2Cl2) to afford 4.9 mg
(29%) of 172. 1H-NMR (400 MHz, CD3OD) δ 8.41 (d, IH), 7.85 (dd, IH), 7.64 (s, IH), 7.42
(m, IH), 7.33 (m, 4H), 7.16 (t, 2H), 6.49 (d, IH), 4.25 (t, 2H), 4.01 (s, 3H), 3.65 - 3.78 (m, 7H),
2.64 (t, 2H),
2.54 (m, 3H), 2.0 - 2.32 (m, 7H); 19F NMR (376 MHz, CD3OD) δ -117.2. LRMS (ESI pos) m/e
647 (M+l).
[00513] Example 73 Preparation of l-(3-fluoro-4-(6-methoxy-7-(3- morpholinopropoxy)quinolin-4-yloxy)phenyl)-N-(4-fluorophenyl)-2-oxo-l,2-dihydropyridine-3- carboxamide 173
Figure imgf000135_0001
[00514] Step A: Preparation of methyl l-(3-fluoro-4-(6-methoxy-7-(3- morpholinopropoxy)quinolin-4-yloxy)phenyl)-2-oxo- 1 ,2-dihydropyridine-3-carboxylate: To a solution of methyl 2-oxo-2H-pyran-3-carboxylate (9.7 mg, 0.042 mmol) in a mixture of THF (2 mL) and DMF (0.5 mL) at room temperature was added 3-fluoro-4-(6-methoxy-7-(3- morpholinopropoxy)quinolin-4-yloxy)aniline (prepared according to Example 72, steps C-F) (18 mg, 0.042 mmol), and the reaction mixture was stirred for 2.5 hours. To the aniline adduct intermediate formed via Michael addition was added in situ EDCI (13 mg, 0.066 mmol) and DMAP (0.57 mg, 0.0047 mmol) at room temperature. The reaction mixture was stirred at room 111-03-PCT- P2338R1 - 02120.004WO1
135 temperature for 5 days. To the reaction mixture were added aqueous 1 N NaHCO3, extracted with EtOAc, dried over MgSO4, and concentrated to give the crude material that was purified by silica gel flash column chromatography (10% MeOH in CH2Cl2) to afford 3.5 mg (13%) of the desired product. 1H-NMR (400 MHz, CD3OD) δ 8.47 (d, IH), 8.35 (dd, IH), 7.98 (dd, IH), 7.65 (s, IH), 7.59 (m, 2H), 7.40 (m, 2H), 6.65 (d, IH), 6.57 (t, IH), 4.27 (t, 2H), 4.02 (s, 3H), 3.87 (s, 3H), 3.75 (t, 4H), 2.74 (t, 2H), 2.64 (m, 4H), 2.17 (m, 2H); 19F NMR (376 MHz, CD3OD) δ - 129.1. LRMS (ESI pos) m/e 564 (M+ 1).
[00515] Step B: Preparation of l-(3-fluoro-4-(6-methoxy-7-(3- morpholinopropoxy)quinolin-4-yloxy)phenyl)-2-oxo-l,2-dihydropyridine-3-carboxylic acid: LiOH (0.012 mL, 0.012 mmol, 1.0 M in H2O) was added to a solution of methyl l-(3-fluoro-4-(6- methoxy-7-(3-morpholinopropoxy)quinolin-4-yloxy)phenyl)-2-oxo-l,2-dihydropyridine-3- carboxylate in a mixture of THF (1.5 mL) and MeOH (0.5 mL) at room temperature for 6 hours. The reaction mixture was acidified to pH 1 with aqueous 1 N HCl solution (0.012 mL) and then concentrated to afford the desired product salt. LRMS (ESI pos) m/e 550 (M+l). [00516] Step C: Preparation of l-(3-fluoro-4-(6-methoxy-7-(3- morpholinopropoxy)quinolin-4-yloxy)phenyl)-N-(4-fluorophenyl)-2-oxo-l,2-dihydropyridine-3- carboxamide: EDCI (3.6 mg, 0.019 mmol) was added to a mixture of l-(3-fluoro-4-(6-methoxy- 7-(3-morpholinopropoxy)quinolin-4-yloxy)phenyl)-2-oxo- 1 ,2-dihydropyridine-3-carboxylic acid (3.4 mg, 0.0062 mmol) and HOBt (2.5 mg, 0.019 mmol) in DMF (0.5 mL) and was stirred at room temperature for 1 hour. 4-Fluoroaniline (2.1 mg, 0.019 mmol) was added followed by Et3N (1.9 mg, 0.019 mmol). After stirring 17 hours, the reaction mixture was diluted with EtOAc and washed with saturated aqueous NH4Cl, saturated aqueous NaHCO3, and brine. The organic layer was dried over MgSO4 and concentrated under reduced pressure to give the crude material that was purified by silica gel flash column chromatography (5% MeOH in CH2Cl2) to afford 0.7 mg (18%) of 173. LRMS (ESI pos) m/e 643 (M+l).
[00517] Example 74 Preparation of N-(3-fluoro-4-(6-methoxy-7-(3- morpholinopropoxy)quinolin-4-yloxy)phenyl)quinoline-8-carboxamide 174
Figure imgf000136_0001
111-03-PCT - P2338R1 - 02120.004WO1
136
[00518] EDCI (27 mg, 0.14 mmol) was added to a mixture of quinoline-8-carboxylic acid
(8.1 mg, 0.047 mmol) and HOBt (19 mg, 0.14 mmol) in DMF (2 mL) was stirred at room temperature for 1 hour. 3-Fluoro-4-(6-methoxy-7-(3-morpholinopropoxy)quinolin-4- yloxy)aniline (prepared in Example 72, steps C-F) (10 mg, 0.023 mmol) was added followed by Et3N (0.020 mL, 0.14 mmol). After stirring 17 hours, the reaction mixture was diluted with EtOAc and washed with saturated aqueous NH4Cl, saturated aqueous NaHCO3, and brine. The organic layer was dried over MgSO4 and concentrated under reduced pressure to give the crude material that was purified by silica gel flash column chromatography (5% MeOH in CH2Cl2) to afford 9.6 mg (70 %) of 174. 1H-NMR (400 MHz, CD3OD/CDCI3) δ 9.11 (dd, IH), 8.83 (dd, IH), 8.50 (dd, IH), 8.42 (d, IH), 8.18 (d, IH), 8.13 (dd, IH), 7.79 (t, IH), 7.66 (m, 3H), 7.39 (t, IH), 7.36 (s, IH), 6.54 (d, IH), 4.27 (t, 2H), 4.04 (s, 3H), 3.75 (t, 4H), 2.66 (t, 2H), 2.56 (m, 4H), 2.15 (m, 2H); 19F NMR (376 MHz, CD3OD/CDCI3) δ -129.0. LRMS (ESI pos) m/e 583 (M+ 1). [00519] Example 75 Preparation of N-(3-fluoro-4-(6-methoxy-7-(3- moφholinopropoxy)quinolin-4-yloxy)phenyl)-l-(4-fluorophenyl)-2-oxopyrrolidine-3- carboxamide 175
Figure imgf000137_0001
[00520] EDCI (67 mg, 0.35 mmol) was added to a mixture of l-(4-fluorophenyl)-2- oxopyrrolidine-3-carboxylic acid (31 mg, 0.14 mmol) and HOBt (47 mg, 0.35 mmol) in DMF (3 mL) and was stirred at room temperature for 30 minutes. 3-fluoro-4-(6-methoxy-7-(3- morpholinopropoxy)quinolin-4-yloxy)aniline (prepared in Example 72, steps C-F) (30 mg, 0.070 mmol) was added followed by Et3N (0.049 mL, 0.35 mmol). After stirring 2 hours, the reaction mixture was diluted with EtOAc and washed with saturated aqueous NH4Cl, saturated aqueous NaHCO3, and brine. The organic layer was dried over MgSO4 and concentrated under reduced pressure to give the crude material that was purified by silica gel flash column chromatography (7% MeOH in CH2Cl2) to afford 29 mg (65 %) of 175. 1H-NMR (400 MHz, CDCl3) δ 9.85 (s, IH), 8.48 (d, IH), 7.82 (dd, IH), 7.57 (m, 3H), 7.44 (s, IH), 7.32 (m, IH), 7.22 (t, IH), 7.13 (t, 2H), 6.39 (d, IH), 4.28 (t, 2H), 4.04 (s, 3H), 3.91 (m, 2H), 3.72 (m, 5H), 2.71 (m, IH), 2.58 (m, 3H), 2.49 (m, 4H), 2.14 (m, 2H); 19F NMR (376 MHz, CDCl3) δ -115.9, -127.1. LRMS (ESI pos) m/e 633 (M+l). 111-03-PCT - P2338R1 - 02120.004WO1
137
[00521] Example 76 Preparation of l-(4-chlorophenyl)-N-(3-fluoro-4-(6-methoxy-7-(3- morpholinopropoxy)quinolin-4-yloxy)phenyl)-2-oxopyrrolidine-3-carboxamide 176
Figure imgf000138_0001
[00522] Prepared from 3-fluoro-4-(6-methoxy-7-(3-morpholinopropoxy)quinolin-4- yloxy)aniline (prepared in Example 72, steps C-F) and l-(4-chlorophenyl)-2-oxopyrrolidine-3- carboxylic acid according to the procedure for Example 75. The crude was purified by silica gel flash column chromatography (5% MeOH in CH2Cl2) to afford 25 mg (55%) of 176. 1H-NMR (400 MHz, CDCl3) δ 9.85 (s, IH), 8.48 (d, IH), 7.82 (dd, IH), 7.57 (m, 3H), 7.40 (m, 3H), 7.32 (m, IH), 7.22 (t, IH), 6.39 (d, IH), 4.28 (t, 2H), 4.04 (s, 3H), 3.91 (m, 2H), 3.73 (m, 5H), 2.71 (m, IH), 2.58 (m, 3H), 2.49 (m, 4H), 2.13 (m, 2H); 19F NMR (376 MHz, CDCl3) δ -127.0. LRMS (ESI pos) m/e 649, 650 (M+, Cl pattern).
[00523] Example 77 Preparation of N-(3-fluoro-4-(6-methoxy-7-(3- morpholinopropoxy)quinolin-4-yloxy)phenyl)-2-oxo- 1 -phenylpyrrolidine-3-carboxamide 177
Figure imgf000138_0002
[00524] Prepared from 3-fluoro-4-(6-methoxy-7-(3-morpholinopropoxy)quinolin-4- yloxy)aniline (prepared in Example 72, steps C-F) and 2-oxo-l-phenylpyrrolidine-3-carboxylic acid according to the procedure for Example 75. The crude was purified by silica gel flash column chromatography (7% MeOH in CH2Cl2) to afford 6.5 mg (5%) of 177. 1H-NMR (400
MHz, CD3OD) δ 8.42 (d, IH), 7.88 (d, IH), 7.63 (m, 3H), 7.40 (m, 5H), 7.22 (t, IH), 6.50 (d,
IH), 4.24 (t, 2H), 4.0
(m, 5H), 3.72 (t, 4H), 2.64 (t, 2H), 2.54 (m, 6H), 2.13 (m, 2H); 19F NMR (376 MHz, CD3OD) δ -
130.1. LRMS (ESI neg) m/e 613 (M-I).
[00525] Example 78 Preparation of N-(3-fluoro-4-(6-methoxy-7-(3- morpholinopropoxy)quinolin-4-yloxy)phenyl)-l-(4-fluorophenyl)-3-methyl-2-oxopyrrolidine-3- carboxamide 178 111-03-PCT - P2338R1 - 02120.004WO1
138
Figure imgf000139_0001
[00526] Step A: Preparation of methyl l-(4-fluorophenyl)-2-oxopyrrolidine-3- carboxylate: To a solution of l-(4-fluorophenyl)-2-oxopyrrolidine-3-carboxylic acid (0.20 g, 0.90 mmol) in a mixture of Et2O (6 mL), MeOH (2 mL), and THF (2 mL) was added (diazomethyl)trimethylsilane (1.1 mL, 2.0 M) at 00C. The resulting mixture was stirred for 30 minutes at room temperature, quenched with AcOH, and diluted with EtOAc. The organic layer was washed with water, NaHCO3 solution (2x), and brine, dried over MgSO4, and concentrated under reduced pressure to give the desired product (0.206 g, 98%). LRMS (ESI pos) m/e 238 (M+l).
[00527] Step B: Preparation of methyl l-(4-fluorophenyl)-3-methyl-2-oxopyrrolidine-3- carboxylate: LiH (13.8 mg, 1.737 mmol) was added to the solution of methyl l-(4-fluorophenyl)- 2-oxopyrrolidine-3-carboxylate (0.206 g, 0.868 mmol) in DMF (5 mL) at O0C. After 30 minutes stirring, iodomethane (0.16 mL, 2.61 mmol) was added to the reaction mixture at O0C, and then the reaction was warmed to room temperature. The reaction mixture was stirred 17 hours and heated at 4O0C for 3 hours. After cooled to room temperature, the mixture was treated with EtOAc, quenched with ice water, extracted with EtOAc, washed with brine, dried over MgSO4, and concentrated to give the crude material that was purified by silica gel flash column chromatography (19:1 CH2Cl2/EtOAc) to afford 0.149 g (68%) of the desired product. 1H-NMR (400 MHz, CDCl3) δ 7.61 (m, 2H), 7.07 (m, 2H), 3.94 (m, IH), 3.78 (m, IH), 3.75 (s, 3H), 2.68 (m, IH), 2.06 (m, IH), 1.55 (s, 3H); 19F NMR (376 MHz, CDCl3) δ -117.6. LRMS (ESI pos) m/e 252 (M+l).
[00528] Step C: Preparation of l-(4-fluorophenyl)-3-methyl-2-oxopyrrolidine-3- carboxylic acid: LiOH (1.2 mL, 1.19 mmol, 1.0 M in H2O) was added to a solution of methyl 1- (4-fluorophenyl)-3-methyl-2-oxopyrrolidine-3-carboxylate (0.149 g, 0.593 mmol) in a mixture of THF (4.5 mL) and MeOH (1.5 mL) at room temperature for 1 hour. The reaction mixture was acidified with aqueous 1 N HCl solution (1.4 mL), extracted with EtOAc, washed with brine, dried over MgSO4, and concentrated to afford the desired product (0.13 g, 92%). 1H-NMR (400 MHz, CD3OD) δ 7.62 (m, 2H), 7.13 (t, 2H), 3.97 (m, IH), 3.86 (td, IH), 2.63 (m, IH), 2.13 (m, IH), 1.47 (s, 3H); 19F NMR (376 MHz, CD3OD) δ -119.3. 111-03-PCT- P2338R1 - 02120.004WO1
139
[00529] Step D: Preparation of N-(3-fluoro-4-(6-methoxy-7-(3- morpholinopropoxy)quinolin-4-yloxy)phenyl)-l-(4-fluorophenyl)-3-methyl-2-oxopyiτolidine-3- carboxamide: Prepared from 3-fluoro-4-(6-methoxy-7-(3-morpholinopropoxy)quinolin-4- yloxy)aniline (prepared in Example 72, steps C-F) and l-(4-fluorophenyl)-3-methyl-2- oxopyrrolidine-3-carboxylic acid according to the procedure for Example 75. The crude was purified by silica gel flash column chromatography (5% MeOH in CH2Cl2) to afford 66 mg (62%) of 178, as a racemic mixture. 1H-NMR (400 MHz, CD3OD) δ 8.41 (d, IH), 7.87 (dd, IH), 7.69 (m, 2H), 7.63 (s, IH), 7.45 (m, IH), 7.35 (t, 2H), 7.16 (t, 2H), 6.49 (d, IH), 4.25 (t, 2H), 4.01 (s, 3H), 3.92 (m, 2H), 3.72 (t, 4H), 2.81 (m, IH), 2.64 (t, 2H), 2.54 (m, 4H), 2.19 (m, IH), 2.13 (m, 2H), 1.66 (s, 3H); 19F NMR (376 MHz, CD3OD) δ -119.0, -130.1. LRMS (ESI pos) m/e 647 (M+l).
[00530] Examples 79 and 80 Preparation . of (S)-N-(3-fluoro-4-(6-methoxy-7-(3- moφholinopropoxy)quinolin-4-yloxy)phenyl)-l-(4-fluorophenyl)-3-methyl-2-oxopyrrolidine-3- carboxamide 179
Figure imgf000140_0001
[00531] and (R)-N-(3-fluoro-4-(6-methoxy-7-(3-morpholinopropoxy)quinolin-4- yloxy)phenyl)-l-(4-fluorophenyl)-3-methyl-2-oxopyrrolidine-3-carboxamide 180
Figure imgf000140_0002
[00532] The title compounds were isolated from the racemic mixture of 178 from
Example 78 N-(3-fluoro-4-(6-methoxy-7-(3-morpholinopropoxy)quinolin-4-yloxy)phenyl)- 1 -(4- fluorophenyl)-3-methyl-2-oxopyrrolidine-3-carboxamide by chiral Prep HPLC (Agilent 1100 MSD prep, Fifi) with 40% EtOH and 60% Hexane using Chiralpak IA 25Ox 10 mm column. [00533] Example 81 N-(3-fluoro-4-(6-methoxy-7-(3-morpholinopropoxy)quinolin-4- yloxy)phenyl)-3-(4-fluorophenyl)-2-oxo-3-azabicyclo[3.1.0]hexane-l-carboxamide 181 111-03-PCT - P2338R1 - 02120 004WO1
140
Figure imgf000141_0001
[00534] Step A: Preparation of N-allyl-4-fluoroaniline: To a stirred solution of p- fluoroaniline (1.92 mL, 20 mmol) in 60 mL THF at -780C was added n-BuLi (12.5 mL, 20 mmol, 1.6 M in hexanes) dropwise by syringe. After 30 minutes, allyl bromide (1.69 mL, 20 mmol) was added neat by syringe. After 2 hours at -780C, the reaction was allowed to warm to O0C, was quenched by pouring into 50 mL H2O and then excess THF was removed by rotovap. The residual material was extracted 2 x 50 mL with EtOAc. The combined organics were dried (MgSO4), filtered and concentrated to a crude oil that was purified by silica gel flash column chromatography (5/95 Et2O/hexanes). Product containing fractions were pooled and concentrated to an orange oil (1.9 g, 63% yield). 1H NMR (400 MHz, CDCl3) δ 6.88 (m, 2H), 6.56 (m, 2H), 5.95 (m, IH), 5.28 (m, IH), 5.17 (m, IH), 3.74 (br d, 2H), 3.66 (br s, IH).
[00535] Step B: Preparation of methyl 3-(allyl(4-fluorophenyl)amino)-3-oxopropanoate:
To a stirred solution of N-allyl-4-fluoroaniline (207 mg, 1.37 mmol) in 3 mL CH2Cl2 at O0C under nitrogen was added DIEA (262 uL, 1.5 mmol) followed by DMAP (17 mg, 0.14 mmol) and the methyl malonyl chloride (161 uL, 1.5 mmol) as a solution in 1 mL CH2Cl2 dropwise by syringe. After 1 hour at O0C, the reaction was diluted to 30 mL with CH2Cl2 and washed 2 x 30 mL with 2N HCl and 2 x 30 mL with saturated NaHCO3. The organics were dried (MgSO4), filtered and concentrated to a yellow oil that was used as is in the next reaction (260 mg, 75%). 1H NMR (400 MHz, CDCl3) δ 7.18 (m, 2H), 7.09 (m, 2H), 5.85 (m, IH), 5.13 (m, 2H), 4.29 (m, 2H), 3.68 (s, 3H), 3.19 (s, 2H).
[00536] Step C: Preparation of methyl 3-(4-fluorophenyl)-2-oxo-3- azabicyclo[3.1.0]hexane-l-carboxylate: To a stirred suspension of manganese triacetate dihydrate (557 mg, 2.08 mmol) and copper diacetate monohydrate (207 mg, 1.04 mmol) in 6 mL glacial acetic acid at room temperature under nitrogen was added a solution of methyl 3-(allyl(4- fluorophenyl)amino)-3-oxopropanoate (261 mg, 1.04 mmol) in 1 mL acetic acid. After stirring overnight at room temperature, a solution of 10% aqueous sodium bisulfite was added (40 mL). After stirring for a few minutes, the suspension was extracted 3 x 50 mL with EtOAc. The combined organics were washed 3 x 50 mL with H2O and 3 x 50 mL with saturated NaHCO3. The organics were dried (MgSO4), filtered and concentrated. The residue was purified by silica 111-03-PCT - P2338R1 - 02120.004WO1
141 gel flash column chromatography (3/7 EtOAc/hexanes) to give after pooling and concentration of product containing fractions a clear solid (26 mg, 10%). 1H NMR
(400 MHz, CDCl3) δ 7.51 (m, 2H), 7.04 (m, 2H), 4.04 (m, IH), 3.82 (s, 3H), 3.71 (d, IH), 2.50 (m, IH), 2.05 (m, IH), 1.31 (m, IH).
[00537] Step D: Preparation of 3-(4-fluorophenyl)-2-oxo-3-azabicyclo[3.1.0]hexane-l- carboxylic acid: To a stirred solution of methyl 3-(4-fluorophenyl)-2-oxo-3- azabicyclo[3.1.0]hexane-l-carboxylate (26 mg, 0.1 mmol) in 1 mL 3:2 THF:H2O at room temperature under nitrogen was added lithium hydroxide powder (4.8 mg, 0.2 mmol). After stirring overnight, the reaction was partitioned between EtOAc (30 mL) and 2N HCl (30 mL). The organics were washed 1 x 30 mL with brine, was dried (MgSO4), filtered and concentrated to a brown solid (20 mg, 85%). 1H NMR (400 MHz, CDCl3) δ 7.47 (m, 2H), 7.08 (m, 2H), 4.10 (m, IH), 3.77 (m, IH), 2.75 (m, IH), 2.05 (m, IH), 1.44 (m, IH).
[00538] Step E: To a stirred solution of 3-(4-fluorophenyl)-2-oxo-3- azabicyclo[3.1.0]hexane-l-carboxylic acid (20 mg, 0.85 mmol) in 850 uL CH2Cl2 at room temperature under nitrogen was added DIEA (44 uL, 0.26 mmol) followed by EDCI (24 mg, 0.13 mmol) and HOBt (17 mg, 0.13 mmol). After 30 minutes, 3-fluoro-4-(6-methoxy-7-(3- morpholinopropoxy)quinolin-4-yloxy)aniline (prepared in Example 72, steps C-F) (36 mg, 0.85 mmol) was added as a solid. After stirring overnight, the reaction was diluted to 30 mL with CH2Cl2 and stirred with 10 mL 10% Na2CO3. The layers were separated and the aqueous solution extracted 1 x 10 mL with CH2Cl2. The combined organics were dried (MgSO4), filtered and concentrated. The crude product was purified by silica gel flash column chromatography, loading with CH2Cl2 and eluted with 100 mL CH2Cl2 and then 5/95 MeOH/CH2Cl2. Fractions were pooled and concentrated to give 181 as a yellow oil (30 mg, 55%). 1H NMR (400 MHz, CDCl3) δ 10.50 (s, IH), 8.48 (d, IH), 7.82 (m, IH), 7.57 (s, IH), 7.48 (m, 2H), 7.44 (s, IH), 7.31 (m, IH), 7.22 (m, IH), 7.11 (m, 2 H), 6.40 (m, IH), 5.30 (s, IH), 4.28 (m, 2H), 4.11 (m, IH),
4.04 (s, 3H), 3.77 (m, IH), 3.72 (m, 4H), 2.81 (m, IH), 2.58 (m, 2H), 2.49 (m, 4H), 2.13 (m, 2H),
2.05 (m, IH), 1.39 (m, IH).
[00539] Example 82 Preparation of N-(3-fluoro-4-(6-methoxy-7-(3- morpholinopropoxy)quinolin-4-yloxy)phenyl)- 1 -(4-fluorophenyl)-2-oxo- 1 ,2-dihydropyridine-3- carboxamide 182 111-03-PCT - P2338R1 - 02120.004WO1
142
Figure imgf000143_0001
[00540] Prepared from 3-fluoro-4-(6-methoxy-7-(3-morpholinopropoxy)quinolin-4- yloxy)aniline (prepared in Example 72, steps C-F) and l-(4-fluorophenyl)-2-oxo-l,2- dihydropyridine-3-carboxylic acid (prepared from methyl 2-oxo-2H-pyran-3-carboxylate with 4- fluoroaniline and followed by hydrolysis using the methods described in US 2005/0239820) according to the procedure for Example 75. The crude was purified by silica gel flash column chromatography (7% MeOH in CH2Cl2) to afford 8.1 mg (49%) of 182. 1H-NMR (400 MHz, CD3OD) δ 8.66 (dd, IH), 8.42 (d, IH), 8.01 (dd, IH), 7.97 (dd, IH), 7.64 (s, IH), 7.54 (m, 2H), 7.43 (m, IH), 7.34 (m, 4H), 6.74 (t, IH), 6.51 (d, IH), 4.25 (t, 2H), 4.01 (s, 3H), 3.72 (t, 4H), 2.64 (t, 2H), 2.54 (m, 4H), 2.13 (m, 2H); 19F NMR (376 MHz, CD3OD) δ -114.5, -129.8. LRMS (ESI pos) m/e 643 (M+l).
[00541] Example 83 Preparation of N-(3-fluoro-4-(6-methoxy-7-(3- morpholinopropoxy)quinolin-4-yloxy)phenyl)-l-(4-fluorobenzyl)-2-oxo-l,2-dihydropyridine-3- carboxamide 183
Figure imgf000143_0002
[00542] Step A: Preparation of methyl l-(4-fluorobenzyl)-2-oxo-l,2-dihydropyridine-3- carboxylate: LiH (7.8 mg, 0.980 mmol) was added to the solution of methyl 2-oxo-l,2- dihydropyridine-3-carboxylate (50 mg, 0.327 mmol) in DMF (3 mL) at O0C. After 30 minutes stirring, l-(bromomethyl)-4-fluorobenzene (9.3 mg, 0.490 mmol) was added to the reaction mixture at O0C, and then the reaction was warmed to room temperature. After 4 hours stirring, the reaction mixture was quenched with ice water, extracted with EtOAc, washed with brine, dried over MgSO4, and concentrated to give the crude material that was purified by silica gel flash column chromatography (100% Et2O and then 3: I=Et2OrEtOAc) to afford both of 23.2 mg (27%) of methyl l-(4-fluorobenzyl)-2-oxo-l,2-dihydropyridine-3-carboxylate and 25.5 mg (22%) of 4-fluorobenzyl l-(4-fluorobenzyl)-2-oxo-l,2-dihydropyridine-3-carboxylate. For methyl l-(4- 1-11-03-PCT - P2338R1 - 02120.004WO1
143 fluorobenzyl)-2-oxo-l,2-dihydropyridine-3-carboxylate: 1H-NMR (400 MHz, CDCl3) δ 8.16 (dd, IH), 7.52 (dd, IH), 7.33 (dd, 2H), 7.02 (t, 2H), 6.22 (t, IH), 5.13 (s, 2H), 3.91 (s, 3H); 19F NMR (376 MHz, CDCl3) δ -113.9. LRMS (ESI pos) m/e 262 (M+l). For 4-fluorobenzyl l-(4- fluorobenzyl)-2-oxo-l,2-dihydropyridine-3-carboxylate: 1H-NMR (400 MHz, CDCl3) δ 8.12 (dd, IH), 7.51 (dd, IH), 7.45 (m, 2H), 7.33 (m, 2H), 7.04 (m, 4H), 6.21 (t, IH), 5.31 (s, 2H), 5.14 (s, 2H); 19F NMR (376 MHz, CDCl3) δ -1 13.8, -114.4. LRMS (ESI pos) m/e 356 (M+l). [00543] Step B: Preparation of l-(4-fluorobenzyl)-2-oxo-l,2-dihydropyridine-3- carboxylic acid: LiOH (0.14 mL, 0.14 mmol, 1.0 M in H2O) was added to a solution of 4- fluorobenzyl l-(4-fluorobenzyl)-2-oxo-l,2-dihydropyridine-3-carboxylate (24 mg, 0.068 mmol) in a mixture of THF (1.5 mL) and MeOH (0.5 mL) at room temperature for 2 hours. The reaction mixture was acidified to pH 1 with aq 1 N HCl solution (0.14 mL) and then concentrated to afford the desired product salt. 1H-NMR (400 MHz, CD3OD) δ 8.46 (dd, IH), 8.16 (dd, IH), 7.45 (m, 2H), 7.10 (m, 2H), 6.68 (t, IH), 5.31 (s, 2H); 19F NMR (376 MHz, CD3OD) δ -115.5. LRMS (ESI neg) m/e 246 (M-I).
[00544] Step C: Preparation of N-(3-fluoro-4-(6-methoxy-7-(3- morpholinopropoxy)quinolin-4-yloxy)phenyl)-l-(4-fluorobenzyl)-2-oxo-l,2-dihydropyridine-3- carboxamide: Prepared from 3-fluoro-4-(6-methoxy-7-(3-morpholinopropoxy)quinolin-4- yloxy)aniline (prepared in Example 72, steps C-F) and l-(4-fluorobenzyl)-2-oxo-l,2- dihydropyridine-3-carboxylic acid according to the procedure for Example 72. The crude was purified by silica gel flash column chromatography (7% MeOH in CH2Cl2) to afford 6.9 mg (46%) of 183. 1H-NMR (400 MHz, CD3OD/CDC13) δ 8.58 (dd, IH), 8.41 (d, IH), 8.07 (dd, IH), 8.0 (dd, IH), 7.64 (s, IH), 7.45 (m, 3H), 7.34 (m, 2H), 7.10 (t, 2H), 6.64 (t, IH), 6.51 (d, IH), 5.32 (s, 2H), 4.26 (t, 2H), 4.03 (s, 3H), 3.73 (t, 4H), 2.65 (t, 2H), 2.55 (m, 4H), 2.14 (m, 2H); 19F NMR (376 MHz, CD3OD/CDC13) δ -115.9, -129.3. LRMS (ESI pos) m/e 657 (M+l). [00545] Example 84 Preparation of l-(4-chlorobenzyl)-N-(3-fluoro-4-(6-methoxy-7-(3- moφholinopropoxy)quinolin-4-yloxy)phenyl)-2-oxo-l,2-dihydropyridine-3-carboxamide 184
Figure imgf000144_0001
[00546] Prepared from 3-fluoro-4-(6-methoxy-7-(3-morpholinopropoxy)quinolin-4- yloxy)aniline (prepared in Example 72, steps C-F) and l-(4-chlorobenzyl)-2-oxo-l,2- 111-03-PCT- P2338R1 - 02120.004WO1
144 dihydropyridine-3-carboxylic acid (prepared from methyl 2-oxo-l,2-dihydropyridine-3- carboxylate with l-(bromomethyl)-4-chlorobenzene and followed by hydrolysis using the methods described in Example 83, steps A and B) according to the procedure for Example 72. The crude was purified by silica gel flash column chromatography (7% MeOH in CH2Cl2) to afford 9 mg (57%) of 184. 1H-NMR (400 MHz, CD3OD/CDCI3) δ 8.59 (dd, IH), 8.41 (d, IH), 8.05 (dd, IH), 7.99 (dd, IH), 7.64 (s, IH), 7.49 (dd, IH), 7.34 (m, 6H), 6.65 (t, IH), 6.41 (d, IH), 5.32 (s, 2H), 4.27 (t, 2H), 4.03 (s, 3H), 3.74 (t, 4H), 2.66 (t, 2H), 2.56 (m, 4H), 2.15 (m, 2H); 19F NMR (376 MHz, CD3OD/CDCI3) δ -129.0. LRMS (ESI pos) m/e 673 (M+l). [00547] Example 85 Preparation of l-benzyl-N-(3-fluoro-4-(6-methoxy-7-(3- morpholinopropoxy)quinolin-4-yloxy)phenyl)-2-oxo- 1 ,2-dihydropyridine-3-carboxamide 185
Figure imgf000145_0001
[00548] Prepared from 3-fluoro-4-(6-methoxy-7-(3-morpholinopropoxy)quinolin-4- yloxy)aniline (prepared in Example 72, steps C-F) and l-benzyl-2-oxo-l,2-dihydropyridine-3- carboxylic acid according to the procedure for Example 72. The crude was purified by silica gel flash column chromatography (5% MeOH in CH2Cl2) to afford 9.2 mg (62%) of 185. 1H-NMR (400 MHz, CD3OD/CDCI3) δ 8.59 (dd, IH), 8.41 (d, IH), 8.02 (m, 2H), 7.64 (s, IH), 7.46 (m, IH), 7.34 (m, 7H), 6.63 (t, IH), 6.50 (d, IH), 5.35 (s, 2H), 4.26 (t, 2H), 4.03 (s, 3H), 3.74 (t, 4H), 2.65 (t, 2H), 2.55 (m, 4H), 2.14 (m, 2H); 19F NMR (376 MHz, CD3OD/CDCI3) δ -129.1. LRMS (ESI pos) m/e 639 (M+l).
[00549] Example 86 Preparation of N-(3-fluoro-4-(6-methoxy-7-(3- morpholinopropoxy)quinolin-4-yloxy)phenyl)- 1 -methyl-2-oxo- 1 ,2-dihydropyridine-3- carboxamide 186
Figure imgf000145_0002
[00550] Prepared from 3-fluoro-4-(6-methoxy-7-(3-morpholinopropoxy)quinolin-4- yloxy)aniline (prepared in Example 72, steps C-F) and l-(4-chlorobenzyl)-2-oxo-l,2- 111-03-PCT - P2338R1 - 02120.004WO1
145 dihydropyridine-3-carboxylic acid (prepared from methyl 2-oxo-l,2-dihydropyridine-3- carboxylate with iodomethane and followed by hydrolysis using the methods described in Example 83, steps A and B) according to the procedure for Example 72. The crude was purified by silica gel flash column chromatography (7% MeOH in CH2Cl2) to afford 8.5 mg (65%) of 186. 1H-NMR (400 MHz, CD3OD/CDCI3) δ 8.57 (dd, IH), 8.41 (d, IH), 8.0 (m, IH), 7.64 (s, IH), 7.46 (m, IH), 7.36 (m, 2H), 6.61 (t, IH), 6.51 (d, IH), 4.26 (t, 2H), 4.02 (s, 3H), 3.73 (t, 4H), 3.72 (s, 3H), 2.65 (t, 2H), 2.55 (m, 4H), 2.14 (m, 2H); 19F NMR (376 MHz, CD3OD/CDCI3) δ -129.3. LRMS (ESI pos) m/e 563 (M+l).
[00551] Example 87 Preparation of N-(3-fluoro-4-(6-methoxy-7-(3- morpholinopropoxy)quinolin-4-yloxy)phenyl)-2-oxo- 1 -(pyrimidin-4-ylmethyl)- 1 ,2- dihydropyridine-3-carboxamide 187
Figure imgf000146_0001
[00552] Prepared from 3-fluoro-4-(6-methoxy-7-(3-morpholinopropoxy)quinolin-4- yloxy)aniline (prepared in Example 72, steps C-F) and l-(4-chlorobenzyl)-2-oxo-l,2- dihydropyridine-3-carboxylic acid (prepared from methyl 2-oxo-l,2-dihydropyridine-3- carboxylate with 4-(chloromethyl)pyrimidine and followed by hydrolysis using the methods described in Example 83 steps A and B) according to the procedure for Example 72. The crude was purified by silica gel flash column chromatography (7% MeOH in CH2Cl2) to afford 10.5 mg (70%) of 187. 1H-NMR (400 MHz, CD3OD/CDC13) δ 9.11 (s, IH), 8.77 (d, IH), 8.67 (dd, IH), 8.41 (dd, IH), 8.09 (dd, IH), 7.96 (dd, IH), 7.63 (s, IH), 7.51 (d, IH), 7.42 (d, IH), 7.36 (s, IH), 7.29 (t, IH), 6.72 (t, IH), 6.48 (d, IH), 5.44 (s, 2H), 4.28 (t, 2H), 4.04 (s, 3H), 3.76 (t, 4H), 2.67 (t, 2H), 2.57 (m, 4H), 2.16 (m, 2H); 19F NMR (376 MHz, CD3OD/CDCI3) δ -128.5. LRMS (ESI pos) m/e 641 (M+l).
[00553] Example 88 Preparation of 4-benzyl-N-(3-fluoro-4-(6-methoxy-7-(3- morpholinopropoxy)quinolin-4-yloxy)phenyl)-3-oxo-3,4-dihydropyrazine-2-carboxamide 188 111-03-PCT - P2338R1 - 02120.004WO1
146
Figure imgf000147_0001
[00554] Step A: Preparation of methyl 4-benzyl-3-oxo-3,4-dihydropyrazine-2- carboxylate: LiH (7.8 mg, 0.980 mmol) was added to the solution of methyl 3-oxo-3,4- dihydropyrazine-2-carboxylate (100 mg, 0.65 mmol) in DMF (3 mL) at O0C. After 30 minutes stirring, (chloromethyl)benzene (0.15 mL, 1.30 mmol) was added to the reaction mixture at O0C, and then the reaction was warmed to room temperature. After 4 hours stirring, the reaction mixture was quenched with ice water, extracted with EtOAc, washed with brine, dried over MgSO4, and concentrated to give the crude material that was purified by silica gel flash column chromatography (2% MeOH in CH2Cl2) to afford 0.102 g (64%) of the desired product. 1H-NMR (400 MHz, CDCl3) δ 7.38 (m, 6H), 7.29 (d, IH), 5.14 (s, 2H), 3.98 (s, 3H). [00555] Step B: Preparation of 4-benzyl-3-oxo-3,4-dihydropyrazine-2-carboxylic acid:
LiOH (0.82 mL, 0.82 mmol, 1.0 M in H2O) was added to a solution of methyl 4-benzyl-3-oxo- 3,4-dihydropyrazine-2-carboxylate (100 mg, 0.41 mmol) in a mixture of THF (4.5 mL) and MeOH (1.5 mL) at room temperature for 4 hours. The reaction mixture was acidified to pH 1 with aq. 1 N HCl solution and treated with water (5 mL), extracted with EtOAc, washed with brine, dried over MgSθ4, and concentrated to afford 77 mg (82%) of the desired product. 1H- NMR (400 MHz, CD3OD) δ 8.0 (d, IH), 7.68 (d, IH), 7.36 - 7.42 (m, 5H), 5.29 (s, 2H). [00556] Step C: Preparation of 4-benzyl-N-(3-fluoro-4-(6-methoxy-7-(3- moφholinopropoxy)quinolin-4-yloxy)phenyl)-3-oxo-3,4-dihydropyrazine-2-carboxamide: Prepared from 3-fluoro-4-(6-methoxy-7-(3-morpholinopropoxy)quinolin-4-yloxy)aniline (prepared in Example 72, steps C-F) and 4-benzyl-3-oxo-3,4-dihydropyrazine-2-carboxylic acid according to the procedure for Example 72. The crude was purified by silica gel flash column chromatography (7% MeOH in CH2Cl2) to afford 24.8 mg (83%) of 188. 1H-NMR (400 MHz, CD3OD) δ 8.43 (d, IH), 8.06 (dd, IH), 8.01 (d, IH), 7.73 (d, IH), 7.65 (s, IH), 7.55 (d, IH), 7.45 (m, IH), 7.38 (m, 6H), 6.52 (d, IH), 5.33 (s, 2H), 4.25 (t, 2H), 4.01 (s, 3H), 3.72 (t, 4H), 2.64 (t, 2H), 2.54 (m, 4H), 2.13 (m, 2H). LRMS (APCI pos) m/e 640 (M+l).
[00557] Example 89 Preparation of N-(3-fluoro-4-(6-methoxy-7-(3- morpholinopropoxy)quinolin-4-yloxy)phenyl)-2-(4-fluorophenyl)-3-oxo-2,3-dihydropyridazine- 4-carboxamide 189 111-03-PCT - P2338R1 - 02120.004WO1
147
Figure imgf000148_0001
[00558] Step A: Preparation of (E)-2-(2-(4-fluorophenyl)hydrazono)acetaldehyde: A mixture of the 4-fluorophenylhydrazine HCl salt (5.0 g, 30.75 mmol), water (20 mL), and acetic acid (20 mL) was added with stiring to a 40% aqueous solution of glyoxal (17.6 mL, 153.8 mmol) during 20 minutes. Stirring was continued for 2 hours and the mixture was then filtered. The precipitate was washed with water and dried to afford 5.0 g (98%) of the desired product. 1H-NMR (400 MHz, CDCl3) δ 9.56 (d, IH), 8.63 (br. s, IH), 7.24 (m, IH), 7.16 (m, 2H), 7.06 (m, 2H); 19F NMR (376 MHz, CDCl3) δ -120.3. LRMS (ESI pos) m/e 151 (M-16). [00559] Step B: Preparation of (E)-5-(2-(2-(4-fluorophenyl)hydrazono)ethylidene)-2,2- dimethyl-l,3-dioxane-4,6-dione: A suspension of the dioxan-dione (1.44 g, 10.0 mmol) and (E)- 2-(2-(4-fluorophenyl)hydrazono)acetaldehyde (1.66 g, 10.0 mmol) in toluene (15 mL) was treated with acetic acid (5 drops) and with piperidine (5 drops). The reaction mixture was then stirred at room temp for 17 hours. The precipitated condensation product was filtered off and thoroughly washed with light petroleum to afford 2.87 g (98%) of the desired product. 1H-NMR (400 MHz, CD3OD/CDCI3) δ 8.72 (d, IH), 8.24 (d, IH), 7.32 (m, 2H), 7.08 (t, 2H), 1.76 (s, 6H); 19F NMR (376 MHz, CD3OD/CDCI3) δ -119.1.
[00560] Step C: Preparation of 2-(4-fluorophenyl)-3-oxo-2,3-dihydropyridazine-4- carboxylic acid: A mixture of (E)-5-(2-(2-(4-fluorophenyl)hydrazono)ethylidene)-2,2-dimethyl- l,3-dioxane-4,6-dione (0.60 g, 2.05 mmol) and NaOMe (0.133 g, 2.46 mmol) in MeOH (10 mL) was heated under reflux for 15 hours. The salt was treated with cold 1 N HCl solution, extrated with DCM, dried over MgSO4, and concentrated to afford 0.42 g (87%) ot the desired product. 1H-NMR (400 MHz, CDCl3) δ 13.57 (br. s, IH), 8.29 (m, 2H), 7.63 (m, 2H), 7.24 (m, 2H); 19F NMR (376 MHz, CDCl3) δ -110.7. LRMS (ESI pos) m/e 235 (M+l).
[00561] Step D: Preparation of N-(3-fluoro-4-(6-methoxy-7-(3- morpholinopropoxy)quinolin-4-yloxy)phenyl)-2-(4-fluorophenyl)-3-oxo-2,3-dihydropyridazine- 4-carboxamide: Prepared from 3-fluoro-4-(6-methoxy-7-(3-morpholinopropoxy)quinolin-4- yloxy)aniline (prepared in Example 72, steps C-F) and 2-(4-fluorophenyl)-3-oxo-2,3- dihydropyridazine-4-carboxylic acid according to the procedure for Example 72. The crude was purified by silica gel flash column chromatography (5% MeOH in CH2Cl2) to afford 10 mg 111-03-PCT- P2338R1 - 02120.004WO1
148
(66%) of 189. 1H-NMR (400 MHz, CDCI3/CD3OD) δ 8.41 (d, IH), 8.38 (d, IH), 8.32 (d, IH), 8.01 (dd, IH), 7.66 (m, 2H), 7.63 (s, IH), 7.43 (m, IH), 7.34 (m, 2H), 7.28 (t, 2H), 6.50 (d, IH), 4.26 (t, 2H), 4.03 (s, 3H), 3.74 (t, 4H), 2.65 (t, 2H), 2.56 (m, 4H), 2.15 (m, 2H); 19F NMR (376 MHz, CDCI3/CD3OD) δ -113.7, -128.6. LRMS (ESI pos) m/e 644 (M+l). [00562] Example 90 Preparation of N-(3-fluoro-4-(6-methoxy-7-(3-(4- methylpiperazin-l-yl)propoxy)quinolin-4-yloxy)phenyl)-2-(4-fluorophenyl)-3-oxo-2,3- dihydropyridazine-4-carboxamide 190
Figure imgf000149_0001
[00563] Step A: Preparation of 4-(2-fluoro-4-nitrophenoxy)-6-methoxy-7-(3-(4- methylpiperazin-l-yl)propoxy)quinoline: To a stirred suspension of the 4-(2-fluoro-4- nitrophenoxy)-6-methoxyquinolin-7-ol (Example 72, step D, 0.15 g, 0.454 mmol) in CH2Cl2 (4 mL) at room temperature under nitrogen was added the 3-(4-methylpiperazin-l-yl)propan-l-ol (0.086 g, 0.545 mmol) followed by PPh3 (0.191 g, 0.727 mmol) and (E)-diethyl diazene-1,2- dicarboxylate (0.127 g, 0.727 mmol). After 17 hours stirring, the reaction was concentrated to a residue under reduced pressure. The crude was purified by silica gel flash column chromatography (10% MeOH in CH2Cl2) to afford 0.185 mg (87%) of the desired product. LRMS (ESI pos) m/e 471 (M+l).
[00564] Step B: Preparation of 3-fluoro-4-(6-methoxy-7-(3-(4-methylpiperazin-l- yl)propoxy)quinolin-4-yloxy)aniline: 10% Pd/C (0.105 g, 0.197 mmol, 20% Wt) was added to a solution of 4-(2-fluoro-4-nitrophenoxy)-6-methoxy-7-(3-(4-methylpiperazin- 1 - yl)propoxy)quinoline in a mixture of THF (6 mL) and EtOH (3 mL) at room temperature and then the mixture was held under 1 atmosphere of hydrogen gas pressure. After 17 hours stirring, the mixture was filtered with MeOH and concentrated under reduced pressure to give the desired product (0.17 g, 98%). 1H-NMR (400 MHz, CD3OD) δ 8.39 (d, IH), 7.63 (s, IH), 7.33 (s, IH), 7.04 (t, IH), 6.62 (dd, IH), 6.57 (m, IH), 6.46 (d, IH), 4.24 (t, 2H), 4.0 (s, 3H), 2.68 (t, 2H), 2.62 (m, 8H), 2.35 (s, 3H), 2.12 (m, 2H); 19F NMR (376 MHz, CD3OD) δ -132.4. LRMS (ESI pos) m/e 441 (M+l).
[00565] StepC: Preparation of N-(3-fluoro-4-(6-methoxy-7-(3-(4-methylpiperazin-l- yl)propoxy)quinolin-4-yloxy)phenyl)-2-(4-fluorophenyl)-3-oxo-2,3-dihydropyridazine-4- 1 11-03-PCT - P2338R1 - 02120.004WO1
149 carboxamide: Prepared from 3-fluoro-4-(6-methoxy-7-(3-(4-methylpiperazin-l- yl)propoxy)quinolin-4-yloxy)aniline and 2-(4-fluorophenyl)-3-oxo-2,3-dihydropyridazine-4- carboxylic acid (Example 89, step C) according to the procedure for Example 72. The crude was purified by silica gel flash column chromatography (10 to 20% MeOH in CH2Cl2) to afford 9.2 mg (31%) of 190. 1H-NMR (400 MHz, CD3OD) δ 8.42 (d, IH), 8.35 (d, IH), 8.30 (d, IH), 8.04 (dd, IH), 7.68 (m, 2H), 7.64 (s, IH), 7.50 (m, IH), 7.38 (t, IH), 7.35 (s, IH), 7.29 (t, 2H), 6.51 (d, IH), 4.24 (t, 2H), 4.0 (s, 3H), 2.65 (t, 2H), 2.55 (m, 6H), 2.30 (s, 3H), 2.12 (m, 2H); 19F NMR (376 MHz, CD3OD) δ -114.8, -129.6. LRMS (APCI pos) m/e 657 (M+l). [00566] Example 91 Preparation of N-(3-fluoro-4-(6-methoxy-7-(3-(piperidin-l- yl)propoxy)quinolin-4-yloxy)phenyl)-2-(4-fluorophenyl)-3-oxo-2,3-dihydropyridazine-4- carboxamide 191
Figure imgf000150_0001
[00567] Prepared from 3-fluoro-4-(6-methoxy-7-(3-(piperidin-l-yl)propoxy)quinolin-4- yloxy)aniline (prepared from 4-(2-fluoro-4-nitrophenoxy)-6-methoxyquinolin-7-ol with 3- (piperidin-l-yl)propan-l-ol and followed by hydrogenation using the methods described in Example 90, steps A and B) and Example 89C 2-(4-fluorophenyl)-3-oxo-2,3-dihydropyridazine- 4-carboxylic acid according to the procedure for Example 72. The crude was purified by silica gel flash column chromatography (10 to 20% MeOH in CH2Cl2) to afford 20 mg (66%) of 191. 1H-NMR (400 MHz, CDC13/CD3OD) δ 8.43 (d, IH), 8.38 (d, IH), 8.32 (d, IH), 8.03 (dd, IH), 7.68 (m, 2H), 7.65 (s, IH), 7.49 (m, IH), 7.36 (t, 2H), 7.29 (t, 2H), 6.52 (d, IH), 4.27 (t, 2H), 4.03 (s, 3H), 2.89 (m, 2H), 2.80 (m, 4H), 2.23 (m, 2H), 1.74 (m, 4H), 1.59 (m, 2H); 19F NMR (376 MHz, CDC13/CD3OD) δ -114.0, -128.8. LRMS (APCI pos) m/e 642 (M+l). [00568] Example 92 Preparation of 2-(4-fluorophenyl)-N-(6-(6-methoxy-7-(3- morpholinopropoxy)quinolin-4-yloxy)pyridin-3-yl)-3-oxo-2,3-dihydropyridazine-4-carboxamide 192 111-03-PCT - P2338R1 - 02120.004WO1
150
Figure imgf000151_0001
[00569] Step A: Preparation of 7-(benzyloxy)-6-methoxy-4-(5-nitropyridin-2- yloxy)quinoline: To a stirred solution of 7-(benzyloxy)-6-methoxyquinolin-4-ol (562 mg, 2 mmol) (reference for preparation given in Example 73, step C)in 20 mL CH3CN at room temperature under nitrogen was added cesium carbonate (716 mg, 2.2 mmol). After 5 minutes 2- chloro-5-nitropyridine (384 mg, 2.2 mmol) was added. The reaction was allowed to proceed overnight. The reaction was diluted to 30 mL with EtOAc and washed 4 x 30 mL with H2O/brine and then 1 x 30 mL with brine. The organics were dried (MgSO4), filtered and concentrated. The residue was purified by silica gel flash column chromatography (1/1 EtOAc/hexanes). Product containing fractions were pooled and concentrated to a brown solid (177 mg, 22%). 1H-NMR (400 MHz, CDCl3) δ 9.06 (d, IH), 8.71 (d, IH), 8.58 (m, IH), 7.52 (s, IH), 7.50 (d, IH), 7.39 (m, 2H), 7.33 (m, IH), 7.24(d, IH), 7.16 (s, IH), 7.05 (d, IH), 5.32 (s, 2H), 3.94 (s, 3H).
[00570] Step B: Preparation of 6-methoxy-4-(5-nitropyridin-2-yloxy)quinolin-7-ol: A suspension of 7-(benzyloxy)-6-methoxy-4-(5-nitropyridin-2-yloxy)quinoline (130 mg, 0.32 mmol) was stirred in 300 uL 33 wt % HBr in acetic acid. After 4 hours a tan precipitate had formed. The reaction was diluted with 5 mL diethyl ether and filtered, rinsing with ether. The isolated solid is presumably the di HBr salt. This material was dissolved in 20 mL 4:1 CH2Cl2:Me0H and stirred with 20 mL of water (pH was <3). The pH was raised to approximately 6-7 with saturated NaHCO3. A little more MeOH was added to make the mixture a biphasic solution with no precipitate present. The organics were isolated, dried (MgSO4), filtered and concentrated to a yellow solid (80 mg, 79%). HBr Salt: 1H-NMR (400 MHz, d6- DMSO) δ 9.18 (d, IH), 9.00 (d, IH), 8.89 (m, IH), 7.76 (d, IH), 7.66 (d, IH), 7.65 (s, IH), 7.56 (s, IH), 4.00 (s, 3H).
[00571] Step C: Preparation of 6-methoxy-7-(3-morpholinopropoxy)-4-(5-nitropyridin-2- yloxy)quinoline: To a stirred suspension of 6-methoxy-4-(5-nitropyridin-2-yloxy)quinolin-7-ol (150 mg, 0.48 mmol) in 1.5 mL CH2Cl2 at room temperature under nitrogen was added 3- morpholinopropan-1-ol (106 uL, 0.77 mmol) followed by triphenylphosphine (201 mg, 0.77 mmol) and finally DEAD (121 uL, 0.77 mmol). After stirring overnight, the reaction was concentrated to a residue by rotovap and purified directly by silica gel flash column 111-03-PCT- P2338R1 - 02120.004WO1
151 chromatography (9/1 EtOAc/MeOH). Product containing fractions were pooled and concentrated to a tan foam (100 mg, 47%). 1H-NMR (400 MHz, CDCl3) δ 9.07 (d, IH), 8.73 (d, IH), 8.58 (m, IH), 7.50 (s, IH), 7.25 (d, IH), 7.15 (s, IH), 7.06 (d, IH), 4.28 (m, 2H), 3.92 (s, 3H), 3.72 (m, 4H), 2.57 (m, 2H), 2.48 (m, 4H), 2.13 (m, 2H).
[00572] Step D: Preparation of 6-(6-methoxy-7-(3-morpholinopropoxy)quinolin-4- yloxy)pyridin-3-amine: A solution of 6-methoxy-7-(3-morpholinopropoxy)-4-(5-nitropyridin-2- yloxy)quinoline (100 mg, 0.23 mmol) was formed in 25 mL of 95% EtOH and 25 mL EtOAc in a 250 mL Parr bottle. Pearlman's catalyst (160 mg, 0.23 g/atom) was added and the reaction put through a vacuum/purge cycle three times with hydrogen gas and then held under 50 psi hydrogen and shaken overnight. The reaction was filtered through GF/F filter paper with 95% EtOH and concentrated to a yellow foam (93 mg, 100%). 1H-NMR (400 MHz, CDCl3) δ 8.49 (d, IH), 7.84 (d, IH), 7.54 (s, IH), 7.44 (s, IH), 7.21 (m, IH), 6.98 (d, IH), 6.63 (d, IH), 4.28 (m, 2H), 4.00 (s, 3H), 3.81 (m, 4H), 2.73 (m, 2H), 2.65 (m, 4H), 2.22 (m, 2H). [00573] Step E: Prepared from 6-(6-methoxy-7-(3-morpholinopropoxy)quinolin-4- yloxy)pyridin-3-amine and 2-(4-fluorophenyl)-3-oxo-2,3-dihydropyridazine-4-carboxylic acid (Example 89C) according to the procedure for Example 72. The crude was purified by silica gel flash column chromatography (7% MeOH in CH2Cl2) to afford 10 mg (33%) of 192. 1H-NMR (400 MHz, CD3OD) δ 8.63 (d, IH), 8.53 (d, IH), 8.41 (d, IH), 8.38 (dd, IH), 8.32 (d, IH), 7.64 (m, 2H), 7.47 (s, IH), 7.38 (s, IH), 7.27 (m, 3H), 6.87 (d, IH), 4.28 (t, 2H), 3.99 (s, 3H), 3.76 (t, 4H), 2.66 (t, 2H), 2.57 (m, 4H), 2.16 (m, 2H); 19F NMR (376 MHz, CD3OD) δ -112.4. LRMS (APCI pos) m/e 627 (M+ 1).
[00574] Example 93 Preparation of N-(3-fluoro-4-(6-methoxy-7-(3-(4- methylpiperazin-l-yl)propoxy)quinolin-4-yloxy)phenyl)-2-(4-fluorophenyl)-6-methyl-3-oxo-2,3- dihydropyridazine-4-carboxamide 193
Figure imgf000152_0001
[00575] Step A: Preparation of (E)-2-(2-(4-fluorophenyl)hydrazono)propanal and l-(2-(4- fluorophenyl)hydrazono)propan-2-one: A mixture of (4-fluorophenyl)hydrazine HCl salt (2.0 g, 12.30 mmol), water (10 mL), and acetic acid (10 mL) was added with stiring to a 40% aqueous solution of 2-oxopropanal (9.41 mL, 61.5 mmol) during 20 minutes. Stirring was continued for 4 111-03-PCT- P2338R1 - 02120.004WO1
152 hours and the mixture was then filtered. The precipitate was washed with water and dried to afford the desired products. The crude was purified by silica gel flash column chromatography (1 :50 to 1 : 10 EtOAc/CH2Cl2) to afford 2.05 g (93%) of both desired products. [00576] (E)-2-(2-(4-fluorophenyl)hydrazono)propanal: 1H-NMR (400 MHz, CDCl3) δ
9.47 (s, IH), 8.09 (br. s, IH), 7.24 (m, 2H), 7.06 (t, 2H), 1.98 (s, 3H); 19F NMR (376 MHz, CDCl3) δ -121.0.
[00577] l-(2-(4-fluorophenyl)hydrazono)propan-2-one (two isomers - cis and trans): 1H-
NMR (400 MHz, CDCl3) δ 8.23 (br. s, IH), 7.22 (m, 2H isomer b), 7.13 (m, Tϋisomer b), 7.04 (m, misomer a), 6.96 (s, IH), 2.44 (s, VAisomer a), 2.67 (s, 3H isomer b); 19F NMR (376 MHz, CDCl3) δ -120.2, -121.4.
[00578] Step B: Preparation of 2-(4-fluorophenyl)-6-methyl-3-oxo-2,3-dihydropyridazine-
4-carboxylic acid: A suspension of 2,2-dim ethyl- l,3-dioxane-4,6-dione (0.71 g, 4.93 mmol) and (E)-2-(2-(4-fluorophenyl)hydrazono)propanal (0.889 g, 4.934 mmol) in toluene (20 mL) was treated with acetic acid (5 drops) and with piperidine (5 drops). The reaction mixture was then stirred at room temperature for 17 hours. The precipitated condensation - cyclization product (2 steps in one pot reaction) was filtered off and thoroughly washed with light petroleum to afford 0.709 g (58%) of the desired product. 1H-NMR (400 MHz, CD3OD) δ 7.96 (s, IH), 7.61 (m, 2H), 7.24 (t, 2H), 2.45 (s, 3H); 19F NMR (376 MHz, CD3OD) δ -115.1. LRMS (ESI pos) m/e 249 (M+l).
[00579] Step C: Preparation of N-(3-fluoro-4-(6-methoxy-7-(3-(4-methylpiperazin-l- yl)propoxy)quinolin-4-yloxy)phenyl)-2-(4-fluorophenyl)-6-methyl-3-oxo-2,3-dihydropyridazine- 4-carboxamide: Prepared from 3-fluoro-4-(6-methoxy-7-(3-(4-methylpiperazin-l- yl)propoxy)quinolin-4-yloxy)aniline (prepared in example 72, steps C-F) and 2-(4-fluorophenyl)- 6-methyl-3-oxo-2,3-dihydropyridazine-4-carboxylic acid according to the procedure for Example 72. The crude was purified by silica gel flash column chromatography (10% MeOH in CH2Cl2) to afford 7.4 mg (19%) of 193. 1H-NMR (400 MHz, CD3OD) δ 8.40 (d, IH), 8.26 (s, IH), 8.02 (dd, IH), 7.65 (m, 2H), 7.62 (s, IH), 7.46 (m, IH), 7.37 (t, IH), 7.34 (s, IH), 7.27 (t, 2H), 6.49 (d, IH), 4.23 (t, 2H), 3.99 (s, 3H), 2.65 (t, 2H), 2.55 (m, 6H), 2.49 (s, 3H), 2.31 (s, 3H), 2.11 (m, 2H); 19F NMR (376 MHz, CD3OD) δ -115.0, -129.5. LRMS (ESI pos) m/e 671 (M+l). [00580] Example 94 N-(3-Fluoro-4-(6-methoxy-7-(3-morpholinopropoxy)quinolin-4- yloxy)phenyl)pyridin-2-amine 194 111-03-PCT - P2338R1 - 02120.004WO1
Figure imgf000154_0001
[00581] Step A: Preparation of N-(3-fluoro-4-methoxyphenyl)pyridin-2 -amine: A mixture of pyridin-2 -amine (0.433 g, 4.60 mmol), 4-bromo-2-fluoro-l-methoxybenzene (1.23 g, 5.98 mmol), Pd2(dba)3 (0.421 g, 0.460 mmol), 4,5-bis(diphenylphosphino)-9,9-dimethyl-9H-xanthene (0.799 g, 1.38 mmol), Cs2CO3 (3.00 g, 9.20 mmol), in dioxane (25 mL) was stirred at 1000C for 16 hours. Water (25 mL) was added and extracted with CH2Cl2 (3 x 100 mL). The combined organic layers were dried over Na2SCV Concentration and purification by silica gel flash column chromatography afforded the desired product (0.59 g, 59%). 1H NMR (400 MHz, CDCl3) δ 8.17 (m, IH), 7.48 (m, IH), 7.22 (dd, J = 13.0, 2.6 Hz, IH), 7.01 (m, IH), 6.92 (t, J - 9.0 Hz, IH), 6.82 (s, br, IH, NH), 6.69-6.75 (m, 2H). LRMS (ESI pos) m/e 219 (M+l). [00582] Step B: Preparation of 2-fluoro-4-(pyridin-2-ylamino)phenol: A mixture of N-(3- fluoro-4-methoxyphenyl)pyridin-2-amine (0.587g, 2.690 mmol) and tribromoborane (3.369 g, 13.45 mmol) in CH2Cl2 (50 mL) was stirred at O0C for 4 hours. Saturated NaHCO3 was added and then extracted with CH2Cl2 (3 x 100 mL). The combined organic layers were dried over Na2SO4. Concentration afforded the crude product (0.48 g, 88%), which was used in the next step without further purification. 1H NMR (400 MHz, CDCl3) δ 8.17 (m, IH), 7.48 (m, IH), 7.17 (dd, J - 12.0, 2.4 Hz, IH), 6.88-6.97 (m, 2H), 6.70-6.74 (m, 2H), 6.45 (br s, IH). LRMS (ESI pos) m/e 205(M+l).
[00583] Step C: Preparation of N-(3-fluoro-4-(6-methoxy-7-(3- moipholinopropoxy)quinolin-4-yloxy)phenyl)pyridin-2-amine: A mixture of 4-chloro-6- methoxy-7-(3-morpholinopropoxy)quinoline (prepared according to WO 01/55116, Example 2, 20 mg, 0.059 mmol), 2-fluoro-4-(pyridin-2-ylamino)phenol (14.6 mg, 0.0713 mmol) and N,N- dimethylpyridin-4-amine (0.725 mg, 0.00594 mmol) in bromobenzene (10 mL) was stirred at
15O0C for 2 days. Water (10 mL) was added and the aqueous extracted with CH2Cl2 (3 x 50 mL). The organic layers were combined and dried over Na2SO4. The crude product was concentrated and purified by silica gel flash column chromatography to afford 194 (15.8 mg, 53%). 1H NMR (400 MHz, CDCl3)..δ 8.49 (d, J = 5.2 Hz, IH), 8.27 (dd, J = 5.2, 1.6 Hz, IH), 7.63 (dd, J = 12.4, 2.4 Hz, IH), 7.59 (s, IH), 7.55-7.58 (m, IH), 7.44 (s, IH), 7.14-7.22 (m, 2H), 6.82-6.85 (m, 2H), 6.58 (s, IH, NH), 6.45 (d, J = 5.2 Hz, IH), 4.28 (t, J = 6.8 Hz, 2H), 4.05 (s, 111-03-PCT - P2338R1 - 02120.004WO1
154
3H), 3.73 (t, J = 4.6 Hz, 4H), 2.58 (t, J = 7.2 Hz, 2H), 2.49 (m, 4H), 2.10-2.17 (m, 2H). LRMS (APCI neg) m/z 503 (M-I).
[00584] Example 95 N-(3-Fluoro-4-(6-methoxy-7-(3-morpholinopropoxy)quinolin-4- yloxy)phenyl)- 1 -methyl-2-oxopyrrolidine-3-carboxamide 195
Figure imgf000155_0001
[00585] Step A: Preparation of methyl l-methyl-2-oxopyrrolidine-3-carboxylate: LDA
(43.77 niL, 78.79 mmol) was added into a solution of l-methylpyrrolidin-2-one (5.2Og, 52.53 mmol) in THF (125 mL) under -780C. The mixture was stirred for 30 minutes, and then methyl carbonochloridate (7.45g, 78.79 mmol) was added. The mixture was stirred for four hours at room temperature. Water (150 mL) was added and the aqueous phase was extracted with CH2Cl2 (3 x 150 mL). The organic layers were combined and dried over Na2SO4. Concentration to afford the crude desired product (7.35g, 89%) without further purification. 1H NMR (400 MHz, CD3Cl)..δ 3.37 (s, 3H), 3.28-3.25 (m, 2H), 2.85 (s, 3H), 2.62-2.67 (m, IH), 2.13-2.22 (m, IH), 1.99-2.06 (m, IH).
[00586] Step B: Preparation of l-methyl-2-oxopyrrolidine-3-carboxylic acid: A mixture of methyl l-methyl-2-oxopyrrolidine-3-carboxylate (1.89 g, 12.04 mmol) and TMSOK (4.64 g, 36.13 mmol) in THF (100 mL) was stirred overnight at room temperature. HCl (50 mL, 100 mmol, 2.0 M in Et2O) was added and the mixture was stirred for 20 minutes. The solid was removed by filtration with Et2O. The filtrated was then concentrated under reduced pressure to afford the crude product (1.28 g, 74.2%), which was used for next step without further purification. 1H NMR (400 MHz, CDCl3).δ 3.40-3.49 (m, 3H), 2.94 (s, 3H), 2.37-2.47 (m, 2H). [00587] Step C: Preparation of N-(3-fluoro-4-(6-methoxy-7-(3- morpholinopropoxy)quinolin-4-yloxy)phenyl)- 1 -methyl-2-oxopyrrolidine-3-carboxamide: A mixture of 3-fluoro-4-(6-methoxy-7-(3-morpholinopropoxy)quinolin-4-yloxy)aniline (prepared in Example 72, steps C-F) (10.0 mg, 0.0234 mmol), l-methyl-2-oxopyrrolidine-3-carboxylic acid (16.7 mg, 0.117 mmol), N1-((ethylimino)methylene)-N3,N3-dimethylpropane-l,3-diamine hydrochloride (22.4 mg, 0.117 mmol), lH-benzo[d][l,2,3]triazol-l-ol (15.8 mg, 0.117 mmol) and N-ethyl-N-isopropylpropan-2 -amine (0.0204 ml, 0.117 mmol) in CH2Cl2 (10 mL) was stirred at room temperature for 2 days. Water (10 mL) was added and the aqueous phase was extracted with CH2Cl2 (3 x 50 mL). The organic layers were combined and dried over Na2SO4. 111-03-PCT - P2338R1 - 02120.004WO1
155
Concentration and purification by silica gel flash column chromatography afforded 195 (12.4 mg, 96%). 1H NMR (400 MHz, CDCl3) δ 10.02 (s, IH, NH), 8.48 (d, J = 5.2 Hz, IH), 7.80 (dd, J = 12.0, 2.4 Hz, IH), 7.57 (s, IH), 7.44 (s, IH), 7.30 (m, IH), 7.21 (t, J = 8.8 Hz, IH), 6.39 (d, J - 5.2 Hz, IH), 4.28 (t, J - 6.8 Hz, 2H), 4.04 (s, 3H), 3.73 (t, J = 4.8 Hz, 4H), 3.43-3.49 (m, 3H), 2.95 (s, 3H), 2.58 (t, J = 7.2 Hz, 2H), 2.40-2.54 (m, 6H), 2.10-2.17 (m, 2H). LRMS (APCI neg) m/z 551 (M-I).
[00588] Example 96 N-(3-Fluoro-4-(6-methoxy-7-(3-morpholinopropoxy)quinolin-4- yloxy)phenyl)-2-oxopyrrolidine-3-carboxamide 196
Figure imgf000156_0001
[00589] Step A: Preparation of 2-oxopyrrolidine-3-carboxylic acid: A mixture of ethyl 1- methyl-2-oxopyrrolidine-3-carboxylate (0.50 g, 3.18 mmol) and TMSOK (1.34 g, 10.48 mmol) in THF (10 mL) was stirred overnight at room temperature. HCl (20 mL, 100 mmol, 2.0 M in Et20) was added and the mixture stirred for 20 minutes. The solid was removed by filtration with Et2O and the filtrate was concentrated under reduced pressure to afford the crude product (0.19 g, 42%), which was used for next step without further purification.
[00590] Step B: Preparation of N-(3-fluoro-4-(6-methoxy-7-(3- moφholinopropoxy)quinolin-4-yloxy)phenyl)-2-oxopyrrolidine-3-carboxamide: A mixture of 3- fluoro-4-(6-methoxy-7-(3-morpholinopropoxy)quinolin-4-yloxy)benzenamine (prepared in Example 72, steps C-F) (15.0 mg, 0.0351 mmol), 2-oxopyrrolidine-3-carboxylic acid (22.7 mg, 0.175 mmol), N1-((ethylimino)methylene)-N3,N3-dimethylpropane-l,3-diamine hydrochloride (33.6 mg, 0.175 mmol), ^-benzo^HU^triazol-l-ol (23.7 mg, 0.175 mmol) and N-ethyl-N- isopropylpropan-2-amine (22.7 mg, 0.175 mmol) in THF (10 mL) was stirred at room temperature for 16 hours. Water (10 mL) was added and the aqueous phase extracted with CH2Cl2 (3 x 50 mL). The combined organic layers were dried over Na2SO4. Concentration and purification by silica gel flash column chromatography afforded 196 (17.3 mg, 92%). 1H NMR (400 MHz, CDCl3).δ 9.84 (s, IH, NH), 8.48 (d, J = 5.6 Hz, IH), 7.80 (dd, J - 12.0, 2.4 Hz, IH), 7.57 (s, IH), 7.44 (s, IH), 7.30 (m, IH), 7.21 (t, J = 8.8 Hz, IH), 6.39 (d, J = 5.6 Hz, IH), 5.77 (s, IH, NH), 4.28 (t, J = 6.6 Hz, 2H), 4.04 (s, 3H), 3.73 (t, J = 4.6 Hz, 4H), 3.38-3.51 (m, 3H), 2.53- 2.73 (m, 2H), 2.58 (t, J = 7.2 Hz, 2H), 2.49 (m, 4H), 2.10-2.17 (m, 2H). LRMS (APCI neg) m/z 537 (M-I). 111-03-PCT- P2338R1 - 02120.004WO1
156
[00591] Example 97 N-(3-fluoro-4-(6-methoxy-7-(3-morpholinopropoxy)quinolin-4- yloxy)phenyl)-7,7-dimethyl-2-oxobicyclo[2.2.1]heptane-l-carboxamide 197
Figure imgf000157_0001
[00592] A mixture of 3-fiuoro-4-(6-methoxy-7-(3-morpholinopropoxy)quinolin-4- yloxy)benzenamine (prepared in Example 72, steps C-F) (10.0 mg, 0.0234 mmol), (lS)-7,7- dimethyl-2-oxobicyclo[2.2.1]heptane-l-carboxylic acid (21.3 mg, 0.117 mmol), N1- ((ethylimino)methylene)-N3,N3-dimethylpropane-l,3-diamine hydrochloride (22.4 mg, 0.117 mmol), 1H-benzo[d][l,2,3]triazol-l-ol (15.8 mg, 0.117 mmol) and N-ethyl-N-isopropylpropan-2- amine (0.0204 ml, 0.117 mmol) in THF (10 mL) was stirred at room temperature for 4 days. Water (10 mL) was added and the aqueous phase extracted with CH2Cl2 (3 x 50 mL). The organic layers were combined and dried over Na2SO4. Concentration and purification by silica gel flash column chromatography afforded 197 (2.4 mg, 17%). 1H NMR (400 MHz, CDCl3)..δ 9.94 (s, IH, NH), 8.47 (d, J = 5.2 Hz, IH), 7.84 (dd, J = 12.4, 2.4 Hz, IH), 7.58 (s, IH), 7.43 (s, IH), 7.32 (m, IH), 7.21 (t, J = 8.8 Hz, IH), 6.39 (d, J = 5.2 Hz, IH), 4.28 (t, J = 6.6 Hz, 2H), 4.04 (s, 3H), 3.73 (t, J = 4.6 Hz, 4H), 2.58 (t, J = 7.2 Hz, 2H), 2.49 (m, 4H), 2.12-2.17 (m, 2H), 1.56 (s, 3H), 1.18-1.32 (m, 3H), 1.26 (s, 3H), 0.78-0.94 (m, 4H). LRMS (APCI neg) m/z 590 (M-I). [00593] Example 98 N-(3-Fluoro-4-(6-methoxy-7-(3-morpholinopropoxy)quinolin-4- yloxy)phenyl)-3-(pyridin-2-yloxy)pyridin-2-amine 198
Figure imgf000157_0002
[00594] Step A: Preparation of 3-(pyridin-2-yloxy)pyridin-2-amine: A mixture of 2- aminopyridin-3-ol (0.50g, 4.54 mmol), Cs2CO3 (4.44 g, 13.6 mmol) and 2-fluoropyridine (0.441 g, 4.54 mmol) in DMF (25 mL) was stirred at 1000C for 4 hours. The reaction mixture was cooled to room temperature, water (25 mL) was added and then the aqueous phase extracted with 111-03-PCT - P2338R1 - 02120.004WO1
157
CH2Cl2 (3 x 100 mL). The combined organic layers were dried over Na2SO4. Concentration and purification by silica gel flash column chromatography afforded the desired product (0.823 g, 97%). 1H NMR (400 MHz, CDCl3) δ 8.20 (m, IH), 7.95 (dd, J = 5.0, 1.4 Hz, IH), 7.71 (m, IH), 7.29 (dd, J
= 7.8, 1.4 Hz, IH), 7.03 (m, IH), 6.94 (d, J = 8.0 Hz, IH), 6.71 (dd, J = 8.0, 4.8, IH), 4.63 (s, 2H). LRMS (ESI pos) m/e 188 (M+l).
[00595] Step B: Preparation of N-(3-fluoro-4-(6-methoxy-7-(3- morpholinopropoxy)quinolin-4-yloxy)phenyl)-3-(pyridin-2-yloxy)pyridin-2-amine: A mixture of 4-(4-bromo-2-fluorophenoxy)-6-methoxy-7-(3-morpholinopropoxy)quinoline (Example 45) (20.0 mg, 0.0407 mmol), 3-(pyridin-2-yloxy)pyridin-2-amine (22.9 mg, 0.122 mmol), Pd2(dba)3 (7.45 mg, 0.00814 mmol), 4,5-bis(diphenylphosphino)-9,9-dimethyl-9H-xanthene (14.1 mg, 0.0244 mmol) and Cs2CO3 (39.8 mg, 0.122 mmol) in dioxane (10 mL) was stirred at 1000C for 1 hour. The reaction mixture was cooled to room temperature, water (10 mL) was added and the aqueous layer extracted with CH2Cl2 (3 x 100 mL). The combined organic layers were dried over Na2SO^ Concentration and purification by silica gel flash column chromatography afforded 198 (13.8 mg, 57%). 1H NMR (400 MHz, CDCl3)..δ 8.50 (d, J = 5.2 Hz, IH), 8.29 (dd, J = 4.8, 1.2 Hz, IH), 8.05 (dd, J = 4.4, 1.6 Hz, IH), 7.63 (m, IH), 7.56 (s, IH), 7.43 (m, 2H), 7.16-7.24 (m, 3H), 7.08- 7.11 (m, IH), 6.95-6.98 (m, IH), 6.85 (d, J = 8.4 Hz, IH), 6.52 (d, J = 5.2 Hz, IH), 4.27 (t, J = 6.6 Hz, 2H), 4.03 (s, 3H), 3.72 (t, J = 4.4 Hz, 4H), 2.58 (t, J - 7.0 Hz, 2H), 2.48 (m, 4H), 2.10-2.18 (m, 2H). LRMS (APCI neg) m/z 596 (M-I).
[00596] Example 99 3-(Benzyloxy)-N-(3-fluoro-4-(6-methoxy-7-(3- morpholinopropoxy)quinolin-4-yloxy)phenyl)pyridin-2-amine 199
Figure imgf000158_0001
[00597] A mixture of 4-(4-bromo-2-fluorophenoxy)-6-methoxy-7-(3- morpholinopropoxy)quinoline (Example 45) (20.0 mg, 0.0407 mmol), 3-(benzyloxy)pyridin-2- amine (40.8 mg, 0.204 mmol), Pd2(dba)3 (7.45 mg, 0.00814 mmol), 4,5-bis(diphenylphosphino)- 9,9-dimethyl-9H-xanthene (14.1 mg, 0.0244 mmol) and Cs2CO3 (39.8 mg, 0.122 mmol) in dioxane (10 mL) was stirred at 1000C for 1 hour. The reaction mixture was cooled to room 11 1-03-PCT - P2338R1 - 02120.004WO1
158 temperature, water (10 niL) was added and the aqueous layer was extracted with CH2Cl2 (3 x 50 mL). The combined organic layers were dried over Na2SO4. Concentration and purification by silica gel chromatography afforded 199 (16.8 mg, 68%). 1H NMR (400 MHz, CDCl3) δ 8.47 (d, J = 5.2 Hz, IH), 8.07 (d, J = 12.4 Hz, IH), 7.89 (d, J = 4.4 Hz, IH), 7.60 (s, IH), 7.45 (m, 5H), 7.31 (d, J = 8.8 Hz, IH), 7.16 (m, 2H), 7.09 (d, J - 7.2 Hz, IH), 6.76 (m, IH), 6.44 (d, J - 4.8 Hz, IH), 5.18 (s, 2H), 4.28 (t, J = 6.4 Hz, 2H), 4.05 (s, 3H), 3.73 (m, 4H), 2.58 (t, J = 6.8 Hz, 2H), 2.49 (m, 4H), 2.13 (m, 2H). LRMS (APCI neg) m/z 609 (M-I).
[00598] Example 100 3-Fluoro-4-(6-methoxy-7-(3-morpholinopropoxy)quinolin-4- yloxy)-N-phenylaniline 200
Figure imgf000159_0001
[00599] A mixture of 4-(4-bromo-2-fluorophenoxy)-6-methoxy-7-(3- morpholinopropoxy)quinoline (Example 45) (10.0 mg, 0.0204 mmol), aniline (9.48 mg, 0.102 mmol), Pd2(dba)3 (3.73 mg, 0.00407 mmol), 4,5-bis(diphenylphosphino)-9,9-dimethyl-9H- xanthene (7.07 mg, 0.0122 mmol) and Cs2CO3 (33.2 mg, 0.102 mmol) in dioxane (10 mL) was stirred at 1000C for 4 hours. The reaction mixture was cooled to room temperature, water (10 mL) was added and the aqueous phase was extracted with CH2Cl2 (3 x 50 mL). The combined organic layers were dried over Na2SO4. Concentration and purification by silica gel flash column chromatography afforded 200 (9.9 mg, 97%). 1H NMR (400 MHz, CDCl3).δ 8.49 (d, J - 5.2 Hz, IH), 7.59 (s, IH), 7.43 (s, IH), 7.34 (m, 2H), 7.12-7.16 (m, 3H), 7.04 (t, J = 7.2 Hz, IH), 6.97 (dd, J = 12.4, 2.8 Hz, IH), 6.85 (m, IH), 6.45 (dd, J = 5.2, 0.8Hz, IH), 5.83 (s, IH, NH), 4.28 (t, J - 6.8 Hz, 2H), 4.04 (s, 3H), 3.73 (t, J = 4.6 Hz, 4H), 2.58 (t, J = 7.2 Hz, 2H), 2.49 (m, 4H), 2.07- 2.17 (m, 2H). LRMS (APCI neg) m/z 502 (M-I).
[00600] Example 101 N-(3-Fluoro-4-(6-methoxy-7-(3-morpholinopropoxy)quinolin-4- yloxy)phenyl)-2,3'-bipyridin-2'-amine 201 111 -03-PCT - P2338R1 - 02120.004WO1
159
Figure imgf000160_0001
[00601] Step A: Preparation of 2'-fluoro-2,3'-bipyridine: A mixture of 2-fluoropyridin-3- ylboronic acid (200 mg, 1.419 mmol), 2-iodopyridine (291.0 mg, 1.419 mmol) and Tetrakis(triphenylphosphine)palladium(0) (328.0 mg, 0.2839 mmol) in 2 M aqueous Na2CO3 (3.5 mL) and DME (10 mL) was stirred at 8O0C for 8 hours. Water (10 mL) was added and the aqueous phase was extracted with CH2Cl2 (3 x 50 mL). The combined organic layers were dried over Na2 SO4. Concentration and purification by silica gel chromatography afforded the product (124.6 mg, 50%). 1H NMR (400 MHz, CDCl3)..δ 8.72-8.75 (m, IH), 8.8.50-8.57 (m, IH), 8.24- 8.27 (m, IH), 7.88-7.92 (m, IH), 7.77-7.82 (m, IH), 7.28-7.36 (m, 2H). LRMS (ESI pos) m/e 175 (M+l).
[00602] Step B: Prearation of N-(3-fluoro-4-(6-methoxy-7-(3- morpholinopropoxy)quinolin-4-yloxy)phenyl)-2,3'-bipyridin-2'-amine: A mixture of 3-fluoro-4- (6-methoxy-7-(3-morpholinopropoxy)quinolin-4-yloxy)benzenamine (prepared in Example 72, steps C-F) (10.0 mg, 0.0234 mmol), NaH (2.81 mg, 0.117 mmol) and 2-fluoro-3-(pyridin-2- yl)pyridine (4.89 mg, 0.0281 mmol) in DMF (10 mL) was stirred at 7O0C for 16 hours. Water (10 mL) was added and the aqueous phase was extracted with CH2Cl2 (3 x 50 mL). The combined organic layers were dried over Na2SO4. Concentration and purification by silica gel flash column chromatography afforded 201 (10.8 mg, 79%). 1H NMR (400 MHz, CDCl3) δ 8.72 (m, IH), 8.47 (d, J = 5.2 Hz, IH), 8.34 (dd, J - 4.8, 1.6 Hz, IH), 8.07 (dd, J = 13.2, 2.8 Hz, IH), 8.03 (dd, J = 8.0, 2.0 Hz, IH), 7.83-7.89 (m, 2H), 7.62 (s, IH), 7.44 (s, IH), 7.40-7.43 (m, IH), 7.31-7.34 (m, IH), 7.18 (t, J = 8.8 Hz, IH), 6.90 (m, IH), 6.47 (m, IH), 4.28 (t, J = 6.8
Hz, 2H), 4.05 (s, 3H), 3.73 (t, J = 4.6 Hz, 4H), 2.58 (t, J = 7.2 Hz, 2H), 2.49 (m, 4H), 2.10-2.17 (m, 2H). LRMS (APCI neg) m/z 580 (M-I).
[00603] Example 102 Preparation of 6-(4-(6,7-dimethoxyquinolin-4-yloxy)-3- fluorophenyl)-N-phenylpyridin-3-amine 202 111-03-PCT - P2338R1 - 02120.Q04WO1
160
Figure imgf000161_0001
[00604] Step A: Preparation of 2-(4-(benzyloxy)-3-fluorophenyl)-5-bromopyridine: To a mixture of 2,5-dibromopyridine (3.00 g, 12.7 mmol), 4-(benzyloxy)-3-fluorophenylboronic acid (3.74 g, 15.2 mmol), Pd(PPh3)4 (0.73 g, 0.63 mmol) in a 3:1 mixture of toluenerethanol (60 mL total) was added a 2M aqueous solution OfNa2CO3 (15 mL). The reaction mixture was stirred at 105°C for 3 hours. After cooling to room temperature, the mixture was partitioned between ethyl acetate (100 mL) and water (50 mL). The organic layer was separated, washed with brine, dried over Na2SO4, and evaporated to dryness. The crude material was purified by silica gel flash column chromatography (70:30 hexanes:EtOAc). Insoluble material was removed by filtration prior to chromatography. The desired product (200 mg, 0.4%) was obtained as an off-white solid. 1H-NMR (400 MHz; DMSO-d6) δ 8.74 (d, IH), 8.11 (d, IH), 7.92-7.98 (m, 3H), 7.89 (d, IH), 7.49 (d, 2H) 7.34-7.44 (m, 3H), 5.26 (s, 2H). LRMS (ESI pos) m/e 360 (M+l). [00605] Step B: Preparation of 6-(4-(benzyloxy)-3-fluorophenyl)-N-phenylpyridin-3- amine: To a mixture of 2-(4-(benzyloxy)-3-fiuorophenyl)-5-bromopyridine (200 mg, 0.56 mmol), Pd2dba3 (5.11 mg, 0.0056 mmol), (S)(-)2,2'-Bis(diphenylphosphino)-l,l'-binaphthyl (5.22 mg, 0.0084 mmol) in dry THF (2 mL) was added NaOtBu (75.12 mg, 0.78 mmol) and aniline (0.053 ml, 0.586 mmol). The mixture was degassed by successive evacuation and back-filling with nitrogen (3x). The mixture was heated to reflux temperature and stirred under nitrogen for 18 hours. The reaction mixture was cooled to room temperature and was partitioned between ethyl acetate (15 mL) and water (15 mL). The organic layer was separated, washed with brine, dried over Na2SO4, and evaporated to give a crude black oil. The crude product was purifed by silica gel flash column chromatography loading with dichloromethane and eluting with 80:20 hexanes:EtOAc to give the desired product (87 mg, 42%) as an off-white crystalline solid. 1H- NMR (400 MHz, DMSO-d6) δ 8.50 (s, IH), 8.40 (d, IH), 7.86 (d, IH), 7.79 (m, 2H), 7.53-7.47 (m, 3H), 7.42 (t, 2H), 7.36 (t, IH), 7.29 (t, 3H), 7.12 (d, 2H), 6.90 (t, IH), 5.23 (s, 2H). LRMS (APCI pos) m/e 371 (M+l).
[00606] Step C: Preparation of 2-fluoro-4-(5-(phenylamino)pyridin-2-yl)phenol: 6-(4-
(benzyloxy)-3-fluorophenyl)-N-phenylpyridin-3 -amine (62 mg, 0.167 mmol) was dissolved in TFA (1.29 ml, 16.7 mmol) and the reaction mixture was stirred at 70°C for 18 hours. The 111-03-PCT - P2338R1 - 02120.004WO1
161 mixture was cooled to room temperature and the excess TFA was removed by evacuation. The crude product was partitioned between dichloromethane and saturated Na2CO3 solution. The layers were separated and the organic layer was dried over Na2SO4 and evaporated to give a dark yellow oil which was purified by silica gel flash column chromatography, loading with DCM and eluting with 80/20 hexanes/EtOAc to give the desired product (42 mg, 89%) as yellow glass. LRMS (APCI pos) m/e 281 (M+ 1).
[00607] Step D: Preparation of 6-(4-(6,7-dimethoxyquinolin-4-yloxy)-3-fluorophenyl)-N- phenylpyridin-3-amine: 2-fiuoro-4-(5-(phenylamino)pyridin-2-yl)phenol (42 mg, 0.150 mmol), 4- chloro-6,7-dimethoxyquinoline (Example 5) (40.2 mg, 0.180 mmol) and DMAP (5.49 mg, 0.045 mmol) were added to a small sealable glass reaction tube. The mixture was suspended in bromobenzene (1.5 mL) and stirred at 150°C for 36 hours. The reaction mixture was cooled to room temperature, the solvent was evaporated, and the crude dissolved in MeOH and absorbed onto silica gel and purifed by silica gel flash column chromatography, eluting with 60/40 hexanes/EtOAc to yield 202 (29 mg, 36%) as a pale yellow solid. 1H-NMR (400 MHz, CDCl3) δ 8.49 (d, 2H), 7.92-7.79 (m, 2H), 7.62 (s, IH), 7.55-7.41 (m, 2H), 7.39-7.24 (m, 3H), 7.15 (d, 2H), 7.05 (t, IH), 6.49 (s, IH), 5.98 (s, IH), 4.06 (d, 6H). LRMS (APCI pos) m/e 468 (M+l). [00608] Example 103 6-(4-(6,7-dimethoxyquinolin-4-yloxy)-3-fluorophenyl)-N-methyl-
N-phenylpyridin-3-amine 203
Figure imgf000162_0001
[00609] Step A: Preparation of 6-(4-(6,7-dimethoxyquinolin-4-yloxy)-3-fluorophenyl)-N- methyl-N-phenylpyridin-3-amine: 6-(4-(6,7-dimethoxyquinolin-4-yloxy)-3-fluorophenyl)-N- phenylpyridin-3 -amine (Example 102, 22 mg, 0.047 mmol) was dissolved in DMF (0.5 mL) and cooled to 00C. NaH, 60% dispersion in oil (26 mg, 0.056 mmol) was added and the mixture was allowed to warm to room temperature and stirred for 10 minutes. Iodomethane (0.012 ml, 0.188 mmol) was added and the reaction was stirred at 25°C for 18 hours. The solvent was evaporated and the crude product purified by silica gel flash column chromatography, loading with DCM and eluting with 80/20 EtOAc/hexane to give 203 (5 mg, 19%) as pale yellow glass. 1H-NMR (400 MHz, CDCl3) δ 8.50 (s, IH), 8.37 (s, IH), 7.89 (d, IH), 7.79 (d, IH), 7.61-7.58 (m, 2H), 7.45 (s, 111-03-PCT- P2338R1 - 02120.004WO1
162
IH), 7.39 (t, 2H), 7.33-7.14 (m, 5H), 6.49 (d, IH), 4.07 (s, 3H), 4.06 (s, 3H), 3.41 (s, 3H). LRMS (APCI pos) m/e 482 (M+l).
[00610] Example 104 5-(4-(6,7-dimethoxyquinolin-4-yloxy)-3-fluorophenyl)-N- phenylpyrimidin-2 -amine 204
Figure imgf000163_0001
[00611] Step A: Preparation of 5-bromo-N-phenylpyrimidin-2 -amine: To a sealable glass reaction tube was 5-bromo-2-chloropyrimidine (1.00 g, 5.17 mmol) dissolved in 1-propanol (10 mL). DIEA (1.08 ml, 6.20 mmol) and aniline (0.565 ml, 6.20 mmol) were added and the tube was sealed and stirred at 1000C - 120°C for 18 hours. The mixture was cooled in an ice-water bath and a white precipitate formed. The mixture was diluted with ethyl acetate (10 mL), washed with brine (20 mL), water (20 mL), and brine (20 mL). The organic layer was isolated and evaporated to give 1.18g of crude material. The crude product was triturated with 90/10 hexane/EtOAc and the solid isolated by filtration to give the desired product (0.74 g, 57%) as a pale brown solid. 1H-NMR (400 MHz, DMSOd6) δ 9.85 (s, IH), 8.59 (s, 2H), 7.70 (d, J = 7.8 Hz, 2H), 7.29 (t, J = 8.0 Hz, 2H), 6.98 (t, J = 7.2 Hz, IH). LRMS (ESI pos) m/e 252 (M+l). [00612] Step B: Preparation of 5-(4-(benzyloxy)-3-fluorophenyl)-N-phenylpyrimidin-2- amine: A mixture of 5-bromo-N-phenylpyrimidin-2-amine (0.500 g, 2.00 mmol), 4-(benzyloxy)- 3-fluorophenylboronic acid (0.984 g, 4.00 mmol), Pd(PPh3)4 (0.116 g, 0.1000 mmol) and lithium chloride (0.170 g, 4.00 mmol) in dioxane (10 mL) and 2M aqueous Na2CO3 (2 mL) was stirred at 9O0C for 2 hours and then at room temperature for 1 hour. Water (50 mL) and ethyl acetate (100 mL) were added and the organic layer was separated, washed with brine (100 mL), dried over MgSO4, filtered and evaporated to give 1.3 g crude solid. The crude material was triturated with dichloromethane:MeOH and the resulting white solid was filtered and washed with dichloromethane to give the desired product (480 mg, 65%) as a white solid. 1H-NMR (400 MHz, DMSO-d6) δ 9.77 (s, IH), 8.82 (s, 2H), 7.79 (d, 2H), 7.67 (d, IH), 7.50-7.47 (m, 3H), 7.42 (t, 3H), 7.37-7.27 (m, 3H), 6.96 (t, IH), 5.24 (s, 2H). LRMS (ESI pos) m/e 372 (M+l). [00613] Step C: Preparation of 2-fluoro-4-(2-(phenylamino)pyrimidin-5-yl)phenol: 5-(4-
(benzyloxy)-3-fluorophenyl)-N-phenylpyrimidin-2-amine (280 mg, 0.754 mmol) was suspended in TFA (5 mL) and the suspension was stirred at 700C for 18 hours. The excess solvent was 111-03-PCT- P2338R1 - 02120.004WO1
163 removed under reduced pressure and the residue dissolved in dichloromethane (15 mL). The organic layer was washed with water (15 mL), aqueous NaHCO3 solution, separated and evaporated to give the desired product (120 mg, 57%) as a yellow solid. 1H-NMR (400 MHz, DMSO-d6) δ 10.01 (s, IH), 9.73 (s, IH), 8.78 (s, 2H), 7.79 (d, 2H), 7.56 (d, IH), 7.36 (d, IH), 7.29 (t, 2H), 7.04 (t, IH), 6.96 (t, IH). LRMS (ESI pos) m/e 282 (M+ 1).
[00614] Step D: Preparation of 5-(4-(6,7-dimethoxyquinolin-4-yloxy)-3-fluorophenyl)-N- phenylpyrimidin-2-amine: 2-fluoro-4-(2-(phenylamino) pyrimidin-5-yl)phenol (51.0 mg, 0.181 mmol), 4-chloro-6,7-dimethoxyquinoline (Example 5, 48.7 mg, 0.218 mmol), and DMAP (6.65 mg, 0.054 mmol) were added to a sealable glass reaction tube. The mixture was suspended in bromobenzene (2 mL) and stirred at 150°C for 18 hours. The reaction mixture was cooled to room temperature and the solvent was evaporated. The crude product was triturated with EtOAc:MeOH and the resulting solid filtered to give 204 (52 mg, 61%) as a white solid. 1H- NMR (400 MHz, DMSOd6) δ 9.88 (s, IH), 8.94 (s, 2H), 8.51 (d, IH), 7.96 (d, IH), 7.80 (s, 2H), 7.75 (d, IH), 7.57 (m, IH), 7.43 (s, IH), 7.32 (t, 2H), 6.99 (t, IH), 6.55 (d, IH), 3.96 (s, 6H). LRMS (APCI pos) m/e 469 (M+l).
[00615] Example 105 5-(4-(6,7-dimethoxyquinolin-4-yloxy)-3-fluorophenyl)-N-methyl-
N-phenylpyrimidin-2 -amine 205
Figure imgf000164_0001
[00616] Step A: Preparation of 5-(4-(6,7-dimethoxyquinolin-4-yloxy)-3-fluorophenyl)-N- methyl-N-phenylpyrimidin-2-amine: 5-(4-(6,7-dimethoxyquinolin-4-yloxy)-3-fiuorophenyl)-N- phenylpyrimidin-2 -amine (Example 104, 35 mg, 0.0747 mmol) was dissolved in DMF (0.5 mL) and cooled to 00C. NaH, 60% dispersion in oil (3.59 mg, 0.0897 mmol) was added and the mixture was allowed to warm to room temperature and stirred overnight. The mixture was partitioned between ethyl acetate (10 mL) and water (10 mL). The organic layer was washed with brine, dried over Na2SO4, and evaporated to give 205 (32 mg, 89%) as an off-white glass. 1H-NMR (400 MHz, DMSO-d6) δ 8.82 (s, 2H), 8.50 (d, IH), 7.89 (d, IH), 7.66 (d, IH), 7.58- 7.52 (m, 2H), 7.45-7.37 (m, 5H), 7.25 (t, IH), 6.52 (d, IH), 3.96 (s, 6H), 3.54 (s, 3H). LRMS (APCI pos) m/e 483 (M+l). [00617] Example 106 c-Met enzyme assay 111-03-PCT - P2338R1 - 02120.004WO1
164
[00618] The assay for the determination of c-Met kinase activity is based on an enzyme linked immunosorbant assay (ELISA). A compound of Formula I, 50 pM c-Met (His-tagged recombinant human Met (amino acids 974-end), expressed by baculovirus), and 5 μM ATP in assay buffer (25 mM MOPS, pH 7.4, 5 mM MgCl2, 0.5 raM MnCl2, 100 μM Sodium Orthovanadate, 0.01% Triton X-IOO, 1 mM DTT, final DMSO concentration 1% (v/v)) are incubated on a 0.25 mg/mL PGT coated plates for 20 minutes at room temperature. The reaction mixture is removed by washing and the phosphorylated polymer substrate is detected with 0.2 μg/mL phosphotyrosine specific monoclonal antibody (PY20) conjugated to horseradish peroxidase (HRP). After the addition of IM phosphoric acid to stop the development, the chromogenic substrate (TMB) color is quantitated by spectrophotometry at 450 nm. [00619] Example 107 in vitro cell proliferation assay
[00620] The cellular activity of the compounds of the present invention may be determined by the following procedure. MKN45 cells were plated in Costar 3904 96-well plates in growth media (RPMI, 10% FBS) at a density of 15000 cells/well and incubated at 37° C, 5% CO2 overnight. The following day, one-tenth volume of a 1OX concentration of compounds was added to the wells in a 11 -point dilution series. The dilutions series was composed of an initial 1:3 dilution in DMSO, followed by a 1:20 dilution in HBSS, for a final DMSO concentration on cells of 0.5%. Control wells were treated with 0.5% DMSO. The typical range of dilution was 5 μM to 0.3 nM, which was expanded to 25 μM depending on the potency of the compound. Once compound was added to the cells, plates were incubated for one hour at 370C, 5% CO2. Plates were then washed in PBS, fixed in 4% formaldehyde and rehydrated with a 10% methanol solution. The plates were then blocked with Licor blocking buffer. The total phosphorylated c- Met levels were measured by incubating with a rabbit polyclonal antibody against phosphorylated c-Met followed by an anti-rabbit antibody conjugated to Alexa Fluor 680. Signal was normalized for differences in cell number by reference to the levels of the housekeeping protein GAPDH. Cells were incubated with a mouse monoclonal antibody against GAPDH followed by an anti- mouse antibody labeled with IRdye 800. Signal was measured on the Licor. The overall fluorescent signal from the Alexa Fluor 680 is normalized by dividing the value by the fluorescent value of the IRdye 800 signal. The fluorescent signal of the control wells was defined as 100% and the percent of inhibition of phosphorylated c-Met was expressed as percent of control. IC50 values were determined from the percent of control data using a standard A- parameter logistical model.
[00621] The foregoing description is considered as illustrative only of the principles of the invention. Further, since numerous modifications and changes will be readily apparent to those 111-03-PCT - P2338R1 - 02120 004WO1
165 skilled in the art, it is not desired to limit the invention to the exact construction and process shown as described above. Accordingly, all suitable modifications and equivalents may be considered to fall within the scope of the invention as defined by the claims that follow. [00622] The words "comprise," "comprising," "include," "including," and "includes" when used in this specification and in the following claims are intended to specify the presence of stated features, integers, components, or steps, but they do not preclude the presence or addition of one or more other features, integers, components, steps, or groups thereof.

Claims

111-03-PCT - P2338R1 - 02120 004WO1
166
What is claimed is: 1. A compound having Formula I:
Figure imgf000167_0001
and stereoisomers, geometric isomers, tautomers, solvates, metabolites, and salts thereof, wherein:
R1, R2 and R4 are independently selected from H, F, Cl, Br, I, CN,
-(CR14R15^NR10R1 \ -C(=Y)R10, -C(=Y)OR10, -CC=Y)NR10R1 \
-C(=O)NR12(CR14R15)tNR10Rn, -NO2, -NR10R11, -NR10CC=Y)R1 \ -NR10CC=Y)OR1 ',
-NR12CC=Y)NR10R11, -NR12SO2NR10R11, OR10, OC(=Y)R10, OC(^Y)OR10, OC(=Y)NR10Rπ, -OP(=Y)(OR10)(OR11), -OP(OR10XOR11), -P(^Y)(OR10XOR11), -SR10, -S(O)R10,
-S(O)2R10, -S(O)2NR10R11, -SC(=Y)R10, -SC(=Y)OR10, Cj-C12 alkyl, C2-C8 alkenyl, C2-C8 alkynyl, C3-C12 carbocyclyl, C2-C20 heterocyclyl, CO-C20 aryl, and Ci-C20 heteroaryl, where said alkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl and heteroaryl are optionally substituted with one or more groups independently selected from F, Cl, Br, I, CN, CF3, -NO2, oxo, -C(=Y)R10, -C(=Y)OR10, -Q=Y)NR10R1 ', -(CR14R15VNR10R1 \ -NR10C(=Y)R10,
-NR10C(=Y)ORπ, -NR12Q=Y)NR10R11, -NR12SO2R10, =NR10, OR10, -OC(=Y)R10,
-OC(=Y)OR10, -OC(=Y)NR10Rn, -OS(O)2(OR10), -OP(=Y)(OR10)(ORn),
-OP(OR10XOR11), SR10, -S(O)R10, -S(O)2R10, -S(O)2NR10R11, -S(O)(OR10), -S(O)2(OR10),
-SC(=Y)R10, -SC(=Y)OR10, -SC(=Y)NR10Rn, C1-C12 alkyl, C2-C8 alkenyl, C2-C8 alkynyl, C3-Ci2 carbocyclyl, C2-C20 heterocyclyl, C6-C20 aryl, Ci-C20 heteroaryl,
-(CR14R15)t-NR12C(=O)(CR14R15)NR10Rπ, and (CR4R5X-NR10R11, with the proviso that at least one of R1 and R2 is not H;
L is C3-Ci2 carbocyclyl, C2-C20 heterocyclyl, CO-C20 aryl or Ci-C20 heteroaryl, wherein said carbocyclyl, heterocyclyl, aryl and heteroaryl are optionally substituted with one or more groups independently selected from R4 and R10, with the proviso that L is not naphthyl;
R5 is -C(=Y)R13, -C(=Y)NR10R13, -NR10R13, -NR10C(=Y)R13, -NR10C(=Y)OR13,
-NR12SO2R10, -NR12C(=Y1)(CR14R15)C(-Y2)NR10R11, C3-Ci2 carbocyclyl, C2-C 2o 111-03-PCT - P2338R1 - 02120.004WO1
167 heterocyclyl, C6-C2O aryl, or C]-C20 heteroaryl, wherein said carbocyclyl, heterocyclyl, aryl, and heteroaryl are optionally substituted with one or more groups independently selected from oxo, F, Cl, Br, I, SO2RC, CN, ORa, (CH2)n-NRaRb, C(=O)NRaRb, C(=O)Ra, CRaC(=O)Rb, NHSO2RC, CF3, Ci-C12 alkyl, C2-C8 alkenyl, C2-C8 alkynyl, (CH2)n-(C6-C20 aryl), (CH2)n-cycloalkyl, (CH2)n-cycloalkyl, CH(OH)-aryl, CH(CO2CH3)aryl, and (CH2)n- (Ci-C2O heteroaryl), and wherein any aryl or heteroaryl of the one or more groups is optionally substituted with one or more Rd;
R10, R11 and R12 are independently H, C1-Ci2 alkyl, C2-C8 alkenyl, C2-C8 alkynyl, C3-Ci2 carbocyclyl, C2-C20 heterocyclyl, C6-C20 aryl, or Ci-C20 heteroaryl, wherein said alkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl, and heteroaryl are optionally substituted with one or more groups independently selected from F, Cl, Br, I, SO2R0, CN, ORa, NRaRb, C(=O)NRaRb, CRaC(=O)Rb, Ci-Q2 alkyl, C2-C8 alkenyl, C2-C8 alkynyl, C3-Ci2 carbocyclyl, C2-C20 heterocyclyl optionally substituted with Ci-C6 alkyl, CH2OH or SO2Me, C6-C20 aryl, and Ci-C20 heteroaryl optionally substituted with Ci-C6 alkyl, or R10 and R11 together with the nitrogen to which they are attached optionally form a saturated, partially unsaturated or fully unsaturated C3-C20 heterocyclic ring optionally containing one or more additional ring atoms selected from N, O or S, wherein said heterocyclic ring is optionally substituted with one or more groups independently selected from oxo, (CH2)nORa, NRaRb, CF3, F, Cl, Br, I, SO2Ra, C(=O)Ra, NR10C(=Y)RH, C(^Y)NR10R11, Ci-Ci2 alkyl, C2-C8 alkenyl, C2-C8 alkynyl, C3-Ci2 carbocyclyl, C2-C20 heterocyclyl, C6-C20 aryl and Ci-C20 heteroaryl;
R13 is H, C1-C6 alkyl, -(CR14R15)n-cycloalkyl, -(CR14R15)n-heterocyclyl, -(CR14RI5)n-aryl, -(CR14R15)n-heteroaryl, (CR14R15)n-O-(CR14R15)m-aryl, (CR14R15)- N(SO2Ra)-(CR14R15)Rπ, (CRI4R15)n-heterocyclyl-(CR14R15)t-aryl, or (CR14R15)- NR10C(=O)aryl, where said cycloalkyl, heterocyclyl, aryl, and heteroaryl portions are optionally substituted with one or more groups independently selected from F, Cl, Br, I, oxo, SO2RC, CN, ORa, C(=O)Ra, C(=O)ORa, NRaRb, NRaC(=O)Rb, O-(CH2)-aryl, Cj-Ci2 alkyl, C2-C8 alkenyl, C2-C8 alkynyl, C3-Ci2 carbocyclyl, C2-C20 heterocyclyl, C6-C20 aryl, and Ci-C20 heteroaryl; each R14 and R15 is independently H, Ci-Ci2 alkyl, or (CH2)t-aryl, or R14 and R15 together with the atoms to which they are attached form a saturated or partially unsaturated C3-Ci2 carbocyclic ring, 111-03-PCT- P2338R1 - 02120.004WO1
168 or R10 and R15 together with the atoms to which they are attached form a saturated or partially unsaturated C2-Ci2 heteorcyclic ring, or R14 is null and R10 and R15 together with the atoms to which they are attached form a 5-6 membered heteroaryl ring, or R12 and R14 together with the atoms to which they are attached form a saturated or partially unsaturated C2-Ci2 heteorcyclic ring;
Ra and Rb are independently H, Ci-Ci2 alkyl, C2-C8 alkenyl, C2-C8 alkynyl, C3-C12 carbocyclyl, C2-C20 heterocyclyl, C6-C20 aryl, or Ci-C2O heteroaryl, wherein said alkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl, and heteroaryl are optionally substituted with one or more alkyl groups;
Rc is Ci-Ci2 alkyl or C6-C2O aryl, wherein said alkyl and aryl are optionally substituted with one or more groups independently selected from F, Cl, Br, I, ORa and C(=O)NRaRb;
Rd is F, Cl, Br, I, CF3, SO2RC, CN, ORa, NRaRb, C(=O)NRaRb, CRaC(=O)Rb, CI-CJ2 alkyl, C2-C8 alkenyl, C2-C8 alkynyl, C6-C20 aryl, or Ci-C20 heteroaryl; Y, Y1 and Y2 are independently O or S; t is 1, 2, 3, 4, 5 or 6; and n and m are independently 0, 1, 2, 3, 4, 5 or 6.
2. The compound of claim 1, wherein one or both of R1 and R2 is -OR10 where R10 is Ci-C12 alkyl.
3. The compound of claim 1, wherein one of R1 and R2 is methoxy.
4. The compound of claim 1, wherein both of R1 and R2 are methoxy.
5. The compound of claim 1, wherein one or both of R1 and R2 is -OR10 where R10 is CI-CJ2 alkyl substituted with NRaRb.
6. The compound of claim 1, wherein one or both of R1 and R2 is -OR10 where
R10 is Ci-Ci2 alkyl substituted with C2-C20 heterocyclyl optionally substituted with Ci-C6 alkyl, CH2OH or SO2Me.
7. The compound of claim 6 wherein -OR10 is selected from the structures: 111-03-PCT- P2338R1 - 02120.004WO1
169
Figure imgf000170_0001
where the wavy line is the attachment site to the quinoline ring.
8. The compound of claim 1 wherein R1 is methoxy and R2 is 3- morpholinopropoxy.
9. The compound of claim 1, wherein one or both of R1 and R2 is -OR10 where R10 is Ci-Ci2 alkyl substituted with C]-C20 heteroaryl, wherein said heteroaryl is optionally substituted with CI-CO alkyl.
10. The compound of claim 9 wherein -OR!0 is selected from the structures:
Figure imgf000170_0002
where the wavy line is the attachment site to the quinoline ring.
11. The compound of claim 1, wherein one or both of R1 and R2 are independently selected from alkynyl substituted by -(CR14R15)t-NR12C(=O)(CR14R15)NR10Rn or -(CR4R5^NR10R1 \
12. The compound of claim 11 wherein one or both of R1 and R2 are independently selected from the structures:
Figure imgf000170_0003
111-03-PCT - P2338R1 - 02120.004WO1
170
Figure imgf000171_0001
13. The compound of claim 1 wherein one or both of R1 and R2 are independently selected from optionally substituted aryl or heteroaryl.
14. The compound of claim 13 wherein one or both of R1 and R2 are independently selected from:
Figure imgf000171_0002
15. The compound of claim 1, wherein one or both of R1 and R2 are independently selected from -CC=O)NR10R11 or -(CR14R15^NR10R11.
16. The compound of claim 1 wherein one or both of R1 and R2 are independently selected from alkyl optionally substituted with one or more groups independently selected from OR10, NR10R11, heterocyclyl and heteroaryl. 111-03-PCT - P2338R1 - 02120.004WO1
171
17. The compound of claim 16 wherein one or both of R1 and R2 are independently selected from methyl, -CH2OH, -CH2CH2OH, -CH2CH2CH2OH, and -CH(OH)CH2OH.
18. The compound of claim 1 wherein each R4 is H.
19. The compound of claim 1 where L-R5 is (C3-Ci2 carbocyclyty-R5.
20. The compound of claim 19 wherein L-R5 is selected from the structures:
Figure imgf000172_0001
where the wavy line indicates the point of attachment to the 4-oxy position of the quinoline ring.
21. The compound of claim 1 where L-R5 is (C2-C2O heterocyclyl)-R5.
22. The compound of claim 21 wherein L-R5 is selected from the structures:
Figure imgf000172_0002
where the wavy line indicates the point of attachment to the 4-oxy position of the quinoline ring.
23. The compound of claim 1 where L-R5 is (CO-C2O aryl)-R5.
24. The compound of claim 23 wherein L-R5 is selected from the structures:
Figure imgf000172_0003
111-Q3-PCT- P2338R1 - 02120.004WO1
172 where the wavy line indicates the point of attachment to the 4-oxy position of the quinoline ring and each R4 is independent of the other.
25. The compound of claim 24 where L-R5 is selected from the structures:
Figure imgf000173_0001
26. The compound of claim 24 where L-R5 is selected from the structures:
Figure imgf000173_0002
111-03-PCT - P2338R1 - 02120.004WO1
173
27. The compound of claim 24 where L-R5 is selected from the structures:
Figure imgf000174_0001
28. The compound of claim 23 where L-R5 is selected from the structures:
Figure imgf000174_0002
29. The compound of claim 1 where L-R5 is (C]-C2O heteroaryl)-R5.
30. The compound of claim 29 wherein L-R5 is selected from the structures:
Figure imgf000174_0003
where the wavy line indicates the point of attachment to the 4-oxy position of the quinoline ring.
31. The compound of claim 30 wherein L-R5 is selected from the structures
Figure imgf000174_0004
111-03-PCT- P2338R1 - 02120.004WO1
174
32. The compound of claim 29 wherein L-R5 is selected from the structures:
Figure imgf000175_0001
33. The compound of claim 29 wherein L is selected from the structures:
Figure imgf000175_0002
34. The compound of claim 24 wherein R5 is -C(=Y)R13.
35. The compound of claim 34 wherein R5 is selected from the structures:
Figure imgf000175_0003
where the wavy line indicates the point of attachment to L.
36. The compound of claim 24 wherein R5 is -C(=Y)NR10R13. 111-03-PCT- P2338R1 - 02120.004WO1
175
37. The compound of claim 36 wherein R5 is selected from the structures:
Figure imgf000176_0001
where the wavy line indicates the point of attachment to L.
38. The compound of claim 1 wherein R5 is -NR10R13.
39. The compound of claim 38 wherein R5 is selected from the structures:
Figure imgf000176_0002
where the wavy line indicates the point of attachment to L.
40. The compound of claim 24 wherein R5 is NR12C(=Y1)(CR14R15)C(=Y2)NR10R11, wherein R15 and R10 optionally together with the atoms to which they are attached form a 5-6 membered heterocyclic ring, and wherein R14 and the adjacent saturated ring carbon together with the atoms to which they are attached optionally form a fused cyclopropyl ring.
41. The compound of claim 40 wherein R5 is selected from the structures:
Figure imgf000176_0003
where the wavy line indicates the point of attachment to L.
42. The compound of claim 41 wherein R5 is selected from the structures:
Figure imgf000176_0004
111-03-PCT- P2338R1 - 02120.004WO1
Figure imgf000177_0001
43. The compound of claim 40 wherein R4 is null and R10 and R15 together with the nitrogen atom to which they are attached form a heteroaryl ring optionally having an additional ring nitrogen atom.
44. The compound of claim 43, wherein R5 is selected from the structures:
Figure imgf000177_0002
where Y1 and Y2 are independently selected from O and S; and where the wavy line indicates the point of attachment to L.
45. The compound of claim 43 wherein R5 is selected from the structures:
Figure imgf000177_0003
111-03-PCT- P2338R1 - 02120.004WO1
177
Figure imgf000178_0001
wherein the cyclohexyl and phenyl groups are optionally substituted with one or more R groups independently selected from F, Cl, Br, I, SO2RC, CN, ORa, NRaRb, C(=O)NRaRb,
CRaC(=O)Rb, Ci-Ci2 alkyl, C2-C8 alkenyl, C2-C8 alkynyl, C6-C20 aryl, and Q-C 20 heteroaryl.
46. The compound of claim 24, wherein R5 is -NRI2C(=Y1)(CR14R15)C(=Y2)NR10R1 \ wherein R12 and R14 together with the atoms to which they are attached form a 5-6 membered heterocyclic ring.
47. The compound of claim 46, wherein R5 is
Figure imgf000178_0002
48. The compound of claim 1, wherein R5 is -NR10CO=Y)R13, wherein R13 is Ci- C6 alkyl, (CR14R15)n-O-(CR14R15)m-aryl, (CR14R15)-aryl, (CR14R15)-heteroaryl, (CR14R15)- heterocyclyl, (CR14R15)-N(SO2Ra)(CR14R15)Rπ, or (CR14R15)NR10C(=O)-aryl, wherein said alkyl, aryl, heteroaryl and heterocyclyl portions are optionally substituted.
49. The compound of claim 48 wherein R5 is selected from the structures:
o -PCT-P2338R1 -02120.004WO1
178
Figure imgf000179_0001
111-03-PCT - P2338R1 - 02120.004WO1
179
Figure imgf000180_0001
where the wavy line indicates the point of attachment to L.
50. The compound of claim 1 wherein R5 is -NR10C(=Y)OR13.
51. The compound of claim 50 wherein R5 is selected from the structures:
Figure imgf000180_0002
where the wavy line indicates the point of attachment to L.
52. The compound of claim 1 wherein R5 is -NR12SO2R10.
53. The compound of claim 52 wherein R10 is alkyl or optionally substituted aryl.
54. The compound of claim 53 wherein R5 is selected from the structures:
Figure imgf000180_0003
where the wavy line indicates the point of attachment to L.
55. The compound of claim 1 wherein R5 is a substituted carbocyclyl.
56. The compound of claim 55 wherein R5 is selected from the structures:
Figure imgf000180_0004
where the wavy line indicates the point of attachment to L. 111-03-PCT - P2338R1 - 02120.004WO1
180
57. The compound of claim 1 wherein R5 is a substituted heterocyclyl.
58. The compound of claim 57 wherein R5 is selected from the structures:
Figure imgf000181_0001
where the wavy line indicates the point of attachment to L.
59. The compound of claim 1 wherein R5 is an optionally substituted aryl.
60. The compound of claim 59 wherein R5 is selected from the structures:
Figure imgf000181_0002
where the wavy line indicates the point of attachment to L.
61. The compound of claim 1 wherein R5 is an optionally substituted heteroaryl.
62. The compound of claim 61 wherein R5 is selected from the structures: 111-03-PCT- P2338R1 - 02120.004WO1
181
Figure imgf000182_0001
where R >2o is H, Ci-Ci2 alkyl, C3-Ci2 cycloalkyl, C6-C20 aryl, or Ci-C20 heteroaryl, and R21 and R22 are independently selected from H or C]-Ci2 alkyl, wherein said alkyl, cycloalkyl, aryl, heteroaryl are optionally substituted with one or more groups independently selected from F, Cl, Br, I and Ci-Ci2 alkyl; each Re is independently H or Ci-C4 alkyl; and where the wavy line indicates the point of attachment to L.
63. The compound of claim 62 wherein R20 is H.
64. The compound of claim 61 wherein R5 is selected from the structures:
Figure imgf000182_0002
111-03-PCT- P2338R1 - 02120.004WO1
Figure imgf000183_0001
where the phenyl groups are optionally substituted with one or more Rd groups independently selected from F, Cl, Br, I, CF3, SO2RC, CN, ORa, NRaRb, C(=O)NRaRb, CRaC(=O)Rb, C1-C12 alkyl, C2-C8 alkenyl, C2-C8 alkynyl, C6-C20 aryl, and C1-C20 heteroaryl; and each Re is independently H or C1-C4 alkyl.
65. The compound of claim 61, wherein R5 is selected from:
Figure imgf000183_0002
66. The compound of claim 1 selected from:
N-(4-(7-(3-morpholinopropoxy)-6-methoxyquinolin-4-yloxy)-3-fluorophenyl)pyridin- 2-amine; 3-(4-(7-(3-morpholinopropoxy)-6-methoxyquinolin-4-yloxy)-3-fluorophenyl)-6- benzyl-l-methylpyridin-2(lH)-one; l-(4-(7-(3-morpholinopropoxy)-6-methoxyquinolin-4-yloxy)-3-fluorophenyl)-4-(2- methyl)benzyl)-5-methyl-pyrimidin-6-one;
5-(4-(7-(3-moφholinopropoxy)-6-methoxyquinolin-4-yloxy)-3-fluorophenyl)-3- ethyl-2-(phenylamino)pyrimidin-4(3H)-one; 111-03-PCT - P2338R1 - 02120.004WO1
183
N-(4-(7-(3-(piperidin-l-yl)propoxy)-6-methoxyquinolin-4-yloxy)-3-fluorophenyl)-2- (4-fluorophenyl)-2,3-dihydro-3-oxopyridazine-4-carboxamide;
N-(4-(7-(3-moφholinopropoxy)-6-methoxyquinolin-4-yloxy)-3-fluorophenyl)-l- methyl-2-oxopyrrolidine-3-carboxamide; N-(4-(7-(3-raorpholinopropoxy)-6-methoxyquinolin-4-yloxy)-3-fluorophenyl)-4- benzyl-3,4-dihydro-3-oxopyrazine-2-carboxamide;
N-(6-(7-(3-morpholinopropoxy)-6-methoxyquinolin-4-yloxy)pyridin-3-yl)-2-(4- fluorophenyl)-2, 3-dihydro-3 -oxopyridazine-4-carboxam ide ;
N-(4-(7-(3-(4-methylpiperazin-l-yl)propoxy)-6-methoxyquinolin-4-yloxy)-3- fluorophenyl)-2-(4-fluorophenyl)-2,3-dihydro-3-oxopyridazine-4-carboxamide;
2-(4-fluorophenylamino)-5-(4-(7-(3-morpholinopropoxy)-6-methoxyquinolin-4- yloxy)-3-fluorophenyl)-3-methylpyrimidin-4(3H)-one;
5-(4-(7-(3-morpholinopropoxy)-6-methoxyquinolin-4-yloxy)-3-fluorophenyl)-2- (cyclopropylmethylamino)-3-methylpyrimidin-4(3H)-one; 5-(4-(7-(3-morpholinopropoxy)-6-methoxyquinolin-4-yloxy)-3-fluorophenyl)-2- benzyl-3-methylpyrimidin-4(3H)-one; l-(4-(7-(3-morpholinopropoxy)-6-methoxyquinolin-4-yloxy)-3-fluorophenyl)-N-(4- fluorophenyl)-l,2-dihydro-2-oxopyridine-3-carboxamide;
3-(4-(7-(3-moφholinopropoxy)-6-methoxyquinolin-4-yloxy)-3-fluorophenyl)-5- benzylpyrimidin-4(3H)-one;
N-(4-(7-(3-moφholinopropoxy)-6-methoxyquinolin-4-yloxy)-3-fluorophenyl)-2- oxopyrrolidine-3-carboxamide;
3-(4-(7-(3-moφholinopropoxy)-6-methoxyquinolin-4-yloxy)-3-fluorophenyl)-5- methyl-6-(phenylamino)pyrimidin-4(3H)-one; 3-(4-(7-(3-moφholinopropoxy)-6-methoxyquinolin-4-yloxy)-3-fluorophenyl)-6- benzylpyrim idin-4(3H)-one ;
3-(4-(7-(3-moφholinopropoxy)-6-methoxyquinolin-4-yloxy)-3-fluorophenyl)-6- benzyl-5-methylpyrimidin-4(3H)-one;
(4-(7-(3-moφholinopropoxy)-6-methoxyquinolin-4-yloxy)-3-fluorophenyl)(3- benzylpiperidin- 1 -yl)methanone;
N-(4-(7-(3-moφholinopropoxy)-6-methoxyquinolin-4-yloxy)-3-fluorophenyl)-l,2- dihydro-l-methyl-2-oxopyridine-3-carboxamide;
N-(4-(7-(3-moφholinopropoxy)-6-methoxyquinolin-4-yloxy)-3-fluorophenyl)-7,7- dimethyl^-oxobicycloP^.llheptane-l-carboxamide; 111-03-PCT - P2338R1 - 02120.004WO1
184
N-(4-(7-(3-moφholinopropoxy)-6-methoxyquinolin-4-yloxy)-3-fluorophenyl)-2- (pyridin-2-yl)acetamide;
N-(4-(7-(3-moφholinopropoxy)-6-methoxyquinolin-4-yloxy)-3-fluorophenyl)-2-(4- fluorophenyl)-2,3-dihydro-3-oxopyridazine-4-carboxamide; N-(4-(7-(3-morpholinopropoxy)-6-methoxyquinolin-4-yloxy)-3- fluorophenyl)quinoline-8-carboxamide;
N-(4-(7-(3-morpholinopropoxy)-6-methoxyquinolin-4-yloxy)-3-fluorophenyl)-l,2- dihydro-2-oxo- 1 -((pyrimidin-4-yl)methyl)pyridine-3-carboxamide; l-(4-chlorobenzyl)-N-(4-(7-(3-morpholinopropoxy)-6-methoxyquinolin-4-yloxy)-3- fluorophenyl)- 1 ,2-dihydro-2-oxopyridine-3-carboxamide;
N-(4-(7-(3-morpholinopropoxy)-6-methoxyquinolin-4-yloxy)-3-fluorophenyl)-l- benzyl- 1 ,2-dihydro-2-oxopyridine-3-carboxamide;
5-(4-(7-(3-moφholinopropoxy)-6-methoxyquinolin-4-yloxy)-3-fluorophenyl)-3- methyl-2-(phenylamino)pyrimidin-4(3H)-one; 3-(4-(7-(3-morpholinopropoxy)-6-methoxyquinolin-4-yloxy)phenyl)-5-methyl-6-
(phenylamino)pyrimidin-4(3H)-one;
N-(4-(7-(3-morpholinopropoxy)-6-methoxyquinolin-4-yloxy)-3-fluorophenyl)-l-(4- fluorophenyl)-2-oxopiperidine-3-carboxamide;
N-(4-(7-(3-morpholinopropoxy)-6-methoxyquinolin-4-yloxy)-3-fluorophenyl)-l-(4- fluorophenyl)- 1 ,2-dihydro-2-oxopyridine-3-carboxamide;
3-(4-(7-(3-morpholinopropoxy)-6-methoxyquinolin-4-yloxy)-3-fluorophenyl)-3,4- dihydroquinazolin-2(lH)-one;
N-(4-(7-(3-morpholinopropoxy)-6-methoxyquinolin-4-yloxy)-3-fluorophenyl)-l-(4- fluorophenyl)-3-methyl-2-oxopyrrolidine-3-carboxamide; l-(4-fluorobenzyl)-N-(4-(7-(3-morpholinopropoxy)-6-methoxyquinolin-4-yloxy)-3- fluorophenyl)- 1 ,2-dihydro-2-oxopyridine-3-carboxamide;
N-(4-(7-(3-moφholinopropoxy)-6-methoxyquinolin-4-yloxy)-3-fluorophenyl)-2-oxo- l-phenylpyrrolidine-3-carboxamide;
5-(4-(7-(3-moφholinopropoxy)-6-methoxyquinolin-4-yloxy)phenyl)-2- benzylpyrimidin-4(3H)-one;
N-(4-(7-(3-moφholinopropoxy)-6-methoxyquinolin-4-yloxy)-3-fluorophenyl)-l-(4- chlorophenyl)-2-oxopyrrolidine-3-carboxamide;
N-(4-(7-(3-moφholinopropoxy)-6-methoxyquinolin-4-yloxy)-3-fluorophenyl)-l-(4- fluorophenyl)-2-oxopyrrolidine-3-carboxamide; 111-03-PCT - P2338R1 - 02120.004WO1
185
5-(4-(7-(3-(piperidin-l-yl)propoxy)-6-methoxyquinolin-4-yloxy)-3-fluorophenyl)-3- benzylpyrimidin-4(3H)-one;
5-(4-(7-(3-(4-methylpiperazin-l-yl)propoxy)-6-methoxyquinolin-4-yloxy)-3- fluorophenyl)-3-benzylpyrimidin-4(3H)-one; 3-(4-chloro-2-fluorobenzyl)-5-(4-(6,7-dimethoxyquinolin-4-yloxy)-3- fluorophenyl)pyrimidin-4(3H)-one; l-(4-(6,7-dimethoxyquinolin-4-yloxy)-3-fluorophenyl)-N-(4-fluorophenyl)-2- oxopyrrolidine-3-carboxamide;
3-(4-fluoro-3-methylbenzyl)-5-(4-(6,7-dimethoxyquinolin-4-yloxy)-3- fluorophenyl)pyrimidin-4(3H)-one;
3-(3,4-dimethylbenzyl)-5-(4-(6,7-dimethoxyquinolin-4-yloxy)-3- fluorophenyl)pyrimidin-4(3H)-one;
3-(4-chloro-2,6-difluorobenzyl)-5-(4-(6,7-dimethoxyquinolin-4-yloxy)-3- fluorophenyl)pyrimidin-4(3H)-one; 3-(2-chloro-4-fluorobenzyl)-5-(4-(6,7-dimethoxyquinolin-4-yloxy)-3- fluorophenyl)pyrimidin-4(3H)-one;
3-(3,4-dichlorobenzyl)-5-(4-(6,7-dimethoxyquinolin-4-yloxy)-3- fluorophenyl)pyrimidin-4(3H)-one;
N-(2-(7-(3-moφholinopropoxy)-6-methoxyquinolin-4-yloxy)naphthalen-6- yl)thiophene-3-carboxamide;
5-(4-(7-(2-(lH-imidazol-l-yl)ethoxy)-6-methoxyquinolin-4-yloxy)-3-fluorophenyl)- 3-benzylpyrimidin-4(3H)-one;
3-(4-(trifluoromethyl)benzyl)-5-(4-(6,7-dimethoxyquinolin-4-yloxy)-3- fluorophenyl)pyrimidin-4(3H)-one; 3-(4-tolyl)-5-(4-(6,7-dimethoxyquinolin-4-yloxy)-3-fluorophenyl)pyrimidin-4(3H)- one;
3-(4-fluorobenzyl)-5-(4-(6,7-dimethoxyquinolin-4-yloxy)-3-fluorophenyl)pyrimidin- 4(3H)-one;
3-(2-fluorobenzyl)-5-(4-(6,7-dimethoxyquinolin-4-yloxy)-3-fluorophenyl)pyrimidin- 4(3H)-one;
3-(4-chlorobenzyl)-5-(4-(7-(3-morpholinopropoxy)-6-methoxyquinolin-4-yloxy)-3- fluorophenyl)pyrim idin-4(3H)-one ;
5-(4-(7-(3-morpholinopropoxy)-6-methoxyquinolin-4-yloxy)-3-fluorophenyl)-3- benzylpyrimidin-4(3H)-one; 111-03-PCT - P2338R1 - 02120.004WO1
186
(4-(6,7-dimethoxyquinolin-4-yloxy)-3-fluorophenyl)(4-benzylpiperidin-l- yl)methanone;
(4-(6,7-dimethoxyquinolin-4-yloxy)-3-fluorophenyl)(3-benzylpiperidin-l- yl)methanone; 3-(4-chlorobenzyl)-5-(4-(6,7-dimethoxyquinolin-4-yloxy)-3-fluorophenyl)pyrimidin-
4(3H)-one;
3-(3-chlorobenzyl)-5-(4-(6,7-dimethoxyquinolin-4-yloxy)-3-fluorophenyl)pyrimidin- 4(3H)-one;
3-(2-methylbenzyl)-5-(4-(6,7-dimethoxyquinolin-4-yloxy)-3-fluorophenyl)pyrimidin- 4(3H)-one;
3-(2-chlorobenzyl)-5-(4-(6,7-dimethoxyquinolin-4-yloxy)-3-fluorophenyl)pyrimidin- 4(3H)-one;
5-(4-(7-(3-moφholinopropoxy)-6-methoxyquinolin-4-yloxy)-3-fluorophenyl)-2- benzylpyrimidin-4(3H)-one; 3-(4-(7-(3-morpholinopropoxy)-6-methoxyquinolin-4-yloxy)-3-fluorophenyl)-6- benzylpyridin-2( 1 H)-one ; l-(4-(7-(3-morpholinopropoxy)-6-methoxyquinolin-4-yloxy)-3-fluorophenyl)-4- benzylpyridin-2( 1 H)-one ;
5-(4-(6,7-dimethoxyquinolin-4-yloxy)-3-fluorophenyl)-N-methyl-N-phenylpyrimidin- 2-amine;
5-(4-(6,7-dimethoxyquinolin-4-yloxy)-3-fluorophenyl)-N-phenylpyrimidin-2 -amine; 4-((6,7-dimethoxyquinolin-4-yloxy)-3-fluorophenyl)-(2-tetrahydro-2H-pyranyl)- phenol;
4-((6,7-dimethoxyquinolin-4-yloxy)-3-fluorophenyl)-N-cyclopropylbenzamide; 4-((6,7-dimethoxyquinolin-4-yloxy)-3-fluorophenyl)-benzyl-lH-pyrazole;
4-((6,7-dimethoxyquinolin-4-yloxy)-3-fluorophenyl)-cyclohexylbenzene; 4-((6,7-dimethoxyquinolin-4-yloxy)-3-fluorophenyl)-N-phenylmethylsulfonamide; 4-((6,7-dimethoxyquinolin-4-yloxy)-3-fluorophenyl)-phenoxybenzene; 4-(2-fluoro-4-(6-methoxypyridin-3-yl)phenoxy)-6,7-dimethoxyquinoline; tert-butyl 2-(4-(6,7-dimethoxyquinolin-4-yloxy)-3-fluorophenyl)-lH-pyrrole-l- carboxylate;
5-(4-(6,7-dimethoxyquinolin-4-yloxy)-3-fluorophenyl)-2-benzylpyrimidin-4(3H)-one; 5-(4-(6,7-dimethoxyquinolin-4-yloxy)-3-fluorophenyl)-3-benzylpyrimidin-4(3H)-one; 5-(4-(6,7-dimethoxyquinolin-4-yloxy)-3-fluorophenyl)pyrimidin-4(3H)-one; 3-(4-(6,7-dimethoxyquinolin-4-yloxy)-3-fluorophenyl)-6-benzylpyridin-2(lH)-one; 111-03-PCT - P2338R1 -02120.004WO1
187
(l-(4-(6,7-dimethoxyquinolin-4-yloxy)-3-fluorophenyl)-l,2-dihydro-2-oxopyridin-4- yl)(phenyl)methyl acetate; l-(4-(6,7-dimethoxyquinolin-4-yloxy)-3-fluorophenyl)-4- (hydroxy(phenyl)methyl)pyridin-2(lH)-one; 1 -(4-(6,7-dimethoxyquinolin-4-yloxy)-3-fluorophenyl)-(4-phenylmethanone) pyridin-
2(lH)-one; l-(4-(6,7-dimethoxyquinolin-4-yloxy)-3-fluorophenyl)-3-methylpyridin-2(lH)-one; l-(4-(6,7-dimethoxyquinolin-4-yloxy)-3-fluorophenyl)-4-methylpyridin-2(lH)-one; l-(4-(6,7-dimethoxyquinolin-4-yloxy)-3-fluorophenyl)-4-benzylpyridin-2(lH)-one; and l-(4-(6,7-dimethoxyquinolin-4-yloxy)-3-fluorophenyl)pyridin-2(lH)-one.
67. A pharmaceutical composition comprised of a compound of claim 1.
68. The composition according to claim 67, further comprising an additional therapeutic agent selected from an anti-proliferative agent, an anti-inflammatory agent, an immunomodulatory agent, a neurotropic factor, an agent for treating cardiovascular disease, an agent for treating liver disease, an anti-viral agent, an agent for treating blood disorders, an agent for treating diabetes, or an agent for treating immunodeficiency disorders.
69. A composition comprising a compound of claim 1 in an amount to detectably inhibit Met kinase activity and a pharmaceutically acceptable carrier, adjuvant, or vehicle.
70. A method of treating or lessening the severity of a disease or condition selected from the group consisting of cancer, stroke, diabetes, hepatomegaly, cardiovascular disease, Alzheimer's disease, cystic fibrosis, viral disease, autoimmune diseases, atherosclerosis, restenosis, psoriasis, allergic disorders, inflammation, neurological disorders, a hormone-related disease, conditions associated with organ transplantation, immunodeficiency disorders, destructive bone disorders, proliferative disorders, infectious diseases, conditions associated with cell death, thrombin-induced platelet aggregation, chronic myelogenous leukemia (CML), liver disease, pathologic immune conditions involving T cell activation, and CNS disorders in a patient, comprising the step of administering to said patient a compound of claim 1.
71. A method of treating cancer in a mammal in need of such treatment which is comprised of administering to said mammal a therapeutically effective amount of a compound of claim 1.
72 The method of claim 71 wherein the cancer is selected from breast, ovary, cervix, prostate, testis, genitourinary tract, esophagus, larynx, glioblastoma, neuroblastoma, stomach, skin, keratoacanthoma, lung, epidermoid carcinoma, large cell carcinoma, non-small 111-03-PCT- P2338R1 - 02120.004WO1
188 cell lung carcinoma (NSCLC), small cell carcinoma, lung adenocarcinoma, bone, colon, adenoma, pancreas, adenocarcinoma, thyroid, follicular carcinoma, undifferentiated carcinoma, papillary carcinoma, seminoma, melanoma, sarcoma, bladder carcinoma, liver carcinoma and biliary passages, kidney carcinoma, myeloid disorders, lymphoid disorders, hairy cells, buccal cavity and pharynx (oral), lip, tongue, mouth, pharynx, small intestine, colon-rectum, large intestine, rectum, brain and central nervous system, Hodgkin's and leukemia.
73. A process for making a pharmaceutical composition which comprises combining a compound of claim 1 with a pharmaceutically acceptable carrier.
74. The use of a compound according to claim 1 in the manufacture of a medicament for the prophylactic or therapeutic treatment of cancer.
75. The use of a compound according to claim 1 for the treatment of cancer.
76. A method for inhibiting or modulating receptor tyrosine kinase activity, comprising contacting the kinase with an effective inhibitory amount of a compound of claim 1.
77. The method of claim 76 wherein the kinase is c-Met.
78. A method for inhibiting or modulating receptor tyrosine kinase activity in a mammal, comprising administering to the mammal a therapeutically effective amount of a compound of claim 1.
79. The method according to claim 78 wherein the receptor tyrosine kinase is c-
Met.
80. A kit for treating a c-Met-mediated condition, comprising: a) a first pharmaceutical composition comprising a compound of claim 1; and b) instructions for use.
81. The kit of claim 80 further comprising (c) a second pharmaceutical composition, wherein the second pharmaceutical composition comprises a second compound having anti-hyperproliferative activity.
82. The compound of claim 1 wherein -OR10 is selected from the structure:
Figure imgf000189_0001
83. The compound of claim 1 selected from 3-benzyl-5-(4-(7-(benzyloxy)-6- methoxyquinolin-4-yloxy)-3-fluorophenyl)pyrimidin-4(3H)-one.
PCT/US2007/070787 2006-06-08 2007-06-08 Quinoline compounds and methods of use WO2007146824A2 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
EP07798333A EP2032538A2 (en) 2006-06-08 2007-06-08 Quinoline compounds and methods of use
CA002655128A CA2655128A1 (en) 2006-06-08 2007-06-08 Quinoline compounds and methods of use
US12/303,930 US20110053931A1 (en) 2006-06-08 2007-06-08 Quinoline compounds and methods of use
JP2009514553A JP2009539878A (en) 2006-06-08 2007-06-08 Quinoline compounds and methods of use

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US81190906P 2006-06-08 2006-06-08
US60/811,909 2006-06-08

Publications (2)

Publication Number Publication Date
WO2007146824A2 true WO2007146824A2 (en) 2007-12-21
WO2007146824A3 WO2007146824A3 (en) 2008-04-10

Family

ID=38658188

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2007/070787 WO2007146824A2 (en) 2006-06-08 2007-06-08 Quinoline compounds and methods of use

Country Status (6)

Country Link
US (1) US20110053931A1 (en)
EP (1) EP2032538A2 (en)
JP (1) JP2009539878A (en)
CN (1) CN101528702A (en)
CA (1) CA2655128A1 (en)
WO (1) WO2007146824A2 (en)

Cited By (34)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2038272A2 (en) * 2006-06-30 2009-03-25 Sunesis Pharmaceuticals, Inc. Pyridinonyl pdk1 inhibitors
FR2940289A1 (en) * 2008-12-23 2010-06-25 Biopharmed AMINO HYDROXYQUINOLINE CLASS DERIVATIVES FOR THE TREATMENT OF PANCREATIC CANCER
WO2010056527A3 (en) * 2008-10-30 2010-07-08 Gilead Palo Alto, Inc. B-biphenyl sulfonamide compounds as ion channel modulators
JP2011517689A (en) * 2008-04-16 2011-06-16 マックス プランク ゲゼルシャフト ツゥアー フェデルゥン デル ヴィッセンシャフテン エー フォー Quinoline derivatives as AXL kinase inhibitors
EP2423208A1 (en) * 2010-08-28 2012-02-29 Lead Discovery Center GmbH Pharmaceutically active compounds as Axl inhibitors
WO2012068096A2 (en) 2010-11-15 2012-05-24 Exelixis, Inc. Benzoxazepines as inhibitors of pi3k/mtor and methods of their use and manufacture
US8404846B2 (en) 2007-08-29 2013-03-26 Methylgene Inc. Inhibitors of protein tyrosine kinase activity
US8569319B2 (en) 2010-04-29 2013-10-29 Deciphera Pharmaceuticals, LLS Pyridone amides and analogs exhibiting anti-cancer and anti-proliferative activities
WO2014001247A1 (en) 2012-06-26 2014-01-03 Bayer Pharma Aktiengesellschaft N-[4-(Quinolin-4-yloxy)cyclohexyl(methyl)](hetero)arylcarboxamides as androgen receptor antagonists, production and use thereof as medicinal products
CN103739596A (en) * 2013-12-10 2014-04-23 刘磊 Quinazoline derivative used for cardio cerebrovascular diseases
WO2014194975A1 (en) * 2013-06-06 2014-12-11 Merck Patent Gmbh A quinoline inhibitor of the macrophage stimulating 1 receptor mst1 r
US20140364431A1 (en) * 2011-12-30 2014-12-11 Shenyang Pharmaceutical University Quinoline and cinnoline derivatives and their applications
WO2015012298A1 (en) 2013-07-24 2015-01-29 小野薬品工業株式会社 Quinoline derivative
EP2852388A4 (en) * 2012-05-23 2016-01-13 Univ Johns Hopkins Compounds and methods of use thereof for treating neurodegenerative disorders
US9745283B2 (en) 2011-11-14 2017-08-29 Ignyta, Inc. Uracil derivatives as AXL and c-MET kinase inhibitors
WO2017168448A1 (en) 2016-03-30 2017-10-05 Council Of Scientific & Industrial Research Silicon incorporated quinolines with anti-malarial and anti-toxoplasmosis activity
AU2014219024B2 (en) * 2013-02-20 2018-04-05 KALA BIO, Inc. Therapeutic compounds and uses thereof
WO2018108671A1 (en) 2016-12-16 2018-06-21 Basf Se Pesticidal compounds
WO2018177781A1 (en) 2017-03-28 2018-10-04 Basf Se Pesticidal compounds
US10208034B2 (en) 2014-12-25 2019-02-19 Ono Pharmaceutical Co., Ltd. Quinoline derivative
WO2019121159A1 (en) 2017-12-21 2019-06-27 Basf Se Pesticidal compounds
WO2019124548A1 (en) 2017-12-22 2019-06-27 住友化学株式会社 Heterocyclic compound and harmful arthropod controlling agent containing same
WO2019231942A1 (en) * 2018-06-01 2019-12-05 Rigel Pharmaceuticals, Inc. Quinoline derivatives useful as tyrosine kinase inhibitors
WO2020027723A1 (en) * 2018-08-01 2020-02-06 Agency For Science, Technology And Research Bicyclic compounds as kinase modulators, methods and uses thereof
WO2020058062A1 (en) 2018-09-19 2020-03-26 Bayer Aktiengesellschaft Herbicidally active substituted phenylpyrimidine hydrazides
EP3643705A1 (en) 2018-10-24 2020-04-29 Basf Se Pesticidal compounds
WO2020141470A1 (en) * 2019-01-03 2020-07-09 Array Biopharma Inc. Quinoline compounds as inhibitors of tam and met kinases
WO2020154610A1 (en) * 2019-01-25 2020-07-30 Exelixis, Inc. Compounds for the treatment of kinase-dependent disorders
US10836747B2 (en) 2017-01-26 2020-11-17 Ono Pharmaceutical Co., Ltd. Ethane-sulfonate salt of quinoline derivative
EP3766879A1 (en) 2019-07-19 2021-01-20 Basf Se Pesticidal pyrazole derivatives
WO2021025407A1 (en) * 2019-08-02 2021-02-11 웰마커바이오 주식회사 Oxo-pyridine fusion ring derivative and pharmaceutical composition comprising same
EP3795567A4 (en) * 2018-05-15 2022-03-02 Beijing InnoCare Pharma Tech Co., Ltd. Indoline-1-formamide compound, preparation method therefor, and medical use thereof
US11753395B2 (en) 2021-12-16 2023-09-12 Kinnate Biopharma Inc. Inhibitors of MET kinase
US11826363B2 (en) 2017-10-13 2023-11-28 Ono Pharmaceutical Co., Ltd. Therapeutic agent for solid cancers, which comprises Axl inhibitor as active ingredient

Families Citing this family (25)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2008236670B2 (en) * 2007-04-05 2011-12-01 Amgen Inc. Aurora kinase modulators and method of use
SG188802A1 (en) * 2008-03-06 2013-04-30 Genentech Inc Combination therapy with c-met and egfr antagonists
WO2012011548A1 (en) * 2010-07-23 2012-01-26 国立大学法人 東京大学 Nitrogen-containing heterocyclic derivative
CN102558147B (en) * 2010-12-23 2014-09-17 江苏先声药物研究有限公司 Compound, and preparation method and application thereof
ES2590778T3 (en) 2011-02-28 2016-11-23 Calitor Sciences, Llc Quinoline Substituted Compounds   
CN103965107B (en) * 2013-02-06 2016-08-17 沈阳药科大学 2-aryl substituted quinoline derivatives and application thereof
AU2014216178B2 (en) 2013-02-15 2018-06-28 KALA BIO, Inc. Therapeutic compounds and uses thereof
US9688688B2 (en) 2013-02-20 2017-06-27 Kala Pharmaceuticals, Inc. Crystalline forms of 4-((4-((4-fluoro-2-methyl-1H-indol-5-yl)oxy)-6-methoxyquinazolin-7-yl)oxy)-1-(2-oxa-7-azaspiro[3.5]nonan-7-yl)butan-1-one and uses thereof
CN104072480B (en) * 2013-03-27 2016-12-28 沈阳药科大学 Quinolines and its preparation method and application
AU2014260011A1 (en) * 2013-05-01 2015-11-12 Celgene Corporation Synthesis of 3-(5-amino-2-methyl-4-oxoquinazolin-3(4H)-yl)piperidine-2-6-dione
MX355330B (en) 2013-11-01 2018-04-16 Kala Pharmaceuticals Inc CRYSTALLINE FORMS OF THERAPEUTIC COMPOUNDS and USES THEREOF.
US9890173B2 (en) 2013-11-01 2018-02-13 Kala Pharmaceuticals, Inc. Crystalline forms of therapeutic compounds and uses thereof
MX2016012285A (en) 2014-03-24 2017-01-23 Genentech Inc Cancer treatment with c-met antagonists and correlation of the latter with hgf expression.
CN104086529B (en) * 2014-05-18 2015-12-30 马宝花 A kind of quinazoline derivative, Its Preparation Method And Use
CN104817497B (en) * 2015-03-20 2017-03-08 南京众睿缘生物科技有限公司 A kind of alkynes is for quinoline and its production and use
CN104926789A (en) * 2015-05-20 2015-09-23 沈阳药科大学 4-phenoxyl substituted quinoline compound containing imidazolone and application thereof
CN107474039B (en) * 2016-06-08 2020-05-01 沈阳药科大学 4-phenoxy substituted quinoline compound containing triazazolone and imidazole and application thereof
EP3509421A4 (en) 2016-09-08 2020-05-20 Kala Pharmaceuticals, Inc. Crystalline forms of therapeutic compounds and uses thereof
AU2017324716B2 (en) 2016-09-08 2020-08-13 KALA BIO, Inc. Crystalline forms of therapeutic compounds and uses thereof
EP3509422A4 (en) 2016-09-08 2020-05-20 Kala Pharmaceuticals, Inc. Crystalline forms of therapeutic compounds and uses thereof
CN108299292B (en) * 2018-02-13 2021-03-26 安徽工业大学 Application of 5, 7-dibromo-8- (methoxymethoxy) -2-methylquinoline or medicinal salt thereof in treating breast cancer
CN108707145B (en) * 2018-07-05 2021-02-26 沈阳药科大学 Quinoline compound containing five-membered heterocyclic structure and preparation and application thereof
EP3845527A4 (en) * 2018-08-27 2022-06-08 Beijing Yuezhikangtai Biomedicines Co., Ltd. Multi-substituted pyridone derivatives and medical use thereof
CN111303024B (en) * 2018-12-12 2023-03-28 安徽中科拓苒药物科学研究有限公司 Quinoline-structured pan-KIT kinase inhibitor and application thereof
CN117362275A (en) * 2022-06-29 2024-01-09 广州百霆医药科技有限公司 Tyrosine protein kinase inhibitor and application thereof

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1548008A1 (en) * 2002-08-23 2005-06-29 Kirin Beer Kabushiki Kaisha Compound having tgf-beta inhibitory activity and medicinal composition containing the same
WO2005070891A2 (en) * 2004-01-23 2005-08-04 Amgen Inc Compounds and methods of use
WO2006060318A2 (en) * 2004-11-30 2006-06-08 Amgen Inc. Quinolines and quinazoline analogs and their use as medicaments for treating cancer
WO2006117552A1 (en) * 2005-05-05 2006-11-09 Chroma Therapeutics Ltd Quinoline and quinoxaline derivatives as inhibitors of kinase enzymatic activity

Family Cites Families (50)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5880141A (en) * 1995-06-07 1999-03-09 Sugen, Inc. Benzylidene-Z-indoline compounds for the treatment of disease
US20030162795A1 (en) * 1998-10-22 2003-08-28 Pfizer Inc. Thienopyrimidine and thienopyridine derivatives useful as anticancer agents
GB9906566D0 (en) * 1999-03-23 1999-05-19 Zeneca Ltd Chemical compounds
US6297238B1 (en) * 1999-04-06 2001-10-02 Basf Aktiengesellschaft Therapeutic agents
US20020004511A1 (en) * 2000-06-28 2002-01-10 Luzzio Michael Joseph Thiophene derivatives useful as anticancer agents
US6599902B2 (en) * 2001-05-30 2003-07-29 Sugen, Inc. 5-aralkysufonyl-3-(pyrrol-2-ylmethylidene)-2-indolinone derivatives as kinase inhibitors
AU2002345792A1 (en) * 2001-06-21 2003-01-08 Pfizer Inc. Thienopyridine and thienopyrimidine anticancer agents
EP1411046B1 (en) * 2001-06-22 2009-09-16 Kirin Pharma Kabushiki Kaisha Quinoline derivative and quinazoline derivative inhibiting self-phosphorylation of hepatocytus proliferator receptor, and medicinal composition containing the same
US7495104B2 (en) * 2001-10-17 2009-02-24 Kirin Beer Kabushiki Kaisha Quinoline or quinazoline derivatives inhibiting auto-phosphorylation of fibroblast growth factor receptors
EP1483268A2 (en) * 2002-03-01 2004-12-08 Pfizer Inc. Indolyl-urea derivatives of thienopyridines useful as anti-angiogenic agents
US6790852B2 (en) * 2002-04-18 2004-09-14 Hoffmann-La Roche Inc. 2-(2,6-dichlorophenyl)-diarylimidazoles
WO2004031184A1 (en) * 2002-10-01 2004-04-15 Johnson & Johnson Pharmaceutical Research & Development, Inc. 4,6-diaminosubstituted-2-[oxy or aminoxy]-[1,3,5]triazines as protein tyrosine kinase inhibitors
CL2003002287A1 (en) * 2002-11-25 2005-01-14 Wyeth Corp COMPOUNDS DERIVED FROM TIENO [3,2-b] -PIRIDINA-6-CARBONITRILOS AND TIENEO [2,3-b] -PIRIDINA-5-CARBONITRILS, PHARMACEUTICAL COMPOSITION, PROCEDURE OF PREPARATION AND INTERMEDIARY COMPOUNDS, AND THEIR USE IN THE TREATMENT OF CANCER, APOPLEJIA, OSTEOPOROSIS
US7276519B2 (en) * 2002-11-25 2007-10-02 Wyeth Thieno[3,2-b]pyridine-6-carbonitriles and thieno[2,3-b]pyridine-5-carbonitriles as protein kinase inhibitors
MXPA05008521A (en) * 2003-02-13 2005-10-20 Pharmacia Corp Antibodies to c-met for the treatment of cancers.
PL216368B1 (en) * 2003-02-26 2014-03-31 Sugen Aminoheteroaryl compounds as protein kinase inhibitors
EP1615906A1 (en) * 2003-04-03 2006-01-18 Vertex Pharmaceuticals Incorporated Compositions useful as inhibitors of protein kinases
KR20110117728A (en) * 2003-06-06 2011-10-27 제넨테크, 인크. Modulating the interaction between hgf beta chain and c-met
US20060009493A1 (en) * 2003-07-02 2006-01-12 Sugen, Inc. Indolinone hydrazides as c-Met inhibitors
US7122548B2 (en) * 2003-07-02 2006-10-17 Sugen, Inc. Triazolotriazine compounds and uses thereof
CA2536470A1 (en) * 2003-08-15 2005-02-24 Vertex Pharmaceuticals Incorporated Pyrrole compositions useful as inhibitors of c-met
WO2005021554A1 (en) * 2003-08-29 2005-03-10 Pfizer Inc. Thienopyridine-phenylacet amides and their derivatives useful as new anti-angiogenic agents
MXPA06002256A (en) * 2003-08-29 2006-05-17 Pfizer Naphthalene carboxamides and their derivatives useful as new anti-angiogenic agents.
WO2005040345A2 (en) * 2003-09-24 2005-05-06 Vertex Pharmceuticals Incorporated 4-azole substituted imidazole compositions useful as inhibitors or c-met receptor tyrosine kinase
EP2213661B1 (en) * 2003-09-26 2011-07-20 Exelixis Inc. c-Met Modulators and Methods of Use
JP2007518823A (en) * 2004-01-23 2007-07-12 アムゲン インコーポレイテッド Quinoline, quinazoline, pyridine, and pyrimidine compounds and their use in the treatment of inflammation, angiogenesis, and cancer
TW200538120A (en) * 2004-02-20 2005-12-01 Kirin Brewery Compound having TGF-beta inhibitory activity and pharmaceutical composition containing same
US7459562B2 (en) * 2004-04-23 2008-12-02 Bristol-Myers Squibb Company Monocyclic heterocycles as kinase inhibitors
TW200538453A (en) * 2004-04-26 2005-12-01 Bristol Myers Squibb Co Bicyclic heterocycles as kinase inhibitors
AU2005245386B2 (en) * 2004-05-07 2008-11-27 Amgen Inc. Nitrogenated heterocyclic derivatives as protein kinase modulators and use for the treatment of angiogenesis and cancer
EP1753428A4 (en) * 2004-05-14 2010-09-15 Abbott Lab Kinase inhibitors as therapeutic agents
US7452887B2 (en) * 2004-06-04 2008-11-18 Amphora Discovery Corporation Quinoline- and isoquinoline-based compounds exhibiting ATP-utilizing enzyme inhibitory activity, and compositions, and uses thereof
US7439246B2 (en) * 2004-06-28 2008-10-21 Bristol-Myers Squibb Company Fused heterocyclic kinase inhibitors
US20050288290A1 (en) * 2004-06-28 2005-12-29 Borzilleri Robert M Fused heterocyclic kinase inhibitors
JP5368701B2 (en) * 2004-07-02 2013-12-18 エクセリクシス、インコーポレイテッド c-Met Modulator and Method of Use
EP1781664B1 (en) * 2004-07-30 2013-09-04 MethylGene Inc. Inhibitors of vegf receptor and hgf receptor signaling
AU2006229343A1 (en) * 2005-03-28 2006-10-05 Kirin Pharma Kabushiki Kaisha Thienopyridine derivative, or quinoline derivative, or quinazoline derivative, having c-Met autophosphorylation inhibiting potency
JP2008537748A (en) * 2005-04-06 2008-09-25 エクセリクシス、インコーポレイテッド c-Met Modulator and Method of Use
US7470693B2 (en) * 2005-04-21 2008-12-30 Bristol-Myers Squibb Company Oxalamide derivatives as kinase inhibitors
JO2787B1 (en) * 2005-04-27 2014-03-15 امجين إنك, Substituted Amid derivatives & methods of use
BRPI0610382A2 (en) * 2005-05-20 2010-06-15 Methylgene Inc vegf receptor and hgf receptor signaling inhibitors
CN101796055B (en) * 2005-05-20 2013-09-04 梅特希尔基因公司 Inhibitors of VEGF receptor and HGF receptor signaling
US7732613B2 (en) * 2005-09-14 2010-06-08 Bristol-Myers Squibb Company Met kinase inhibitors
US7880004B2 (en) * 2005-09-15 2011-02-01 Bristol-Myers Squibb Company Met kinase inhibitors
US7547782B2 (en) * 2005-09-30 2009-06-16 Bristol-Myers Squibb Company Met kinase inhibitors
CN101522687A (en) * 2006-01-30 2009-09-02 阿雷生物药品公司 Heterobicyclic thiophene compounds and methods of use
US7723330B2 (en) * 2006-03-07 2010-05-25 Array Biopharma Inc. Heterobicyclic pyrazole compounds and methods of use
ATE545417T1 (en) * 2007-03-14 2012-03-15 Ranbaxy Lab Ltd PYRAZOLOA3,4-BUPYRIDINE DERIVATIVES AS PHOSPHODIESTERASE INHIBITORS
CN101918403A (en) * 2007-09-06 2010-12-15 阵列生物制药公司 Pyrazolo-pyridines as tyrosine kinase inhibitor
EP2225239B1 (en) * 2007-12-21 2014-10-22 F. Hoffmann-La Roche AG Heterocyclic antiviral compounds

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1548008A1 (en) * 2002-08-23 2005-06-29 Kirin Beer Kabushiki Kaisha Compound having tgf-beta inhibitory activity and medicinal composition containing the same
WO2005070891A2 (en) * 2004-01-23 2005-08-04 Amgen Inc Compounds and methods of use
WO2006060318A2 (en) * 2004-11-30 2006-06-08 Amgen Inc. Quinolines and quinazoline analogs and their use as medicaments for treating cancer
WO2006117552A1 (en) * 2005-05-05 2006-11-09 Chroma Therapeutics Ltd Quinoline and quinoxaline derivatives as inhibitors of kinase enzymatic activity

Cited By (69)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9873693B2 (en) 2006-06-30 2018-01-23 Sunesis Pharmaceuticals, Inc. Methods of treatment using pyridinonyl PDK1 inhibitors
EP2038272A4 (en) * 2006-06-30 2011-01-05 Sunesis Pharmaceuticals Inc Pyridinonyl pdk1 inhibitors
EP2038272A2 (en) * 2006-06-30 2009-03-25 Sunesis Pharmaceuticals, Inc. Pyridinonyl pdk1 inhibitors
US8778977B2 (en) 2006-06-30 2014-07-15 Sunesis Pharmaceuticals, Inc. Pyridinonyl PDK1 inhibitors
US8846927B2 (en) 2007-08-29 2014-09-30 Methylgene Inc. Inhibitors of protein tyrosine kinase activity
US8404846B2 (en) 2007-08-29 2013-03-26 Methylgene Inc. Inhibitors of protein tyrosine kinase activity
JP2011517689A (en) * 2008-04-16 2011-06-16 マックス プランク ゲゼルシャフト ツゥアー フェデルゥン デル ヴィッセンシャフテン エー フォー Quinoline derivatives as AXL kinase inhibitors
WO2010056527A3 (en) * 2008-10-30 2010-07-08 Gilead Palo Alto, Inc. B-biphenyl sulfonamide compounds as ion channel modulators
US8389500B2 (en) 2008-10-30 2013-03-05 Gilead Sciences, Inc. Fused heterocyclic compounds as ion channel modulators
FR2940289A1 (en) * 2008-12-23 2010-06-25 Biopharmed AMINO HYDROXYQUINOLINE CLASS DERIVATIVES FOR THE TREATMENT OF PANCREATIC CANCER
WO2010073235A1 (en) * 2008-12-23 2010-07-01 Biopharmed Ppar agonist compositions and methods of use
US8569319B2 (en) 2010-04-29 2013-10-29 Deciphera Pharmaceuticals, LLS Pyridone amides and analogs exhibiting anti-cancer and anti-proliferative activities
RU2573834C2 (en) * 2010-08-28 2016-01-27 Лид Дискавери Сентер Гмбх PHARMACEUTICALLY ACTIVE COMPOUNDS AS Axl INHIBITORS
US8999982B2 (en) 2010-08-28 2015-04-07 Lead Discovery Center Gmbh Pharmaceutically active compounds as Axl inhibitors
WO2012028332A1 (en) 2010-08-28 2012-03-08 Lead Discovery Center Gmbh Pharmaceutically active compounds as axl inhibitors
EP2423208A1 (en) * 2010-08-28 2012-02-29 Lead Discovery Center GmbH Pharmaceutically active compounds as Axl inhibitors
AU2011297889B2 (en) * 2010-08-28 2015-12-24 Lead Discovery Center Gmbh Pharmaceutically active compounds as Axl inhibitors
WO2012068096A2 (en) 2010-11-15 2012-05-24 Exelixis, Inc. Benzoxazepines as inhibitors of pi3k/mtor and methods of their use and manufacture
US10017496B2 (en) 2011-11-14 2018-07-10 Ignyta, Inc. Uracil derivatives as AXL and c-MET kinase inhibitors
US9745283B2 (en) 2011-11-14 2017-08-29 Ignyta, Inc. Uracil derivatives as AXL and c-MET kinase inhibitors
EP2799437A4 (en) * 2011-12-30 2015-08-19 Univ Shenyang Pharmaceutical Quinoline and cinnoline compounds and use thereof
US20140364431A1 (en) * 2011-12-30 2014-12-11 Shenyang Pharmaceutical University Quinoline and cinnoline derivatives and their applications
US9382232B2 (en) 2011-12-30 2016-07-05 Shenyang Pharmaceutical University Quinoline and cinnoline derivatives and their applications
EP2852388A4 (en) * 2012-05-23 2016-01-13 Univ Johns Hopkins Compounds and methods of use thereof for treating neurodegenerative disorders
US9539259B2 (en) 2012-05-23 2017-01-10 The Johns Hopkins University Compounds and methods of use thereof for treating neurodegenerative disorders
US9428460B2 (en) 2012-06-26 2016-08-30 Bayer Pharma Aktiengesellschaft N-[4-(quinolin-4-yloxy)cyclohexyl(methyl)](hetero)arylcarboxamides as androgen receptor antagonists, production and use thereof as medicinal products
WO2014001247A1 (en) 2012-06-26 2014-01-03 Bayer Pharma Aktiengesellschaft N-[4-(Quinolin-4-yloxy)cyclohexyl(methyl)](hetero)arylcarboxamides as androgen receptor antagonists, production and use thereof as medicinal products
AU2014219024B2 (en) * 2013-02-20 2018-04-05 KALA BIO, Inc. Therapeutic compounds and uses thereof
US9481668B2 (en) 2013-06-06 2016-11-01 Merck Patent Gmbh Quinoline inhibitor of the macrophage stimulating 1 receptor MSTR1
JP2016520627A (en) * 2013-06-06 2016-07-14 メルク パテント ゲゼルシャフト ミット ベシュレンクテル ハフツングMerck Patent Gesellschaft mit beschraenkter Haftung Quinoline inhibitor of 1 receptor MSTR1 that stimulates macrophages
WO2014194975A1 (en) * 2013-06-06 2014-12-11 Merck Patent Gmbh A quinoline inhibitor of the macrophage stimulating 1 receptor mst1 r
AU2014277262B2 (en) * 2013-06-06 2019-10-03 Lead Discovery Center Gmbh A quinoline inhibitor of the macrophage stimulating 1 receptor MST1 R
JP2018024691A (en) * 2013-07-24 2018-02-15 小野薬品工業株式会社 Quinoline derivative
JPWO2015012298A1 (en) * 2013-07-24 2017-03-02 小野薬品工業株式会社 Quinoline derivatives
US9573935B2 (en) 2013-07-24 2017-02-21 Ono Pharmaceutical Co., Ltd. Quinoline derivative
KR20160033707A (en) 2013-07-24 2016-03-28 오노 야꾸힝 고교 가부시키가이샤 Quinoline derivative
US9994549B2 (en) 2013-07-24 2018-06-12 Ono Pharmaceutical Co., Ltd. Quinoline derivative
KR102251609B1 (en) 2013-07-24 2021-05-13 오노 야꾸힝 고교 가부시키가이샤 Quinoline derivative
WO2015012298A1 (en) 2013-07-24 2015-01-29 小野薬品工業株式会社 Quinoline derivative
US10676462B2 (en) 2013-07-24 2020-06-09 Ono Pharmaceutical Co., Ltd. Quinoline derivative
EP3415501A1 (en) 2013-07-24 2018-12-19 ONO Pharmaceutical Co., Ltd. Quinoline derivative
US10208022B2 (en) 2013-07-24 2019-02-19 Ono Pharmaceutical Co., Ltd. Quinoline derivative
US10501442B2 (en) 2013-07-24 2019-12-10 Ono Pharmaceuticals Co., Ltd. Quinoline derivative
CN103739596A (en) * 2013-12-10 2014-04-23 刘磊 Quinazoline derivative used for cardio cerebrovascular diseases
US10208034B2 (en) 2014-12-25 2019-02-19 Ono Pharmaceutical Co., Ltd. Quinoline derivative
WO2017168448A1 (en) 2016-03-30 2017-10-05 Council Of Scientific & Industrial Research Silicon incorporated quinolines with anti-malarial and anti-toxoplasmosis activity
US10550135B2 (en) 2016-03-30 2020-02-04 Council Of Scientific & Industrial Research Silicon incorporated quinolines with anti-malarial and anti-toxoplasmosis activity
WO2018108671A1 (en) 2016-12-16 2018-06-21 Basf Se Pesticidal compounds
US10836747B2 (en) 2017-01-26 2020-11-17 Ono Pharmaceutical Co., Ltd. Ethane-sulfonate salt of quinoline derivative
WO2018177781A1 (en) 2017-03-28 2018-10-04 Basf Se Pesticidal compounds
US11826363B2 (en) 2017-10-13 2023-11-28 Ono Pharmaceutical Co., Ltd. Therapeutic agent for solid cancers, which comprises Axl inhibitor as active ingredient
WO2019121159A1 (en) 2017-12-21 2019-06-27 Basf Se Pesticidal compounds
WO2019124548A1 (en) 2017-12-22 2019-06-27 住友化学株式会社 Heterocyclic compound and harmful arthropod controlling agent containing same
EP3795567A4 (en) * 2018-05-15 2022-03-02 Beijing InnoCare Pharma Tech Co., Ltd. Indoline-1-formamide compound, preparation method therefor, and medical use thereof
WO2019231942A1 (en) * 2018-06-01 2019-12-05 Rigel Pharmaceuticals, Inc. Quinoline derivatives useful as tyrosine kinase inhibitors
US10851093B2 (en) 2018-06-01 2020-12-01 Rigel Pharmaceuticals, Inc. Tyrosine kinase inhibitors
WO2020027723A1 (en) * 2018-08-01 2020-02-06 Agency For Science, Technology And Research Bicyclic compounds as kinase modulators, methods and uses thereof
US11702425B2 (en) 2018-08-01 2023-07-18 Agency For Science, Technology And Research Bicyclic compounds as kinase modulators, methods and uses thereof
WO2020058062A1 (en) 2018-09-19 2020-03-26 Bayer Aktiengesellschaft Herbicidally active substituted phenylpyrimidine hydrazides
WO2020083733A1 (en) 2018-10-24 2020-04-30 Basf Se Pesticidal compounds
EP3643705A1 (en) 2018-10-24 2020-04-29 Basf Se Pesticidal compounds
TWI745824B (en) * 2019-01-03 2021-11-11 美商亞雷生物製藥股份有限公司 Quinoline compounds as inhibitors of tam and met kinases
WO2020141470A1 (en) * 2019-01-03 2020-07-09 Array Biopharma Inc. Quinoline compounds as inhibitors of tam and met kinases
WO2020154610A1 (en) * 2019-01-25 2020-07-30 Exelixis, Inc. Compounds for the treatment of kinase-dependent disorders
CN113710322A (en) * 2019-01-25 2021-11-26 埃克塞里艾克西斯公司 Compounds for the treatment of kinase-dependent disorders
EP3766879A1 (en) 2019-07-19 2021-01-20 Basf Se Pesticidal pyrazole derivatives
WO2021013561A1 (en) 2019-07-19 2021-01-28 Basf Se Pesticidal pyrazole and triazole derivatives
WO2021025407A1 (en) * 2019-08-02 2021-02-11 웰마커바이오 주식회사 Oxo-pyridine fusion ring derivative and pharmaceutical composition comprising same
US11753395B2 (en) 2021-12-16 2023-09-12 Kinnate Biopharma Inc. Inhibitors of MET kinase

Also Published As

Publication number Publication date
US20110053931A1 (en) 2011-03-03
JP2009539878A (en) 2009-11-19
CA2655128A1 (en) 2007-12-21
EP2032538A2 (en) 2009-03-11
WO2007146824A3 (en) 2008-04-10
CN101528702A (en) 2009-09-09

Similar Documents

Publication Publication Date Title
US20110053931A1 (en) Quinoline compounds and methods of use
US8003662B2 (en) Heterobicyclic thiophene compounds and methods of use
US7723330B2 (en) Heterobicyclic pyrazole compounds and methods of use
US20110130406A1 (en) Pyrazolo-pyridines as tyrosine kinase inhibitors
RU2592703C2 (en) INHIBITORS OF TYPE ErbB INHIBITORS
JP6026427B2 (en) Substituted 6,6-fused nitrogen heterocyclic compounds and uses thereof
JP5677945B2 (en) Diazacarbazole and method of use thereof
US20070049603A1 (en) Raf inhibitor compounds and methods of use thereof
JP5689069B2 (en) Pyrazolopyridine PI3K inhibitor compounds and methods of use
EP2440529A1 (en) Janus kinase inhibitor compounds and methods
JPWO2005080377A1 (en) Compound having TGFβ inhibitory activity and pharmaceutical composition comprising the same
TW202039490A (en) Heterobicyclic inhibitors of mat2a and methods of use for treating cancer
EP3319602B1 (en) Fused quinoline compounds as pi3k/mtor inhibitors
CN104822658A (en) Fused tricyclic amide compounds as multiple kinase inhibitors
TWI565698B (en) Quinoxaline compounds, method for preparing the same and use thereof
JP6943886B2 (en) Condensed pyrimidinopiperidin derivative, and its production method and application

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 200780029441.2

Country of ref document: CN

121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 07798333

Country of ref document: EP

Kind code of ref document: A2

WWE Wipo information: entry into national phase

Ref document number: 2009514553

Country of ref document: JP

ENP Entry into the national phase

Ref document number: 2655128

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 5043/KOLNP/2008

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: 2007798333

Country of ref document: EP

NENP Non-entry into the national phase

Ref country code: RU