WO2013177092A1 - Substituted alkynyl pyridine compounds and methods of use - Google Patents

Substituted alkynyl pyridine compounds and methods of use Download PDF

Info

Publication number
WO2013177092A1
WO2013177092A1 PCT/US2013/041918 US2013041918W WO2013177092A1 WO 2013177092 A1 WO2013177092 A1 WO 2013177092A1 US 2013041918 W US2013041918 W US 2013041918W WO 2013177092 A1 WO2013177092 A1 WO 2013177092A1
Authority
WO
WIPO (PCT)
Prior art keywords
alkylene
cancer
compound
cycloalkyl
heterocyclyl
Prior art date
Application number
PCT/US2013/041918
Other languages
French (fr)
Inventor
Ning Xi
Lihua Yin
Xiaobo Li
Na YU
Yanjun Wu
Original Assignee
Sunshine Lake Pharma Co., Ltd.
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 Sunshine Lake Pharma Co., Ltd. filed Critical Sunshine Lake Pharma Co., Ltd.
Publication of WO2013177092A1 publication Critical patent/WO2013177092A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/60Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D213/72Nitrogen atoms
    • C07D213/73Unsubstituted amino or imino radicals
    • 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
    • C07D401/06Heterocyclic 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 carbon chain containing only aliphatic carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/02Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings
    • C07D405/06Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms

Definitions

  • This invention relates to novel substituted alkynyl pyridine compounds, and salts thereof, which are useful in the treatment of hyperproliferative diseases, such as cancers, in mammals.
  • the invention relates to compounds that inhibit the protein tyrosine kinase activity, resulting in the inhibition of inter- and/or intra-cellular signaling.
  • This invention also relates to a method of using such compounds in the treatment of hyperproliferative diseases in mammals, especially humans, and to pharmaceutical compositions containing such compounds.
  • Protein kinases are key regulators of cell function that constitute one of the largest and most functionally diverse gene families. By adding phosphate groups to substrate proteins, they direct the activity, localization and overall function of many proteins, and serve to orchestrate the activity of many cellular processes. Kinases are particularly prominent in signal transduction and co-ordination of complex functions such as the cell cycle. Of the 518 human protein kinases, 478 belong to a single superfamily whose catalytic domains are related in sequence. These can be clustered into groups, families and sub-families, of increasing sequence similarity and biochemical function.
  • a partial list of such kinases includes abl, AATK, ALK, Akt, Axl, bmx, bcr-abl, Blk, Brk, Btk, csk, c-kit, c-Met, c-src, c-fins, CDK1, CDK2, CDK3, CDK4, CDK5, CDK6, CDK7, CDK8, CDK9, CDK10, cRafl, CSF1R, CSK, DDR1, DDR2, EPHA, EPHB, EGFR, ErbB2, ErbB3, ErbB4, Erk, Fak, fes, FER, FGFR1, FGFR2, FGFR3, FGFR4, FGFR5, Fgr, flt-l, Fps, Frk, Fyn, GSG2, GSK, Hck, ILK, INSRR, IRAK4, ITK, IGF-1R, INS-R, Jak, KSR1, K
  • Receptor tyrosine kinases are a diverse group of transmembrane proteins that act as receptors for cytokines, growth factors, hormones and other signaling molecules. Receptor tyrosine kinases (RTKs) are expressed in many cell types and play important roles in a wide variety of cellular processes, including growth, differentiation and angiogenesis. Activation of the kinase is effected by binding of a ligand to the extracellular domain, which induces dimerization of the receptors. Activated receptors auto-phosphorylate tyrosine residues outside the catalytic domain via cross-phosphorylation. This auto-phosphorylation stabilizes the active receptor conformation and creates phosphotyrosine docking sites for proteins that transduce signals within the cell.
  • RTKs Receptor tyrosine kinases
  • Receptor tyrosine kinases are hyper-activated (through receptor activating mutations, gene amplification, growth factor activation, etc.) in many human solid tumors and hematological malignancies. RTK's elevated activation contributes to tumourigenesis factors such as hyperplasia, survival, invasion, metastasis and angiogenesis. Inhibition of receptor tyrosine kinases proved to be effective strategies in cancer therapy (Sharma et al., "Receptor tyrosine kinase inhibitors as potent weapons in war against cancers", Curr. Pharm. Des., 2009, 15, 758).
  • ALK Anaplastic lymphoma kinase
  • NPM nucleophosmin
  • ALK anaplastic large -cell lymphoma
  • ALK fusions were also found in the human sarcomas called inflammatory myofibroblastic tumors (IMTs). Studies suggested that the ALK fusion, TPM4-ALK, may be involved in the genesis of a subset of esophageal squamous cell carcinomas. Moreover, studies have implicated various mutations of the ALK gene in both familial and sporadic cases of neuroblastoma. ALK mutations in neuroblastoma cells results in constitutive ALK phosphorylation and attenuation.
  • EML4-ALK fusion gene comprised of portions of the echinoderm microtubule-associated protein-like 4 (EML4) gene and the ALK gene were identified in NSCLC cells.
  • EML4-ALK fusion transcript was detected in approximately 3- 7% of NSCLC patients examined.
  • Experimental evidences from in vitro and in vivo studies demonstrated oncogenic transforming activity of the EML4-ALK fusion proteins and reinforced the pivotal role of EML4-ALK in the pathogenesis of NSCLC in humans (Soda et al., "Identification of the transforming EML4-ALK fusion gene in non-small-cell lung cancer", Nature, 2007, 448, 561).
  • JAK3-STAT3 pathway and the PI3K-Akt pathway have been shown to be vital primarily for cell survival and phenotypic changes (Chiarle et al., "The anaplastic lymphoma kinase in the pathogenesis of cancer", Nat. Rev. Cancer, 2008, 8, 11 ; Barreca et al., "Anaplastic lymphoma kinase (ALK) in human cancer", J. Mol. Endocrinol., 2011, 47, Rl 1).
  • c-Met also referred to as hepatocyte growth factor receptor (HGFR)
  • HGFR hepatocyte growth factor receptor
  • HGF hepatocyte growth factor
  • SF scatter factor
  • c-Met In both embryos and adults, activated c-Met promotes a morphogenetic program, known as invasive growth, which induces cell spreading, the disruption of intercellular contacts, and the migration of cells towards their surroundings (Peschard et al., "From Tpr-Met to Met, tumorigenesis and tubes” Oncogene, 2007, 26, 1276; Stellrecht et al., "Met Receptor Tyrosine Kinase as a Therapeutic Anticancer Target", Cancer Letter, 2009, 280, 1). [013] A wide variety of human malignancies exhibit sustained c-Met stimulation, overexpression, or mutation, including carcinomas of the breast, liver, lung, ovary, kidney, thyroid, colon, renal, glioblastomas, and prostate, etc.
  • c-Met is also implicated in atherosclerosis and lung fibrosis. Invasive growth of certain cancer cells is drastically enhanced by tumor- stromal interactions involving the HGF/c-Met pathway. Thus, extensive evidence indicates that c-Met signaling is involved in the progression and spread of several cancers and an enhanced understanding of its role in disease has generated considerable interest in c-Met as major targets in cancer drug development (Migliore et al., "Molecular cancer therapy: can our expectation be MET", Eur. J. Cancer, 2008, 44, 641; Peruzzi et al., “Targeting the c-Met Signaling Pathway in Cancer", Clinical Cancer Research, 2006, 12, 3657).
  • Crizotinib is an ATP-competitive small molecule ALK inhibitor, which also displays activity against the c-Met receptor tyrosine kinase.
  • the FDA recently approved crizotinib (Pfizer' s XALKORI ® , originally known as PF-02341066) for treatment of patients with locally advanced or metastatic non-small cell lung cancer (NSCLC), in which tumor cells exhibit rearrangements in the anaplastic lymphoma kinase (ALK) gene.
  • Crizotinib is administered 250 mg twice daily. Following oral single-dose administration, crizotinib was absorbed with median time to achieve peak concentration from 4 to 6 hours. Following crizotinib 250 mg twice daily, steady state was reached within 15 days and remained stable, with a median accumulation ratio of 4.8 (XALKORI ® FDA-Approved Patient Labeling, Pfizer Inc. February 2012).
  • crizotinib As seen with other targeted cancer drugs, patients with ALK-positive NSCLC eventually relapse on crizotinib. The development of acquired resistance is clearly the major hurdle preventing targeted therapies such as crizotinib from having an even more substantial impact on patients (Nature Reviews Drug Discovery, 2011, 10, 897).
  • the present invention provides novel compounds believed to have clinical use for treatment of cancer through inhibiting ALK and/or c-Met. Preferred compounds of the present invention are also believed to provide an improvement in potency, pharmacokinetic properties, and/or toxicity profile over certain other ALK and/or c-Met inhibitor compounds found in the art.
  • the present invention provides new compounds and methods for treating cell proliferative diseases.
  • the compounds of the invention are inhibitors of protein tyrosine kinases.
  • the compounds of the invention are capable of inhibiting, for example, ALK (including ALK fusions such as EML4-ALK, NPM-ALK, etc.), and c-Met receptor (hepatocyte growth factor receptor) signaling.
  • ALK including ALK fusions such as EML4-ALK, NPM-ALK, etc.
  • c-Met receptor hepatocyte growth factor receptor
  • each R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , X and Z is as defined herein.
  • each R 1 , R 2 , R 3 , R 4 , R 5 and R 6 is independently H, D or F;
  • Z is Ci_ 6 alkyl, C3_ 6 cycloalkyl, -(Ci ⁇ alkylene)-(C 3 _ 6 cycloalkyl), C 3 - 6 heterocyclyl,
  • each R and R b is independently H, Ci- 6 aliphatic, C 3 _ 6 cycloalkyl, -(Ci_ 4 alkylene)-(C 3 _ 6 cycloalkyl), C 3 _ 6 heterocyclyl, -(Ci_ 4 alkylene)-(C 3 _ 6 heterocyclyl), C6-ioaryl, 5-10 membered heteroaryl or -(5-10membered heteroaryl), provided that when Z is Ci_ 6 alkyl, NR R b is not H 2 or Me 2 , or when R and R b are bonded to the same nitrogen atom, R and R b , together with the nitrogen atom they are attached to, optionally form a 3-8 membered heterocyclyl, wherein each of the Ci_ 6 aliphatic, C 3 _ 6 cycloalkyl, -(Ci_ 4 alkylene)-(C 3 _ 6 cycloalkyl), C 3 _ 6 heterocyclyl,
  • each R 1 , R 2 , R 3 , R 4 , R 5 and R 6 is independently H or D.
  • X is phenyl optionally substituted with 1, 2, 3 or 4 substituents independently selected from D, F, CI, Br,
  • Z is C 3 _ 6 cycloalkyl, C 3 _ 6 heterocyclyl, -(Ci_ 4 alkylene)- (C 3 _ 6 heterocyclyl), or -(Ci ⁇ alkylene)-(C5-i 2 spirobicyclyl), wherein the Ci- 6 alkyl, C 3 _ 6 cycloalkyl, C 3 _ 6 heterocyclyl, -(Ci_ 4 alkylene)-(C 3 _ 6 heterocyclyl), bicyclyl), C5- i 2 spirobicyclyl and -(Ci ⁇ alkylene)-(C5_i 2 spirobicyclyl), each of which is optionally substituted with 1, 2, 3 or 4 substituents independently selected from D, F, CI, Br, I, OR , NR R b ; each of the cyclic structures in the bicyclyl groups is a carbocyclic ring or a heterocyclic ring.
  • each of R and R b is independently H, Ci_ 3 alkyl, C3_ 6 cycloalkyl or -(Ci- 3 alkylene)-(C 3 _ 6 cycloalkyl), provided that when Z is NR R b is not NH 2 or Me 2 , wherein each of the C3_ 6 cycloalkyl or -(Ci_ 3 alkylene)-(C 3 _ 6 cycloalkyl) is optionally substituted with 1, 2, 3 or 4 substituents independently selected from D, F, OH, NH 2 ,
  • Z is selected from the following substructures:
  • each W and W is independently O or NH;
  • each hydrogen in Z is optionally substituted with 1, 2, 3 or 4 substituents independently selected from D, F, CI, Br, I, C ⁇ alkyl, d_ 6 haloalkyl, OR a and NR a R b , provided that NR a R b is not NH 2 or NMe 2 in structures (Z25) to (Z32).
  • compositions comprising a compound disclosed herein, or a stereoisomer, geometric isomer, tautomer, solvate, metabolite, pharmaceutically acceptable salt or prodrug thereof, and an optional pharmaceutically acceptable carrier, excipient, diluent, adjuvant, vehicle or a combination thereof.
  • the compound is an inhibitor of protein tyrosine kinase.
  • the compound is an inhibitor of ALK receptor signaling and HGF receptor signaling.
  • the pharmaceutical composition disclosed herein further comprises an additional therapeutic agent.
  • the therapeutic agent is a chemotherapeutic agent, an anti-proliferative agent, an agent for treating atherosclerosis, an agent for treating lung fibrosis, and combinations thereof.
  • the therapeutic agent is chlorambucil, melphalan, cyclophosphamide, ifosfamide, busulfan, carmustine, lomustine, streptozocin, cisplatin, carboplatin, oxaliplatin, dacarbazine, temozolomide, procarbazine, methotrexate, fluorouracil, cytarabine, gemcitabine, mercaptopurine, fludarabine, vinblastine, vincristine, vinorelbine, paclitaxel, docetaxel, topotecan, irinotecan, etoposide, trabectedin, dactinomycin, doxorubicin, epirubicin, daunorubicin, mitoxantrone, bleomycin, mitomycin, ixabepilone, tamoxifen, flutamide, gonadorelin analogues, megestrol
  • kits for preventing, managing, treating or lessening the severity of a proliferative disorder in a patient infected with the proliferative disorder which comprises administrating a pharmaceutically effective amount of a compound disclosed herein, or the pharmaceutical composition disclosed herein to the patient.
  • provided herein is use of the compound disclosed herein, or the pharmaceutical composition disclosed herein in the manufacture of a medicament for preventing, managing, treating or lessening the severity of a proliferative disorder in a patient.
  • the proliferative disorder is metastatic cancer.
  • the proliferative disorder is colon cancer, gastric adenocarcinoma, bladder cancer, breast cancer, kidney cancer, liver cancer, lung cancer, skin cancer, thyroid cancer, cancer of the head and neck, prostate cancer, pancreatic cancer, cancer of the CNS, glioblastoma or a myeloproliferative disorder.
  • the proliferative disorder is atherosclerosis or lung fibrosis.
  • a method of inhibiting or modulating the activity of a protein kinase in a biological sample comprising contacting a biological sample with the compound disclosed herein, or the pharmaceutical composition disclosed herein.
  • the protein kinase is a receptor tyrosine kinase.
  • the receptor tyrosine kinase is ALK and/or c-Met.
  • a method of inhibiting protein tyrosine kinase comprises contacting the kinase with the compound disclosed herein, or with the composition disclosed herein.
  • a method of inhibiting ALK receptor signaling and/or HGF receptor signaling comprises contacting the receptor with the compound disclosed herein, or with the pharmaceutical composition disclosed herein.
  • inhibition of receptor protein kinase activity can be in a cell or a multicellular organism. If in a multicellular organism, the method disclosed herein may comprise administering to the organism the compound disclosed herein, or the pharmaceutical composition disclosed herein. In some embodiments, the organism is a mammal. In other embodiments, the organism is a human. In still other embodiments, the method further comprises contacting the kinase with an additional therapeutic agent.
  • a method of inhibiting proliferative activity of a cell comprising contacting the cell with an effective proliferative inhibiting amount of the compound disclosed herein or the pharmaceutical composition disclosed herein. In some embodiments, the method further comprises contacting the cell with an additional therapeutic agent.
  • a method of treating a cell proliferative disease in a patient comprising administering to the patient in need of such treatment an effective therapeutic amount of the compound disclosed herein or the pharmaceutical composition disclose herein. In other embodiments, the method further comprises administering an additional therapeutic agent.
  • a method of inhibiting tumor growth in a patient comprises administering to the patient in need thereof an effective therapeutic amount of a compound disclosed herein or a composition thereof. In other embodiments, the method further comprises administering an additional therapeutic agent.
  • provided herein include methods of preparing, methods of separating, and methods of purifying compounds of Formula (I).
  • compounds of the invention may optionally be substituted with one or more substituents, such as those illustrated below, or as exemplified by particular classes, subclasses, and species of the invention.
  • substituents such as those illustrated below, or as exemplified by particular classes, subclasses, and species of the invention.
  • the phrase “optionally substituted” is used interchangeably with the phrase “substituted or unsubstituted”.
  • substituted whether proceeded by the term “optionally” or not, refers to the replacement of one or more hydrogen radicals in a given structure with the radical of a specified substituent.
  • an optionally substituted group may have a substituent at each substitutable position of the group.
  • substituent When more than one position in a given structure can be substituted with more than one substituent selected from a specified group, the substituent may be either the same or different at each position.
  • substituent include, but are not limited to, D, F, CI, Br, I, alkyl, haloalkyl, alkenyl, alkynyl, OH and NH 2 .
  • aliphatic refers to a straight-chain (i.e., unbranched) or branched, substituted or unsubstituted hydrocarbon chain that is completely saturated or that contains one or more units of unsaturation. Unless otherwise specified, aliphatic groups contain
  • aliphatic groups contain 1- 10 carbon atoms. In other embodiments, aliphatic groups contain 1 -8 carbon atoms. In still other embodiments, aliphatic groups contain 1 -6 carbon atoms, and in yet other embodiments, aliphatic groups contain 1-4 carbon atoms.
  • Suitable aliphatic groups include, but are not limited to, linear or branched, substituted or unsubstituted alkyl, alkenyl, or alkynyl groups. For example, groups include unbranched or branched, unsubstituted or suitably substituted C 2 - 6 alkenyl or C 2 - 6 alkynyl groups.
  • alkyl refers to a saturated linear or branched-chain monovalent hydrocarbon radical of one to twenty carbon atoms, wherein the alkyl radical may be optionally substituted independently with one or more substituents described below. Unless otherwise specified, alkyl groups contain 1 -20 carbon atoms. In some embodiments, alkyl groups contain 1 -10 carbon atoms. In other embodiments, alkyl groups contain 1 -8 carbon atoms. In still other embodiments, alkyl groups contain 1 -6 carbon atoms, and in yet other embodiments, alkyl groups contain 1-4 carbon atoms.
  • alkyl groups include methyl (Me, -CH 3 ), ethyl (Et, - CH 2 CH 3 ), 1 -propyl ( «-Pr, w-propyl, -CH 2 CH 2 CH 3 ), 2-propyl (/-Pr, -propyl, -CH(CH 3 ) 2 ), 1 -butyl (H-BU, H-butyl, -CH2CH2CH2CH3), 2-methyl-l-propyl (/-Bu, /-butyl, -CH 2 CH(CH 3 ) 2 ), 2-butyl (s- Bu, s-butyl, -CH(CH 3 )CH 2 CH 3 ), 2-methyl-2-propyl (/-Bu, /-butyl, -C(CH 3 ) 3 ), 1 -pentyl (w-pentyl, -CH 2 CH 2 CH 2 CH 3 ), 2-pentyl
  • alkylene refers to a saturated divalent hydrocarbon group derived from a straight or branched chain saturated hydrocarbon by the removal of two hydrogen atoms, and is exemplified by methylene (-CH 2 -), etliylidene (-CH(CH 3 )-), ethylene (-CH 2 CH 2 -), isopropylene (-CH(CH 3 )CH 2 -), and the like.
  • 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.
  • 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.
  • Some non-limiting examples include ethynyl (-C ⁇ CH), propynyl (propargyl, -CH 2 C ⁇ CH), -C ⁇ C-CH 3 , and the like.
  • alkoxy refers to an alkyl group, as previously defined, attached to the principal carbon atom through an oxygen atom. Unless otherwise specified, alkoxy groups contain 1-20 carbon atoms. In some embodiments, alkoxy groups contain 1-10 carbon atoms. In other embodiments, alkoxy groups contain 1-8 carbon atoms. In still other embodiments, alkoxy groups contain 1-6 carbon atoms, and in yet other embodiments, alkoxy groups contain 1-4 carbon atoms.
  • alkoxy groups include methoxy (MeO, -OCH 3 ), ethoxy (EtO, -OCH 2 CH 3 ), 1-propoxy (w-PrO, w-propoxy, -OCH 2 CH 2 CH 3 ), 2-propoxy (z ' -PrO, z ' -propoxy, -OCH(CH 3 ) 2 ), 1-butoxy ( «-BuO, «-butoxy, -OCH 2 CH 2 CH 2 CH 3 ), 2-methyl-l-propoxy (z-BuO, i- butoxy, -OCH 2 CH(CH 3 ) 2 ), 2-butoxy (s-BuO, s-butoxy, -OCH(CH 3 )CH 2 CH 3 ), 2-methyl-2- propoxy (z-BuO, z-butoxy, -OC(CH 3 ) 3 ), 1-pentoxy ( «-pentoxy, -OCH 2 CH 2 CH 2 CH 2 CH 3 ), 2-
  • haloalkyl or haloalkoxy refers to alkyl, or alkoxy, as the case may be, substituted with one or more halogen atoms.
  • Some non-limiting examples include -CF 3 , - CFHCH 3 , -OCF 3 , -CCI 3 , 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, bicyclic, or tricyclic ring system.
  • suitable cycloaliphatic groups include cycloalkyl, cycloalkenyl, and cycloalkynyl.
  • cycloaliphatic groups include cyclopropyl, cyclobutyl, cyclopentyl, 1-cyclopent-l- enyl, l-cyclopent-2-enyl, l-cyclopent-3-enyl, cyclohexyl, 1 -cyclohex-l-enyl, l-cyclohex-2-enyl, 1- cyclohex-3-enyl, cyclohexadienyl, and the like.
  • cycloalkyl refers to a monovalent or multivalent saturated ring having 3 to 12 carbon atoms as a monocyclic ring system.
  • heterocycle refers to a monocyclic, bicyclic, or tricyclic ring system in which one or more ring members are an independently selected heteroatom and that is completely saturated or that contains one or more units of unsaturation, but not aromatic having a single point of attachment to the rest of the molecule.
  • One or more ring atoms are optionally substituted independently with one or more substituents described herein.
  • the "heterocycle”, “heterocyclyl”, or “heterocyclic” group is 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 S or P is optionally substituted with one or more oxo to provide the group SO or S0 2 , PO or P0 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 or P is optionally substituted with one or more oxo to provide the group SO or SO 2 , PO or PO 2 ).
  • the heterocyclyl may be a carbon radical or heteroatom radical.
  • Some non-limiting examples of heterocyclic rings include pyrrolidinyl, tetrahydrofuranyl, dihydrofuranyl, tetrahydrothienyl, tetrahydropyranyl, dihydropyranyl, tetrahydrothiopyranyl, piperidino, morpholino, thiomorpholino, thioxanyl, piperazinyl, homo-piperazinyl, azetidinyl, oxetanyl, thietanyl, homopiperidinyl, oxepanyl, thiepanyl, oxazepinyl, diazepinyl, thiazepinyl, 2- pyrrolinyl, 3-pyrrolinyl, indolinyl, 2H-pyranyl, ⁇ H-pyranyl, dioxanyl, 1,3-dioxolan
  • the heterocycle groups herein are optionally substituted independently with one or more substituents described herein.
  • heteroatom refers to one or more of oxygen, sulfur, nitrogen, phosphorus, or silicon, including any oxidized forms of nitrogen, sulfur, or phosphorus; the quaternized forms of any basic nitrogen; or a substitutable nitrogen of a heterocyclic ring, for example N (as in 3,4- dihydro-2H-pyrrolyl), ⁇ (as in pyrrolidinyl) or NR (as in N- substituted pyrrolidinyl).
  • halogen refers to F, CI, Br or I.
  • refers to a single hydrogen atom. This radical may be attached, for example, to an oxygen atom to form a hydroxyl radical.
  • D denotes a single deuterium atom.
  • One of this radical may be attached, for example, to a methyl group to form a mono-deuterated methyl group (-CDH 2 ), two of deuterium atoms may be attached to a methyl group to form a di-deuterated methyl (-CD 2 H), and three of deuterium atoms may be attached to a methyl group to form a tri-deuterated methyl group (-CD 3 ).
  • ⁇ 3 refers to an azide moiety. This radical may be attached, for example, to a methyl group to form azidomethane (methyl azide, Me s); or attached to a phenyl group to form phenyl azide (PI1N 3 ).
  • aryl used alone or as part of a larger moiety as in “aralkyl”, “aralkoxy” or “aryloxyalkyl” refers to monocyclic, bicyclic, and tricyclic carbocyclic ring systems having a total of six to fourteen ring members, wherein at least one ring in the system is aromatic, wherein each ring in the system contains 3-7 ring members and that has a single point of attachment to the rest of the molecule.
  • aryl may be used interchangeably with the term “aryl ring”. Examples of aryl rings would include phenyl, naphthyl, and anthracene.
  • heteroaryl used alone or as part of a larger moiety as in “heteroaralkyl” or “heteroarylalkoxy” refers to monocyclic, bicyclic, and tricyclic ring systems having a total of five to fourteen ring members, wherein at least one ring in the system is aromatic, at least one ring in the system contains one or more heteroatoms, wherein each ring in the system contains 5- 7 ring members and that has a single point of attachment to the rest of the molecule.
  • heteroaryl may be used interchangeably with the term “heteroaryl ring” or the term “heteroaromatic”.
  • heteroaryl rings include the following monocycles: 2- furanyl, 3-furanyl, N-imidazolyl, 2-imidazolyl, 4-imidazolyl, 5-imidazolyl, 3-isoxazolyl, 4- isoxazolyl, 5-isoxazolyl, 2-oxazolyl, 4-oxazolyl, 5-oxazolyl, N-pyrrolyl, 2-pyrrolyl, 3- pyrrolyl, 2-pyridyl, 3-pyridyl, 4-pyridyl, 2-pyrimidinyl, 4-pyrimidinyl, 5-pyrimidinyl, pyridazinyl (e.g., 3- pyridazinyl), 2-thiazolyl, 4-thiazolyl, 5-thiazolyl, tetrazolyl (e.g., 5- tetrazolyl), triazolyl (e.g., 2- triazolyl and 5-triazolyl), 2-thiazolyl, 4-thiazolyl
  • carboxy or “carboxyl”, whether used alone or with other terms, such as “carboxyalkyl”, refers to -CO 2 H.
  • alkylamino embraces “N-alkylamino” and “N,N-dialkylamino” where amino groups are independently substituted with one alkyl radical and with two alkyl radicals, respectively. More preferred alkylamino radicals are “lower alkylamino” radicals having one or two alkyl radicals of one to six carbon atoms, attached to a nitrogen atom. Suitable alkylamino radicals may be mono or dialkylamino such as N-methylamino, N-ethylamino, N,N- dimethylamino, N,N-diethylamino, and the like.
  • arylamino refers to amino groups substituted with one or two aryl radicals, such as N-phenylamino.
  • the arylamino radical may be further substituted on the aryl ring portion of the radical.
  • aminoalkyl refers to linear or branched alkyl radicals having one to about ten carbon atoms any one of which may be substituted with one or more amino radicals. More preferred aminoalkyl radicals are "lower aminoalkyl” radicals having one to six carbon atoms and one or more amino radicals. Some non-limiting examples of such radicals include aminomethyl, aminoethyl, aminopropyl, aminobutyl and aminohexyl.
  • fused bicyclic refers to saturated bridged ring system which has a C-C bond shared between two five-membered rings (Structure a), two six-membered rings (Structure b) and one five-membered ring and one six- membered ring (Structure c), as depicted in Structures a-c.
  • Each cyclic ring in a fused bicyclyl group is a carbocyclic or a heterocyclic.
  • fused bicyclic ring system examples include hexahydrofuro[3,2- &]furan, hexahydrofuro[2,3-£]furan, octahydrocyclopenta[c]pyrrole, hexahydro-lH-pyrrolizine and octahydro- lH-pyrido [ 1 ,2-a]pyrazine.
  • spirocyclyl refers to a ring originating from a particular annular carbon of another ring.
  • a saturated bridged ring system (ring B and B') is termed as "fused bicyclic"
  • ring A and ring B share an atom between the two saturated ring system, which terms as a "spirocyclyl” or "spiro bicyclyl”.
  • Each cyclic ring in a spirocyclyl group is a carbocyclic or a heterocyclic.
  • a bond drawn from a substituent to the center of one ring within a ring system represents substitution of the substituent at any substitutable position on the rings to which it is attached.
  • Structure e represents possible substitution in any of the positions on the B ring shown in Structure f.
  • structures depicted herein are also meant to include all isomeric (e.g., enantiomeric, diastereomeric, and geometric (or conformational)) forms of the structure; for example, the R and S configurations for each asymmetric center, (Z) and (E) double bond isomers, and (Z) and (E) conformational isomers. Therefore, single stereochemical isomers as well as enantiomeric, diastereomeric, and geometric (or conformational) mixtures of the present compounds are within the scope of the invention.
  • 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.
  • prodrug refers to a compound that is transformed in vivo into a compound of formula (I). Such a transformation can be affected, for example, by hydrolysis in blood or enzymatic transformation of the prodrug form to the parent form in blood or tissue.
  • Prodrugs of the compounds of the invention may be, for example, esters. Esters that may be utilized as prodrugs in the present invention are phenyl esters, aliphatic C 1-24 esters, acyloxymethyl esters, carbonates, carbamates, and amino acid esters. For example, a compound of the invention that contains an OH group may be acylated at this position in its prodrug form.
  • prodrug forms include phosphates, such as, for example those phosphates resulting from the phosphonation of an OH group on the parent compound.
  • phosphates such as, for example those phosphates resulting from the phosphonation of an OH group on the parent compound.
  • a thorough discussion of prodrugs is provided in Higuchi et al., Pro-drugs as Novel Delivery Systems, Vol. 14 A.C.S. Symposium Series; Roche et al., ed., Bioreversible Carriers in Drug Design, American Pharmaceutical Association and Pergamon Press, 1987; Rautio et al., Prodrugs: Design and Clinical Applications, Nature Reviews Drug Discovery, 2008, 7, 255-270, and Hecker et al., Prodrugs of Phosphates and Phosphonates, J. Med. Chem., 2008, 51, 2328-2345, all of which areincorporated herein by reference.
  • 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 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 term "racemic mixture” or “racemate” refers to an equimolar mixture of two enantiomeric species, devoid of optical activity.
  • a "pharmaceutically acceptable salt” refers to organic or inorganic salts of a compound of the invention.
  • Pharmaceutically acceptable salts are well known in the art. For example, Berge et al., describe pharmaceutically acceptable salts in detail in J. Pharmacol Sci, 1977, 66, 1-19, which is incorporated herein by reference.
  • non-limiting examples of pharmaceutically acceptable, nontoxic salts include salts of an amino group formed with inorganic acids such as hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid and perchloric acid or with organic acids such as acetic acid, oxalic acid, maleic acid, tartaric acid, citric acid, succinic acid or malonic acid or by using other methods used in the art such as ion exchange.
  • inorganic acids such as hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid and perchloric acid
  • organic acids such as acetic acid, oxalic acid, maleic acid, tartaric acid, citric acid, succinic acid or malonic acid or by using other methods used in the art such as ion exchange.
  • salts include adipate, alginate, ascorbate, aspartate, benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphorate, camphorsulfonate, citrate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, formate, fumarate, glucoheptonate, glycerophosphate, gluconate, hemisulfate, heptanoate, hexanoate, hydroiodide, 2-hydroxy-ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, malate, maleate, malonate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oleate, oxalate, palmitate, pamoate, pectinate,
  • Salts derived from appropriate bases include alkali metal, alkaline earth metal, ammonium and N + (Ci_4 alkyl)4 salts.
  • This invention also envisions the quaternization of any basic nitrogen-containing groups of the compounds disclosed herein. Water or oil-soluble or dispersable products may be obtained by such quaternization.
  • Representative alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, and the like.
  • Further pharmaceutically acceptable salts include, when appropriate, nontoxic ammonium, quaternary ammonium, and amine cations formed using counterions such as halide, hydroxide, carboxylate, sulfate, phosphate, nitrate, C 1-8 sulfonate and aryl sulfonate.
  • 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 limited to, water, isopropanol, ethanol, methanol, DMSO, ethyl acetate, acetic acid and ethanolamine.
  • hydrate refers to the complex where the solvent molecule is water.
  • protecting group refers to a substituent that is commonly employed to block or protect a particular functionality while reacting with 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, i-butoxycarbonyl (BOC, Boc), benzyloxycarbonyl (CBZ, 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 S0 2 Ph, cyanoethyl, 2-(trimethylsilyl)ethyl, 2- (trimethylsilyl) ethoxy-methyl, 2-(p-toluenesulfonyl) ethyl, 2-(p-nitrophenylsulfenyl)-ethyl, 2- (diphenylphosphino)-ethyl, nitroethyl and the like.
  • the present invention provides pyridine compounds, salts, and pharmaceutical formulations thereof, which are potentially useful in the treatment of diseases, conditions and disorders modulated by receptor tyrosine kinases, especially ALK and c-Met receptor. More specifically, the present invention provides a compound of Formula (I):
  • R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , X and Z is as defined herein.
  • each R 1 , R 2 , R 3 , R 4 , R 5 and R 6 is independently H, D or F;
  • Z is Ci_ 6 alkyl, C3_ 6 cycloalkyl, -(Ci ⁇ alkylene)-(C 3 _ 6 cycloalkyl), C 3 - 6 heterocyclyl,
  • each of the Ci- 6 alkyl, C 3 _ 6 cycloalkyl, -(Ci_ 4 alkylene)-(C 3 - 6 cycloalkyl), C 3 _ 6 heterocyclyl, -(Ci ⁇ alkylene)-(C 3 _ 6 heterocyclyl), Cs- ⁇ fused bicyclyl, and -(Ci_ 4 alkylene)-(C5_ i 2 spirobicyclyl) is optionally substituted with 1, 2, 3 or 4 substituents independently selected from D, F, CI, Br, I, C ⁇ alkyl, Ci_ 6 haloalkyl, OR a , -(C M alkylene)-OR a , -(C M alkylene)-NR a R b , NR a R b -C( 0)NR
  • each R and R b is independently H, Ci- 6 aliphatic, C 3 _ 6 cycloalkyl, -(Ci_ 4 alkylene)-(C 3 _ 6 cycloalkyl), C3_6heterocyclyl, -(Ci_ 4 alkylene)-(C3_6heterocyclyl), C6-ioaryl, 5-10 membered heteroaryl or -(5-10membered heteroaryl), provided that when Z is Ci_ 6 alkyl, NR R b is not NH 2 or Me 2 , and when R and R b are bonded to the same nitrogen atom, R and R b , together with the nitrogen atom they are attached to, optionally form a 3-8 membered heterocyclyl, wherein each of the C 3 _ 6 cycloalkyl, -(Ci- 4 alkylene)-(C 3 _ 6 cycloalkyl), C 3 _ 6 heterocyclyl, -(Ci_
  • bicyclyl groups disclosed herein include
  • R 1 , R 2 , R 3 , R 4 , R 5 and R 6 is independently H or D.
  • X is phenyl optionally substituted with 1, 2, 3 or 4 substituents independently selected from D, F, CI, Br, and Ci_ 3 haloalkyl.
  • Z is Ci_ 6 alkyl, C 3 _ 6 cycloalkyl, C 3 _ 6 heterocyclyl, -(Ci_ 4 alkylene)- (C3_ 6 heterocyclyl), or -(Ci ⁇ alkylene)-(C5-i 2 spirobicyclyl), wherein the Ci- 6 alkyl, C 3 _ 6 cycloalkyl, C 3 - 6 heterocyclyl, -(Ci_ 4 alkylene)-(C 3 _ 6 heterocyclyl), bicyclyl), C5- i 2 spirobicyclyl and -(Ci ⁇ alkylene)-(C5_i 2 spirobicyclyl), each of which is optionally substituted with 1, 2, 3 or 4 substituents independently selected from D, F, CI, Br, I,
  • each of the cyclic structures in the bicyclyl groups is a carbocyclic ring or a heterocyclic ring.
  • each R and R b is independently H, C 3 _ 6 cycloalkyl or - (Ci_ 3 alkylene)-(C 3 - 6 cycloalkyl), provided that when Z is Ci_ 6 alkyl, NR R b is not NH 2 or NMe 2 , wherein each of the C 3 _ 6 cycloalkyl and -(Ci_ 3 alkylene)-(C 3 _ 6 cycloalkyl) is optionally substituted with 1, 2, 3 or 4 substituents independently selected from D, F, OH, NH 2 , and Ci- 3 alkylamino.
  • Z is selected from the following substructures:
  • n 0, 1, 2 or 3;
  • each W and W is independently O or NH;
  • each hydrogen in Z is optionally substituted with 1, 2, 3 or 4 substituents independently selected from D, F, CI, Br, I, d_ 3 alkyl, Ci_ 6 haloalkyl, OR a and NR a R b , provided that NR a R b is not NH 2 or NMe 2 in structures (Z25) to (Z32).
  • the present invention also comprises the use of a compound of the invention, or pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment either acutely or chronically of a hyperproliferative disease state and/or an angiogenesis mediated disease state, including those described previously.
  • the compounds of the present invention are useful in the manufacture of an anti-cancer medicament.
  • the compounds of the present invention are also useful in the manufacture of a medicament to attenuate or prevent disorders through inhibition of protein kinases.
  • the present invention comprises a
  • composition comprising a therapeutically effective amount of a compound of Formula (I) in association with at least one pharmaceutically acceptable carrier, adjuvant or diluent.
  • the present invention also comprises a method of treating hyperproliferating and angiogenesis related disorders in a subject having or susceptible to such disorder, the method comprising treating the subject with a therapeutically effective amount of a compound of Formula (I).
  • 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 the invention 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).
  • 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 -toluenesulfonic acid or ethanesulfonic acid, or the like.
  • an inorganic acid such as hydrochloric acid, hydrobromic
  • the invention features pharmaceutical compositions that include a compound of formula (I), a compound listed in table 1, and a pharmaceutically acceptable carrier, adjuvant, or vehicle.
  • the amount of compound in the compositions of the invention is such that is effective to detectably inhibit a protein kinase in a biological sample or in a patient.
  • a pharmaceutically acceptable derivative includes, but is not limited to, pharmaceutically acceptable prodrugs, salts, esters, salts of such esters, or any other adducts or derivatives which upon administration to a patient in need is capable of providing, directly or indirectly, a compound as otherwise described herein, or a metabolite or residue thereof.
  • compositions of the present invention additionally comprise a pharmaceutically acceptable carrier, adjuvant, or vehicle, which, as used herein, includes any and all solvents, diluents, or other liquid vehicle, dispersion or suspension aids, surface active agents, isotonic agents, thickening or emulsifying agents, preservatives, solid binders, lubricants and the like, as suited to the particular dosage form desired.
  • a pharmaceutically acceptable carrier, adjuvant, or vehicle which, as used herein, includes any and all solvents, diluents, or other liquid vehicle, dispersion or suspension aids, surface active agents, isotonic agents, thickening or emulsifying agents, preservatives, solid binders, lubricants and the like, as suited to the particular dosage form desired.
  • Some non-limiting examples of materials which can serve as pharmaceutically acceptable carriers include ion exchangers, alumina, aluminum stearate, lecithin, serum proteins, such as human serum albumin, buffer substances such as phosphates, glycine, sorbic acid or potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, polyacrylates, waxes, polyethylene-polyoxypropylene-block polymers, wool fat, sugars such as lactose, glucose and sucrose; starches such as corn starch and potato starch; cellulose and its derivatives such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; powdered tragacanth; malt; gelatin; talc
  • compositions of the present invention may be administered orally, parenterally, by inhalation spray, topically, rectally, nasally, buccally, vaginally or via an implanted reservoir.
  • parenteral includes subcutaneous, intravenous, intramuscular, intra-articular, intra- synovial, intrasternal, intrathecal, intraocular, intrahepatic, intralesional and intracranial injection or infusion techniques.
  • the compositions are administered orally, intraperitoneally or intravenously.
  • Sterile injectable forms of the compositions of this invention may be aqueous or oleaginous suspension. These suspensions may be formulated according to techniques known in the art using suitable dispersing or wetting agents and suspending agents.
  • the sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally acceptable diluent or solvent, for example as a solution in 1,3- butanediol.
  • a non-toxic parenterally acceptable diluent or solvent for example as a solution in 1,3- butanediol.
  • acceptable vehicles and solvents that may be employed are water, Ringer's solution and isotonic sodium chloride solution.
  • sterile, fixed oils are conventionally 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 and its glyceride derivatives are useful in the preparation of injectables, as are natural pharmaceutically-acceptable oils, such as olive oil or castor oil, especially in their polyoxyethylated versions.
  • These oil solutions or suspensions may also contain a long-chain alcohol diluent or dispersant, such as carboxymethyl cellulose or similar dispersing agents that are commonly used in the formulation of pharmaceutically acceptable dosage forms including emulsions and suspensions.
  • surfactants such as TWEEN ® , SPAN ® and other emulsifying agents or bioavailability enhancers which are commonly used in the manufacture of pharmaceutically acceptable solid, liquid, or other dosage forms may also be used for the purposes of formulation.
  • compositions of this invention may be orally administered in any orally acceptable dosage form including capsules, tablets, aqueous suspensions or solutions.
  • carriers commonly used include lactose and corn starch.
  • Lubricating agents, such as magnesium stearate, are also typically added.
  • useful diluents include lactose and dried cornstarch.
  • aqueous suspensions are required for oral use, the active ingredient is combined with emulsifying and suspending agents. If desired, certain sweetening, flavoring or coloring agents may also be added.
  • compositions of this invention may be administered in the form of suppositories for rectal administration.
  • suppositories for rectal administration.
  • suppositories can be prepared by mixing the agent with a suitable non-irritating excipient that is solid at room temperature but liquid at rectal temperature and therefore will melt in the rectum to release the drug.
  • suitable non-irritating excipient include cocoa butter, beeswax and polyethylene glycols.
  • compositions of this invention may also be administered topically, especially when the target of treatment includes areas or organs readily accessible by topical application, including diseases of the eye, the skin, or the low intestinal tract. Suitable topical formulations are readily prepared for each of these areas or organs.
  • Topical application for the lower intestinal tract can be effected in a rectal suppository formulation (see above) or in a suitable enema formulation. Topically-transdermal patches may also be used.
  • the pharmaceutically acceptable compositions may be formulated in a suitable ointment containing the active component suspended or dissolved in one or more carriers.
  • Some non-limiting examples of carriers for topical administration of the compounds of this invention include mineral oil, liquid petrolatum, white petrolatum, propylene glycol, polyoxy ethylene, polyoxypropylene compound, emulsifying wax and water.
  • the pharmaceutically acceptable compositions can be formulated in a suitable lotion or cream containing the active components suspended or dissolved in one or more pharmaceutically acceptable carriers.
  • suitable carriers include mineral oil, sorbitan monostearate, polysorbate 60, cetyl esters wax, cetearyl alcohol, 2- octyldodecanol, benzyl alcohol and water.
  • the pharmaceutically acceptable compositions may be formulated, e.g., as micronized suspensions in isotonic, pH adjusted sterile saline or other aqueous solution, or, preferably, as solutions in isotonic, pH adjusted sterile saline or other aqueous solution, either with or without a preservative such as benzylalkonium chloride.
  • the pharmaceutically acceptable compositions may be formulated in an ointment such as petrolatum.
  • the pharmaceutically acceptable compositions of this invention may also be administered by nasal aerosol or inhalation.
  • compositions are prepared according to techniques well-known in the art of pharmaceutical formulation and may be prepared as solutions in saline, employing benzyl alcohol or other suitable preservatives, absorption promoters to enhance bioavailability, fluorocarbons, and/or other conventional solubilizing or dispersing agents.
  • liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs.
  • the liquid dosage forms may contain inert diluents commonly used in the art such as, for example, water or other solvents, solubilizing agents and emulsifiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, dimethylformamide, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor, and sesame oils), glycerol, tetrahydrofurfuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof.
  • the oral compositions can also include adjuvants
  • Injectable preparations for example, sterile injectable aqueous or oleaginous suspensions may be formulated according to the known art using suitable dispersing or wetting agents and suspending agents.
  • the sterile injectable preparation may also be a sterile injectable solution, suspension or emulsion in a nontoxic parenterally acceptable diluent or solvent, for example, as a solution in 1, 3-butanediol.
  • acceptable vehicles and solvents that may be employed are water, Ringer's solution, U.S. P. and isotonic sodium chloride solution.
  • sterile, fixed oils are conventionally employed as a solvent or suspending medium.
  • any bland fixed oil can be employed including synthetic mono- or diglycerides.
  • fatty acids such as oleic acid are used in the preparation of injectables.
  • the injectable formulations can be sterilized, for example, by filtration through a bacterial-retaining filter, or by incorporating sterilizing agents in the form of sterile solid compositions which can be dissolved or dispersed in sterile water or other sterile injectable medium prior to use.
  • dissolving or suspending the compound in an oil vehicle accomplishes delayed absorption of a parenterally administered compound form.
  • Injectable depot forms are made by forming microencapsule matrices of the compound in biodegradable polymers such as polylactide-polyglycolide. Depending upon the ratio of compound to polymer and the nature of the particular polymer employed, the rate of compound release can be controlled. Examples of other biodegradable polymers include poly(orthoesters) and poly(anhydrides). Depot injectable formulations are also prepared by entrapping the compound in liposomes or microemulsions that are compatible with body tissues.
  • biodegradable polymers such as polylactide-polyglycolide.
  • Depot injectable formulations are also prepared by entrapping the compound in liposomes or microemulsions that are compatible with body tissues.
  • compositions for rectal or vaginal administration are preferably suppositories which can be prepared by mixing the compounds of this invention with suitable non- irritating excipients or carriers such as cocoa butter, polyethylene glycol or a suppository wax which are solid at ambient temperature but liquid at body temperature and therefore melt in the rectum or vaginal cavity and release the active compound.
  • suitable non- irritating excipients or carriers such as cocoa butter, polyethylene glycol or a suppository wax which are solid at ambient temperature but liquid at body temperature and therefore melt in the rectum or vaginal cavity and release the active compound.
  • Solid dosage forms for oral administration include capsules, tablets, pills, powders, and granules.
  • the active compound is mixed with at least one inert, pharmaceutically acceptable excipient or carrier such as sodium citrate or dicalcium phosphate and/or a) fillers or extenders such as starches, lactose, sucrose, glucose, mannitol, and silicic acid, b) binders such as, for example, carboxymethylcellulose, alginates, gelatin, polyvinylpyrrolidinone, sucrose, and acacia, c) humectants such as glycerol, d) disintegrating agents such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate, e) solution retarding agents such as paraffin, f) absorption accelerators such as quaternary ammonium compounds, g) wetting agents such as, for example, cetyl alcohol
  • Solid compositions of a similar type may also be employed as fillers in soft and hard- filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polyethylene glycols and the like.
  • the solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings and other coatings well known in the pharmaceutical formulating art. They may optionally contain opacifying agents and can also be of a composition that they release the active ingredient(s) only, or preferentially, in a certain part of the intestinal tract, optionally, in a delayed manner. Examples of embedding compositions that can be used include polymeric substances and waxes. Solid compositions of a similar type may also be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polythylene glycols and the like.
  • the active compounds can also be in micro-encapsulated form with one or more excipients as noted above.
  • the solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings, release controlling coatings and other coatings well known in the pharmaceutical formulating art.
  • the active compound may be admixed with at least one inert diluent such as sucrose, lactose or starch.
  • Such dosage forms may also comprise, as is normal practice, additional substances other than inert diluents, e.g., tableting lubricants and other tableting aids such a magnesium stearate and microcrystalline cellulose.
  • the dosage forms may also comprise buffering agents. They may optionally contain pacifying agents and can also be of a composition that they release the active ingredient(s) only, or preferentially, in a certain part of the intestinal tract, optionally, in a delayed manner.
  • buffering agents include polymeric substances and waxes.
  • Dosage forms for topical or transdermal administration of a compound of this invention include ointments, pastes, creams, lotions, gels, powders, solutions, sprays, inhalants or patches.
  • the active component is admixed under sterile conditions with a pharmaceutically acceptable carrier and any needed preservatives or buffers as may be required.
  • Ophthalmic formulation, eardrops, and eye drops are also contemplated as being within the scope of this invention.
  • the present invention contemplates the use of transdermal patches, which have the added advantage of providing controlled delivery of a compound to the body.
  • Such dosage forms can be made by dissolving or dispensing the compound in the proper medium.
  • Absorption enhancers can also be used to increase the flux of the compound across the skin. The rate can be controlled by either providing a rate controlling membrane or by dispersing the compound in a polymer matrix or gel.
  • the compounds of the invention are preferably formulated in dosage unit form for ease of administration and uniformity of dosage.
  • dosage unit form refers to a physically discrete unit of agent appropriate for the patient to be treated. It will be understood, however, that the total daily usage of the compounds and compositions of the present invention will be decided by the attending physician within the scope of sound medical judgment.
  • the specific effective dose level for any particular patient or organism will depend upon a variety of factors including the disorder being treated and the severity of the disorder; the activity of the specific compound employed; the specific composition employed; the age, body weight, general health, sex and diet of the patient; the time of administration, route of administration, and rate of excretion of the specific compound employed; the duration of the treatment; drugs used in combination or coincidental with the specific compound employed, and like factors well known in the medical arts.
  • compositions should be formulated so that a dosage of between 0.01 - 200 mg/kg body weight/day of the inhibitor can be administered to a patient receiving these compositions.
  • Compounds of this invention can be administered as the sole pharmaceutical agent or in combination with one or more other additional therapeutic (pharmaceutical) agents where the combination causes no unacceptable adverse effects. This may be of particular relevance for the treatment of hyper-proliferative diseases such as cancer.
  • the compound of this invention can be combined with known cytotoxic agents, signal transduction inhibitors, or with other anti-cancer agents, as well as with admixtures and combinations thereof.
  • additional therapeutic agents that are normally administered to treat a particular disease, or condition, are known as "appropriate for the disease, or condition, being treated”.
  • additional therapeutic agents is meant to include chemotherapeutic agents and other antiproliferative agents.
  • chemotherapeutic agents or other antiproliferative agents may be combined with the compounds of this invention to treat proliferative disease or cancer.
  • Some non-limiting examples of chemotherapeutic agents or other antiproliferative agents include HDAC inhibitors including SAHA, MS-275, MGO 103, and those described in WO 2006/010264, WO 03/024448, WO 2004/069823, US 2006/0058298, US 2005/0288282, WO 00/71703, WO 01/38322, WO 01/70675, WO 03/006652, WO 2004/035525, WO 2005/030705, WO 2005/092899, and demethylating agents including, but not limited to, 5-aza-dC, Vidaza and Decitabine and those described in US 6,268137, US 5,578,716, US 5,919,772, US 6,054,439, US 6,184,211, US 6,020,318, US 6,066,625, US 6,506,735, US 6,
  • chemotherapeutic agents or other anti -proliferative agents may be combined with the compounds of this invention to treat proliferative diseases and cancer.
  • chemotherapeutic agents include other therapies or anticancer agents that may be used in combination with the inventive anticancer agents of the present invention and include surgery, radiotherapy (in but a few examples, gamma radiation, neutron beam radiotherapy, electron beam radiotherapy, proton therapy, brachytherapy, and systemic radioactive isotopes, to name a few), endocrine therapy, taxanes (paclitaxel, taxotere), platinum derivatives (cisplatin, carboplatin, oxaliplatin), biologic response modifiers (interferons, interleukins), tumor necrosis factor (TNF, TRAIL receptor targeting agents, to name a few), hyperthermia and cryotherapy, agents to attenuate any adverse effects (e.g., antiemetics), and other approved
  • the compounds of the present invention can be combined, with cytotoxic anti-cancer agents.
  • cytotoxic anti-cancer agents examples include, by no way of limitation, asparaginase, bleomycin, carboplatin, carmustine, chlorambucil, cisplatin, colaspase, cyclophosphamide, cytarabine, dacarbazine, dactinomycin, daunorubicin, doxorubicin (adriamycine), epirubicin, etoposide, 5- fluorouracil, hexamethylmelamine, hydroxyurea, ifosfamide, irinotecan, leucovorin, lomustine, mechlorethamine, 6-mercaptopurine, mesna, methotrexate, mitomycin C, mitoxantrone, prednisolone, prednisone, proc
  • cytotoxic drugs suitable for use with the compounds of the invention include, but are not limited to, those compounds acknowledged to be used in the treatment of neoplastic diseases, such as those for example in Goodman and Gilman's The Pharmacological Basis of Therapeutics (Ninth Edition, 1996, McGraw-Hill).
  • agents include, by no way of limitation, aminoglutethimide, L-asparaginase, azathioprine, 5-azacytidine cladribine, busulfan, diethylstilbestrol, 2,2'-difluorodeoxycytidine, docetaxel, erythrohydroxynonyladenine, ethinyl estradiol, 5-fluorodeoxyuridine, 5-fluorodeoxyuridine monophosphate, fludarabine phosphate, fluoxymesterone, flutamide, hydroxyprogesterone caproate, idarubicin, interferon, medroxyprogesterone acetate, megestrol acetate, melphalan, mitotane, paclitaxel, pentostatin, N- phosphonoacetyl-L-aspartate (PALA), plicamycin, semustine, teniposide, testosterone propionate, thiotepa, trimethylmelamine,
  • cytotoxic anti-cancer agents suitable for use in combination with the compounds of the invention also include newly discovered cytotoxic principles such as oxaliplatin, gemcitabine, capecitabine, epothilone and its natural or synthetic derivatives, temozolomide (Quinn et al, J. Clin. Oncol, 2003, 21(4), 646-651), tositumomab (BEXXAR ® ), trabedectin (Vidal et al., Proceedings of the American Society for Clinical Oncology 2004, 23, abstract 3181), and the inhibitors of the kinesin spindle protein Eg5 (Wood, et al., Curr. Opin. Pharmacol, 2001, 1, 370-377).
  • cytotoxic principles such as oxaliplatin, gemcitabine, capecitabine, epothilone and its natural or synthetic derivatives, temozolomide (Quinn et al, J. Clin. Oncol, 2003, 21(4), 646-6
  • the compounds of the present invention can be combined with other signal transduction inhibitors.
  • examples of such agents include, by no way of limitation, antibody therapies such as trastuzumab (HERCEPTIN ® ), cetuximab (ERBITUX ® ), ipilimumab
  • YERVOY ® and pertuzumab.
  • therapies also include, by no way of limitation, small-molecule kinase inhibitors such as imatinib (GLEEVEC ® ), sunitinib (SUTENT ® ), sorafenib (NEXAVAR ® ), erlotinib (TARCEVA ® ), gefitinib (IRESSA ® ), dasatinib
  • VOTRIENT ® afatinib, alisertib, amuvatinib, axitinib, bosutinib, brivanib, canertinib, cabozantinib, cediranib, crenolanib, dabrafenib, dacomitinib, danusertib, dovitinib, foretinib, ganetespib, ibrutinib, iniparib, lenvatinib, linifanib, linsitinib, masitinib, momelotinib, motesanib, neratinib, niraparib, oprozomib, olaparib, pictilisib, ponatinib, quizartinib, regorafenib, rigosertib, rucaparib, saracatinib, saridegib, a
  • the compounds of the present invention can be combined with inhibitors of histone deacetylase.
  • inhibitors of histone deacetylase include, by no way of limitation, suberoylanilide hydroxamic acid (SAHA), LAQ-824 (Ottmann et al., Proceedings of the American Society for Clinical Oncology, 2004, 23, abstract 3024), LBH-589 (Beck et al., Proceedings of the American Society for Clinical Oncology, 2004, 23, abstract 3025), MS-275 (Ryan et al., Proceedings of the American Association of Cancer Research 2004, 45, abstract 2452), FR-901228 (Piekarz et al., Proceedings of the American Society for Clinical Oncology, 2004, 23, abstract 3028) and MGCDOl 03 (US 6,897,220).
  • SAHA suberoylanilide hydroxamic acid
  • LAQ-824 Ottmann et al., Proceedings of the American Society for Clinical Oncology, 2004, 23, abstract 3024
  • LBH-589 Beck et
  • the compounds of the present invention can be combined with other anti-cancer agents such as proteasome inhibitors, and m-TOR inhibitors. These include, by no way of limitation, bortezomib, and CCI-779 (Wu et al., Proceedings of the American Association of Cancer Research, 2004, 45, abstract 3849).
  • the compounds of the present invention can be combined with other anti-cancer agents such as topoisomerase inhibitors, including but not limited to camptothecin.
  • those additional agents may be administered separately from the compound- containing composition, as part of a multiple dosage regimen.
  • those agents may be part of a single dosage form, mixed together with the compound of this invention in a single composition. If administered as part of a multiple dosage regimen, the two active agents may be submitted simultaneously, sequentially or within a period of time from one another which would result in the desired activity of the agents.
  • the amount of both the compound and the additional therapeutic agent (in those compositions which comprise an additional therapeutic agent as described above) that may be combined with the carrier materials to produce a single dosage form will vary depending upon the host treated and the particular mode of administration. Normally, the amount of additional therapeutic agent present in the compositions of this invention will be no more than the amount that would normally be administered in a composition comprising that therapeutic agent as the only active agent. Preferably the amount of additional therapeutic agent in the presently disclosed compositions will range from about 50% to 100% of the amount normally present in a composition comprising that agent as the only therapeutically active agent. In those compositions which comprise an additional therapeutic agent, that additional therapeutic agent and the compound of this invention may act synergistically.
  • the invention features pharmaceutical compositions that include a compound of formula (I), or a compound listed in Table 1, and a pharmaceutically acceptable carrier, adjuvant, or vehicle.
  • the amount of compound in the compositions of the invention is such that is effective to detectably inhibit a protein kinase, such as ALK and c-Met inhibitory activity.
  • the compounds of the invention are useful in therapy as antineoplasia agents or to minimize deleterious effects of ALK and c-Met signaling.
  • Compounds of the present invention would be useful for, but not limited to, the prevention or treatment of proliferative diseases, condition, or disorder in a patient by administering to the patient a compound or a composition of the invention in an effective amount.
  • diseases, conditions, or disorders include cancer, particularly metastatic cancer, atherosclerosis and lung fibrosis.
  • neoplasm including cancer and metastasis, including, but not limited to: carcinoma such as cancer of the bladder, breast, colon, kidney, liver, lung (including small cell lung cancer), esophagus, gall-bladder, ovary, pancreas, stomach, cervix, thyroid, prostate, and skin (including squamous cell carcinoma); hematopoietic tumors of lymphoid lineage (including leukemia, acute lymphocytic leukemia, acute lymphoblastic leukemia, B-cell lymphoma, T-cell-lymphoma, Hodgkin's lymphoma, non-Hodgkin's lymphoma, hairy cell lymphoma and Burkett's lymphoma); hematopoietic tumors of myeloid lineage (including acute and chronic myelogenous leukemias, myelodysplastic syndrome and promyelocytic leukemia); tumors
  • tumors of the central and peripheral nervous system including astrocytoma, neuroblastoma, glioma and schwannomas); and other tumors (including melanoma, seminoma, teratocarcinoma, osteosarcoma, xeroderoma pigmentosum, keratoacanthoma, thyroid follicular cancer and Kaposi's sarcoma).
  • the compounds also would be useful for treatment of ophthalmological conditions such as corneal graft rejection, ocular neovascularization, retinal neovascularization including neovascularization following injury or infection, diabetic retinopathy, retrolental fibroplasia and neovascular glaucoma; retinal ischemia; vitreous hemorrhage; ulcerative diseases such as gastric ulcer; pathological, but non-malignant, conditions such as hemangiomas, including infantile hemaginomas, angiofibroma of the nasopharynx and avascular necrosis of bone; and disorders of the female reproductive system such as endometriosis.
  • the compounds are also useful for the treatment of edema, and conditions of vascular hyperpermeability.
  • the compounds of the present invention are also useful in the treatment of diabetic conditions such as diabetic retinopathy and microangiopathy.
  • the compounds of the present invention are also useful in the reduction of blood flow in a tumor in a subject.
  • the compounds of the present invention are also useful in the reduction of metastasis of a tumor in a subject.
  • these compounds are also useful for veterinary treatment of companion animals, exotic animals and farm animals, including mammals, rodents, and the like. More preferred animals include horses, dogs, and cats.
  • the compounds of the present invention include the pharmaceutically acceptable derivatives thereof.
  • the treatment method that includes administering a compound or composition of the invention can further include administering to the patient an additional therapeutic agent (combination therapy) selected from: a chemotherapeutic or anti-proliferative agent, or an antiinflammatory agent, wherein the additional therapeutic agent is appropriate for the disease being treated and the additional therapeutic agent is administered together with a compound or composition of the invention as a single dosage form or separately from the compound or composition as part of a multiple dosage form.
  • the additional therapeutic agent may be administered at the same time as a compound of the invention or at a different time. In the latter case, administration may be staggered by, for example, 6 hours, 12 hours, 1 day, 2 days, 3 days, 1 week, 2 weeks, 3 weeks, 1 month, or 2 months.
  • the invention also features a method of inhibiting the growth of a cell that expresses ALK or c-Met, that includes contacting the cell with a compound or composition of the invention, thereby causing inhibition of growth of the cell.
  • a cell whose growth can be inhibited include: a breast cancer cell, a colorectal cancer cell, a lung cancer cell, a papillary carcinoma cell, a prostate cancer cell, a lymphoma cell, a colon cancer cell, a pancreatic cancer cell, an ovarian cancer cell, a cervical cancer cell, a central nervous system cancer cell, an osteogenic sarcoma cell, a renal carcinoma cell, a hepatocellular carcinoma cell, a bladder cancer cell, a gastric carcinoma cell, a head and neck squamous carcinoma cell, a melanoma cell, or a leukemia cell.
  • the invention provides a method of inhibiting ALK or c-Met kinase activity in a biological sample that includes contacting the biological sample with a compound or composition of the invention.
  • biological sample means a sample outside a living organism and includes, without limitation, cell cultures or extracts thereof; biopsied material obtained from a mammal or extracts thereof; and blood, saliva, urine, feces, semen, tears, or other body fluids or extracts thereof.
  • Inhibition of kinase activity, particularly ALK or c-Met kinase activity, in a biological sample is useful for a variety of purposes known to one of skill in the art. Examples of such purposes include, but are not limited to, blood transfusion, organ-transplantation, biological specimen storage, and biological assays.
  • an "effective amount” or “effective dose” of the compound or pharmaceutically acceptable composition is that amount effective for treating or lessening the severity of one or more of the aforementioned disorders.
  • the compounds and compositions, according to the method of the present invention may be administered using any amount and any route of administration effective for treating or lessening the severity of the disorder or disease. The exact amount required will vary from subject to subject, depending on the species, age, and general condition of the subject, the severity of the infection, the particular agent, its mode of administration, and the like.
  • a compound or composition can also be administered with one or more other therapeutic agents, as discussed above.
  • the compounds of this invention or pharmaceutical compositions thereof may also be used for coating an implantable medical device, such as prostheses, artificial valves, vascular grafts, stents and catheters.
  • an implantable medical device such as prostheses, artificial valves, vascular grafts, stents and catheters.
  • Vascular stents for example, have been used to overcome restenosis (re-narrowing of the vessel wall after injury).
  • patients using stents or other implantable devices risk clot formation or platelet activation. These unwanted effects may be prevented or mitigated by pre-coating the device with a pharmaceutically acceptable composition comprising a compound of this invention.
  • Suitable coatings and the general preparation of coated implantable devices are described in U.S. Patent Nos. 6,099,562; 5,886,026; and 5,304,121, all of which are herein incorporated by reference in their entireties.
  • the coatings are typically biocompatible polymeric materials such as a hydrogel polymer, polymethyldisiloxane, polycaprolactone, polyethylene glycol, polylactic acid, ethylene vinyl acetate, and mixtures thereof.
  • the coatings may optionally be further covered by a suitable topcoat of fluorosilicone, polysaccarides, polyethylene glycol, phospholipids or combinations thereof to impart controlled release characteristics into the composition.
  • Implantable devices coated with a compound of this invention are another embodiment of the present invention.
  • the compounds may also be coated on implantable medical devices, such as beads, or co- formulated with a polymer or other molecule, to provide a "drug depot" thus permitting the drug to be released over a longer time period than administration of an aqueous solution of the drug.
  • the compounds in this invention may be prepared by methods described herein, wherein the substituents are as defined for formula (I), above, except where further noted.
  • the following non-limiting schemes and examples are presented to further exemplify the invention.
  • Persons skilled in the art will recognize that the chemical reactions described herein may be readily adapted to prepare a number of other compounds of the invention, and alternative methods for preparing the compounds of this invention are deemed to be within the scope of this invention.
  • 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.
  • other reactions disclosed herein or known in the art will be recognized as having applicability for preparing other compounds of the invention.
  • 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.
  • MS data were generally determined on an Agilent 1200 Series LCMS (Zorbax SB-C18, 2.1 x 30 mm, 4 micorn, 10 minutes run, 0.6 mL/min flow rate, 5 to 95% (0.1% formic acid in CH 3 CN) in (0.1% formic acid in H 2 0)) with UV detection at 210/254 nm and a low resonance electrospray mode (ESI) and Agilent 7890A-5975C Series GCMS (DB-624, 30 mmx0.32 mm, 1.8mm) with electron impact mode (EI) 0
  • the compounds disclosed herein may also be prepared by the method as described in Scheme 2.
  • an amine (5) is reacted with (Boc ⁇ O in the presence of a base such as Et 3 N to give an N-protected compound (8).
  • Sonogashira coupling of the N-protected compound (8) with an alkynol (9) in the presence of an appropriate Pd catalyst such as Pd(dppf)Cl2 affords an alcohol (10).
  • the alcohol (10) is then activated to a methanesulfonate derivative (11) with the aid of a base such as Et 3 N or i-Pr 2 NEt.
  • Boc-protecting groups can be removed under acidic conditions, for example, trifluoroacetic acid (TFA) in DCM, or HCl in ethyl acetate or ethyl ether to afford a desired kinase inhibitor (14) disclosed herein.
  • TFA trifluoroacetic acid
  • Step 4) 4-(6-amino-5-((R -l-(2,6-dichloro-3-fluorophenyl ethoxy pyridin-3-yl but-3-yn-2-ol
  • Step 1) l. l . l-trifluoro-2-methylpent-4-yn-2-ol
  • Step 2) 5-(6-amino-5-((R)-l-(2,6-dichloro-3-fluorophenyl)ethoxy)pyridin-3-yl)-l J -trifmoro-2- methylpent-4-yn-2-ol
  • Example 5 4- 6-3 ⁇ -5- /?)-1- 2,6-( ⁇ € ⁇ -3- ⁇ 6 ⁇ 1)6 ⁇ ) ⁇ ( ⁇ -3- ⁇ 1)-1,1,1- trifluoro-2-methylbut-3-vn-2-ol
  • Step 3) 4-(6-amino-5-((R)-l-(2.6-dichloro-3-fluorophenyl)ethoxy)pyridin-3-yl)-l. l .l-trifluoro-2- methylbut-3-yn-2-ol
  • Step 1) 1 -(ter?-butoxycarbonyl)-4-((trimethylsilyl)ethvnyl)piperidin-4-ol
  • Step 2) l-(ter/-butoxycarbonyl)-4-ethvnylpiperidin-4-ol
  • Step 1) 1 -(ferf-butoxycarbonyl)-3 -((trimethylsilyl)ethynyl)pyrrolidin-3 -ol
  • Step 2) l-(ter?-butoxycarbonyl)-3-ethynylpyrrolidin-3-ol
  • Step 3 3-((6-amino-5-((R -l-(2,6-dichloro-3-fluorophenyl ethoxy pyridin-3-yl ethvnyl -l-(ter?- butoxycarbonyl)pyrrolidin-3 -ol
  • Step 1) (R)-5-bromo-N.N-bis(te ⁇ butoxycarbonyl)-3-(l-(2.6-dichloro-3-fluorophenyl)ethoxy) pyridin-2-amine
  • Step 2) 4-(6-(bis(ter/-butoxycarbonyl)amino)-5-((R)-l-(2,6-dichloro-3-fluorophenyl)ethoxy) pyridin-3 -yl)but-3 -yn-2 - ol
  • Step 3 4-(6-(bis(te ⁇ butoxycarbonyl amino -5-( ' ( ' R -l-( ' 2,6-dichloro-3-fluorophenyl ethoxy pyridin-3-yl)but-3-yn-2-yl methanesulfonate
  • Step 4) N.N-bis(ferf-butoxycarbonyl)-3 -((R)- 1 -(2.6-dichloro-3 -fluorophenyl)ethoxy)-5-(3 - morpholinobut- 1 -yn- 1 -yl)pyridin-2-amine
  • Step 5 3 -((R)- 1 -(2 ,6-dichloro-3 -fluorophenyl)ethoxy)-5 -(3 -morpholinobut- 1 -yn- 1 -yl)pyridin-2- amine
  • Step 1) NN-bis(ter/-butoxycarbonyl)-3-((R)-l-(2,6-dichloro-3-fluorophenyl)ethoxy)-5-(3- (piperazin- 1 -yPbut- 1 -yn- 1 -yl)pyridin-2 -amine
  • Step 2) 3 -((R)- 1 -(2 ,6-dichloro-3 -fluorophenyl)ethoxy)-5-(3 -(piperazin- 1 -yl)but- 1 -yn- 1 -yl) pyridin-2-amine
  • Step 1) N.N-bis(ter?-butoxycarbonyl)-3-((R)-l-(2.6-dichloro-3-fluorophenyl)ethoxy)-5-(3-(4- methylpiperazin- 1 -vDbut- 1 -yn- 1 -yl)pyridin-2-amine
  • Step 2) 3 -((R)- 1 -(2,6-dichloro-3 -fluorophenyl)ethoxy)-5 -(3 -(4-methylpiperazin- 1 -yl)but- 1 -yn- 1 - vDpyridin-2-amine
  • Step 5 5-(4-(6-(bis(ter?-butoxycarbonyl)amino)-5-((R)-l-(2,6-dichloro-3-fluorophenyl)ethoxy) pyridin-3-yl)but-3-vn-2-yl)-N-(ter?-butoxycarbonyl)-5-azaspiro[2.4]heptan-7-amine
  • Step 1) 5-(4-(6-(bis(te ⁇ butoxycarbonyl amino -5-( ' ( ' R -l-( ' 2,6-dichloro-3-fluorophenyl ethoxy pyridin-3-yl)but-3-yn-2-yl)-5-azaspiro[2.4]heptan-7-ol
  • Step 2) 5-(4-(6-amino-5-((R)-l-(2.6-dichloro-3-fluorophenyl)ethoxy)pyridin-3-yl)but-3-yn-2-yl)- 5 -azaspiro ⁇ 2.4 "
  • Step 1) (R -3-(6-(bis(te ⁇ butoxycarbonyl amino -5-( ' l-( ' 2,6-dichloro-3-fluorophenyl ethoxy pyridin-3 -yl)prop-2-yn- 1 -ol
  • Step 2 (R)-3-(6-(bis(ter/-butoxycarbonyl)amino)-5-(l-(2,6-dichloro-3-fluorophenyl)ethoxy) pyridin-3 -yl)prop-2-yn- 1 -yl methanesulfonate
  • the LC/MS/MS system used in the analysis consists of an Agilent 1200 Series vacuum degasser, binary pump, well-plate autosampler, thermostatedcolumn compartment, the Agilent G6430 TripleQuadrupole Mass Spectrometer with an electrosprayionization (ESI) source. Quantitative analysis was carried out using MRM mode. The parameters for MRM transitions are in the Table A.
  • the mobile phase was 5mM ammonia acetate, 0.1% MeOH in water (A) : 5mM ammonia acetate, 0.1% MeOH in acetonitrile (B) (70:30, v/v).
  • the flow rate was 0.6 mL/min. Column was maintained at ambient temperature. 20 ⁇ ⁇ of the samples were injected.
  • Human or rat liver microsomes incubations were conducted in duplicate in polypropylene tubes.
  • the typical incubation mixtures consisted of human liver microsomes (0.5 mg protein/mL), compounds of interest (5 ⁇ ) and NADPH (1.0 mM) in a total volume of 200 ⁇ , potassium phosphate buffer (PBS, 100 mM, pH7.4).
  • PBS potassium phosphate buffer
  • Compounds were dissolved in DMSO and diluted with PBS such that the final concentration of DMSO was 0.05%.
  • the enzymatic reactions were commenced with the addition of protein after a 3 -min preincubation and incubated in a water bath open to the air at 37°C. Reactions were terminated at various time points (0, 5, 10, 15, 30, 60 min) by adding equal volume of ice-cold acetonitrile.
  • the samples were stored at -80°C until LC/MS/MS assays.
  • concentrations of compounds in the incubation mixtures of human liver microsomes were determined by a LC/MS/MS method.
  • the ranges of the linearity in the concentration range were determined for each tested compounds.
  • Dextromethorphan 70 ⁇ was selected as the positive control, and reactions were terminated at various time points (0, 5, 10, 15, 30, 60 min) after incubation at 37°C. Both positive and negative control samples were included in each assay to ensure the integrity of the microsomal incubation system.
  • the stability of some of the compounds disclosed herein in human (or rat) liver microsomes were also conducted in the following protocol.
  • the incubations were conducted in duplicate in polypropylene tubes.
  • the typical incubation mixtures consisted of human (or rat) liver microsomes (final concentration: 0.5 mg protein/mL), compounds (final concentration: 1.5 ⁇ ) in a total volume of 30 ⁇ ⁇ K-buffer (contain 1.0 mM EDTA, 100 mM, pH7.4).
  • Compounds were dissolved in DMSO and diluted with K-buffer such that the final concentration of DMSO was 0.2%.
  • ketanserin (1 ⁇ ) was selected as the positive control, and reactions were terminated at various time points (0, 15, 30, 60 min) after incubation at 37°C. Both positive control samples were included in each assay to ensure the integrity of the microsomal incubation system.
  • mice and dogs and monkeys are assessed in pharmacokinetic studies in mice, rats, dogs or monkeys.
  • the compounds are administered as a water solution, 2% HPMC + 1% TWEEN ® 80 in water solution, 5% DMSO + 5% solutol in saline, 4% MC suspension or capsule.
  • the animals are generally given at 1 or 2 mg/kg dose.
  • mice and rats are generally given 5 or 10 mg/kg dose
  • dogs and monkeys are generally given 10 mg/kg dose.
  • the blood samples (0.3 mL) are drawn at 0.25, 0.5, 1.0, 2.0, 3.0, 4.0, 6.0, 8.0, 12 and 24 h time points or 0.083, 0.25, 0.5, 1.0, 2.0, 4.0, 6.0, 8.0 and 24 h time points and centrifuged at 3,000 or 4000 rpm for 2 to 10 min.
  • the plasma solutions are collected and stored at -20 °C or -70 °C before analyzed by LC/MS/MS as described above.
  • the compounds disclosed herein exhibited optimized pharmacokinetic properties with desirable clearance (CI) and half-life (T 1/2 ) when the compounds were administered intravenously.
  • Kinase assays can be performed by measurement of incorporation of ⁇ - 33 ⁇ ATP into immobilized myelin basic protein (MBP).
  • MBP myelin basic protein
  • High binding white 384 well plates (Greiner) are coated with MBP (Sigma #M-1891) by incubation of 60 ⁇ /well of 20 ⁇ g/mL MBP in Tris- buffered saline (TBS; 50 mM Tris pH 8.0, 138 mM NaCl, 2.7 mM KC1) for 24 h at 4°C. Plates are washed 3 x with 100 ⁇ ⁇ TBS.
  • TBS Tris- buffered saline
  • Kinase reactions are carried out in a total volume of 34 ⁇ ⁇ in kinase buffer (5 mM Hepes pH 7.6, 15 mM NaCl, 0.01% bovine gamma globulin (Sigma #1- 5506), 10 mM MgCl 2 , 1 mM DTT, 0.02% TritonX-100).
  • Compound dilutions are performed in DMSO and added to assay wells to a final DMSO concentration of 1%. Each data point is measured in duplicate, and at least two duplicate assays are performed for each individual compound determination.
  • Enzyme is added to final concentrations of 10 nM or 20 nM, for example.
  • a mixture of unlabeled ATP and ⁇ - 33 ⁇ ATP is added to start the reaction (2 x 10 6 cpm of ⁇ - 33 ⁇ ATP per well (3000 Ci/mmole) and 10 ⁇ unlabeled ATP, typically.
  • the reactions are carried out for 1 h at rt with shaking. Plates are washed 7x with TBS, followed by the addition of 50 ⁇ scintillation fluid (Wallac). Plates are read using a Wallac Trilux counter. This is only one format of such assays; various other formats are possible, as known to one skilled in the art.
  • the above assay procedure can be used to determine the IC5 0 for inhibition and/or the inhibition constant, 3 ⁇ 4.
  • the IC5 0 is defined as the concentration of compound required to reduce the enzyme activity by 50% under the condition of the assay.
  • the IC5 0 value is estimated by preparing a 10 point curve using a 1 ⁇ 2 log dilution series (for example, a typical curve may be prepared using the following compound concentrations: 10 ⁇ , 3 ⁇ , 1 ⁇ , 0.3 ⁇ , 0.1 ⁇ , 0.03 ⁇ , 0.01 ⁇ , 0.003 ⁇ , 0.001 ⁇ and 0 ⁇ ).
  • ALK (h) is incubated with 8 mM MOPS pH 7.0, 0.2 mM EDTA, 250 ⁇ KKKSPGEYV IEFG, 10 mM MgAcetate and [ ⁇ - 33 ⁇ - ⁇ ] (specific activity aprrox. 500 pcm/pmol, concentration as required (10 ⁇ )).
  • the reaction is initiated by the addition of the MgATO mix. After incubation for 40 min at room temperature, the reaction is stopped by the addition of 3% phosphoric acid solution. 10 ⁇ ⁇ of the reaction is then spotted onto a P30 filter mat and washed three times for 5 min in 75 mM phosphoric acid and once in methanol prior to drying and scintillation counting.
  • Met (h) is incubated with 8 mM MOPS pH 7.0, 0.2 mM EDTA, 250 ⁇
  • mice When tumors reach a volume of 100-250 mm 3 , animals are randomly divided into vehicle control (for example, 2% HPMC + 1% TWEEN ® in water) and compound groups. Subsequent administration of compound by oral gavage (for example, 3-50 mpk/dose, dissolved in 2% HPMC + 1% Tween-80 in water) begins anywhere from day 0 to day 15 post tumor cell challenge and generally continues with once a day for the duration of the experiment.
  • vehicle control for example, 2% HPMC + 1% TWEEN ® in water
  • Subsequent administration of compound by oral gavage for example, 3-50 mpk/dose, dissolved in 2% HPMC + 1% Tween-80 in water
  • TGI Tumor Growth Inhibition
  • TGI tumor volume
  • the compouds described herein were also administrated orally (p.o.) once a day (QD), for 13-21 days in U87MG xenograft animal model. At doses of 60 mg/kg, the compouds produced statistically significant inhibition of growth of certain tumors grown subcutaneously in athymic nude mice.

Abstract

The present invention provides novel substituted alkynyl pyridine compounds, pharmaceutical acceptable salts and formulations thereof useful in modulating the protein tyrosine kinase activity, and in modulating cellular activities such as proliferation, differentiation, apoptosis, migration and invasion. The invention also provides pharmaceutically acceptable compositions comprising such compounds and methods of using the compositions in the treatment of hyperproliferative disorders in mammals, especially humans.

Description

SUBSTITUTED ALKYNYL PYRIDINE COMPOUNDS AND METHODS OF USE
CROSS-REFERENCE TO RELATED APPLICATION
[001] This application claims the benefit of U.S. Provisional Application Serial Number 61/650,513 filed May, 23, 2012, the contents of which is hereby incorporated by reference in its entirety.
FIELD OF THE INVENTION
[002] This invention relates to novel substituted alkynyl pyridine compounds, and salts thereof, which are useful in the treatment of hyperproliferative diseases, such as cancers, in mammals. In particular, the invention relates to compounds that inhibit the protein tyrosine kinase activity, resulting in the inhibition of inter- and/or intra-cellular signaling. This invention also relates to a method of using such compounds in the treatment of hyperproliferative diseases in mammals, especially humans, and to pharmaceutical compositions containing such compounds.
BACKGROUND OF THE INVENTION
[003] Protein kinases are key regulators of cell function that constitute one of the largest and most functionally diverse gene families. By adding phosphate groups to substrate proteins, they direct the activity, localization and overall function of many proteins, and serve to orchestrate the activity of many cellular processes. Kinases are particularly prominent in signal transduction and co-ordination of complex functions such as the cell cycle. Of the 518 human protein kinases, 478 belong to a single superfamily whose catalytic domains are related in sequence. These can be clustered into groups, families and sub-families, of increasing sequence similarity and biochemical function.
[004] A partial list of such kinases includes abl, AATK, ALK, Akt, Axl, bmx, bcr-abl, Blk, Brk, Btk, csk, c-kit, c-Met, c-src, c-fins, CDK1, CDK2, CDK3, CDK4, CDK5, CDK6, CDK7, CDK8, CDK9, CDK10, cRafl, CSF1R, CSK, DDR1, DDR2, EPHA, EPHB, EGFR, ErbB2, ErbB3, ErbB4, Erk, Fak, fes, FER, FGFR1, FGFR2, FGFR3, FGFR4, FGFR5, Fgr, flt-l, Fps, Frk, Fyn, GSG2, GSK, Hck, ILK, INSRR, IRAK4, ITK, IGF-1R, INS-R, Jak, KSR1, KDR, LMTK2, LMTK3, LTK, Lck, Lyn, MATK, MERTK, MLTK, MST1R, MUSK, NPR1, NTRK, MEK, MER, PLK4, PTK, p38, PDGFR, PIK, PKC, PYK2, RET, ROR1, ROR2, RYK, ros, Ron, SGK493, SRC, SRMS, STYK1, SYK, TEC, TEK, TEX 14, TNK1, TNK2, TNNI3K, TXK, TYK2, Tyro-3, tie, tie2, TRK, Yes and Zap70.
[005] Receptor tyrosine kinases (RTKs) are a diverse group of transmembrane proteins that act as receptors for cytokines, growth factors, hormones and other signaling molecules. Receptor tyrosine kinases (RTKs) are expressed in many cell types and play important roles in a wide variety of cellular processes, including growth, differentiation and angiogenesis. Activation of the kinase is effected by binding of a ligand to the extracellular domain, which induces dimerization of the receptors. Activated receptors auto-phosphorylate tyrosine residues outside the catalytic domain via cross-phosphorylation. This auto-phosphorylation stabilizes the active receptor conformation and creates phosphotyrosine docking sites for proteins that transduce signals within the cell.
[006] Receptor tyrosine kinases (RTKs) are hyper-activated (through receptor activating mutations, gene amplification, growth factor activation, etc.) in many human solid tumors and hematological malignancies. RTK's elevated activation contributes to tumourigenesis factors such as hyperplasia, survival, invasion, metastasis and angiogenesis. Inhibition of receptor tyrosine kinases proved to be effective strategies in cancer therapy (Sharma et al., "Receptor tyrosine kinase inhibitors as potent weapons in war against cancers", Curr. Pharm. Des., 2009, 15, 758).
[007] Anaplastic lymphoma kinase (ALK), a membrane associated tyrosine kinase receptor from the insulin receptor superfamily, has been implicated in oncogenesisin several human tumors. Indeed, ALK was initially identified in constitutively activated and oncogenic fusion forms (the most common being nucleophosmin (NPM)-ALK) in a non-Hodgkin's lymphoma (NHL) known as anaplastic large -cell lymphoma (ALCL) (Morris et al, "Fusion of a kinase gene, ALK, to a nucleolar protein gene, NPM, in non-Hodgkin's lymphoma", Science, 1994, 263, 1281).
[008] ALK fusions were also found in the human sarcomas called inflammatory myofibroblastic tumors (IMTs). Studies suggested that the ALK fusion, TPM4-ALK, may be involved in the genesis of a subset of esophageal squamous cell carcinomas. Moreover, studies have implicated various mutations of the ALK gene in both familial and sporadic cases of neuroblastoma. ALK mutations in neuroblastoma cells results in constitutive ALK phosphorylation and attenuation. Conversely, inhibition of ALK by sRNA and small molecule ALK inhibitors resulted in profound growth inhibition in those cell lines (Palmer et al., "Anaplastic lymphoma kinase: signalling in development and disease", Biochem. J., 2009, 420, 345).
[009] More recently, various isoforms of a fusion gene comprised of portions of the echinoderm microtubule-associated protein-like 4 (EML4) gene and the ALK gene were identified in NSCLC cells. The EML4-ALK fusion transcript was detected in approximately 3- 7% of NSCLC patients examined. Experimental evidences from in vitro and in vivo studies demonstrated oncogenic transforming activity of the EML4-ALK fusion proteins and reinforced the pivotal role of EML4-ALK in the pathogenesis of NSCLC in humans (Soda et al., "Identification of the transforming EML4-ALK fusion gene in non-small-cell lung cancer", Nature, 2007, 448, 561).
[010] Fusions of ALK have clear oncogenic potential as its aberrant tyrosine kinase activity enhances cell proliferation and survival and leads to cytoskeletal rearrangements and changes in cell shape. Oncogenic ALK transformation is mediated by interactions with downstream molecules that trigger substantial intracellular signaling cascades. Similarly to most normal and oncogenic tyrosine kinases, ALK fusions activate many different pathways that are strictly interconnected and overlapping. The most relevant and better characterized pathways are reported: the Ras-extracellular signal-regulated kinase (ERK) pathway, the Janus kinase 3 (JAK3)-STAT3 pathway and the phosphatidylinositol 3-kinase (PBK)-Akt pathway. These three pathways have many points of interaction to mediate the effects of ALK activity. Overall, the JAK3-STAT3 pathway and the PI3K-Akt pathway have been shown to be vital primarily for cell survival and phenotypic changes (Chiarle et al., "The anaplastic lymphoma kinase in the pathogenesis of cancer", Nat. Rev. Cancer, 2008, 8, 11 ; Barreca et al., "Anaplastic lymphoma kinase (ALK) in human cancer", J. Mol. Endocrinol., 2011, 47, Rl 1).
[011] The involvement of the full-length, normal ALK receptor in the genesis of additional malignancies including glioblastoma, neuroblastoma, breast cancer, and others has also been implicated. In a survey of a collection of human cancer cell lines, Dirks, et al. confirmed the expression of ALK transcripts in nervous system-derived lines, including retinoblastoma, and a large percentage of cell lines derived from solid cancers of ectodermal origin, including melanoma and breast carcinoma (Dirks et al., "Cancer's source in the peripheral nervous system" Nature Medicine, 2008, 14, 373).
[012] c-Met, also referred to as hepatocyte growth factor receptor (HGFR), is expressed predominantly in epithelial cells but has also been identified in endothelial cells, myoblasts, hematopoietic cells and motor neurons. The natural ligand for c-Met is hepatocyte growth factor (HGF), also known as scatter factor (SF). In both embryos and adults, activated c-Met promotes a morphogenetic program, known as invasive growth, which induces cell spreading, the disruption of intercellular contacts, and the migration of cells towards their surroundings (Peschard et al., "From Tpr-Met to Met, tumorigenesis and tubes" Oncogene, 2007, 26, 1276; Stellrecht et al., "Met Receptor Tyrosine Kinase as a Therapeutic Anticancer Target", Cancer Letter, 2009, 280, 1). [013] A wide variety of human malignancies exhibit sustained c-Met stimulation, overexpression, or mutation, including carcinomas of the breast, liver, lung, ovary, kidney, thyroid, colon, renal, glioblastomas, and prostate, etc. c-Met is also implicated in atherosclerosis and lung fibrosis. Invasive growth of certain cancer cells is drastically enhanced by tumor- stromal interactions involving the HGF/c-Met pathway. Thus, extensive evidence indicates that c-Met signaling is involved in the progression and spread of several cancers and an enhanced understanding of its role in disease has generated considerable interest in c-Met as major targets in cancer drug development (Migliore et al., "Molecular cancer therapy: can our expectation be MET", Eur. J. Cancer, 2008, 44, 641; Peruzzi et al., "Targeting the c-Met Signaling Pathway in Cancer", Clinical Cancer Research, 2006, 12, 3657). Agents targeting c-Met signaling pathway are now under clinical investigation (Eder et al., "Novel Therapeutic Inhibitors of the c-Met Signaling Pathway in Cancer", Clinical Cancer Research, 2009, 15, 2207; Paolo et al., "Drug development of MET inhibitors: targeting oncogene addiction and expedience", Nature Reviews Drug Discovery, 2008, 7, 504).
[014] Many ALK and/or c-Met inhibitors are now under clinical development for the treatment of various human cancers. Crizotinib is an ATP-competitive small molecule ALK inhibitor, which also displays activity against the c-Met receptor tyrosine kinase. The FDA recently approved crizotinib (Pfizer' s XALKORI®, originally known as PF-02341066) for treatment of patients with locally advanced or metastatic non-small cell lung cancer (NSCLC), in which tumor cells exhibit rearrangements in the anaplastic lymphoma kinase (ALK) gene. These rearrangements of the ALK gene (EML4-ALK) constitute driver mutations that are critical for the malignant phenotype of lung adenocarcinomas that have the mutations. Thus, the inhibition of mutated kinase ALK for the treatment of cancer is validated.
[015] Crizotinib is administered 250 mg twice daily. Following oral single-dose administration, crizotinib was absorbed with median time to achieve peak concentration from 4 to 6 hours. Following crizotinib 250 mg twice daily, steady state was reached within 15 days and remained stable, with a median accumulation ratio of 4.8 (XALKORI® FDA-Approved Patient Labeling, Pfizer Inc. February 2012).
[016] As seen with other targeted cancer drugs, patients with ALK-positive NSCLC eventually relapse on crizotinib. The development of acquired resistance is clearly the major hurdle preventing targeted therapies such as crizotinib from having an even more substantial impact on patients (Nature Reviews Drug Discovery, 2011, 10, 897).
[017] There is, therefore, still a need for effective therapies for use in proliferative disease, including treatments for primary cancers, metastatic disease, and for targeted therapies, including tyrosine kinase inhibitors, such as ALK and/or c-Met inhibitors, dual inhibitors, selective inhibitors, and for potent, orally bioavailable, and efficacious inhibitors, and for inhibitors that provide optimized dosing schedule, such as once daily oral administration.
[018] The present invention provides novel compounds believed to have clinical use for treatment of cancer through inhibiting ALK and/or c-Met. Preferred compounds of the present invention are also believed to provide an improvement in potency, pharmacokinetic properties, and/or toxicity profile over certain other ALK and/or c-Met inhibitor compounds found in the art.
SUMMARY OF THE INVENTION
[019] The present invention provides new compounds and methods for treating cell proliferative diseases. The compounds of the invention are inhibitors of protein tyrosine kinases. Preferably, the compounds of the invention are capable of inhibiting, for example, ALK (including ALK fusions such as EML4-ALK, NPM-ALK, etc.), and c-Met receptor (hepatocyte growth factor receptor) signaling. Accordingly, the invention provides new inhibitors of protein tyrosine kinase receptor signaling, for example, ALK receptor signaling and c-Met receptor signaling.
[020] Specifically, it has been found that compounds of this invention, and pharmaceutically acceptable compositions thereof, are effective as inhibitors of receptor tyrosine kinases such as ALK and c-Met. Accordingly, the invention provides compounds having the formula (I):
Figure imgf000006_0001
or a stereoisomer, a geometric isomer, a tautomer, an N-oxide, a solvate, a metabolite, a pharmaceutically acceptable salt or a prodrug thereof, wherein each R1, R2, R3, R4, R5, R6, X and Z is as defined herein.
[021] In certain embodiments, each R1, R2, R3, R4, R5 and R6 is independently H, D or F;
X is C6-ioaryl or 5-10 membered heteroaryl comprising 1, 2, 3 or 4 heteroatoms independently selected from O, S or Ν, wherein each of the C6-ioaryl and 5-10 membered heteroaryl is optionally substituted with 1, 2, 3 or 4 substituents independently selected from D, F, CI, Br, I, CN, N02, N3, ORa, SRa, NRaRb, -C(=0)NRaRb, Ci_6alkyl, Ci_6haloalkyl, C2_6alkenyl, C2_6alkynyl, -(CMalkylene)-CN, -(Ci_4alkylene)-ORa, -(CMalkylene)-NRaRb, C6-ioaryl and 5-10 membered heteroaryl;
Z is Ci_6alkyl, C3_6cycloalkyl, -(Ci^alkylene)-(C3_6cycloalkyl), C3-6heterocyclyl,
Figure imgf000007_0001
(C3_6heterocyclyl), Cs-^fused bicyclyl, -(Ci_4alkylene)-(C5_i2fused bicyclyl), Cs-^spirobicyclyl or
Figure imgf000007_0002
C3_6cycloalkyl, -(Ci_
4alkylene)-(C3-6cycloalkyl), C3_6heterocyclyl, -(Ci_4alkylene)-(C3_6heterocyclyl), Cs-^fused bicyclyl,
Figure imgf000007_0003
i2spirobicyclyl) is optionally substituted with 1, 2, 3 or 4 substituents independently selected from D, F, CI, Br, I, C^alkyl, Ci_6haloalkyl, ORa, -(CMalkylene)-ORa, -(Ci_4alkylene)-NRaRb, NRaRb, -C(=0)NRaRb and -OC(=0)NRaRb; and each of the cyclic structures in the bicyclyl groups is a carbocyclic ring or a heterocyclic ring; and
each R and Rb is independently H, Ci-6aliphatic, C3_6cycloalkyl, -(Ci_4alkylene)-(C3_6cycloalkyl), C3_6heterocyclyl, -(Ci_4alkylene)-(C3_6heterocyclyl), C6-ioaryl,
Figure imgf000007_0004
5-10 membered heteroaryl or
Figure imgf000007_0005
-(5-10membered heteroaryl), provided that when Z is Ci_ 6alkyl, NR Rb is not H2 or Me2, or when R and Rb are bonded to the same nitrogen atom, R and Rb, together with the nitrogen atom they are attached to, optionally form a 3-8 membered heterocyclyl, wherein each of the Ci_6aliphatic, C3_6cycloalkyl, -(Ci_4alkylene)-(C3_6cycloalkyl), C3_6heterocyclyl, -(Ci_4alkylene)-(C3_6heterocyclyl), C6-ioaryl,
Figure imgf000007_0006
5-10 membered heteroaryl ,
Figure imgf000007_0007
-(5-10membered heteroaryl) and 3-8 membered heterocyclyl is optionally substituted with 1, 2, 3 or 4 substituents independently selected from D, F, CI, CN, N3, OH, NH2, alkoxy and alkylamino.
[022] In certain embodiments, each R1, R2, R3, R4, R5 and R6 is independently H or D.
[023] In another embodiment, X is phenyl optionally substituted with 1, 2, 3 or 4 substituents independently selected from D, F, CI, Br,
Figure imgf000007_0008
[024] In certain embodiments, Z is
Figure imgf000007_0009
C3_6cycloalkyl, C3_6heterocyclyl, -(Ci_4alkylene)- (C3_6heterocyclyl),
Figure imgf000007_0010
or -(Ci^alkylene)-(C5-i2spirobicyclyl), wherein the Ci-6alkyl, C3_6cycloalkyl, C3_6heterocyclyl, -(Ci_ 4alkylene)-(C3_6heterocyclyl),
Figure imgf000007_0011
bicyclyl), C5- i2spirobicyclyl and -(Ci^alkylene)-(C5_i2spirobicyclyl), each of which is optionally substituted with 1, 2, 3 or 4 substituents independently selected from D, F, CI, Br, I,
Figure imgf000007_0012
OR , NR Rb; each of the cyclic structures in the bicyclyl groups is a carbocyclic ring or a heterocyclic ring.
[025] In another embodiment, each of R and Rb is independently H, Ci_3alkyl, C3_6cycloalkyl or -(Ci-3alkylene)-(C3_6cycloalkyl), provided that when Z is
Figure imgf000008_0001
NR Rb is not NH2 or Me2, wherein each of the
Figure imgf000008_0002
C3_6cycloalkyl or -(Ci_3alkylene)-(C3_6cycloalkyl) is optionally substituted with 1, 2, 3 or 4 substituents independently selected from D, F, OH, NH2,
Figure imgf000008_0003
[026] In another embodiment, Z is selected from the following substructures:
Figure imgf000008_0004
(Z34) , (Z35) , (Z36) , (Z37) , (Z38) , (Z39) , (Z40) , (Z41 ) , (Z42) , stereoisomer thereof, wherein n is 0, 1, 2 or 3;
each W and W is independently O or NH;
each hydrogen in Z is optionally substituted with 1, 2, 3 or 4 substituents independently selected from D, F, CI, Br, I, C^alkyl, d_6haloalkyl, ORa and NRaRb, provided that NRaRb is not NH2 or NMe2 in structures (Z25) to (Z32).
[027] Some non-limiting examples of the compound disclosed herein, and their
pharmaceutically acceptable salts and solvates thereof, are shown in the following:
Figure imgf000009_0001
Figure imgf000010_0001
[028] In another aspect, provided herein are pharmaceutical compositions comprising a compound disclosed herein, or a stereoisomer, geometric isomer, tautomer, solvate, metabolite, pharmaceutically acceptable salt or prodrug thereof, and an optional pharmaceutically acceptable carrier, excipient, diluent, adjuvant, vehicle or a combination thereof. In certain embodiments, the compound is an inhibitor of protein tyrosine kinase. In other embodiments, the compound is an inhibitor of ALK receptor signaling and HGF receptor signaling. [029] In some embodiments, the pharmaceutical composition disclosed herein further comprises an additional therapeutic agent. In other embodiments, the therapeutic agent is a chemotherapeutic agent, an anti-proliferative agent, an agent for treating atherosclerosis, an agent for treating lung fibrosis, and combinations thereof.
[030] In certain embodiments, the therapeutic agent is chlorambucil, melphalan, cyclophosphamide, ifosfamide, busulfan, carmustine, lomustine, streptozocin, cisplatin, carboplatin, oxaliplatin, dacarbazine, temozolomide, procarbazine, methotrexate, fluorouracil, cytarabine, gemcitabine, mercaptopurine, fludarabine, vinblastine, vincristine, vinorelbine, paclitaxel, docetaxel, topotecan, irinotecan, etoposide, trabectedin, dactinomycin, doxorubicin, epirubicin, daunorubicin, mitoxantrone, bleomycin, mitomycin, ixabepilone, tamoxifen, flutamide, gonadorelin analogues, megestrol, prednisone, dexamethasone, methylprednisolone, thalidomide, interferon alfa, leucovorin, sirolimus, temsirolimus, everolimus, afatinib, alisertib, amuvatinib, apatinib, axitinib, bortezomib, bosutinib, brivanib, cabozantinib, cediranib, crenolanib, crizotinib, dabrafenib, dacomitinib, danusertib, dasatinib, dovitinib, erlotinib, foretinib, ganetespib, gefitinib, ibrutinib, icotinib, imatinib, iniparib, lapatinib, lenvatinib, linifanib, linsitinib, masitinib, momelotinib, motesanib, neratinib, nilotinib, niraparib, oprozomib, olaparib, pazopanib, pictilisib, ponatinib, quizartinib, regorafenib, rigosertib, rucaparib, ruxolitinib, saracatinib, saridegib, sorafenib, sunitinib, tasocitinib, telatinib, tivantinib, tivozanib, tofacitinib, trametinib, vandetanib, veliparib, vemurafenib, vismodegib, volasertib, alemtuzumab, bevacizumab, brentuximab vedotin, catumaxomab, cetuximab, denosumab, gemtuzumab, ipilimumab, nimotuzumab, ofatumumab, panitumumab, ramucirumab, rituximab, tositumomab, trastuzumab, or a combination thereof.
[031] In another aspect, provided herein are methods for preventing, managing, treating or lessening the severity of a proliferative disorder in a patient infected with the proliferative disorder, which comprises administrating a pharmaceutically effective amount of a compound disclosed herein, or the pharmaceutical composition disclosed herein to the patient.
[032] In another aspect, provided herein is use of the compound disclosed herein, or the pharmaceutical composition disclosed herein in the manufacture of a medicament for preventing, managing, treating or lessening the severity of a proliferative disorder in a patient.
[033] In some embodiments, the proliferative disorder is metastatic cancer. In other embodiments, the proliferative disorder is colon cancer, gastric adenocarcinoma, bladder cancer, breast cancer, kidney cancer, liver cancer, lung cancer, skin cancer, thyroid cancer, cancer of the head and neck, prostate cancer, pancreatic cancer, cancer of the CNS, glioblastoma or a myeloproliferative disorder. In further embodiments, the proliferative disorder is atherosclerosis or lung fibrosis.
[034] In another aspect, provided herein is a method of inhibiting or modulating the activity of a protein kinase in a biological sample comprising contacting a biological sample with the compound disclosed herein, or the pharmaceutical composition disclosed herein.
[035] In some embodiments, the protein kinase is a receptor tyrosine kinase. In other embodiments, the receptor tyrosine kinase is ALK and/or c-Met.
[036] In another aspect, provided herein is a method of inhibiting protein tyrosine kinase, the method comprises contacting the kinase with the compound disclosed herein, or with the composition disclosed herein. In other embodiments, provided herein is a method of inhibiting ALK receptor signaling and/or HGF receptor signaling. The method comprises contacting the receptor with the compound disclosed herein, or with the pharmaceutical composition disclosed herein.
[037] In some embodiments, inhibition of receptor protein kinase activity, such as ALK and/or HGF receptor signaling, can be in a cell or a multicellular organism. If in a multicellular organism, the method disclosed herein may comprise administering to the organism the compound disclosed herein, or the pharmaceutical composition disclosed herein. In some embodiments, the organism is a mammal. In other embodiments, the organism is a human. In still other embodiments, the method further comprises contacting the kinase with an additional therapeutic agent.
[038] In another aspect, provided herein is a method of inhibiting proliferative activity of a cell, wherein the method comprises contacting the cell with an effective proliferative inhibiting amount of the compound disclosed herein or the pharmaceutical composition disclosed herein. In some embodiments, the method further comprises contacting the cell with an additional therapeutic agent.
[039] In another aspect, provided herein is a method of treating a cell proliferative disease in a patient, wherein the method comprises administering to the patient in need of such treatment an effective therapeutic amount of the compound disclosed herein or the pharmaceutical composition disclose herein. In other embodiments, the method further comprises administering an additional therapeutic agent.
[040] In another aspect, provided herein is a method of inhibiting tumor growth in a patient, the method comprises administering to the patient in need thereof an effective therapeutic amount of a compound disclosed herein or a composition thereof. In other embodiments, the method further comprises administering an additional therapeutic agent.
[041] In another aspect, provided herein include methods of preparing, methods of separating, and methods of purifying compounds of Formula (I).
[042] The foregoing merely summarizes certain aspects disclosed herein and is not intended to be limiting in nature. These aspects and other aspects and embodiments are described more fully below.
DETAILED DESCRIPTION OF THE INVENTION
DEFINITIONS AND GENERAL TERMINOLOGY
[043] Reference will now be made in detail to certain embodiments of the invention, examples of which are illustrated in the accompanying structures and formulas. 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 herein. 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.
[044] As used herein, the following definitions shall apply unless otherwise indicated. For purposes of this invention, the chemical elements are identified in accordance with the Periodic Table of the Elements, CAS version, and the Handbook of Chemistry and Physics, 75th Ed. 1994. Additionally, general principles of organic chemistry are described in Sorrell et al., "Organic Chemistry", University Science Books, Sausalito: 1999, and Smith et al., "March's Advanced Organic Chemistry", John Wiley & Sons, New York: 2007, all of which are incorporated herein by reference.
[045] As described herein, compounds of the invention may optionally be substituted with one or more substituents, such as those illustrated below, or as exemplified by particular classes, subclasses, and species of the invention. It will be appreciated that the phrase "optionally substituted" is used interchangeably with the phrase "substituted or unsubstituted". In general, the term "substituted" whether proceeded by the term "optionally" or not, refers to the replacement of one or more hydrogen radicals in a given structure with the radical of a specified substituent. Unless otherwise indicated, an optionally substituted group may have a substituent at each substitutable position of the group. When more than one position in a given structure can be substituted with more than one substituent selected from a specified group, the substituent may be either the same or different at each position. Examples of substituent include, but are not limited to, D, F, CI, Br, I, alkyl, haloalkyl, alkenyl, alkynyl, OH and NH2.
[046] The term "aliphatic" or "aliphatic group" refers to a straight-chain (i.e., unbranched) or branched, substituted or unsubstituted hydrocarbon chain that is completely saturated or that contains one or more units of unsaturation. Unless otherwise specified, aliphatic groups contain
1- 20 carbon atoms. In some embodiments, aliphatic groups contain 1- 10 carbon atoms. In other embodiments, aliphatic groups contain 1 -8 carbon atoms. In still other embodiments, aliphatic groups contain 1 -6 carbon atoms, and in yet other embodiments, aliphatic groups contain 1-4 carbon atoms. Suitable aliphatic groups include, but are not limited to, linear or branched, substituted or unsubstituted alkyl, alkenyl, or alkynyl groups. For example,
Figure imgf000014_0001
groups include unbranched or branched, unsubstituted or suitably substituted
Figure imgf000014_0002
C2-6alkenyl or C2-6alkynyl groups.
[047] The term "alkyl" or "alkyl group" refers to a saturated linear or branched-chain monovalent hydrocarbon radical of one to twenty carbon atoms, wherein the alkyl radical may be optionally substituted independently with one or more substituents described below. Unless otherwise specified, alkyl groups contain 1 -20 carbon atoms. In some embodiments, alkyl groups contain 1 -10 carbon atoms. In other embodiments, alkyl groups contain 1 -8 carbon atoms. In still other embodiments, alkyl groups contain 1 -6 carbon atoms, and in yet other embodiments, alkyl groups contain 1-4 carbon atoms.
[048] Some non-limiting examples of alkyl groups include methyl (Me, -CH3), ethyl (Et, - CH2CH3), 1 -propyl («-Pr, w-propyl, -CH2CH2CH3), 2-propyl (/-Pr, -propyl, -CH(CH3)2), 1 -butyl (H-BU, H-butyl, -CH2CH2CH2CH3), 2-methyl-l-propyl (/-Bu, /-butyl, -CH2CH(CH3)2), 2-butyl (s- Bu, s-butyl, -CH(CH3)CH2CH3), 2-methyl-2-propyl (/-Bu, /-butyl, -C(CH3)3), 1 -pentyl (w-pentyl, -CH2CH2CH2CH2CH3), 2-pentyl (-CH(CH3)CH2CH2CH3), 3-pentyl (-CH(CH2CH3)2), 2-methyl-
2- butyl (-C(CH3)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 (- CH2CH2CH2CH2CH2CH3), 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), 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. [049] The term "alkyl" and the prefix "alk-" are inclusive of both straight chain and branched saturated carbon chain.
[050] The term "alkylene" refers to a saturated divalent hydrocarbon group derived from a straight or branched chain saturated hydrocarbon by the removal of two hydrogen atoms, and is exemplified by methylene (-CH2-), etliylidene (-CH(CH3)-), ethylene (-CH2CH2-), isopropylene (-CH(CH3)CH2-), and the like.
[051] 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. Some non-limiting examples include ethylenyl or vinyl (-CH=CH2), allyl (-CH2CH=CH2), and the like.
[052] 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. Some non-limiting examples include ethynyl (-C≡CH), propynyl (propargyl, -CH2C≡CH), -C≡C-CH3, and the like.
[053] The term "alkoxy" refers to an alkyl group, as previously defined, attached to the principal carbon atom through an oxygen atom. Unless otherwise specified, alkoxy groups contain 1-20 carbon atoms. In some embodiments, alkoxy groups contain 1-10 carbon atoms. In other embodiments, alkoxy groups contain 1-8 carbon atoms. In still other embodiments, alkoxy groups contain 1-6 carbon atoms, and in yet other embodiments, alkoxy groups contain 1-4 carbon atoms.
[054] Some non-limiting examples of alkoxy groups include methoxy (MeO, -OCH3), ethoxy (EtO, -OCH2CH3), 1-propoxy (w-PrO, w-propoxy, -OCH2CH2CH3), 2-propoxy (z'-PrO, z'-propoxy, -OCH(CH3)2), 1-butoxy («-BuO, «-butoxy, -OCH2CH2CH2CH3), 2-methyl-l-propoxy (z-BuO, i- butoxy, -OCH2CH(CH3)2), 2-butoxy (s-BuO, s-butoxy, -OCH(CH3)CH2CH3), 2-methyl-2- propoxy (z-BuO, z-butoxy, -OC(CH3)3), 1-pentoxy («-pentoxy, -OCH2CH2CH2CH2CH3), 2- pentoxy (-OCH(CH3)CH2CH2CH3), 3-pentoxy (-OCH(CH2CH3)2), 2-methyl-2-butoxy (- OC(CH3)2CH2CH3), 3-methyl-2-butoxy (-OCH(CH3)CH(CH3)2), 3-methyl-l-butoxy (- OCH2CH2CH(CH3)2), 2-methyl-l-butoxy (-OCH2CH(CH3)CH2CH3), and the like.
[055] The terms "haloalkyl" or "haloalkoxy" refers to alkyl, or alkoxy, as the case may be, substituted with one or more halogen atoms. Some non-limiting examples include -CF3, - CFHCH3, -OCF3, -CCI3, and the like.
[056] The term "carbocycle", "carbocyclyl", "carbocyclic ring" or "cycloaliphatic" refers to a monovalent or multivalent non-aromatic, saturated or partially unsaturated ring having 3 to 12 carbon atoms as a monocyclic, bicyclic, or tricyclic ring system. Some non-limiting examples of suitable cycloaliphatic groups include cycloalkyl, cycloalkenyl, and cycloalkynyl. Further examples of cycloaliphatic groups include cyclopropyl, cyclobutyl, cyclopentyl, 1-cyclopent-l- enyl, l-cyclopent-2-enyl, l-cyclopent-3-enyl, cyclohexyl, 1 -cyclohex-l-enyl, l-cyclohex-2-enyl, 1- cyclohex-3-enyl, cyclohexadienyl, and the like. The term "cycloalkyl" refers to a monovalent or multivalent saturated ring having 3 to 12 carbon atoms as a monocyclic ring system.
[057] The term "heterocycle", "heterocyclyl" or "heterocyclic" as used interchangeably herein refers to a monocyclic, bicyclic, or tricyclic ring system in which one or more ring members are an independently selected heteroatom and that is completely saturated or that contains one or more units of unsaturation, but not aromatic having a single point of attachment to the rest of the molecule. One or more ring atoms are optionally substituted independently with one or more substituents described herein. In some embodiments, the "heterocycle", "heterocyclyl", or "heterocyclic" group is 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 S or P is optionally substituted with one or more oxo to provide the group SO or S02, PO or P02 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 or P is optionally substituted with one or more oxo to provide the group SO or SO2, PO or PO2).
[058] The heterocyclyl may be a carbon radical or heteroatom radical. Some non-limiting examples of heterocyclic rings include pyrrolidinyl, tetrahydrofuranyl, dihydrofuranyl, tetrahydrothienyl, tetrahydropyranyl, dihydropyranyl, tetrahydrothiopyranyl, piperidino, morpholino, thiomorpholino, thioxanyl, piperazinyl, homo-piperazinyl, azetidinyl, oxetanyl, thietanyl, homopiperidinyl, oxepanyl, thiepanyl, oxazepinyl, diazepinyl, thiazepinyl, 2- pyrrolinyl, 3-pyrrolinyl, indolinyl, 2H-pyranyl, ^H-pyranyl, dioxanyl, 1,3-dioxolanyl, pyrazolinyl, dithianyl, dithiolanyl, dihydropyranyl, dihydrothienyl, dihydrofuranyl, pyrazolidinylimidazolinyl, imidazolidinyl and 1,2,3,4-tetrahydro iso-quinolinyl. Examples of a heterocyclic group wherein 2 ring carbon atoms are substituted with oxo (=0) moieties are pyrimidindionyl and 1, 1 -dioxo-thiomorpholinyl. The heterocycle groups herein are optionally substituted independently with one or more substituents described herein.
[059] The term "heteroatom" refers to one or more of oxygen, sulfur, nitrogen, phosphorus, or silicon, including any oxidized forms of nitrogen, sulfur, or phosphorus; the quaternized forms of any basic nitrogen; or a substitutable nitrogen of a heterocyclic ring, for example N (as in 3,4- dihydro-2H-pyrrolyl), ΝΗ (as in pyrrolidinyl) or NR (as in N- substituted pyrrolidinyl).
[060] The term "halogen" refers to F, CI, Br or I.
[061] The term "Η" refers to a single hydrogen atom. This radical may be attached, for example, to an oxygen atom to form a hydroxyl radical.
[062] The term "D" or "2H" denotes a single deuterium atom. One of this radical may be attached, for example, to a methyl group to form a mono-deuterated methyl group (-CDH2), two of deuterium atoms may be attached to a methyl group to form a di-deuterated methyl (-CD2H), and three of deuterium atoms may be attached to a methyl group to form a tri-deuterated methyl group (-CD3).
[063] The term "Ν3" refers to an azide moiety. This radical may be attached, for example, to a methyl group to form azidomethane (methyl azide, Me s); or attached to a phenyl group to form phenyl azide (PI1N3).
[064] The term "aryl" used alone or as part of a larger moiety as in "aralkyl", "aralkoxy" or "aryloxyalkyl" refers to monocyclic, bicyclic, and tricyclic carbocyclic ring systems having a total of six to fourteen ring members, wherein at least one ring in the system is aromatic, wherein each ring in the system contains 3-7 ring members and that has a single point of attachment to the rest of the molecule. The term "aryl" may be used interchangeably with the term "aryl ring". Examples of aryl rings would include phenyl, naphthyl, and anthracene.
[065] The term "heteroaryl" used alone or as part of a larger moiety as in "heteroaralkyl" or "heteroarylalkoxy" refers to monocyclic, bicyclic, and tricyclic ring systems having a total of five to fourteen ring members, wherein at least one ring in the system is aromatic, at least one ring in the system contains one or more heteroatoms, wherein each ring in the system contains 5- 7 ring members and that has a single point of attachment to the rest of the molecule. The term "heteroaryl" may be used interchangeably with the term "heteroaryl ring" or the term "heteroaromatic".
[066] Some non-limiting examples of heteroaryl rings include the following monocycles: 2- furanyl, 3-furanyl, N-imidazolyl, 2-imidazolyl, 4-imidazolyl, 5-imidazolyl, 3-isoxazolyl, 4- isoxazolyl, 5-isoxazolyl, 2-oxazolyl, 4-oxazolyl, 5-oxazolyl, N-pyrrolyl, 2-pyrrolyl, 3- pyrrolyl, 2-pyridyl, 3-pyridyl, 4-pyridyl, 2-pyrimidinyl, 4-pyrimidinyl, 5-pyrimidinyl, pyridazinyl (e.g., 3- pyridazinyl), 2-thiazolyl, 4-thiazolyl, 5-thiazolyl, tetrazolyl (e.g., 5- tetrazolyl), triazolyl (e.g., 2- triazolyl and 5-triazolyl), 2-thienyl, 3-thienyl, pyrazolyl (e.g., 2-pyrazolyl), isothiazolyl, 1,2,3- oxadiazolyl, 1,2,5-oxadiazolyl, 1,2,4-oxadiazolyl, 1,2,3-triazolyl, 1,2,3-thiadiazolyl, 1,3,4- thiadiazolyl, 1,2,5-thiadiazolyl, pyrazinyl, 1,3,5-triazinyl, and the following bicycles: benzimidazolyl, benzofuryl, benzothiophenyl, indolyl (e.g., 2-indolyl), purinyl, quinolinyl (e.g., 2-quinolinyl, 3-quinolinyl, 4-quinolinyl), and isoquinolinyl (e.g., 1-isoquinolinyl, 3-isoquinolinyl or 4-isoquinolinyl).
[067] The term "carboxy" or "carboxyl", whether used alone or with other terms, such as "carboxyalkyl", refers to -CO2H. The term "carbonyl", whether used alone or with other terms, such as "aminocarbonyl", refers to -(C=0)-.
[068] The term "alkylamino" embraces "N-alkylamino" and "N,N-dialkylamino" where amino groups are independently substituted with one alkyl radical and with two alkyl radicals, respectively. More preferred alkylamino radicals are "lower alkylamino" radicals having one or two alkyl radicals of one to six carbon atoms, attached to a nitrogen atom. Suitable alkylamino radicals may be mono or dialkylamino such as N-methylamino, N-ethylamino, N,N- dimethylamino, N,N-diethylamino, and the like.
[069] The term "arylamino" refers to amino groups substituted with one or two aryl radicals, such as N-phenylamino. The arylamino radical may be further substituted on the aryl ring portion of the radical.
[070] The term "aminoalkyl" refers to linear or branched alkyl radicals having one to about ten carbon atoms any one of which may be substituted with one or more amino radicals. More preferred aminoalkyl radicals are "lower aminoalkyl" radicals having one to six carbon atoms and one or more amino radicals. Some non-limiting examples of such radicals include aminomethyl, aminoethyl, aminopropyl, aminobutyl and aminohexyl.
[071] The term "unsaturated" as used herein, refers to that a moiety has one or more units of unsaturation.
[072] The term "comprising" is meant to be open ended, including the indicated component but not excluding other elements.
[073] The terms "fused bicyclic", "fused cyclic", "fused bicyclyl" or "fused cyclyl" refer to saturated bridged ring system which has a C-C bond shared between two five-membered rings (Structure a), two six-membered rings (Structure b) and one five-membered ring and one six- membered ring (Structure c), as depicted in Structures a-c. Each cyclic ring in a fused bicyclyl group is a carbocyclic or a heterocyclic.
Figure imgf000019_0001
Structure a Structure b Structure c
[074] Some non-limiting examples of fused bicyclic ring system include hexahydrofuro[3,2- &]furan, hexahydrofuro[2,3-£]furan, octahydrocyclopenta[c]pyrrole, hexahydro-lH-pyrrolizine and octahydro- lH-pyrido [ 1 ,2-a]pyrazine.
[075] The term "spirocyclyl", "spirocyclic", "spiro bicyclyl" or "spiro bicyclic" refers to a ring originating from a particular annular carbon of another ring. For example, as depicted in Structure d, a saturated bridged ring system (ring B and B') is termed as "fused bicyclic", whereas ring A and ring B share an atom between the two saturated ring system, which terms as a "spirocyclyl" or "spiro bicyclyl". Each cyclic ring in a spirocyclyl group is a carbocyclic or a heterocyclic.
Figure imgf000019_0002
Structure d
[076] As described herein, a bond drawn from a substituent to the center of one ring within a ring system (as shown below) represents substitution of the substituent at any substitutable position on the rings to which it is attached. For example, Structure e represents possible substitution in any of the positions on the B ring shown in Structure f.
Figure imgf000019_0003
Structure e Structure f
[077] Unless otherwise stated, structures depicted herein are also meant to include all isomeric (e.g., enantiomeric, diastereomeric, and geometric (or conformational)) forms of the structure; for example, the R and S configurations for each asymmetric center, (Z) and (E) double bond isomers, and (Z) and (E) conformational isomers. Therefore, single stereochemical isomers as well as enantiomeric, diastereomeric, and geometric (or conformational) mixtures of the present compounds are within the scope of the invention.
[078] 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.
[079] Unless otherwise stated, all tautomeric forms of the compounds of the invention are within the scope of the invention. Additionally, unless otherwise stated, structures depicted herein are also meant to include compounds that differ only in the presence of one or more isotopically enriched atoms.
[080] The term "prodrug" refers to a compound that is transformed in vivo into a compound of formula (I). Such a transformation can be affected, for example, by hydrolysis in blood or enzymatic transformation of the prodrug form to the parent form in blood or tissue. Prodrugs of the compounds of the invention may be, for example, esters. Esters that may be utilized as prodrugs in the present invention are phenyl esters, aliphatic C1-24 esters, acyloxymethyl esters, carbonates, carbamates, and amino acid esters. For example, a compound of the invention that contains an OH group may be acylated at this position in its prodrug form. Other prodrug forms include phosphates, such as, for example those phosphates resulting from the phosphonation of an OH group on the parent compound. A thorough discussion of prodrugs is provided in Higuchi et al., Pro-drugs as Novel Delivery Systems, Vol. 14 A.C.S. Symposium Series; Roche et al., ed., Bioreversible Carriers in Drug Design, American Pharmaceutical Association and Pergamon Press, 1987; Rautio et al., Prodrugs: Design and Clinical Applications, Nature Reviews Drug Discovery, 2008, 7, 255-270, and Hecker et al., Prodrugs of Phosphates and Phosphonates, J. Med. Chem., 2008, 51, 2328-2345, all of which areincorporated herein by reference.
[081] 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.
[082] Stereochemical definitions and conventions used herein generally follow Parker et al., ed., McGraw-Hill Dictionary of Chemical Terms (1984) McGraw-Hill Book Company, New York and Eliel et al., "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 L or (+) and (-) are employed to designate the sign of rotation of plane-polarized light by the compound, with (-) or L 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 term "racemic mixture" or "racemate" refers to an equimolar mixture of two enantiomeric species, devoid of optical activity.
[083] A "pharmaceutically acceptable salt" refers to organic or inorganic salts of a compound of the invention. Pharmaceutically acceptable salts are well known in the art. For example, Berge et al., describe pharmaceutically acceptable salts in detail in J. Pharmacol Sci, 1977, 66, 1-19, which is incorporated herein by reference. Some non-limiting examples of pharmaceutically acceptable, nontoxic salts include salts of an amino group formed with inorganic acids such as hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid and perchloric acid or with organic acids such as acetic acid, oxalic acid, maleic acid, tartaric acid, citric acid, succinic acid or malonic acid or by using other methods used in the art such as ion exchange. Other pharmaceutically acceptable salts include adipate, alginate, ascorbate, aspartate, benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphorate, camphorsulfonate, citrate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, formate, fumarate, glucoheptonate, glycerophosphate, gluconate, hemisulfate, heptanoate, hexanoate, hydroiodide, 2-hydroxy-ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, malate, maleate, malonate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oleate, oxalate, palmitate, pamoate, pectinate, persulfate, 3-phenylpropionate, phosphate, picrate, pivalate, propionate, stearate, succinate, sulfate, tartrate, thiocyanate, p-toluenesulfonate, undecanoate, valerate salts, and the like. Salts derived from appropriate bases include alkali metal, alkaline earth metal, ammonium and N+(Ci_4 alkyl)4 salts. This invention also envisions the quaternization of any basic nitrogen-containing groups of the compounds disclosed herein. Water or oil-soluble or dispersable products may be obtained by such quaternization. Representative alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, and the like. Further pharmaceutically acceptable salts include, when appropriate, nontoxic ammonium, quaternary ammonium, and amine cations formed using counterions such as halide, hydroxide, carboxylate, sulfate, phosphate, nitrate, C1-8 sulfonate and aryl sulfonate.
[084] 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 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.
[085] The term "protecting group" or "PG" refers to a substituent that is commonly employed to block or protect a particular functionality while reacting with 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, i-butoxycarbonyl (BOC, Boc), benzyloxycarbonyl (CBZ, 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 -CH2CH2S02Ph, cyanoethyl, 2-(trimethylsilyl)ethyl, 2- (trimethylsilyl) ethoxy-methyl, 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 Greene et al., Protective Groups in Organic Synthesis, John Wiley & Sons, New York, 1991 and Kocienski et al. Protecting Groups, Thieme, Stuttgart, 2005.
DESCRIPTION OF COMPOUNDS OF THE INVENTION
[086] The present invention provides pyridine compounds, salts, and pharmaceutical formulations thereof, which are potentially useful in the treatment of diseases, conditions and disorders modulated by receptor tyrosine kinases, especially ALK and c-Met receptor. More specifically, the present invention provides a compound of Formula (I):
Figure imgf000022_0001
or a stereoisomer, a geometric isomer, a tautomer, an N-oxide, a solvate, a metabolite, a pharmaceutically acceptable salt or a prodrug thereof, wherein each of R1, R2, R3, R4, R5, R6, X and Z is as defined herein.
[087] In certain embodiments, each R1, R2, R3, R4, R5 and R6 is independently H, D or F;
X is C6-ioaryl or 5-10 membered heteroaryl comprising 1, 2, 3 or 4 heteroatoms independently selected from O, S and Ν, wherein each of the C6-ioaryl and 5-10 membered heteroaryl is optionally substituted with 1, 2, 3 or 4 substituents independently selected from D, F, CI, Br, I, CN, N02, N3, ORa, SRa, NRaRb, -C(=0)NRaRb, Ci_6alkyl, Ci_6haloalkyl, C2_6alkenyl, C2_6alkynyl, -(CMalkylene)-CN, -(Ci_4alkylene)-ORa, -(CMalkylene)-NRaRb, C6-ioaryl and 5-10 membered heteroaryl;
Z is Ci_6alkyl, C3_6cycloalkyl, -(Ci^alkylene)-(C3_6cycloalkyl), C3-6heterocyclyl,
Figure imgf000023_0001
(C3_6heterocyclyl),
Figure imgf000023_0002
or -(Ci^alkylene)-(C5_i2spirobicyclyl), wherein each of the Ci-6alkyl, C3_6cycloalkyl, -(Ci_ 4alkylene)-(C3-6cycloalkyl), C3_6heterocyclyl, -(Ci^alkylene)-(C3_6heterocyclyl), Cs-^fused bicyclyl,
Figure imgf000023_0003
and -(Ci_4alkylene)-(C5_ i2spirobicyclyl) is optionally substituted with 1, 2, 3 or 4 substituents independently selected from D, F, CI, Br, I, C^alkyl, Ci_6haloalkyl, ORa, -(CMalkylene)-ORa, -(CMalkylene)-NRaRb, NRaRb -C(=0)NRaRb and -OC(=0)NRaRb; and each of the cyclic structures in the bicyclyl groups is a carbocyclic ring or a heterocyclic ring; and
each R and Rb is independently H, Ci-6aliphatic, C3_6cycloalkyl, -(Ci_4alkylene)-(C3_6cycloalkyl), C3_6heterocyclyl, -(Ci_4alkylene)-(C3_6heterocyclyl), C6-ioaryl,
Figure imgf000023_0004
5-10 membered heteroaryl or
Figure imgf000023_0005
-(5-10membered heteroaryl), provided that when Z is Ci_ 6alkyl, NR Rb is not NH2 or Me2, and when R and Rb are bonded to the same nitrogen atom, R and Rb, together with the nitrogen atom they are attached to, optionally form a 3-8 membered heterocyclyl, wherein each of the
Figure imgf000023_0006
C3_6cycloalkyl, -(Ci-4alkylene)-(C3_6cycloalkyl), C3_6heterocyclyl, -(Ci_4alkylene)-(C3_6heterocyclyl), C6-ioaryl,
Figure imgf000023_0007
5-10 membered heteroaryl ,
Figure imgf000023_0008
-(5-10membered heteroaryl) and 3-8 membered heterocyclyl is optionally substituted with 1, 2, 3 or 4 substituents independently selected from D, F, CI, CN, N3, OH, NH2, alkoxy and alkylamino.
[088] The bicyclyl groups disclosed herein include
Figure imgf000023_0009
i2fused bicyclyl), Cs-^spirobicyclyl or -(Ci_4alkylene)-(C5_i2spirobicyclyl).
[089] In certain embodiments, R1, R2, R3, R4, R5 and R6 is independently H or D. [090] In another embodiment, X is phenyl optionally substituted with 1, 2, 3 or 4 substituents independently selected from D, F, CI, Br, and Ci_3haloalkyl.
[091] In certain embodiments, Z is Ci_6alkyl, C3_6cycloalkyl, C3_6heterocyclyl, -(Ci_4alkylene)- (C3_6heterocyclyl),
Figure imgf000024_0001
or -(Ci^alkylene)-(C5-i2spirobicyclyl), wherein the Ci-6alkyl, C3_6cycloalkyl, C3-6heterocyclyl, -(Ci_ 4alkylene)-(C3_6heterocyclyl),
Figure imgf000024_0002
bicyclyl), C5- i2spirobicyclyl and -(Ci^alkylene)-(C5_i2spirobicyclyl), each of which is optionally substituted with 1, 2, 3 or 4 substituents independently selected from D, F, CI, Br, I,
Figure imgf000024_0003
OR and NR Rb; each of the cyclic structures in the bicyclyl groups is a carbocyclic ring or a heterocyclic ring.
[092] In another embodiment, each R and Rb is independently H,
Figure imgf000024_0004
C3_6cycloalkyl or - (Ci_3alkylene)-(C3-6cycloalkyl), provided that when Z is Ci_6alkyl, NR Rb is not NH2 or NMe2, wherein each of the
Figure imgf000024_0005
C3_6cycloalkyl and -(Ci_3alkylene)-(C3_6cycloalkyl) is optionally substituted with 1, 2, 3 or 4 substituents independently selected from D, F, OH, NH2,
Figure imgf000024_0006
and Ci-3alkylamino.
[093] In another embodiment, Z is selected from the following substructures:
Figure imgf000024_0007
(Z1 ) , (Z2) , (Z3) , (Z4) , (Z5) , (Z6) , (Z7) , (Z8) , (Z9) ,
Figure imgf000024_0008
(Z10) , (Z11 ) , (Z12) , (Z13) , (Z14) , (Z15) , (Z16) , (Z17) ,
Figure imgf000024_0009
(Z18) , (Z19) , (Z20) , (Z21 ) , (Z22) , (Z23) , (Z24) ,
Figure imgf000025_0001
(Z25) , (Z26) , (Z27) , (Z28) , (Z29) , (Z30) , (Z31) , (Z32) , (Z33) ,
Figure imgf000025_0002
(Ζ34) , (Ζ35) , (Ζ36) , (Ζ37) , (Ζ38) , (Ζ39) , (Ζ40) , (Ζ41) , (Ζ42) , or a stereoisomer thereof, wherein
n is 0, 1, 2 or 3;
each W and W is independently O or NH;
each hydrogen in Z is optionally substituted with 1, 2, 3 or 4 substituents independently selected from D, F, CI, Br, I, d_3alkyl, Ci_6haloalkyl, ORa and NRaRb, provided that NRaRb is not NH2 or NMe2 in structures (Z25) to (Z32).
[094] Some non-limiting examples of the compound disclosed herein, and their pharmaceutically acceptable salts and solvates thereof, are shown in the following:
Table 1
Figure imgf000025_0003
Figure imgf000026_0001
 [095] The present invention also comprises the use of a compound of the invention, or pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment either acutely or chronically of a hyperproliferative disease state and/or an angiogenesis mediated disease state, including those described previously. The compounds of the present invention are useful in the manufacture of an anti-cancer medicament. The compounds of the present invention are also useful in the manufacture of a medicament to attenuate or prevent disorders through inhibition of protein kinases. The present invention comprises a
pharmaceutical composition comprising a therapeutically effective amount of a compound of Formula (I) in association with at least one pharmaceutically acceptable carrier, adjuvant or diluent.
[096] The present invention also comprises a method of treating hyperproliferating and angiogenesis related disorders in a subject having or susceptible to such disorder, the method comprising treating the subject with a therapeutically effective amount of a compound of Formula (I).
[097] Unless otherwise stated, all stereoisomers, geometric isomers, tautomers, solvates, metabolites, salts, and pharmaceutically acceptable prodrugs of the compounds of the invention are within the scope of the invention.
[098] 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.
[099] The compounds of the invention 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).
[0100] 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 -toluenesulfonic acid or ethanesulfonic acid, or the like. COMPOSITION, FORMULATIONS AND ADMINSTRATION OF COMPOUNDS OF THE INVENTION
[0101] According to one aspect, the invention features pharmaceutical compositions that include a compound of formula (I), a compound listed in table 1, and a pharmaceutically acceptable carrier, adjuvant, or vehicle. The amount of compound in the compositions of the invention is such that is effective to detectably inhibit a protein kinase in a biological sample or in a patient.
[0102] It will also be appreciated that certain of the compounds of present invention can exist in free form for treatment, or where appropriate, as a pharmaceutically acceptable derivative thereof. According to the present invention, a pharmaceutically acceptable derivative includes, but is not limited to, pharmaceutically acceptable prodrugs, salts, esters, salts of such esters, or any other adducts or derivatives which upon administration to a patient in need is capable of providing, directly or indirectly, a compound as otherwise described herein, or a metabolite or residue thereof.
[0103] As described above, the pharmaceutically acceptable compositions of the present invention additionally comprise a pharmaceutically acceptable carrier, adjuvant, or vehicle, which, as used herein, includes any and all solvents, diluents, or other liquid vehicle, dispersion or suspension aids, surface active agents, isotonic agents, thickening or emulsifying agents, preservatives, solid binders, lubricants and the like, as suited to the particular dosage form desired. Troy et al., Remington: The Science and Practice of Pharmacy, 21st ed., 2005, Lippincott Williams & Wilkins, Philadelphia and Swarbrick et al., Encyclopedia of Pharmaceutical Technology, eds. 1988-1999, Marcel Dekker, New York, all of each of which are herein incorporated by reference in there entireties, are disclosed various carriers used in formulating pharmaceutically acceptable compositions and known techniques for the preparation thereof. Except insofar as any conventional carrier medium is in compatible with the compounds of the invention, such as by producing any undesirable biological effect or otherwise interacting in a deleterious manner with any other component(s) of the pharmaceutically acceptable composition, its use is contemplated to be within the scope of this invention.
[0104] Some non- limiting examples of materials which can serve as pharmaceutically acceptable carriers include ion exchangers, alumina, aluminum stearate, lecithin, serum proteins, such as human serum albumin, buffer substances such as phosphates, glycine, sorbic acid or potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, polyacrylates, waxes, polyethylene-polyoxypropylene-block polymers, wool fat, sugars such as lactose, glucose and sucrose; starches such as corn starch and potato starch; cellulose and its derivatives such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; powdered tragacanth; malt; gelatin; talc; excipients such as cocoa butter and suppository waxes; oils such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; glycols; such a propylene glycol or polyethylene glycol; esters such as ethyl oleate and ethyl laurate; agar; buffering agents such as magnesium hydroxide and aluminum hydroxide; alginic acid; pyrogen-free water; isotonic saline; Ringer's solution; ethyl alcohol, and phosphate buffer solutions, as well as other non-toxic compatible lubricants such as sodium lauryl sulfate and magnesium stearate, as well as coloring agents, releasing agents, coating agents, sweetening, flavoring and perfuming agents, preservatives and antioxidants can also be present in the composition, according to the judgment of the formulator.
[0105] The compositions of the present invention may be administered orally, parenterally, by inhalation spray, topically, rectally, nasally, buccally, vaginally or via an implanted reservoir. The term "parenteral" includes subcutaneous, intravenous, intramuscular, intra-articular, intra- synovial, intrasternal, intrathecal, intraocular, intrahepatic, intralesional and intracranial injection or infusion techniques. Preferably, the compositions are administered orally, intraperitoneally or intravenously. Sterile injectable forms of the compositions of this invention may be aqueous or oleaginous suspension. These suspensions may be formulated according to techniques known in the art using suitable dispersing or wetting agents and suspending agents. The sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally acceptable diluent or solvent, for example as a solution in 1,3- butanediol. 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 are conventionally employed as a solvent or suspending medium.
[0106] For this purpose, any bland fixed oil may be employed including synthetic mono- or diglycerides. Fatty acids, such as oleic acid and its glyceride derivatives are useful in the preparation of injectables, as are natural pharmaceutically-acceptable oils, such as olive oil or castor oil, especially in their polyoxyethylated versions. These oil solutions or suspensions may also contain a long-chain alcohol diluent or dispersant, such as carboxymethyl cellulose or similar dispersing agents that are commonly used in the formulation of pharmaceutically acceptable dosage forms including emulsions and suspensions. Other commonly used surfactants, such as TWEEN®, SPAN® and other emulsifying agents or bioavailability enhancers which are commonly used in the manufacture of pharmaceutically acceptable solid, liquid, or other dosage forms may also be used for the purposes of formulation.
[0107] Some non-limiting examples of the pharmaceutically acceptable compositions of this invention may be orally administered in any orally acceptable dosage form including capsules, tablets, aqueous suspensions or solutions. In the case of tablets for oral use, carriers commonly used include lactose and corn starch. Lubricating agents, such as magnesium stearate, are also typically added. For oral administration in a capsule form, useful diluents include lactose and dried cornstarch. When aqueous suspensions are required for oral use, the active ingredient is combined with emulsifying and suspending agents. If desired, certain sweetening, flavoring or coloring agents may also be added.
[0108] Alternatively, the pharmaceutically acceptable compositions of this invention may be administered in the form of suppositories for rectal administration. These can be prepared by mixing the agent with a suitable non-irritating excipient that is solid at room temperature but liquid at rectal temperature and therefore will melt in the rectum to release the drug. Such materials include cocoa butter, beeswax and polyethylene glycols.
[0109] The pharmaceutically acceptable compositions of this invention may also be administered topically, especially when the target of treatment includes areas or organs readily accessible by topical application, including diseases of the eye, the skin, or the low intestinal tract. Suitable topical formulations are readily prepared for each of these areas or organs.
[0110] Topical application for the lower intestinal tract can be effected in a rectal suppository formulation (see above) or in a suitable enema formulation. Topically-transdermal patches may also be used. For topical applications, the pharmaceutically acceptable compositions may be formulated in a suitable ointment containing the active component suspended or dissolved in one or more carriers. Some non-limiting examples of carriers for topical administration of the compounds of this invention include mineral oil, liquid petrolatum, white petrolatum, propylene glycol, polyoxy ethylene, polyoxypropylene compound, emulsifying wax and water. Alternatively, the pharmaceutically acceptable compositions can be formulated in a suitable lotion or cream containing the active components suspended or dissolved in one or more pharmaceutically acceptable carriers. Some non-limiting examples of suitable carriers include mineral oil, sorbitan monostearate, polysorbate 60, cetyl esters wax, cetearyl alcohol, 2- octyldodecanol, benzyl alcohol and water.
[0111] For ophthalmic use, the pharmaceutically acceptable compositions may be formulated, e.g., as micronized suspensions in isotonic, pH adjusted sterile saline or other aqueous solution, or, preferably, as solutions in isotonic, pH adjusted sterile saline or other aqueous solution, either with or without a preservative such as benzylalkonium chloride. Alternatively, for ophthalmic uses, the pharmaceutically acceptable compositions may be formulated in an ointment such as petrolatum. The pharmaceutically acceptable compositions of this invention may also be administered by nasal aerosol or inhalation. Such compositions are prepared according to techniques well-known in the art of pharmaceutical formulation and may be prepared as solutions in saline, employing benzyl alcohol or other suitable preservatives, absorption promoters to enhance bioavailability, fluorocarbons, and/or other conventional solubilizing or dispersing agents.
[0112] Some non-limiting examples of liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs. In addition to the active compounds, the liquid dosage forms may contain inert diluents commonly used in the art such as, for example, water or other solvents, solubilizing agents and emulsifiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, dimethylformamide, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor, and sesame oils), glycerol, tetrahydrofurfuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof. Besides inert diluents, the oral compositions can also include adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents.
[01 13] Injectable preparations, for example, sterile injectable aqueous or oleaginous suspensions may be formulated according to the known art using suitable dispersing or wetting agents and suspending agents. The sterile injectable preparation may also be a sterile injectable solution, suspension or emulsion in a nontoxic parenterally acceptable diluent or solvent, for example, as a solution in 1, 3-butanediol. Among the acceptable vehicles and solvents that may be employed are water, Ringer's solution, U.S. P. and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose any bland fixed oil can be employed including synthetic mono- or diglycerides. In addition, fatty acids such as oleic acid are used in the preparation of injectables.
[01 14] The injectable formulations can be sterilized, for example, by filtration through a bacterial-retaining filter, or by incorporating sterilizing agents in the form of sterile solid compositions which can be dissolved or dispersed in sterile water or other sterile injectable medium prior to use. In order to prolong the effect of a compound of the present invention, it is often desirable to slow the absorption of the compound from subcutaneous or intramuscular injection. This may be accomplished by the use of a liquid suspension of crystalline or amorphous material with poor water solubility. The rate of absorption of the compound then depends upon its rate of dissolution that, in turn, may depend upon crystal size and crystalline form. Alternatively, dissolving or suspending the compound in an oil vehicle accomplishes delayed absorption of a parenterally administered compound form.
[01 15] Injectable depot forms are made by forming microencapsule matrices of the compound in biodegradable polymers such as polylactide-polyglycolide. Depending upon the ratio of compound to polymer and the nature of the particular polymer employed, the rate of compound release can be controlled. Examples of other biodegradable polymers include poly(orthoesters) and poly(anhydrides). Depot injectable formulations are also prepared by entrapping the compound in liposomes or microemulsions that are compatible with body tissues.
[01 16] Compositions for rectal or vaginal administration are preferably suppositories which can be prepared by mixing the compounds of this invention with suitable non- irritating excipients or carriers such as cocoa butter, polyethylene glycol or a suppository wax which are solid at ambient temperature but liquid at body temperature and therefore melt in the rectum or vaginal cavity and release the active compound.
[01 17] Solid dosage forms for oral administration include capsules, tablets, pills, powders, and granules. In such solid dosage forms, the active compound is mixed with at least one inert, pharmaceutically acceptable excipient or carrier such as sodium citrate or dicalcium phosphate and/or a) fillers or extenders such as starches, lactose, sucrose, glucose, mannitol, and silicic acid, b) binders such as, for example, carboxymethylcellulose, alginates, gelatin, polyvinylpyrrolidinone, sucrose, and acacia, c) humectants such as glycerol, d) disintegrating agents such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate, e) solution retarding agents such as paraffin, f) absorption accelerators such as quaternary ammonium compounds, g) wetting agents such as, for example, cetyl alcohol and glycerol monostearate, h) absorbents such as kaolin and bentonite clay, and i) lubricants such as talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate, and mixtures thereof. In the case of capsules, tablets and pills, the dosage form may also comprise buffering agents.
[01 18] Solid compositions of a similar type may also be employed as fillers in soft and hard- filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polyethylene glycols and the like. The solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings and other coatings well known in the pharmaceutical formulating art. They may optionally contain opacifying agents and can also be of a composition that they release the active ingredient(s) only, or preferentially, in a certain part of the intestinal tract, optionally, in a delayed manner. Examples of embedding compositions that can be used include polymeric substances and waxes. Solid compositions of a similar type may also be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polythylene glycols and the like.
[01 19] The active compounds can also be in micro-encapsulated form with one or more excipients as noted above. The solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings, release controlling coatings and other coatings well known in the pharmaceutical formulating art. In such solid dosage forms the active compound may be admixed with at least one inert diluent such as sucrose, lactose or starch. Such dosage forms may also comprise, as is normal practice, additional substances other than inert diluents, e.g., tableting lubricants and other tableting aids such a magnesium stearate and microcrystalline cellulose. In the case of capsules, tablets and pills, the dosage forms may also comprise buffering agents. They may optionally contain pacifying agents and can also be of a composition that they release the active ingredient(s) only, or preferentially, in a certain part of the intestinal tract, optionally, in a delayed manner. Examples of embedding compositions that can be used include polymeric substances and waxes.
[0120] Dosage forms for topical or transdermal administration of a compound of this invention include ointments, pastes, creams, lotions, gels, powders, solutions, sprays, inhalants or patches. The active component is admixed under sterile conditions with a pharmaceutically acceptable carrier and any needed preservatives or buffers as may be required. Ophthalmic formulation, eardrops, and eye drops are also contemplated as being within the scope of this invention. Additionally, the present invention contemplates the use of transdermal patches, which have the added advantage of providing controlled delivery of a compound to the body. Such dosage forms can be made by dissolving or dispensing the compound in the proper medium. Absorption enhancers can also be used to increase the flux of the compound across the skin. The rate can be controlled by either providing a rate controlling membrane or by dispersing the compound in a polymer matrix or gel.
[0121] The compounds of the invention are preferably formulated in dosage unit form for ease of administration and uniformity of dosage. The expression "dosage unit form" as used herein refers to a physically discrete unit of agent appropriate for the patient to be treated. It will be understood, however, that the total daily usage of the compounds and compositions of the present invention will be decided by the attending physician within the scope of sound medical judgment. The specific effective dose level for any particular patient or organism will depend upon a variety of factors including the disorder being treated and the severity of the disorder; the activity of the specific compound employed; the specific composition employed; the age, body weight, general health, sex and diet of the patient; the time of administration, route of administration, and rate of excretion of the specific compound employed; the duration of the treatment; drugs used in combination or coincidental with the specific compound employed, and like factors well known in the medical arts.
[0122] The amount of the compounds of the present invention that may be combined with the carrier materials to produce a composition in a single dosage form will vary depending upon the host treated, the particular mode of administration. Preferably, the compositions should be formulated so that a dosage of between 0.01 - 200 mg/kg body weight/day of the inhibitor can be administered to a patient receiving these compositions.
[0123] Compounds of this invention can be administered as the sole pharmaceutical agent or in combination with one or more other additional therapeutic (pharmaceutical) agents where the combination causes no unacceptable adverse effects. This may be of particular relevance for the treatment of hyper-proliferative diseases such as cancer. In this instance, the compound of this invention can be combined with known cytotoxic agents, signal transduction inhibitors, or with other anti-cancer agents, as well as with admixtures and combinations thereof. As used herein, additional therapeutic agents that are normally administered to treat a particular disease, or condition, are known as "appropriate for the disease, or condition, being treated". As used herein, "additional therapeutic agents" is meant to include chemotherapeutic agents and other antiproliferative agents.
[0124] For example, chemotherapeutic agents or other antiproliferative agents may be combined with the compounds of this invention to treat proliferative disease or cancer. Some non-limiting examples of chemotherapeutic agents or other antiproliferative agents include HDAC inhibitors including SAHA, MS-275, MGO 103, and those described in WO 2006/010264, WO 03/024448, WO 2004/069823, US 2006/0058298, US 2005/0288282, WO 00/71703, WO 01/38322, WO 01/70675, WO 03/006652, WO 2004/035525, WO 2005/030705, WO 2005/092899, and demethylating agents including, but not limited to, 5-aza-dC, Vidaza and Decitabine and those described in US 6,268137, US 5,578,716, US 5,919,772, US 6,054,439, US 6,184,211, US 6,020,318, US 6,066,625, US 6,506,735, US 6,221,849, US 6,953,783, US 11/393,380.
[0125] In another embodiment of the present invention, for example, chemotherapeutic agents or other anti -proliferative agents may be combined with the compounds of this invention to treat proliferative diseases and cancer. Some non-limiting examples of known chemotherapeutic agents include other therapies or anticancer agents that may be used in combination with the inventive anticancer agents of the present invention and include surgery, radiotherapy (in but a few examples, gamma radiation, neutron beam radiotherapy, electron beam radiotherapy, proton therapy, brachytherapy, and systemic radioactive isotopes, to name a few), endocrine therapy, taxanes (paclitaxel, taxotere), platinum derivatives (cisplatin, carboplatin, oxaliplatin), biologic response modifiers (interferons, interleukins), tumor necrosis factor (TNF, TRAIL receptor targeting agents, to name a few), hyperthermia and cryotherapy, agents to attenuate any adverse effects (e.g., antiemetics), and other approved chemotherapeutic drugs, including, but not limited to, alkylating drugs (chlormethine, chlorambucil, cyclophosphamide, ifosfamide, melphalan, etc), anti-metabolites (methotrexate, raltitrexed, pemetrexed, etc), purine antagonists and pyrimidine antagonists (6-mercaptopurine, 5-fluorouracil, cytarabine, gemcitabine), spindle poisons (vinblastine, vincristine, vinorelbine), podophyllotoxins (etoposide, irinotecan, topotecan), antibiotics (doxorubicin, bleomycin, mitomycin), nitrosoureas (carmustine, lomustine), cell cycle inhibitors (KSP mitotic kinesin inhibitors, CENP-E and CDK inhibitors), enzymes (asparaginase), hormones (tamoxifen, leuprolide, flutamide, megestrol, dexamethasone), antiangiogenic agents (avastin and others), monoclonal antibodies (Belimumab (BENLYSTA®), brentuximab (ADCETRIS®), cetuximab (ERBITUX®), gemtuzumab (MYLOTARG®), ipilimumab (YERVOY®), ofatumumab (ARZERRA®), panitumumab (VECTIBIX®), ranibizumab (LUCERTIS®), rituximab (RITUXAN®), tositumomab (BEXXAR®), trastuzumab (HERCEPTIN®)), kinase inhibitors (imatinib (GLEEVEC®), sunitinib (SUTENT®), sorafenib (NEXAVAR®), erlotinib, (TARCEVA®), gefitinib (IRESSA®), dasatinib (SPRYCEL®), nilotinib (TASIGNA®), lapatinib (TYKERB®), crizotinib (XALKORI®), ruxolitinib (JAKAFI®), vemurafenib (ZELBORAF®), vandetanib (CAPRELSA®), pazopanib (VOTRIENT®), and others), and agents inhibiting or activating cancer pathways such as the mTOR, HIF (hypoxia induced factor) pathways (such as everolimus and temsirolimus) and others. For a more comprehensive discussion of updated cancer therapies see, http://www.nci.nih.gov/, a list of the FDA approved oncology drugs at http://www.fda.gov/cder/cancer/druglist-rame.htm, and The Merck Manual, Eighteenth Ed. 2006, tall of which are herein incorporated by reference in their entireties.
[0126] In another embodiment, the compounds of the present invention can be combined, with cytotoxic anti-cancer agents. Examples of such agents can be found in the 13th Edition of the Merck Index (2001). These agents include, by no way of limitation, asparaginase, bleomycin, carboplatin, carmustine, chlorambucil, cisplatin, colaspase, cyclophosphamide, cytarabine, dacarbazine, dactinomycin, daunorubicin, doxorubicin (adriamycine), epirubicin, etoposide, 5- fluorouracil, hexamethylmelamine, hydroxyurea, ifosfamide, irinotecan, leucovorin, lomustine, mechlorethamine, 6-mercaptopurine, mesna, methotrexate, mitomycin C, mitoxantrone, prednisolone, prednisone, procarbazine, raloxifen, streptozocin, tamoxifen, thioguanine, topotecan, vinblastine, vincristine, and vindesine.
[0127] Other cytotoxic drugs suitable for use with the compounds of the invention include, but are not limited to, those compounds acknowledged to be used in the treatment of neoplastic diseases, such as those for example in Goodman and Gilman's The Pharmacological Basis of Therapeutics (Ninth Edition, 1996, McGraw-Hill). These agents include, by no way of limitation, aminoglutethimide, L-asparaginase, azathioprine, 5-azacytidine cladribine, busulfan, diethylstilbestrol, 2,2'-difluorodeoxycytidine, docetaxel, erythrohydroxynonyladenine, ethinyl estradiol, 5-fluorodeoxyuridine, 5-fluorodeoxyuridine monophosphate, fludarabine phosphate, fluoxymesterone, flutamide, hydroxyprogesterone caproate, idarubicin, interferon, medroxyprogesterone acetate, megestrol acetate, melphalan, mitotane, paclitaxel, pentostatin, N- phosphonoacetyl-L-aspartate (PALA), plicamycin, semustine, teniposide, testosterone propionate, thiotepa, trimethylmelamine, uridine, and vinorelbine.
[0128] Other cytotoxic anti-cancer agents suitable for use in combination with the compounds of the invention also include newly discovered cytotoxic principles such as oxaliplatin, gemcitabine, capecitabine, epothilone and its natural or synthetic derivatives, temozolomide (Quinn et al, J. Clin. Oncol, 2003, 21(4), 646-651), tositumomab (BEXXAR®), trabedectin (Vidal et al., Proceedings of the American Society for Clinical Oncology 2004, 23, abstract 3181), and the inhibitors of the kinesin spindle protein Eg5 (Wood, et al., Curr. Opin. Pharmacol, 2001, 1, 370-377).
[0129] In another embodiment, the compounds of the present invention can be combined with other signal transduction inhibitors. Examples of such agents include, by no way of limitation, antibody therapies such as trastuzumab (HERCEPTIN®), cetuximab (ERBITUX®), ipilimumab
(YERVOY®) and pertuzumab. Examples of such therapies also include, by no way of limitation, small-molecule kinase inhibitors such as imatinib (GLEEVEC®), sunitinib (SUTENT®), sorafenib (NEXAVAR®), erlotinib (TARCEVA®), gefitinib (IRESSA®), dasatinib
(SPRYCEL®), nilotinib (TASIGNA®), lapatinib (TYKERB®), crizotinib (XALKORI®), ruxolitinib (JAKAFI®), vemurafenib (ZELBORAF®), vandetanib (CAPRELSA®), pazopanib
(VOTRIENT®), afatinib, alisertib, amuvatinib, axitinib, bosutinib, brivanib, canertinib, cabozantinib, cediranib, crenolanib, dabrafenib, dacomitinib, danusertib, dovitinib, foretinib, ganetespib, ibrutinib, iniparib, lenvatinib, linifanib, linsitinib, masitinib, momelotinib, motesanib, neratinib, niraparib, oprozomib, olaparib, pictilisib, ponatinib, quizartinib, regorafenib, rigosertib, rucaparib, saracatinib, saridegib, tandutinib, tasocitinib, telatinib, tivantinib, tivozanib, tofacitinib, trametinib, vatalanib, veliparib, vismodegib, volasertib, BMS-540215, BMS777607, J J38877605, TKI258, GDC-0941 (Folkes et al., J. Med. Chem., 2008, 51 : 5522), BZE235, and others.
[0130] In another embodiment, the compounds of the present invention can be combined with inhibitors of histone deacetylase. Examples of such agents include, by no way of limitation, suberoylanilide hydroxamic acid (SAHA), LAQ-824 (Ottmann et al., Proceedings of the American Society for Clinical Oncology, 2004, 23, abstract 3024), LBH-589 (Beck et al., Proceedings of the American Society for Clinical Oncology, 2004, 23, abstract 3025), MS-275 (Ryan et al., Proceedings of the American Association of Cancer Research 2004, 45, abstract 2452), FR-901228 (Piekarz et al., Proceedings of the American Society for Clinical Oncology, 2004, 23, abstract 3028) and MGCDOl 03 (US 6,897,220).
[0131] In another embodiment, the compounds of the present invention can be combined with other anti-cancer agents such as proteasome inhibitors, and m-TOR inhibitors. These include, by no way of limitation, bortezomib, and CCI-779 (Wu et al., Proceedings of the American Association of Cancer Research, 2004, 45, abstract 3849). The compounds of the present invention can be combined with other anti-cancer agents such as topoisomerase inhibitors, including but not limited to camptothecin.
[0132] Those additional agents may be administered separately from the compound- containing composition, as part of a multiple dosage regimen. Alternatively, those agents may be part of a single dosage form, mixed together with the compound of this invention in a single composition. If administered as part of a multiple dosage regimen, the two active agents may be submitted simultaneously, sequentially or within a period of time from one another which would result in the desired activity of the agents.
[0133] The amount of both the compound and the additional therapeutic agent (in those compositions which comprise an additional therapeutic agent as described above) that may be combined with the carrier materials to produce a single dosage form will vary depending upon the host treated and the particular mode of administration. Normally, the amount of additional therapeutic agent present in the compositions of this invention will be no more than the amount that would normally be administered in a composition comprising that therapeutic agent as the only active agent. Preferably the amount of additional therapeutic agent in the presently disclosed compositions will range from about 50% to 100% of the amount normally present in a composition comprising that agent as the only therapeutically active agent. In those compositions which comprise an additional therapeutic agent, that additional therapeutic agent and the compound of this invention may act synergistically.
USES OF THE COMPOUNDS AND COMPOSITIONS OF THE INVENTION
[0134] The invention features pharmaceutical compositions that include a compound of formula (I), or a compound listed in Table 1, and a pharmaceutically acceptable carrier, adjuvant, or vehicle. The amount of compound in the compositions of the invention is such that is effective to detectably inhibit a protein kinase, such as ALK and c-Met inhibitory activity. The compounds of the invention are useful in therapy as antineoplasia agents or to minimize deleterious effects of ALK and c-Met signaling.
[0135] Compounds of the present invention would be useful for, but not limited to, the prevention or treatment of proliferative diseases, condition, or disorder in a patient by administering to the patient a compound or a composition of the invention in an effective amount. Such diseases, conditions, or disorders include cancer, particularly metastatic cancer, atherosclerosis and lung fibrosis.
[0136] Compounds of the invention would be useful for the treatment of neoplasm including cancer and metastasis, including, but not limited to: carcinoma such as cancer of the bladder, breast, colon, kidney, liver, lung (including small cell lung cancer), esophagus, gall-bladder, ovary, pancreas, stomach, cervix, thyroid, prostate, and skin (including squamous cell carcinoma); hematopoietic tumors of lymphoid lineage (including leukemia, acute lymphocytic leukemia, acute lymphoblastic leukemia, B-cell lymphoma, T-cell-lymphoma, Hodgkin's lymphoma, non-Hodgkin's lymphoma, hairy cell lymphoma and Burkett's lymphoma); hematopoietic tumors of myeloid lineage (including acute and chronic myelogenous leukemias, myelodysplastic syndrome and promyelocytic leukemia); tumors of mesenchymal origin (including fibrosarcoma and rhabdomyosarcoma, and other sarcomas, e.g. soft tissue and bone); tumors of the central and peripheral nervous system (including astrocytoma, neuroblastoma, glioma and schwannomas); and other tumors (including melanoma, seminoma, teratocarcinoma, osteosarcoma, xeroderoma pigmentosum, keratoacanthoma, thyroid follicular cancer and Kaposi's sarcoma).
[0137] The compounds also would be useful for treatment of ophthalmological conditions such as corneal graft rejection, ocular neovascularization, retinal neovascularization including neovascularization following injury or infection, diabetic retinopathy, retrolental fibroplasia and neovascular glaucoma; retinal ischemia; vitreous hemorrhage; ulcerative diseases such as gastric ulcer; pathological, but non-malignant, conditions such as hemangiomas, including infantile hemaginomas, angiofibroma of the nasopharynx and avascular necrosis of bone; and disorders of the female reproductive system such as endometriosis. The compounds are also useful for the treatment of edema, and conditions of vascular hyperpermeability.
[0138] The compounds of the present invention are also useful in the treatment of diabetic conditions such as diabetic retinopathy and microangiopathy. The compounds of the present invention are also useful in the reduction of blood flow in a tumor in a subject. The compounds of the present invention are also useful in the reduction of metastasis of a tumor in a subject.
[0139] Besides being useful for human treatment, these compounds are also useful for veterinary treatment of companion animals, exotic animals and farm animals, including mammals, rodents, and the like. More preferred animals include horses, dogs, and cats. As used herein, the compounds of the present invention include the pharmaceutically acceptable derivatives thereof.
[0140] Where the plural form is used for compounds, salts, and the like, this is taken to mean also a single compound, salt and the like.
[0141] The treatment method that includes administering a compound or composition of the invention can further include administering to the patient an additional therapeutic agent (combination therapy) selected from: a chemotherapeutic or anti-proliferative agent, or an antiinflammatory agent, wherein the additional therapeutic agent is appropriate for the disease being treated and the additional therapeutic agent is administered together with a compound or composition of the invention as a single dosage form or separately from the compound or composition as part of a multiple dosage form. The additional therapeutic agent may be administered at the same time as a compound of the invention or at a different time. In the latter case, administration may be staggered by, for example, 6 hours, 12 hours, 1 day, 2 days, 3 days, 1 week, 2 weeks, 3 weeks, 1 month, or 2 months.
[0142] The invention also features a method of inhibiting the growth of a cell that expresses ALK or c-Met, that includes contacting the cell with a compound or composition of the invention, thereby causing inhibition of growth of the cell. Examples of a cell whose growth can be inhibited include: a breast cancer cell, a colorectal cancer cell, a lung cancer cell, a papillary carcinoma cell, a prostate cancer cell, a lymphoma cell, a colon cancer cell, a pancreatic cancer cell, an ovarian cancer cell, a cervical cancer cell, a central nervous system cancer cell, an osteogenic sarcoma cell, a renal carcinoma cell, a hepatocellular carcinoma cell, a bladder cancer cell, a gastric carcinoma cell, a head and neck squamous carcinoma cell, a melanoma cell, or a leukemia cell.
[0143] The invention provides a method of inhibiting ALK or c-Met kinase activity in a biological sample that includes contacting the biological sample with a compound or composition of the invention. The term "biological sample" as used herein, means a sample outside a living organism and includes, without limitation, cell cultures or extracts thereof; biopsied material obtained from a mammal or extracts thereof; and blood, saliva, urine, feces, semen, tears, or other body fluids or extracts thereof. Inhibition of kinase activity, particularly ALK or c-Met kinase activity, in a biological sample is useful for a variety of purposes known to one of skill in the art. Examples of such purposes include, but are not limited to, blood transfusion, organ-transplantation, biological specimen storage, and biological assays.
[0144] In certain embodiments of the present invention an "effective amount" or "effective dose" of the compound or pharmaceutically acceptable composition is that amount effective for treating or lessening the severity of one or more of the aforementioned disorders. The compounds and compositions, according to the method of the present invention, may be administered using any amount and any route of administration effective for treating or lessening the severity of the disorder or disease. The exact amount required will vary from subject to subject, depending on the species, age, and general condition of the subject, the severity of the infection, the particular agent, its mode of administration, and the like. A compound or composition can also be administered with one or more other therapeutic agents, as discussed above.
[0145] The compounds of this invention or pharmaceutical compositions thereof may also be used for coating an implantable medical device, such as prostheses, artificial valves, vascular grafts, stents and catheters. Vascular stents, for example, have been used to overcome restenosis (re-narrowing of the vessel wall after injury). However, patients using stents or other implantable devices risk clot formation or platelet activation. These unwanted effects may be prevented or mitigated by pre-coating the device with a pharmaceutically acceptable composition comprising a compound of this invention.
[0146] Suitable coatings and the general preparation of coated implantable devices are described in U.S. Patent Nos. 6,099,562; 5,886,026; and 5,304,121, all of which are herein incorporated by reference in their entireties. The coatings are typically biocompatible polymeric materials such as a hydrogel polymer, polymethyldisiloxane, polycaprolactone, polyethylene glycol, polylactic acid, ethylene vinyl acetate, and mixtures thereof. The coatings may optionally be further covered by a suitable topcoat of fluorosilicone, polysaccarides, polyethylene glycol, phospholipids or combinations thereof to impart controlled release characteristics into the composition. Implantable devices coated with a compound of this invention are another embodiment of the present invention. The compounds may also be coated on implantable medical devices, such as beads, or co- formulated with a polymer or other molecule, to provide a "drug depot" thus permitting the drug to be released over a longer time period than administration of an aqueous solution of the drug.
GENERAL SYNTHETIC PROCEDURES
[0147] 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.
[0148] Generally, the compounds in this invention may be prepared by methods described herein, wherein the substituents are as defined for formula (I), above, except where further noted. The following non-limiting schemes and examples are presented to further exemplify the invention. Persons skilled in the art will recognize that the chemical reactions described herein may be readily adapted to prepare a number of other compounds 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.
[0149] 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, Arco Chemical Company and Alfa Chemical Company, Shanghai Medpep. Co Ltd, Aladdin-Shanghai Jinchun Reagents, Ltd, and were used without further purification unless otherwise indicated. Common solvents were purchased from commercial suppliers such as Shantou XiLong Chemical Factory, Guangdong Guanghua Reagent Chemical Factory Co. Ltd., Guangzhou Reagent Chemical Factory, Tainjin YuYu Fine Chemical Ltd., Qingdao Tenglong Reagent Chemical Ltd., and Qingdao Ocean Chemical Factory.
[0150] Anhydrous THF, dioxane, toluene, and ether were obtained by refluxing the solvent with sodium. Anhydrous CH2CI2 and CHCI3 were obtained by refluxing the solvent with CaH2. EtOAc, PE, hexanes, DMA and DMF were treated with anhydrous a2S04 prior use.
[0151] 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.
[0152] Column chromatography was conducted using a silica gel column. Silica gel (300 - 400 mesh) was purchased from Qingdao Ocean Chemical Factory. JH NMR spectra were recorded with a Bruker 400 MHz spectrometer at ambient temperature. !Η NMR spectra were obtained as CDCI3, ί/6-DMSO, CD3OD or i/6-acetone solutions (reported in ppm), using TMS (0 ppm) or chloroform (7.25 ppm) as the reference standard. 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).
[0153] Low-resolution mass spectral (MS) data were generally determined on an Agilent 1200 Series LCMS (Zorbax SB-C18, 2.1 x 30 mm, 4 micorn, 10 minutes run, 0.6 mL/min flow rate, 5 to 95% (0.1% formic acid in CH3CN) in (0.1% formic acid in H20)) with UV detection at 210/254 nm and a low resonance electrospray mode (ESI) and Agilent 7890A-5975C Series GCMS (DB-624, 30 mmx0.32 mm, 1.8mm) with electron impact mode (EI)0
[0154] Purities of compounds were assessed by Agilent 1100 Series high performance liquid chromatography (HPLC) with UV detection at 210 nm and 254 nm. Column was normally operated at 40 °C.
[0155] The following abbreviations are used throughout the specification:
BBr3 boron tribromide
ΒΓΝΑΡ 2,2'-bis(diphenylphosphino)-l, 1 '-binaphthyl
BOC, Boc butyloxycarbonyl
BSA bovine serum albumin
CDCI3 chloroform deuterated
CHCI3 chloroform
CH2C12, DCM methylene chloride
CH3SO2CI, MsCl methanesulfonyl chloride Cs2C03 cesium carbonate
Cu copper
Cul copper(I) iodide
DAST Diethylaminosulfur trifluoride
DBU l,8-Diazabicyclo[5.4.0]undec-7-ene
DEAD dimethyl azodicarboxylate
DIAD diisopropyl azodicarboxylate
DIBAL diisobutylaluminum hydride
DIEA, DIPEA diisopropylethylamine
DMAP 4-dimethylaminopyridine
DMF dimethylformamide
DMSO dimethylsulfoxide
DPPA diphenylphosphoryl azide
EDCI 1 -(3 -dimethylaminopropyl)-3 -ethylcarbodiimide hydrochloride
EtOAc, EA ethyl acetate
Et20 diethyl ether
Et3N, TEA triethylamine
FBS fetal bovine serum
Fe iron
g gram
h hour
min minute
HATU 0-(7-azabenzotriazol- 1 -yl)-N,N,N',N'-tetramethyluronium hexafluorophosphate HBr hydrobromic acid
HBTU 0-benzotriazol-l-yl-N,N,N',N'-tetramethyluronium hexafluorophosphate HC1 hydrochloric acid
H2 hydrogen H20 water
H202 hydrogen peroxide
HO Ac AcOH acetic acid
HOBt 1 -hydroxybenzotnazole hydrate
K2C03 potassium carbonate
KOH potassium hydroxide
LiHMDS lithium bis(trimethylsilyl)amide
LDA Lithium diisopropylamide
MCPBA meto-chloroperbenzoic acid
MeCN, CH3CN acetonitrile
Mel methyl iodide
MeOH, CH3OH methanol
2-MeTHF 2-methyl tetrahydrofuran
MgS04 magnesium sulfate
mL, ml milliliter
2 nitrogen
NaBH4 sodium borohydride
NaBH3CN Sodium cyanoborohydride
NaBH3CN sodium cyanoborohydride
NaCl sodium chloride
aC102 sodium chlorite
NaH sodium hydride
aHC03 sodium bicarbonate
NaH2P04 sodium biphosphate
Nal sodium iodide
NaO(i-Bu) sodium tert-butoxide
NaOH sodium hydroxide Na2S04 sodium sulfate
NBS N-Bromosuccinimide
Η3 ammonia
NH4C1 ammonium chloride
NMP N-methylpyrrolidinone
PBS phosphate buffered saline
P(7-Bu)3 tri(tert-butyl)phosphine
Pd/C palladium on carbon
Pd2(dba)3 bis(dibenzylideneacetone) palladium
Pd(dppf)Cl2 l,l-bis(diphenylphosphino)ferrocene palladium chloride
Pd(OAc)2 palladium acetate
Pd(OH)2 palladium hydroxide
Pd(PPh3)4 palladium tetrakis triphenylphosphine
Pd(PPh3)2Cl2 Bis(triphenylphosphine)palladium(II) chloride
PE petroleum ether (60-90 °C)
POCI3 phosphorous oxychloride
PyBop benzotriazol- 1 -yl-oxytripyrrolidinophosphonium hexafluorophosphate
RT, rt, r.t. room temperature
Rt retention time
TBAB tetrabutylammonium bromide
TBAHS04 tetrabutylammonium hydrogen sulfate
TBTU O-benzotriazol- 1 -yl-N,N,N ',Ν '-tetramethyluronium tetrafluoroborate
TFA trifluoroacetic acid
TEAC bis(iefr«-ethylammonium)carbonate
THF tetrahydrofuran
[0156] Outlined in Schemes 1 and 2 are representative procedures for preparing the compounds disclosed herein. Each R5, R6, R7, V, X and Z represents appropriate groups. Unless otherwise indicated, R7 is H or Me; V is an optionally substituted heterocycle or spirobicyclyl ring; R5, R6, X and Z carry the definitions set forth above in connection with Formula (I).
Scheme 1
Figure imgf000046_0001
(5) (Z)
[0157] Some compounds with general structures as defined in Formula (I) can be prepared in a general method illustrated in Scheme 1. (R)-(hetero)aryl alcohol (1) and substituted fluoropyridine (2) is treated with a base such as NaH in aprotic solvent such as THF to give a coupled compound (3). The nitro group in the coupled compound (3) is then reduced to an amino group in an amine (4) under acidic conditions using a reducing agent such as Fe powder. Subsequent regio-selective bromination of the pyridine ring can be accomplished with the aid of N-bromo-succinimide to furnish a bromo compound (5). Final coupling of the bromo compound (5) with an appropriate alkyne (6) in the presence of a suitable Pd catalyst affords a desired kinase inhibitor (7) disclosed herein.
Scheme 2
Figure imgf000047_0001
(J!) (13) (14)
[0158] The compounds disclosed herein may also be prepared by the method as described in Scheme 2. Thus, an amine (5) is reacted with (Boc^O in the presence of a base such as Et3N to give an N-protected compound (8). Sonogashira coupling of the N-protected compound (8) with an alkynol (9) in the presence of an appropriate Pd catalyst such as Pd(dppf)Cl2 affords an alcohol (10). The alcohol (10) is then activated to a methanesulfonate derivative (11) with the aid of a base such as Et3N or i-Pr2NEt. Subsequent displacement of the methanesulfonate group in the methanesulfonate derivative (11) with a nitrogen-containing heterocycle or an azaspirobicycle (12) furnishes a nitrogen-compound (13). Boc-protecting groups can be removed under acidic conditions, for example, trifluoroacetic acid (TFA) in DCM, or HCl in ethyl acetate or ethyl ether to afford a desired kinase inhibitor (14) disclosed herein.
Example 1 4-(6-amino-5-((JR)-l-(2,6-dichloro-3-fluorophenyl)ethoxy)pyridin-3-yl)but-3-vn- 2-ol
Figure imgf000047_0002
Stepl) (R)-3-(l-(2,6-dichloro-3-fluorophenyl)ethoxy)-2-nitropyridine [0159] To a solution of (R)-l-(2,6-dichloro-3-fluorophenyl)ethanol (10 g, 47.81 mmol, Zhenjiang Radiant Pharma, Ltd. China) in THF (150 mL) was added NaH (1.40 g, 57.42 mmol) in portions at 0°C in 30 minutes. The mixture was then stirred at room temperature for 2h. A solution of 3-fluoro-2-nitropyridine(8.2 g, 57.43 mmol, Shanghai Link Chem, Ltd. China) in THF (80 mL) was added to the reaction dropwise over 20 min at 0°C. The reaction was then warmed up to rt and stirred further for 3 h. The reaction was quenched with 1 OmL of iced water, and the resulted solution was concentrated in vacuo. The residue was diluted with 150 mL of water, and the resulted aqueous mixture was extracted with EtOAc (150 mL x 3). The combined organic layers were washed with saturated NaHCC (400 mL), brine (400 mL), dried over Na2S04 and concentrated in vacuo. The residue was purified by a silica gel column chromatography (PE/EtOAc = 6/1 to 4/1) to provide the title compound as a white solid (13.4 g, 84.8%).
LC-MS (ESI, pos. ion) m/z: 331 [M + H]+.
Step2) (R)-3-(l-(2.6-dichloro-3-fluorophenyl)ethoxy)pyridin-2-amine
[0160] To a suspension of (R)-3-(l-(2,6-dichloro-3-fluorophenyl)ethoxy)-2-nitropyridine (13.4 g, 39.3 mmol) in 250 mL of EtOH was added iron powder (1 1 g, 197 mmol) in portions. The reaction was stirred at 90°C for 20 min, then was added 4 mL of IN HCl. The reaction was stirred further for 15 min, followed by the addition of another 4 mL of IN HCl. The reaction was subsequently heated at 90°C for 2 h and then cooled down to room temperature. The mixture was filtered through a Celite pad, and the pad was washed with EtOH (80 mL x 3). The filtrate was concentrated in vacuo to give the title compound as a pale brown solid (12 g, 100%).
LC-MS (ESI, pos. ion) m/z: 301 [M + H]+;
!H NMR (400 MHz, i/6-DMSO) δ (ppm): 1.75 (d, J = 6.6 Hz, 3H), 5.67 (brs, 2H), 5.97-5.92 (q, J = 6.6 Hz, 1H), 6.38-6.35 (dd, J= 7.7 Hz, 5.0 Hz, 1H), 6.61 (d, J= 7.1 Hz, 1H), 7.47-7.42 (m, 2H), 7.56-7.52 (dd, J= 7.7Hz, 5.0 Hz, 1H).
Step 3) (R)-5-bromo-3-(l-(2,6-dichloro-3-fluorophenyl)ethoxy)pyridin-2-amine
[0161] To a solution of (R)-3-(l-(2,6-dichloro-3-fluorophenyl)ethoxy)pyridin-2-amine (12 g, 39.3mmol) in 250 mL of CH3CN was added N-bromosuccinimide (9.4 g, 52.3 mmol) in portions in 20 min at 0°C. The mixture was stirred at 0°C for lh, then concentrated in vacuo. The residue was purified by a silica gel column chromatography (PE/EtOAc (v/v) = 4/1 to 3/1) to give the title compound as a pale brown solid (10 g, 68%).
LC-MS (ESI, pos. ion) m/z: 379 [M + H]+; !H NMR (400 MHz, i¾-DMSO) δ (ppm): 1.82 (d, J= 6.6 Hz, 3H), 4.82 (brs, 2H), 6.01-5.96 (q, J = 6.6 Hz, 1H), 6.83 (d, J = 1.8 Hz, 1H), 7.10 (t, J = 8.0 Hz, 1H), 7.33-7.30 (dd, J = 8.9Hz, 4.8 Hz, 2H), 7.66(dd, J= 5.0 Hz, 1.8 Hz, 1H).
Step 4) 4-(6-amino-5-((R -l-(2,6-dichloro-3-fluorophenyl ethoxy pyridin-3-yl but-3-yn-2-ol
[0162] To a mixture of (R)-5-bromo-3-(l-(2,6-dichloro-3-fluorophenyl)ethoxy)pyridin-2- amine (0.19 g, 0.5 mmol), Pd(PPh3)2Cl2 (35 mg, 0.05 mmol) and Cul (9.5 mg, 0.05 mmol) in Et3N (2.5 mL) was added but-3-yn-2-ol (53 mg 0.75 mmol). The reaction was microwaved at 90°C for 80 min, then cooled to rt and concentrated in vacuo. The residue was diluted with DCM (10 mL) and the undissolved solid was removed by filtration. The filtrate was dried over Na2S04 and concentrated in vacuo. The residue was purified by a silica gel column chromatography (DCM/MeOH (v/v) = 10/1) to give the title compound as a yellow solid (0.1 g, 54%).
LC-MS (ESI, pos. ion) m/z: 369 [M + H]+;
¾ NMR (400 MHz, CDC13) δ (ppm): 1.50-1.52 (m, 3H), 1.81 (d, J= 6.7 Hz, 3H), 4.68-4.73 (m, 1H), 5.02 (s, 2H), 5.98-6.03 (q, J = 6.6 Hz, 1H), 6.74 (d, J = 1.4 Hz, 1H), 7.05-7.09 (m, 1H), 7.26-7.32 (m, 1H), 7.77 (d, J= 1.0 Hz, 1H).
Example 2 (j?)-4-(6-amino-5-(l-(2,6-dichloro-3-fluorophenyl)ethoxy)pyridin-3-yl)-2- methylbut-3-yn-2-ol
Figure imgf000049_0001
[0163] To a mixture of (R)-5-bromo-3-(l-(2,6-dichloro-3-fluorophenyl)ethoxy)pyridine-2- amine (0.19 g, 0.5 mmol), Pd(PPh3)2Cl2 (35 mg, 0.05 mmol) and Cul (9.5 mg, 0.05 mmol) in Et3N (2.5 mL) was added 2-methylbut-3-yn-2-ol (63 mg, 0.75 mmol). The reaction was microwaved at 90°C for 1 h, then cooled to rt and concentrated in vacuo. The residue was diluted with DCM (10 mL) and the undissolved solid was removed by filtration. The filtrate was dried over Na2S04 and concentrated in vacuo. The residue was purified by a silica gel column chromatography (DCM/MeOH/Et3N (v/v/v) = 1000/50/1) to give the title compound as a white solid (155 mg, 81%).
LC-MS (ESI, pos. ion) m/z: 384 [M + H]+; !H NMR (400 MHz, CDC13) δ (ppm): 1.63 (s, 6H), 1.80 (d, J = 6.4 Hz, 3H), 2.46 (1H), 4.97 (s, 2H), 6.01 (q, J= 6.4 Hz, 1H), 6.74 (d, J = 1.56 Hz, 1H), 7.07 (m, 1H), 7.29 (m, 1H), 7.74 (s, 1H);
13C NMR (100 MHz, CDC13) δ (ppm): 18.8, 29.7, 31.5, 65.5, 76.7, 93.8, 108.7, 116.8, 1 19.3, 128.9, 130.0, 136.8, 138.8, 143.0, 150.2.
Example 3 j?)-5-mH-pyrazol-4-yl)ethvnyl)-3- i- 2,6-dichloro-3-fluorophenyl)ethoxy) pyridin-2-amine
Figure imgf000050_0001
Step 1) 4-((trimethylsilyl)ethvnyl)-lH-pyrazole
[0164] To a mixture of 4-iodo-lH-pyrazole (2.91 g, 15 mmol), Pd(PPh3)2Cl2 (1 g, 1.5 mmol), Cul (286 mg, 1.5 mmol) and Et3N (6 mL) in EtOH (20 mL) was added trimethylsilylacetylene (3.2 mL, 22.5 mmol). The reaction was stirred at 70°C for 4 h, then cooled to rt and concentrated in vacuo. The residue was diluted with EtOAc (20 mL) and the undissolved solid was removed by filtration. The filtrate was dried over Na2S04 and concentrated in vacuo. The residue was purified by a silica gel column chromatography (PE/EtOAc (v/v) = 5/1) to give the title compound as a white solid (1.3 g, 53%).
LC-MS (ESI, pos. ion) m/z: 165 [M + H]+;
¾ NMR (400 MHz, CDC13) δ (ppm): 0.24 (s, 9H), 7.76 (s, 2H), 11.43 (br, 1H).
Step 2) 4-ethynyl-lH-pyrazole
[0165] To a mixture of 4-((trimethylsilyl)ethynyl)-lH-pyrazole (1 g, 6.1 mmol) in THF (12 mL) and ¾0 (2 mL) was added K2CO3 (1 g, 12.2 mmol) solid. The mixture was stirred at rt for 2 h, then concentrated in vacuo. The residue was used in the next step without purification.
LC-MS (ESI, pos. ion) m/z: 93 [M + H]+;
¾ NMR (400 MHz, CDC13) δ (ppm): 3.04 (s, 1H), 7.75 (s, 2H), 10.85 (br, 1H);
Step 3) (R)-5-((lH-pyrazol-4-yl)ethvnyl)-3-(l-(2,6-dichloro-3-fluorophenyl)ethoxy)pyridin-2- amine [0166] To a mixture of (R)-5-bromo-3-(l-(2,6-dichloro-3-fluorophenyl)ethoxy)pyridin-2- amine (695 mg, 1.83 mmol), Pd(PPh3)2Cl2 (128 mg, 0.18 mmol) and Cul (35 mg, 0.18 mmol) in EtOH (12 mL) and dioxane (12 mL) was added DIPEA (4 mL) and 4-ethynyl- lH-pyrazole (168 mg, 1.83 mmol). The reaction was heated at 80°C for 16 h, then cooled to rt and concentrated in vacuo. The residue was diluted with DCM (25 mL) and the undissolved solid was removed by filtration. The filtrate was dried over a2S04 and concentrated in vacuo. The residue was purified by a silica gel column chromatography (DCM/MeOH/Et3N (v/v/v) = 1000:50: 1) to give the title compound as a white solid (0.25 g, 35%).
LC-MS (ESI, pos. ion) m/z: 393 [M + H]+;
!H NMR (400 MHz, CDC13) δ (ppm): 1.84 (d, J= 6.8 Hz, 1H), 6.09 (q, J= 6.8 Hz, 1H), 6.73 (d, J = 1.6 Hz, 1H), 7.25 (m, 1H), 7.46 (m, 1H), 7.60 (d, J = 1.68 Hz, 1H), 7.62 (s, 1H), 7.83 (s, 1H);
13C NMR (100 MHz, i/6-DMSO) δ (ppm): 18.7, 72.2, 80.8, 87.3, 101.2, 106.8, 117.4, 1 18.1, 120.8, 128.5, 130.5, 136.5, 137.8, 142.5, 150.8, 155.5, 158.0.
Example 4 5- 6-3ΐηίηο-5- /?)-1- 2,6-(ϋ€ΜθΓθ-3-ΑυοΓθρΗ6ην1)6ίΗοχν)ρνΓί(ϋη-3-ν1)-1,1,1- trifluoro-2-methylpent-4-vn-2-ol
Figure imgf000051_0001
Step 1) l. l . l-trifluoro-2-methylpent-4-yn-2-ol
[0167] A mixture of aluminum foil (0.34 g, 12.6 mmol) and mercuric chloride (5 mg, 0.02 mmol) in THF (6 mL) was vigorously stirred at rt for 1 h. Propargyl bromide (1.5 g, 12.6 mmol) was then added dropwise over 30 min, and the reaction was stirred further at 40°C for 2 h, then at rt for 3 h. The resulted mixture was cooled down to -78 °C, followed by the addition of a solution of 1, 1,1-trifluoroacetone (0.24 g, 2.09 mmol) in Et20 (20 mL). The mixture was warmed up to rt and stirred further for 16 h. The reaction was quenched with water (30 mL), and the aqueous phase was extracted with Et20 (40 mL x 3). The combined organic phases were washed with brine (100 mL x 2), dried over anhydrous Na2S04, and concentrated in vacuo below 5 °C to give the title compound as a light-yellow solid (0.46 g, > 100%). The crude product was used for the next reaction without further purification. GC-MS (EI): 152.0, 137.0, 126.8, 113.0.
Step 2) 5-(6-amino-5-((R)-l-(2,6-dichloro-3-fluorophenyl)ethoxy)pyridin-3-yl)-l J -trifmoro-2- methylpent-4-yn-2-ol
[0168] To a suspension of (R)-5-bromo-3-(l-(2,6-dichloro-3-fluorophenyl)ethoxy)pyridin-2- amine (0.3 g, 0.79 mmol), Pd(PPh3)2Cl2 (27 mg, 0.04 mmol) and Cul (7 mg, 0.04 mmol) in DMF (5 mL) was added a solution of l, l, l-trifluoro-2-methylpent-4-yn-2-ol (0.32 g, 2.1 mmol) in DMF (5 mL) followed by Et3N (2.20 mL, 15.8 mmol). The reaction was heated at 75°C for 16 h, then cooled down to rt and diluted with DCM (150 mL). The mixture was washed with water (100 mL x 2) and brine (100 mL x 2), dried over anhydrous a2S04, and concentrated in vacuo. The residue was purified by a silica gel column chromatography (PE/EA (v/v) = 2/1) to give the title compound as a light-yellow solid (0.16 g, 45%).
LC-MS (ESI, pos. ion) m/z: 451.1 [M + H]+;
!H NMR (400 MHz, CDC13) δ (ppm): 1.49 (s, 3H), 1.81 (d, J = 6.68 Hz, 3H), 2.72 (d, J = 17.00 Hz, 1H), 2.89 (d, J = 17.02 Hz, 1H), 5.01 (s, 2H), 5.97-6.03 (q, J = 6.68 Hz, 1H), 6.72 (m, 1H), 7.10 (t, J= 8.48 Hz, 1H), 7.27-7.52 (dd, J= 8.92 Hz, 4.84 Hz, 1H), 7.72 (m, 1H).
Example 5 4- 6-3ΐηίηο-5- /?)-1- 2,6-(ϋ€ΜθΓθ-3-ΑυοΓθρΗ6ην1)6ίΗοχν)ρνΓί(ϋη-3-ν1)-1,1,1- trifluoro-2-methylbut-3-vn-2-ol
Figure imgf000052_0001
Step 1) L L l-trifluoro-2-methyl-4-(trimethylsilyl)but-3-yn-2-ol
[0169] To a suspension of ethynyltrimethylsilane (1 g, 10.2 mmol) in Et20 (10 mL) at -78°C was added n-BuLi (4.1 1 mL, 10.3 mmol, 2.5 M in hexane) via syringe over 30 min. The mixture was stirred at -78 °C for 30 min followed by the addition of a solution of 1, 1,1-trifluoroacetone (1.37 mL, 10.2 mmol) in Et20 (10 mL) over 30 min. The reaction was stirred at rt for 50 min, then quenched with saturated NH4C1 aqueous solution (20 mL). The organic phase was separated and the aqueous phase was extracted with Et20 (30 mL x 4). The combined organic phases were washed with brine (50 mL), dried over anhydrous Na2S04, and concentrated in vacuo below 10 °C to give the title compound as light-yellow oil (2.1 g, 98%). GC-MS (EI): 209.0, 137.0, 1 13.0.
Step 2) U J-trifluoro-2-methylbut-3-vn-2-ol
[0170] To a solution of l, l, l-trifluoro-2-methyl-4-(trimethylsilyl)but-3-yn-2-ol (0.8 g, 3.8 mmol) in MeOH (10 mL) was added KOH (1.28 g, 22.8 mmol) solid in portions. The reaction was stirred at rt for 16 h, then quenched with water (30 mL). The mixture was extracted with Et20 (40 mL x 4). The combined organic phases were washed with brine (100 mL x 2), dried over anhydrous a2S04, and concentrated in vacuo below 10°C. The residue was dried in vacuo at 5 °C to give the title compound as colorless oil (0.31 g, 59%).
GC-MS (EI): 137.0, 1 13.0.
Step 3) 4-(6-amino-5-((R)-l-(2.6-dichloro-3-fluorophenyl)ethoxy)pyridin-3-yl)-l. l .l-trifluoro-2- methylbut-3-yn-2-ol
[0171] To a solution of (R)-5-bromo-3-(l-(2,6-dichloro-3-fluorophenyl)ethoxy)pyridin-2- amine (0.2 g, 0.53 mmol) in DCM (10 mL) was added Pd(PPh3)2Cl2 (18 mg, 0.03 mmol), Cul (5 mg, 0.03 mmol), l, l,l-trifluoro-2-methylbut-3-yn-2-ol (0.31 g, 2.37 mmol) and Et3N (1.47 mL, 10.5 mmol) under N2 atmosphere. The reaction was stirred at 75°C for 2.5 h, then cooled to rt and diluted with DCM (150 mL). The resulted mixture was washed with brine (200 mL x 3), dried over anhydrous Na2S04, and concentrated in vacuo. The residue was purified by a prepared-HPLC to give the title compound as a yellow solid (30 mg, 13%).
LC-MS (ESI, pos. ion) m/z: 437.1 [M + H]+;
!H NMR (400 MHz, CDC13) δ (ppm): 1.22 (s, 3H), 1.75 (d, J = 6.56 Hz, 3H), 5.96-6.02 (q, J = 6.68 Hz, 1H), 6.29 (s, 2H), 6.59 (s, 1H), 7.50 (t, J = 8.60 Hz, 1H), 7.54-7.59 (dd, J = 8.92 Hz, 5.00 Hz, 1H), 7.62 (s, 1H).
Example 6 (j?)-4-((6-amino-5-(l-(2,6-dichloro-3-fluorophenyl)ethoxy)pyridin-3-yl)ethvnyl) piperidin-4-ol
Figure imgf000053_0001
Step 1) 1 -(ter?-butoxycarbonyl)-4-((trimethylsilyl)ethvnyl)piperidin-4-ol [0172] To a mixture of trimethylsilylacetylene (1.7 mL, 12 mmol) in anhydrous THF (20 mL) was added a solution of w-BuLi (4.8 mL, 12 mmol) slowly at -40 °C under 2 atomasphere. The mixture was stirred at -40 °C for 1 h, then cooled to -78 °C. A solution of \-(tert- butoxycarbonyl) piperidin-4-one (2 g, 10 mmol) in anhydrous THF (10 mL) was added to the reaction via a cannula. The reaction was stirred at -78°C for 1 h, then warmed up to rt and stirred further for 72 h. The reaction was quenched with saturated aq. Solution of NH4CI (15 mL) and H2O (50 mL). The resulted mixture was extracted with EtOAc (50 mL x 3). The combined organic phases were washed with H20 (50 mL), followed by brine (50 mL), dried over a2S04 and concentrated in vacuo. The residue was purified by a silica gel column chromatography (PE/EtOAc (v/v) = 5/1) to give the title compound as a white solid (1.93 g, 65%).
!H NMR (400 MHz, CDC13) δ (ppm): 0.17 (s, 9H), 1.46 (s, 9H), 1.68 (m, 2H), 1.87 (m, 2H), 2.19 (s, 1H), 3.22 (m, 2H), 3.79 (br, 2H) ;
13C MPv (100 MHz, CDC13) δ (ppm): 28.5, 39.0 , 67.2, 79.6, 89.9, 107.7, 154.7.
Step 2) l-(ter/-butoxycarbonyl)-4-ethvnylpiperidin-4-ol
[0173] To a mixture of l-(/er/-butoxycarbonyl)-4-((trimethylsilyl)ethynyl)piperidin-4-ol (1.4 g, 6.22 mmol) in MeOH (12 mL) and H20 (2 mL) was added K2C03 (3.4 g, 24.88 mmol). The reaction was stirred at rt for 2 h. The mixture was concentrated in vacuo and the residue was purified by a silica gel column chromatography (PE/EtOAc (v/v) = 5/1) to give the title compound as a white solid (0.95 g, 90%).
!H NMR (400 MHz, CDC13) δ (ppm): 1.46 (s, 9H), 1.68 (m, 2H), 1.87 (m, 2H), 2.53 (s, 1H), 3.27 (m, 2H), 3.50 (s, 1H), 3.75 (br, 2H);
13C NMR (100 MHz, CDC13) δ (ppm): 28.6, 38.9, 40.8, 66.5, 73.1, 79.9, 86.5, 154.8.
Step 3) (R)-4-((6-amino-5-(l-(2,6-dichloro-3-fluorophenyl)ethoxy)pyridin-3-yl)ethvnyl)-l-(ter?- butoxycarbonyl)piperidin-4-ol
[0174] To a mixture of (R)-5-bromo-3-(l-(2,6-dichloro-3-fluorophenyl)ethoxy)pyridine-2- amine (695 mg, 1.83 mmol), Pd(PPh3)2Cl2 (128 mg, 0.18 mmol), Cul (35 mg, 0.18 mmol), K2CO3 (505 mg, 3.66 mmol) in dioxane (12 mL) was added DIPEA (4 mL) and \-(tert- butoxycarbonyl)-4-ethynylpiperidin-4-ol (412 mg, 1.83 mmol). The reaction was stirred at 90°C for 16 h, then cooled to rt. The mixture was concentrated in vacuo and the residue was diluted with DCM (20 mL). The undissolved solid was removed by filtration and the filtrate was dried over Na2S04 and concentrated in vacuo. The residue was purified by a silica gel column chromatography (PE/EtOAc (v/v) = 1/1) to give the title compound as a light yellow solid (0.15 g, 16%).
LC-MS (ESI, pos. ion) m/z: 524 [M + H]+;
!H NMR (400 MHz, CDC13 ) δ (ppm): 1.48 (s, 9H), 1.76 (m, 2H), 1.81 (d, J = 6.8 Hz, 1H), 1.92 (m, 2H), 3.25 (m, 2H), 3.82 (br, 2H), 5.04 (br, 2H), 6.73 (s, 1H), 6.99 (q, J = 6.8 Hz, 1H), 7.30 (m, 1H), 7.74 (m, 1H), 7.79 (br, 1H) ;
13C NMR (100 MHz, CDC13) δ (ppm): 14.1, 28.6, 39.1, 67.1, 72.6, 82.9, 91.5, 116.8, 1 19.2, 122.0, 128.9, 130.0, 136.6, 154.8.
Step 4) (R)-4-((6-amino-5-( 1 -(2.6-dichloro-3 -fluorophenyl)ethoxy)pyridin-3 -yPethynyl) piperidin-4-ol
[0175] To a mixture of (R)-4-((6-amino-5-(l-(2,6-dichloro-3-fluorophenyl)ethoxy)pyridin-3- yl) ethynyl)-l-(tert-butoxycarbonyl)piperidin-4-ol (0.15 g, 0.29 mmol) in DCM (4 mL) was added HCl in EtOAc (3 M, 10 mL) solution at 0°C. The reaction was stirred at rt for 4 h. The product was formed as a solid and was collected by filtration. The solid was treated with H20 (10 mL) and the pH of the resulted mixture was adjusted to 10 with 4 M NaOH aqueous solution. The mixture was extracted with EtOAc (10 mL x 3) and the combined organic phases were dried over Na2S04 and concentrated in vacuo. The residue was purified by a silica gel column chromatography (DCM/MeOH/Et3N (v/v/v) = 1000/50/1) to give the title compound as a white solid (70 mg, 56%).
LC-MS (ESI, pos. ion) m/z: 424 [M + H]+;
¾ NMR (400 MHz, CDC13) δ (ppm): 1.76 (m, 2H), 1.81 (d, J= 6.8 Hz, 1H), 1.92 (m, 2H), 2.86 (m, 2H), 3.12 (br, 2H), 4.96 (br, 2H), 5.99 (q, J= 6.8 Hz, 1H), 6.73 (s, 1H), 7.30 (m, 1H),7.74 (m, 1H), 7.76 (s, 1H);
13C NMR (100 MHz, CDC13) δ (ppm): 11.5, 40.7, 43.7, 67.6, 72.6, 82.7, 91.9, 116.8, 1 19.3, 120.2, 128.9, 136.8, 138.8, 142.9, 150.2.
Example 7 3-((6-3ηΐίηο-5-((/? -1-(2,6-(ϋ€Η1θΓθ-3-ΑυοΓορΗ6ην1 6ίΗοχν ρνΓί(ϋη-3-ν1 6ίΗνην1 tetrahydrofuran-3-ol
Figure imgf000055_0001
Step 1) S-rrtrimethylsilvDethynyDtetrahydrofuran-S-ol
[0176] To a solution of Trimethylsilylacetylene (1 g, 10.2 mmol) in THF (20 mL) was added n- BuLi (4.11 mL, 10.3 mmol) at -78°C. The mixture was stirred at -78°C for 30 min. A solution of dihydrofuran-3(2H)-one (0.87 g, 10.2 mmol) in THF (20 mL) was added to the reaction. The reaction was stirred further at -78 °C for 30 min, then at rt for 50 min. The reaction was quenched with saturated NH4C1 queous solution (20 mL). The mixture was extracted with EtOAc (50 mL x 4) and the combined organic phases were washed with brine (100 mL x 2), dried over anhydrous Na2S04, and concentrated in vacuo to give the title compound as a white solid (1.67 g, 89%). The compound was used in the next step without further purification.
!H NMR (400 MHz, i/6-DMSO) δ (ppm): 0.14 (s, 9H), 2.05-2.08 (m, 2H), 3.62-3.70 (m, 2H), 3.79-3.83 (m, 2H), 5.72 (s, 1H).
Step 2) 3-ethynyltetrahydrofuran-3-ol
[0177] To a solution of 3-((trimethylsilyl)ethynyl)tetrahydrofuran-3-ol (1.67 g, 9.06 mmol) in CH3OH (50 mL) was added KOH (1.01 g, 18.12 mmol). The reaction was stirred at rt for 2 h, then quenched with water (50 mL). The resulted mixture was extracted with EtOAc (100 mL x 3), and the combined organic phases were washed with water (100 ml x2), brine (100 mL x 2), dried over anhydrous Na2S04, and concentrated in vacuo to give the title compound as yellow oil (0.77 g, 75.9%).
LC-MS (ESI, pos. ion) m/z: 113.05 [M + H]+;
!H NMR (400 MHz, i/6-DMSO) δ (ppm): 2.06-2.10 ( m, 2H), 3.41-3.42 (m, 1H), 3.63-3.70 (m, 2H), 3.80-3.83 ( m, 2H), 5.72 (s, 1H).
Step 3) 3-((6-amino-5-((R)-l-(2,6-dichloro-3-fluorophenyl)ethoxy)pyridin-3-yl)ethvnyl) tetrahydrofuran-3 -ol
[0178] To a solution of 3-ethynyltetrahydrofuran-3-ol (0.72 g, 6.43 mmol), (R)-5-bromo-3-(l- (2,6-dichloro-3-fluorophenyl)ethoxy)pyridin-2-amine (1.88 g, 4.95 mmol), Pd(PPh3)2Cl2 (0.17 g, 0.25 mmol), Cul (47 mg, 0.25 mmol) in DMF (20 mL) was added Et3N (9.99 g) under N2 atmosphere. The reaction was stirred at 100°C for 12 h. The mixture was concentrated in vacuo and the residue was diluted with DCM (100 mL), washed with water (100 mL x 2) and brine (100 mL x 2). The organic phase was dried over anhydrous Na2S04 and concentrated in vacuo. The residue was purified by chromatography on silica gel (PE/EtOAc (v/v) = 1/1). The product was recrystallisation in PE/EtOAc (v/v = 10/1) to give the title compound as a light yellow solid (0.3 g, 15%). LC-MS (ESI, pos. ion) m/z: 412.00 [M + H] ;
!H NMR (400 MHz, i¾-DMSO) δ (ppm): 1.74-1.76 (d, 3H), 2.10-2.13 (m, 2H), 3.66-3.74 (m, 2H), 3.82-3.86 (m, 2H), 5.74 (s, 1H), 5.96-6.00 (m, 1H), 6.16 (s, 2H), 6.60-6.61 (m, 1H), 7.44- 7.49 (m, 1H), 7.56-7.57 (m, 1H), 7.59-7.60 (m, 1H).
Example 8 3- 6-3ηΐίηο-5- /?)-1- 2,6-(ϋ€Η1θΓθ-3-ΑυοΓθρΗ6ην1)6ίΗοχν)ρνΓί(ϋη-3-ν1)6ίΗνην1) pyrrolidin-3-ol
Figure imgf000057_0001
Step 1) 1 -(ferf-butoxycarbonyl)-3 -((trimethylsilyl)ethynyl)pyrrolidin-3 -ol
[0179] To a suspension of trimethylsilylacetylene (1 g, 10.2 mmol) in THF (20 mL) was added n-BuLi (4.1 1 mL, 10.3 mmol, 2.5 M in hexane) at -78 °C over 30 min. The mixture was stirred at -78 °C for 30 min. A solution of N-Boc-3-pyrrolidinone (1.89 g, 10.2 mmol) in THF (20 mL) was added to the reaction through an addition funnel over 30 min. The reaction was stirred further at rt for 50 min, then quenched with saturated NH4CI aqueous solution (8 mL). The resulted mixture was extracted with EtOAc (50 mL x 4) and the combined organic phases were washed with brine (100 mL x 2), dried over anhydrous a2S04, and concentrated in vacuo to give the title compound as a light-yellow solid (2.6 g, 90%).
¾ NMR (400 MHz, CDC13) δ (ppm): 0.17 (s, 9H), 1.45 (s, 9H), 2.10-2.24 (m, 2H), 3.44-3.67 (m, 4H).
Step 2) l-(ter?-butoxycarbonyl)-3-ethynylpyrrolidin-3-ol
[0180] To a solution of l-(?ert-butoxycarbonyl)-3-((trimethylsilyl)ethynyl)pyrrolidin-3-ol (1.50 g, 5.29 mmol) in CH3OH (30 mL) was added KOH (0.59 g, 10.6 mmol). The reaction was stirred at rt for 2 h, and then quenched with water (50 mL). The resulted mixture wasd extracted with EtOAc (60 mL x 5) and the combined organic phases were washed with brine (100 mL x 2), dried over anhydrous a2S04, and concentrated in vacuo. The residue was purified by a silica gel colummn chromatography (PE/EtOAc (v/v) = 3/1) to afford the title compound as a white solid (0.86 g, 76%).
LC-MS (ESI, pos. ion) m/z: 156.2 [M + H -56]+; !H NMR (400 MHz, CDC13) δ (ppm): 1.45 (s, 9H), 2.18 (m, 2H), 2.58 (m, 1H), 2.54 (s, 1H), 3.47-3.72 (m, 4H).
Step 3) 3-((6-amino-5-((R -l-(2,6-dichloro-3-fluorophenyl ethoxy pyridin-3-yl ethvnyl -l-(ter?- butoxycarbonyl)pyrrolidin-3 -ol
[0181] To a solution of (R)-5-bromo-3-(l-(2,6-dichloro-3-fluorophenyl)ethoxy)pyridin-2- amine (0.30 g, 0.79 mmol) in DMF (30 mL) was added Pd(PPh3)2Cl2 (28 mg, 0.04 mmol), Cul (7 mg, 0.04 mmol), l-( tert-butoxycarbonyl)-3-ethynylpyrrolidin-3-ol (0.17 g, 0.79 mmol) and Et3 (2.2 mL, 15.8 mmol) under 2 atmosphere. The reaction was heated at 75°C for 16 h, then cooled down to rt and diluted with DCM (300 mL). The mixture was washed with brine (100 mL x 3), dried over anhydrous a2S04, and concentrated in vacuo. The residue was purified by a silica gel colummn chromatography (PE/EtOAc (v/v) = 2/1 to 1/1) to give the title compound as brown-red oil (0.25 g, 62%).
LC-MS (ESI, pos. ion) m/z: 510.0 [M + H]+;
!H NMR (400 MHz, CDC13) δ (ppm): 1.46 (s, 9H), 1.80 (d, J = 6.68 Hz, 3H), 2.20 (m, 2H), 3.47-3.62 (m, 4H), 5.27 (s, 2H), 5.97-6.04 (q, J = 6.72 Hz, 1H), 6.73 (s, 1H), 7.06-7.12 (t, J = 8.32 Hz, 1H), 7.28-7.34 (dd, J = 8.88 Hz, 4.80 Hz, 1H), 7.78 (s, 1H).
Step 4) 3-((6-amino-5-((R)-l-(2.6-dichloro-3-fluorophenyl)ethoxy)pyridin-3-yl)ethynyl) pyrrolidin-3-ol
[0182] To a suspension of 3-((6-amino-5-((R)-l-(2,6-dichloro-3-fluorophenyl)ethoxy)pyridin- 3-yl)ethynyl)-l-(tert-butoxycarbonyl)pyrrolidin-3-ol (0.25 g, 0.49 mmol) in DCM (20 mL) was added a saturated solution of HC1 in EtOAc (5 mL). The reaction mixture was stirred at rt for 16 h, then concentrated in vacuo. The residue was diluted with water (5 mL), and the pH of the resulting solution was adjusted to pH = 12 with saturated Na2C03 aqueous solution. The mixture was extracted with DCM/MeOH (v/v = 10/1) (40 mL x 5) and the combined organic phases were washed with brine (100 mL x 2), dried over anhydrous a2S04, and concentrated in vacuo. The residue was purified by prepared-TLC (DCM/MeOH (v/v) =10/1) to give the titile compound as a yellow solid (0.1 1 g, 54%).
LC-MS (ESI, pos. ion) m/z: 410.0 [M + H]+;
*H NMR (400 MHz, CDCI3+CD3OD) δ (ppm): 1.83 (d, J = 6.68 Hz, 3H), 2.25-2.48 (m, 2H), 3.43-3.58 (m, 4H), 5.88-6.06 (q, J = 6.68 Hz, 1H), 6.72 (d, J = 1.52 Hz, 1H), 7.17 (t, J= 8.80 Hz, 1H), 7.32-7.38 (dd, J= 8.92 Hz, 4.80 Hz, 1H), 7.65 (d, J= 1.56 Hz, 1H). Example 9 3-(( ?)-l-(2,6-dichloro-3-fluorophenyl)ethoxy)-5-(3-morpholinobut-l-vn-l-yl) pyridin-2-amine
Figure imgf000059_0001
Step 1) (R)-5-bromo-N.N-bis(te^butoxycarbonyl)-3-(l-(2.6-dichloro-3-fluorophenyl)ethoxy) pyridin-2-amine
[0183] To a mixture of (R)-5-bromo-3-(l-(2,6-dichloro-3-fluorophenyl)ethoxy)pyridine-2- amine (2.52 g, 6.63 mmol), DMAP (809 mg, 6.63 mmol) in THF(66 mL) was added (Boc)20 (4.7 mL, 19.89 mmol) and Et3N (2.8 mL, 19.89 mmol). The reaction was stirred at 68°C for 1 1 h, then quenched with saturated NaHC(¾ aqueous solution (50 mL). The mixture was extracted with EtOAc (50 mL x 3) and the combined organic phases were washed with water (50 mL), saturated NaHCC^ aqueous solution (50 mL) and brine (50 mL). The solution was dried over anhydrous Na2S04 and concentrated in vacuo. The residue was purified by a silica gel column chromatography (PE/EtOAc (v/v) = 4/1) to give the title compound as a yellow solid (4.4 g, 1 14%).
LC-MS (ESI, pos. ion) m/z: 603 [M + Na .
Step 2) 4-(6-(bis(ter/-butoxycarbonyl)amino)-5-((R)-l-(2,6-dichloro-3-fluorophenyl)ethoxy) pyridin-3 -yl)but-3 -yn-2 - ol
[0184] To a mixture of (R)-5-bromo-N,N-bis(/er/-butoxycarbonyl)-3-(l-(2,6-dichloro-3- fluorophenyl) ethoxy)pyridin-2-amine (0.25 g, 0.43 mmol), Pd(PPh3)2Cl2 (30 mg, 0.04 mmol) and Cul (8.1 mg, 0.04 mmol) in Et3N (2.5 mL) was added but-3-yn-2-ol (0.05 mL, 0.65 mmol). The reaction was microwaved at 90°C for 80 min, then cooled to rt. The mixture was concentrated in vacuo and the residue was diluted with DCM (10 mL). The undissolved solid was removed through filtration, and the filtrate was dried over a2S04 and concentrated in vacuo. The residue was purified by a silica gel column chromatography (PE/EtOAc (v/v) = 2/1) to give the title compound as a yellow solid (201 mg, 82%).
!H NMR (400 MHz, CDC13) δ (ppm): 1.37-1.41 (m, 18H), 1.55 (d, J = 6.6 Hz, 3H), 1.78 (d, J = 6.6 Hz, 3H), 4.72-4.78 (m, 1H), 5.97-5.02 (q, J = 6.7 Hz, 1H), 7.06-7.12 (m, 2H), 7.27-7.32 (m, 1H), 8.12-8.13 (m, 1H). Step 3) 4-(6-(bis(te^butoxycarbonyl amino -5-('('R -l-('2,6-dichloro-3-fluorophenyl ethoxy pyridin-3-yl)but-3-yn-2-yl methanesulfonate
[0185] To a solution of 4-(6-(bis(?ert-butoxycarbonyl)amino)-5-((R)-l-(2,6-dichloro-3- fluorophenyl) ethoxy)pyridin-3-yl)but-3-yn-2-ol (244 mg, 0.43 mmol) in DCM (2.4 mL) was added Et3N (0.14 g, 1.38 mmol) and methanesulfonyl chloride (0.05 mL, 0.65 mmol) at -20°C under 2 atomasphere. The reaction was stirred at -20°C for 3 h, then at rt for 3 h. The mixture was quenched with saturated NaHCC aqueous solution (10 mL) and extracted with DCM (10 mL x 3). The combined organic phases were dried over Na2S04 and concentrated in vacuo to give the title compound as yellow oil (243 mg, 99%). The compound was used in the next step without purification.
LC-MS (ESI, pos. ion) m/z: 647 [M + H]+.
Step 4) N.N-bis(ferf-butoxycarbonyl)-3 -((R)- 1 -(2.6-dichloro-3 -fluorophenyl)ethoxy)-5-(3 - morpholinobut- 1 -yn- 1 -yl)pyridin-2-amine
[0186] To a mixture of 4-(6-(bis(?ert-butoxycarbonyl)amino)-5-((R)-l-(2,6-dichloro-3- fluorophenyl) ethoxy)pyridin-3-yl)but-3-yn-2-yl methanesulfonate (0.28 g, 0.43 mmol) in CH3CN (5 mL) was added morpholine (0.13 g, 1.5 mmol) at rt. The reaction was heated to 60°C for 16 h, then cooled to rt. The reaction mixture was diluted with DCM (50 mL) and H2O (50 mL). The organic phase was separated, dried over Na2S04 and concentrated in vacuo. The residue was purified by a silica gel column chromatography (PE/EtOAc (v/v) = 2/1) to give the title compound as a yellow solid (219 mg, 80%).
LC-MS (ESI, pos. ion) m/z: 638 [M + H]+;
1H NMR (400 MHz, CDC13) δ (ppm): 1.35-1.42 (m, 18H), 1.41-1.44 (m, 3H), 1.81(d, J= 6.7 Hz, 3H), 2.54 -2.56 (m, 2H), 2.71-2.74 (m, 2H), 3.63-3.69 (m, 1H), 3.78-3.80 (m, 4H), 5.98-6.03 (q, J= 6.7 Hz, 1H), 7.06-7.12 (m, 2H), 7.29-7.34 (m, 1H), 8.09 (s, 1H).
Step 5) 3 -((R)- 1 -(2 ,6-dichloro-3 -fluorophenyl)ethoxy)-5 -(3 -morpholinobut- 1 -yn- 1 -yl)pyridin-2- amine
[0187] To a mixture of N,N-bis(?ert-butoxycarbonyl)-3-((R)-l-(2,6-dichloro-3- fluorophenyl)ethoxy)-5-(3-morpholinobut-l-yn-l-yl)pyridin-2-amine (219 mg, 0.34 mmol) in DCM (5 mL) was added HC1 in EtOAc (3 M, 1 mL). The reaction was stirred at rt for 18 h, then quenched with saturated K2CO3 aqueous solution (30 mL). The mixture was extracted with EtOAc (50 mL x 3) and the combined organic phases were washed with water (50 mL) and brine (50 mL), dried over Na2S04 and concentrated in vacuo. The residue was purified by a silica gel column chromatography (PE/EtOAc (v/v) = 1/1) to give the title compound as a yellow solid (134 mg, 90%).
LC-MS (ESI, pos. ion) m/z: 439 [M + H]+;
¾ NMR (400 MHz, CDC13) δ (ppm): 1.37-1.39 (m, 3H), 1.82 (d, J= 6.7 Hz, 3H), 2.51-2.54 (m, 2H), 2.67-2.72 (m, 2H), 3.58-3.64 (m, 1H), 3.71-3.80 (m, 4H), 5.03 (s, 2H), 5.99-6.04 (q, J= 6.6 Hz, 1H), 6.76 (s, 1H), 7.04-7.08 (m, 1H), 7.28-7.32 (m, 1H), 7.72 (d, J= 1.0 Hz, 1H).
Example 10 S- g l- i^-dichloro-S-fluorophenvDethoxy^-S- S- piperazin-l-vDbut-l-yn-l- yl)pyridin-2-amine
Figure imgf000061_0001
Step 1) NN-bis(ter/-butoxycarbonyl)-3-((R)-l-(2,6-dichloro-3-fluorophenyl)ethoxy)-5-(3- (piperazin- 1 -yPbut- 1 -yn- 1 -yl)pyridin-2 -amine
[0188] To a mixture of 4-(6-(bis(/er/-butoxycarbonyl)amino)-5-((R)-l-(2,6-dichloro-3- fluorophenyl) ethoxy)pyridin-3-yl)but-3-yn-2-yl methanesulfonate (0.39 g, 0.6 mmol) in CH3CN (7 mL) was added piperazine (0.52 g, 6 mmol) at rt. The mixture was heated at 60°C for 16 h, then cooled to rt and concentrated in vacuo. The residue was diluted with DCM (50 mL). The resulting mixture was washed with H20 (50 mL), dried over Na2S04 and concentrated in vacuo to give the title compound as a yellow solid (0.23 g, 61%). The compound was used in the next step without purification.
LC-MS (ESI, pos. ion) m/z: 637 [M + H]+.
Step 2) 3 -((R)- 1 -(2 ,6-dichloro-3 -fluorophenyl)ethoxy)-5-(3 -(piperazin- 1 -yl)but- 1 -yn- 1 -yl) pyridin-2-amine
[0189] To a mixture of N,N-bis(?ert-butoxycarbonyl)-3-((R)-l-(2,6-dichloro-3- fluorophenyl)ethoxy)-5-(3-(piperazin-l-yl)but-l-yn-l-yl)pyridin-2-amine (0.2 g, 0.3 mmol) in DCM (5 mL) was added HC1 in EtOAc (3 M, 1 mL). The reaction was stirred at rt for 18 h, then quenched with saturated K2CO3 solution (30 mL). The resulting mixture was extracted with EtOAc (50 mL x 3), and the combined organic phases were washed with water (50 mL), brine (50 mL), dried over Na2S04, and concentrated in vacuo. The residue was purified by a silica gel column chromatography (DMC/MeOH/Et3N (v/v/v) = 200/20/1) to give the title compound as a yellow solid (123 mg, 90%).
LC-MS (ESI, pos. ion) m/z: 219 [M/2 + H]+;
JH NMR (400 MHz, CDC13) δ (ppm): 1.37-1.39 (m, 3H), 1.81 (d, J= 6.64 Hz, 3H), 2.50-2.53 (m, 2H), 2.67-2.71 (m, 2H), 2.91-2.98 (m, 4H), 3.60-3.64 (m, 1H), 4.96 (s, 2H), 5.99-6.04 (q, J = 6.64 Hz, 1H), 6.77 (d, J = 1.5 Hz, 1H), 7.09-7.05 (m, 1H), 7.32-7.26 (m, 1H), 7.73 (d, J= 1.6 Hz, 1H).
Example 11 3-((/?)-l-(2,6-dichloro-3-fluorophenyl)ethoxy)-5-(3-(4-methylpiperazin-l- yl)but-l-yn-l-yl)pyridin-2-amine
Figure imgf000062_0001
Step 1) N.N-bis(ter?-butoxycarbonyl)-3-((R)-l-(2.6-dichloro-3-fluorophenyl)ethoxy)-5-(3-(4- methylpiperazin- 1 -vDbut- 1 -yn- 1 -yl)pyridin-2-amine
[0190] To a mixture of 4-(6-(bis(?ert-butoxycarbonyl)amino)-5-((R)-l-(2,6-dichloro-3- fluorophenyl) ethoxy)pyridin-3-yl)but-3-yn-2-yl methanesulfonate (0.35 g, 0.55 mmol) in CH3CN (4 mL) was added 1 -methylpiperazine (0.22 g, 2.18 mmol) at rt. The reaction was stirred at 60°C for 16 h, then cooled to rt and concentrated in vacuo. The residue was diluted with DCM (50 mL). The resulted mixture was washed with H20 (50 mL), dried over Na2S04 and concentrated in vacuo. The residue was purified by a silica gel column chromatography (PE/EtAOc (v/v) = 2/1) to give the title compound as a yellow solid (0.31 g, 94%).
LC-MS (ESI, pos. ion) m/z: 651 [M + H]+.
Step 2) 3 -((R)- 1 -(2,6-dichloro-3 -fluorophenyl)ethoxy)-5 -(3 -(4-methylpiperazin- 1 -yl)but- 1 -yn- 1 - vDpyridin-2-amine
[0191] To a mixture of N,N-bis(?ert-butoxycarbonyl)-3-((R)-l-(2,6-dichloro-3- fluorophenyl)ethoxy)-5-(3-(4-methylpiperazin-l-yl)but-l-yn-l-yl)pyridin-2-amine (0.31 g, 0.47 mmol) in DCM (5 mL) was added HC1 in EtOAc (3 M, 1 mL). The reaction was stirred at rt for 18 h, then quenched with saturated K2CO3 solution (30 mL). The resulted mixture was extracted with EtOAc (50 mL x 3), and the combined organic phases were washed with water (50 mL) followed by brine (50 mL), dried over Na2S04 and concentrated in vacuo. The residue was purified by a silica gel column chromatography (PE/EtOAc (v/v) = 1/1) to give the title compound as a yellow solid (182 mg, 91%).
LC-MS (ESI, pos. ion) m/z: 451 [M + H]+;
¾ NMR (400 MHz, CDC13) δ (ppm): 1.38-1.40 (dd, J= 7.0 Hz, 1.0 Hz, 3H), 1.80 (d, J= 6.7 Hz, 3H), 2.30 (s, 3H), 2.50-2.58 (m, 6H), 2.74-2.78 (m, 2H), 3.62-3.67 (q, J = 7.0 Hz, 1H), 4.91 (s, 2H), 5.98-6.03 (q, J= 6.64 Hz, 1H), 6.77 (s, 1H), 7.08-7.04 (m, 1H), 7.32-7.26 (m, 1H), 7.72 (d, J= 1.6 Hz, 1H).
Example 12 5-(4-(6-amino-5-((JR)-l-(2,6-dichloro-3-fluorophenyl)ethoxy)pyridin-3-yl)but-3- vn-2-vD-5-azaspiro [2.41 heptan-7-amine
Figure imgf000063_0001
Step 1) (£)-5-benzyl-7-(hydroxyimino)-5-azaspiro[2.41heptan-4-one
[0192] To a mixture of 5-benzyl-5-azaspiro[2.4]heptane-4,7-dione (6 g, 28 mmol) and Et3 (6 mL, 42 mmol) in anhydrous EtOH (144 mL) was added hydroxylammonium chloride (2.92 g, 42 mmol). The reaction was stirred at rt for 24 h, and concentrated in vacuo. The residue was diluted with water (140 mL). The resulted mixture was stirred for 2 h, and then filtered. The solid was dried in vacuo at 45°C for 24 h to give the title compound as a pale yellow solid (5.9 g, 91%).
LC-MS (ESI, pos. ion) m/z: 231.0 [M + H]+;
!H NMR (400 MHz, i/6-DMSO) δ (ppm): 1.22 (d, J= 3.6 Hz, 2H), 1.35 (d, J= 3.6 Hz, 2H), 4.02 (s, 2H), 4.52 (s, 2H), 7.24-7.32 (m, 3H), 7.35-7.39 (m, 2H).
Step 2) 5-benzyl-5-azaspiror2.41heptan-7-amine
[0193] To a mixture of LiAlH4 (0.87 g, 22.8 mmol) in THF (10 mL) was added a solution of (£)-5-benzyl-7-(hydroxyimino)-5-azaspiro[2.4]heptan-4-one (1.31 g, 5.7 mmol) in THF (20 mL) at 0°C. The reaction was stirred at rt for 30 min, then at 60°C for 5 h. The reaction was cooled to 0°C and quenched with EtOAc (40 mL) and ¾0 (20 mL) slowly. The resulting mixture was filtered and the filtrate was concentrated in vacuo. The residue was diluted with EtOAc (40 mL). The resulting solution was washed with H20 (20 mL), dried over anhydrous Na2S04 and concentrated in vacuo to give the title compound as orange oil (1.24 g, 99%).
LC-MS (ESI, pos. ion) m/z: 203.1 [M + H]+.
Step 3) 5-benzyl-N-(ter?-butoxycarbonyl)-5-azaspiror2.41heptan-7-amine
[0194] To a mixture of 5-benzyl-5-azaspiro[2.4]heptan-7-amine(1.2 g, 6 mmol) and Et3 (2.1 mL, 15 mmol) in THF (20 mL) was added (Boc)20 (2.1 mL, 9.0 mmol). The reaction was stirred at rt for 2 h, and then concentrated in vacuo. The residue was diluted with DCM (120 mL), and washed with brine (30 mL x 2). The solution was dried over anhydrous Na2S04 and concentrated in vacuo. The residue was recrystallized in PE to give the title compound as a white solid (1.85 g, 100%).
LC-MS (ESI, pos. ion) m/z: 303.1 [M + H]+;
!H NMR (400 MHz, CDC13) δ (ppm): 0.43-0.45 (m, 2H), 0.58-0.80 (m, 2H), 1.35 (s, 9H), 2.34 (d, J = 8.8 Hz, 1H), 2.66-2.69 (m, 2H), 2.88-2.95 (m, 1H), 3.60 (dd, J = 28.4 Hz, 12.8 Hz, 2H), 3.88 (s, 1H), 4.97 (d, J= 7.6 Hz, 1H), 7.26 (s, 2H), 7.30 (s, 3H).
Step 4) N-(ter/-butoxycarbonyl)-5-azaspiro[2.41heptan-7-amine
[0195] To a mixture of 5-benzyl-N-(/er/-butoxycarbonyl)-5-azaspiro[2.4]heptan-7-amine (0.93 g, 3 mmol) in MeOH (20 mL) was added catalyst Pd/C (10%, 54.5% water). The reaction was purged with H2 and stirred at rt for 24 h. The mixture was filtered and the filtrate was concentrated in vacuo to give the title compound as yellow oil (0.6 g, 100%).
LC-MS (ESI, pos. ion) m/z: 213.1 [M + H]+;
!H NMR (400 MHz, CDC13) δ (ppm): 0.56 (dd, J = 33.2 Hz, 9.6 Hz, 2H), 0.77 (s, 2H), 1.37 (s, 9H), 2.73 (d, J = 10.8 Hz, 1H), 2.93 (dd, J = 11.6 Hz, 3.2 Hz, 1H), 2.99 (d, J = 10.8 Hz, 1H), 3.33 (dd, J= 11.6 Hz, 5.6 Hz, 1H), 3.47 (s, 1H), 3.66 (s, 1H), 7.26 (s, 2H), 4.75 (s, 1H).
Step 5) 5-(4-(6-(bis(ter?-butoxycarbonyl)amino)-5-((R)-l-(2,6-dichloro-3-fluorophenyl)ethoxy) pyridin-3-yl)but-3-vn-2-yl)-N-(ter?-butoxycarbonyl)-5-azaspiro[2.4]heptan-7-amine
[0196] To a mixture of 4-(6-(bis(?ert-butoxycarbonyl)amino)-5-((R)-l-(2,6-dichloro-3- fluorophenyl) ethoxy)pyridin-3-yl)but-3-yn-2-yl methanesulfonate (0.16 g, 0.25 mmol) in CH3CN (5 mL) was added N-(?ert-butoxycarbonyl)-5-azaspiro[2.4]heptan-7-amine (78.5 mg, 0.38 mmol) and Et3N (0.35 mL, 2.5 mmol) at rt. The reaction was stirred at 69°C for 16 h, then cooled to rt and concentrated in vacuo. The residue was diluted with EtOAc (50 mL). The resulted mixture was washed with water (50 mL), dried over Na2S04 and concentrated in vacuo. The residue was purified by a silica gel column chromatography (PE/EtOAc (v/v) = 1/2) to give the title compound as a yellow solid (0.1 1 g, 58%).
LC-MS (ESI, pos. ion) m/z: 764 [M + H]+;
¾ NMR (400 MHz, CDC13) δ (ppm): 0.50-0.70 (m, 2H), 0.73-0.92 (m, 2H), 1.30-1.41 (m, 3H), 1.41-1.46 (m, 27H), 1.79 (d, J = 6.6 Hz, 3H), 2.54-2.64 (m, 1H), 2.69-2.86 (m, 2H), 3.01-3.16 (m, 1H), 3.75-3.76 (m, 1H), 3.84-3.89 (m, 1H), 4.91 -4.95 (m, 1H), 5.97-6.02 (q, J = 6.45 Hz, 1H), 7.06-7.10 (m, 2H), 7.30-7.32 (m, 1H), 8.10 (d, J= 1.5 Hz, 1H).
Step 6) 5-(4-(6-amino-5-((R)-l-(2.6-dichloro-3-fluorophenyl)ethoxy)pyridin-3-yl)but-3-yn-2-yl)- 5 -azaspiro \2.4]heptan-7 -amine
[0197] To a mixture of 5-(4-(6-(bis(/er/-butoxycarbonyl)amino)-5-((R)-l-(2,6-dichloro-3- fluorophenyl)ethoxy)pyridin-3-yl)but-3-yn-2-yl)-N-(/er/-butoxycarbonyl)-5-azaspiro[2.4]heptan- 7-amine (0.4 g, 0.52 mmol) in DCM (15 mL) at 0°C was added HC1 in EtOAc (3 M, 5 mL). The reaction was stirred at rt for 16 h, and then concentrated in vacuo. The residue was diluted with water (50 mL), then adjusted to pHIO with saturated a2C03 aqueous and extracted with DCM (80 mL x 3). The combined organic phases were washed with brine (50 mL), dried over Na2S04 and concentrated in vacuo. The residue was purified by a silica gel column chromatography (DCM/MeOH/Et3N (v/v/v) = 200/20/1) to give the title compound as a yellow solid (294 mg, 90%).
LC-MS (ESI, pos. ion) m/z: 464 [M + H]+;
¾ NMR (400 MHz, CDC13) δ (ppm): 0.41-0.49 (m, 1H), 0.59-0.67 (m, 2H), 0.75-0.78 (m, 1H), 1.40 (d, J = 6.9 Hz, 3H), 1.81 (d, J = 6.7 Hz, 3H), 2.56-2.64 (m, 2H), 2.89-2.80 (m, 1H), 3.07- 3.14 (m, 1H), 3.20-3.25 (m, 1H), 3.70-3.73 (m, 1H), 4.95 (s, 2H), 6.00-6.02 (q, J = 6.5 Hz, 1H), 6.75 (s, 1H), 7.10 (t, J= 8.6 Hz, 1H), 7.28-7.32 (m, 1H), 7.73 (d, J= 1.5 Hz, 1H).
Example 13 5-(4-(6-amino-5-((JR)-l-(2,6-dichloro-3-fluorophenyl)ethoxy)pyridin-3-yl)but-3- yn-2-yl)-5-azaspiro [2.41 heptan-7-ol
Figure imgf000065_0001
Step 1) 5-(4-(6-(bis(te^butoxycarbonyl amino -5-('('R -l-('2,6-dichloro-3-fluorophenyl ethoxy pyridin-3-yl)but-3-yn-2-yl)-5-azaspiro[2.4]heptan-7-ol
[0198] To a mixture of 4-(6-(bis(?ert-butoxycarbonyl)amino)-5-((R)-l-(2,6-dichloro-3- fluorophenyl) ethoxy)pyridin-3-yl)but-3-yn-2-yl methanesulfonate (0.99 g, 1.53 mmol) in CH3CN (13 mL) was added 5-azaspiro[2.4]heptan-7-ol (259 mg, 1.95 mmol) and Et3N (2 mmol) at rt. The mixture was stirred at 69°C for 16 h, then cooled to rt. The mixture was concentrated in vacuo and the residue was diluted with EtOAc (50 mL). The resulted mixture was washed with water (50 mL), dried over Na2S04 and concentrated in vacuo. The residue was purified by a silica gel column chromatography (PE/EtOAc (v/v) = 1/1 to 1/2) to give the title compound as a yellow solid (0.74 g, 74%).
LC-MS (ESI, pos. ion) m/z: 664 [M + H]+;
!H NMR (400 MHz, CDC13) δ (ppm): 0.60-0.72 (m, 2H), 0.73-0.80 (m, 1H), 0.90-0.98 (m, 1H), 1.41-1.48 (m, 21H), 1.79 (d, J= 6.8 Hz, 3H), 2.92-3.10 (m, 2H), 3.72-3.88 (m, 2H), 5.99 (m, 1H), 7.08-7.105 (m, 3H), 8.10 (d, J= 1.7 Hz, 1H).
Step 2) 5-(4-(6-amino-5-((R)-l-(2.6-dichloro-3-fluorophenyl)ethoxy)pyridin-3-yl)but-3-yn-2-yl)- 5 -azaspiro \2.4"|heptan-7 -ol
[0199] To a mixture of 5-(4-(6-(bis(?ert-butoxycarbonyl)amino)-5-((R)-l-(2,6-dichloro-3- fluorophenyl) ethoxy)pyridin-3-yl)but-3-yn-2-yl)-5-azaspiro[2.4]heptan-7-ol (0.42 g, 0.6 mmol) in DCM (15 mL) at 0°C was added HC1 in EtOAc (3 M, 8 mL). The mixture was stirred at rt for 16 h and concentrated in vacuo. The residue was diluted with water (50 mL) and the pH of the resulted solution was adjusted to 10 with saturated Na2C03 aqueous solution. The mixture was extracted with DCM (50 mL x 3) and the combined organic phases were washed with brine (50 mL), dried over Na2S04 and concentrated in vacuo. The residue was purified by a silica gel column chromatography (EtOAc/MeOH/Et3N (v/v/v) = 250/10/1) to give the title compound as a yellow solid (0.16 g, 60%).
LC-MS (ESI, pos. ion) m/z: 464 [M + H]+;
¾ NMR (400 MHz, CDC13) δ (ppm): 0.57-0.58 (m, 1H), 0.63-0.65 (m, 1H), 0.72-0.75 (m, 1H), 0.92-0.94(m, 1H), 1.39-1.42 (m, 3H), 1.81 (d, J = 6.7 Hz, 3H), 2.57-2.64 (m, 1H), 2.84-3.00 (m, 3H), 3.71-3.77 (m, 2H), 5.01 (s, 2H), 6.01 (m, 1H), 6.75 (d, J = 1.4 Hz, 1H), 7.05-7.09 (m, 1H), 7.29-7.31 (m, 1H), 7.72 (s, 1H).
Example 14 5- 3- 6-3ηΊίηο-5- /?)-1- 2,6-άί€ΜθΓθ-3-ΑυοΓθρΗ6ην1)6ίΗοχν)ρνή(ϋη-3-ν1)ρΓθρ- 2-yn-l-yl)-5-azaspiro[2.41heptan-7-ol
Figure imgf000067_0001
Step 1) (R -3-(6-(bis(te^butoxycarbonyl amino -5-('l-('2,6-dichloro-3-fluorophenyl ethoxy pyridin-3 -yl)prop-2-yn- 1 -ol
[0200] To a mixture of (R)-5-bromo-N,N-bis(?ert-butoxycarbonyl)-3-(l-(2,6-dichloro-3- fluorophenyl) ethoxy)pyridin-2 -amine (2 g, 3.5 mmol), Pd(PPh3)2Ci2 (246 mg, 0.35 mmol) and Cul (67 mg, 0.35 mmol) in Et3N (18 mL) was added prop-2-yn-l-ol (0.42 mL, 7 mmol). The reaction was microwaved at 90°C for 80 min, then cooled to rt. The mixture was concentrated in vacuo and the residue was diluted with DCM (100 mL). The undissolved solid was removed through filtration and the filtrate was dried over Na2S04 and concentrated in vacuo. The residue was purified by a silica gel column chromatography (PE/EtOAc (v/v) = 2/1) to give the title compound as a yellow solid (1.52 g, 80%).
!H NMR (400 MHz, CDC13) δ (ppm): 1.37-1.41 (m, 18H), 1.55 (d, J = 6.6 Hz, 3H), 1.78 (d, J = 6.6 Hz, 3H), 4.72-4.78 (m, 1H), 5.97-5.02 (q, J = 6.7 Hz, 1H), 7.06-7.12 (m, 2H), 7.27-7.32 (m, 1H), 8.12-8.13 (m, 1H).
Step 2) (R)-3-(6-(bis(ter/-butoxycarbonyl)amino)-5-(l-(2,6-dichloro-3-fluorophenyl)ethoxy) pyridin-3 -yl)prop-2-yn- 1 -yl methanesulfonate
[0201] To a mixture of (R)-3-(6-((di-/er/-butoxycarbonyl)amino)-5-(l-(2,6-dichloro-3- fluorophenyl) ethoxy)pyridin-3-yl)prop-2-yn-l-ol (0.3 g, 0.54 mmol) in DCM (2.5 mL) was added Et3N (0.22 mL, 1.6 mmol) and methanesulfonyl chloride (63 μί, 0.81 mmol) at 0°C under 2 atmosphere. The reaction was stirred at rt for 1 h, then quenched with saturated NaHC03 aqueous solution (10 mL). The mixture was extracted with DCM (10 mL x 3) and the combined organic phases were dried over Na2S04 and concentrated in vacuo to give the title compound. This compound was used in the next step without purification.
LC-MS (ESI, pos. ion) m/z: 633 [M + H]+.
Step 3) 5-(3-(6-(bis(ter/-butoxycarbonyl)amino)-5-((R)-l-(2,6-dichloro-3-fluorophenyl)ethoxy) pyridin-3-yl)prop-2-yn-l-yl)-5-azaspiro[2.4]heptan-7-ol [0202] To a mixture of (R)-3-(6-((di-?ert-butoxycarbonyl)amino)-5-(l-(2,6-dichloro-3- fluorophenyl) ethoxy)pyridin-3-yl)prop-2-yn-l-yl methanesulfonate (341 mg, 0.54 mmol) in CH3CN (5 mL) was added 5-azaspiro[2.4]heptan-7-ol (92 mg, 1.08 mmol) and Et3 (1 mL) at rt. The reaciton was stirred at 69°C for 16 h, then cooled to rt and concentrated in vacuo. The residue was diluted with EtOAc (50 mL), then washed with H20 (50 mL) and brine (50 mL). The organic phase was dried over Na2S04 and concentrated in vacuo. The residue was purified by a silica gel column chromatography (PE/EtOAc (v/v) = 2/1) to give the title compound as a yellow solid (0.14 g, 40%).
LC-MS (ESI, pos. ion) m/z: 650 [M + H]+;
¾ NMR (400 MHz, CDC13) δ (ppm): 0.61-0.69 (m, 2H), 0.77-0.78 (m, 1H), 0.93-0.96 (m, 1H), 1.38-1.43 (m, 18H), 1.78 (d, J = 6.6 Hz, 3H), 2.57-2.60 (dd, J = 8.8 Hz, 4.8 Hz, 1H), 2.92-2.95 (m, 2H), 3.04-3.07 (dd, J = 10.0 Hz, 1.6 Hz, 1H), 3.65 (s, 2H), 3.77-3.79 (m, 1H), 5.99 (m, 1H), 7.05-7.10 (m, 2H), 7.27-7.32 (m, 1H), 8.10 (d, J= 1.7 Hz, 1H).
Step 4) 5-(3-(6-amino-5-((R)-l-(2,6-dichloro-3-fluorophenyl)ethoxy)pyridin-3-yl)prop-2-vn-l- yl)-5-azaspiror2.41heptan-7-ol
[0203] To a mixture of 5-(3-(6-((di-/er/-butoxycarbonyl)amino)-5-((R)-l-(2,6-dichloro-3- fluorophenyl) ethoxy)pyridin-3-yl)prop-2-yn-l-yl)-5-azaspiro[2.4]heptan-7-ol (0.14 g, 0.22 mmol) in DCM (6 mL) at 0°C was added HC1 in EtOAc (3 M, 2 mL). The reaction was stirred at rt for 16 h and concentrated in vacuo. The residue was diluted with water (50 mL), and the pH of the resulting solution was adjusted to 10 with saturated a2C03 solution. The mixture was extracted with DCM (50 mL x 3) and the combined organic phases were washed with brine (50 mL), dried over Na2S04 and concentrated in vacuo. The residue was purified by a silica gel column chromatography (EtOAc/MeOH/Et3N (v/v/v) = 300/30/1) to give the title compound as a yellow solid (85 mg, 88%).
LC-MS (ESI, pos. ion) m/z: 450 [M + H]+;
¾ NMR (400 MHz, CDC13) δ (ppm): 0.69-0.78 (m, 3H), 0.91-0.95 (m, 1H), 1.81 (d, J = 6.7 Hz, 3H), 2.58 (d, J = 8.6 Hz, 1H), 2.92 (t, J = 8.6 Hz, 2H), 3.02-3.05 (dd, J = 10.1 Hz, 4.9 Hz, 1H), 3.61 (s, 2H), 3.75-3.76 (m, 1H), 4.96 (s, 2H), 5.98-6.03 (q, J = 6.7 Hz, 1H), 6.74 (d, J = 1.3 Hz, 1H), 7.05-7.09 (m, 1H), 7.29-7.32 (m, 1H), 7.73 (s, 1H).
BIOLOGICAL TESTING
[0204] The LC/MS/MS system used in the analysis consists of an Agilent 1200 Series vacuum degasser, binary pump, well-plate autosampler, thermostatedcolumn compartment, the Agilent G6430 TripleQuadrupole Mass Spectrometer with an electrosprayionization (ESI) source. Quantitative analysis was carried out using MRM mode. The parameters for MRM transitions are in the Table A.
Table A
Figure imgf000069_0001
[0205] An Agilent XDB-C18, 2.1 x 30 mm, 3.5 μΜ column was used for the analysis. 5 μΐ^ of the samples were injected. Analysis condition: The mobile phase was 0.1% formic acid in water (A) and 0.1% formic acid in methanol (B). The flow rate was 0.4 mL/min. And the gradient of Mobile phase was in the Table B.
Table B
Figure imgf000069_0002
[0206] Alternatively, an Agilent 6330 series LC/MS/MS spectrometer equipped with G1312A binary pumps, a G1367A autosampler and a G1314C UV detector was used in the analysis. An ESI source was used on the LC/MS/MS spectrometer. The analysis was done in positive ion mode as appropriate and the MRM transition for each analyte was optimized using standard solution. A Capcell MP-C18 100 x 4.6 mm ID., 5 μΜ column (Phenomenex, Torrance, California, USA) was used during the analysis. The mobile phase was 5mM ammonia acetate, 0.1% MeOH in water (A) : 5mM ammonia acetate, 0.1% MeOH in acetonitrile (B) (70:30, v/v). The flow rate was 0.6 mL/min. Column was maintained at ambient temperature. 20 μϊ^ of the samples were injected.
Example A: Compound Stability In Human And Rat Liver Microsomes
[0207] Human or rat liver microsomes incubations were conducted in duplicate in polypropylene tubes. The typical incubation mixtures consisted of human liver microsomes (0.5 mg protein/mL), compounds of interest (5 μΜ) and NADPH (1.0 mM) in a total volume of 200 μΐ, potassium phosphate buffer (PBS, 100 mM, pH7.4). Compounds were dissolved in DMSO and diluted with PBS such that the final concentration of DMSO was 0.05%. The enzymatic reactions were commenced with the addition of protein after a 3 -min preincubation and incubated in a water bath open to the air at 37°C. Reactions were terminated at various time points (0, 5, 10, 15, 30, 60 min) by adding equal volume of ice-cold acetonitrile. The samples were stored at -80°C until LC/MS/MS assays.
[0208] The concentrations of compounds in the incubation mixtures of human liver microsomes were determined by a LC/MS/MS method. The ranges of the linearity in the concentration range were determined for each tested compounds.
[0209] A parallel incubation was performed using denatured microsomes as the negative control, and reactions were terminated at various time points (0, 15, 60 min) after incubation at 37°C.
[0210] Dextromethorphan (70 μΜ) was selected as the positive control, and reactions were terminated at various time points (0, 5, 10, 15, 30, 60 min) after incubation at 37°C. Both positive and negative control samples were included in each assay to ensure the integrity of the microsomal incubation system.
[0211] Alternatively, the stability of some of the compounds disclosed herein in human (or rat) liver microsomes were also conducted in the following protocol. The incubations were conducted in duplicate in polypropylene tubes. The typical incubation mixtures consisted of human (or rat) liver microsomes (final concentration: 0.5 mg protein/mL), compounds (final concentration: 1.5 μΜ) in a total volume of 30 μϊ^ K-buffer (contain 1.0 mM EDTA, 100 mM, pH7.4). Compounds were dissolved in DMSO and diluted with K-buffer such that the final concentration of DMSO was 0.2%. The enzymatic reactions were commenced with the addition of 15 μϊ^ NADPH (final concentration: 2 mM) after 10 min preincubation and incubated in a 37°C incubator. Reactions were terminated at various time points (0, 15, 30, 60 min) by adding 135 μϊ^ acetonitrile (contain IS). Protein is removed by centrifugation with 4000rpm, 10 min. Supernatant was collected for LC-MS/MS analysis
[0212] In the above protocol, ketanserin (1 μΜ) was selected as the positive control, and reactions were terminated at various time points (0, 15, 30, 60 min) after incubation at 37°C. Both positive control samples were included in each assay to ensure the integrity of the microsomal incubation system.
Data Analysis
[0213] The concentrations of compounds in human liver microsome incubations were plotted as a percentage of the relevant zero time point control for each reaction. The in vivo CL;nt were extrapolated (ref : Naritomi et al., Prediction of human hepatic clearance from in vivo animal experiments and in vitro metabolic studies with liver microsomes from animals and humans, Drug Metab. Dispos., 2001, 29: 1316-1324.)
[0214] The compounds disclosed herein exhibited desirable half-life (Ti/2) when the compounds were incubated in human and rat liver microsomes.
Table 2 Stability in human and rat liver microsomes
Figure imgf000071_0001
Example B: Evaluation of Pharmacokinetics After Intravenous and Oral Administration of The
Compounds Disclosed Herein In Mice, Rats, Dogs And Monkeys [0215] Compounds disclosed herein are assessed in pharmacokinetic studies in mice, rats, dogs or monkeys. The compounds are administered as a water solution, 2% HPMC + 1% TWEEN® 80 in water solution, 5% DMSO + 5% solutol in saline, 4% MC suspension or capsule. For the intravenous administration, the animals are generally given at 1 or 2 mg/kg dose. For the oral (p.o.) dosing, mice and rats are generally given 5 or 10 mg/kg dose, and dogs and monkeys are generally given 10 mg/kg dose. The blood samples (0.3 mL) are drawn at 0.25, 0.5, 1.0, 2.0, 3.0, 4.0, 6.0, 8.0, 12 and 24 h time points or 0.083, 0.25, 0.5, 1.0, 2.0, 4.0, 6.0, 8.0 and 24 h time points and centrifuged at 3,000 or 4000 rpm for 2 to 10 min. The plasma solutions are collected and stored at -20 °C or -70 °C before analyzed by LC/MS/MS as described above.
[0216] The compounds disclosed herein exhibited optimized pharmacokinetic properties with desirable clearance (CI) and half-life (T1/2) when the compounds were administered intravenously.
Table 3 Pharmacokinetic profiles in rats
Figure imgf000072_0001
Example C: Kinase Assays
[0217] Kinase assays can be performed by measurement of incorporation of γ-33Ρ ATP into immobilized myelin basic protein (MBP). High binding white 384 well plates (Greiner) are coated with MBP (Sigma #M-1891) by incubation of 60 μΕ/well of 20 μg/mL MBP in Tris- buffered saline (TBS; 50 mM Tris pH 8.0, 138 mM NaCl, 2.7 mM KC1) for 24 h at 4°C. Plates are washed 3 x with 100 μΐ^ TBS. Kinase reactions are carried out in a total volume of 34 μΐ^ in kinase buffer (5 mM Hepes pH 7.6, 15 mM NaCl, 0.01% bovine gamma globulin (Sigma #1- 5506), 10 mM MgCl2, 1 mM DTT, 0.02% TritonX-100). Compound dilutions are performed in DMSO and added to assay wells to a final DMSO concentration of 1%. Each data point is measured in duplicate, and at least two duplicate assays are performed for each individual compound determination. Enzyme is added to final concentrations of 10 nM or 20 nM, for example. A mixture of unlabeled ATP and γ-33Ρ ATP is added to start the reaction (2 x 106 cpm of γ-33Ρ ATP per well (3000 Ci/mmole) and 10 μΜ unlabeled ATP, typically. The reactions are carried out for 1 h at rt with shaking. Plates are washed 7x with TBS, followed by the addition of 50 μΕΛνεΙΙ scintillation fluid (Wallac). Plates are read using a Wallac Trilux counter. This is only one format of such assays; various other formats are possible, as known to one skilled in the art.
[0218] The above assay procedure can be used to determine the IC50 for inhibition and/or the inhibition constant, ¾. The IC50 is defined as the concentration of compound required to reduce the enzyme activity by 50% under the condition of the assay. The IC50 value is estimated by preparing a 10 point curve using a ½ log dilution series (for example, a typical curve may be prepared using the following compound concentrations: 10 μΜ, 3 μΜ, 1 μΜ, 0.3 μΜ, 0.1 μΜ, 0.03 μΜ, 0.01 μΜ, 0.003 μΜ, 0.001 μΜ and 0 μΜ).
[0219] The kinase assays described herein were performed at Millipore UK Ltd, Dundee Technology Park, Dundee DD2 1SW, UK.
ALK (h Kinase Assay
[0220] ALK (h) is incubated with 8 mM MOPS pH 7.0, 0.2 mM EDTA, 250 μΜ KKKSPGEYV IEFG, 10 mM MgAcetate and [γ-33Ρ-ΑΤΡ] (specific activity aprrox. 500 pcm/pmol, concentration as required (10 μΜ)). The reaction is initiated by the addition of the MgATO mix. After incubation for 40 min at room temperature, the reaction is stopped by the addition of 3% phosphoric acid solution. 10 μϊ^ of the reaction is then spotted onto a P30 filter mat and washed three times for 5 min in 75 mM phosphoric acid and once in methanol prior to drying and scintillation counting.
c-Met (h Kinase Assay
[0221] Met (h) is incubated with 8 mM MOPS pH 7.0, 0.2 mM EDTA, 250 μΜ
KKKSPGEYVNIEFG, 10 mM MgAcetate and [γ-33Ρ-ΑΤΡ] (specific activity approx. 500 cpm/pmol, concentration as required (10 μΜ)). The reaction is initiated by the addition of the
MgATP mix. After incubation for 40 min at room temperature, the reaction is stopped by the addition of 3% phosphoric acid solution. 10 μΐ^ of the reaction is then spotted onto a P30 filtermat and washed three times for 5 min in 75 mM phosphoric acid and once in methanol prior to drying and scintillation counting.
[0222] The compounds disclosed herein exhibited potent activities in the ALK and c-Met (h) assays. Table 4 Used the IC50S of some examples described herein in the ALK (h) and c-Met (h) assays.
Table 4 Kinase inhibition data
Figure imgf000074_0001
Example D: Tumor Xenograft Models
[0223] The efficacy of compounds disclosed herein is evaluated in a standard murine model of tumorigenesis. Human tumor cells (such as U87MG glioblastoma cells, MKN45 Gastric Adenocarcinoma cells, MDA-MB-231 breast adenocarcinoma cells, or Caki-1 renal carcinoma cells, all from ATCC) are expended in culture, harvested, and injected subcutaneous ly onto the rear flank of 6-7 week old female athymic nude mice (BALB/cA nu/nu, Shanghai SLAC Laboratory Animal, Co.) (n = 10 for vehicle group, n = 8 for each dosing group). When tumors reach a volume of 100-250 mm3, animals are randomly divided into vehicle control (for example, 2% HPMC + 1% TWEEN® in water) and compound groups. Subsequent administration of compound by oral gavage (for example, 3-50 mpk/dose, dissolved in 2% HPMC + 1% Tween-80 in water) begins anywhere from day 0 to day 15 post tumor cell challenge and generally continues with once a day for the duration of the experiment.
Tumor Growth Inhibition (TGI) Analysis
[0224] Progression of tumor growth is assessed by tumor volumes and recorded as a function of time. The long (L) and short (W) axes of the subcutaneous tumors are measured with calipers twice weekly, and the tumor volume (TV) calculated as (L x W2)/2). TGI is calculated from the difference between the median tumor volumes of vehicle-treated and drug-treated mice, expressed as a percentage of the median tumor volume of the vehicle-treated control group, by the following relation:
Figure imgf000075_0001
Median Tumor Volume control
[0225] Initial statistical analysis is done by repeated measures analysis of variance (RMANOVA), Followed by Scheffe post-hoc testing for multiple comparisons. Vehicle alone (2% HPMC + 1% TWEEN®, or the like) is the negative control.
[0226] The compouds described herein were also administrated orally (p.o.) once a day (QD), for 13-21 days in U87MG xenograft animal model. At doses of 60 mg/kg, the compouds produced statistically significant inhibition of growth of certain tumors grown subcutaneously in athymic nude mice.
[0227] Finally, it should be noted that there are alternative ways of implementing the present invention. Accordingly, the present embodiments are to be considered as illustrative and not restrictive and the invention is not be limited to the details given herein, but may be modified within the scope and equivalents of the appended claims. All publications and patents cited herein are incorporated by reference.

Claims

WHAT IS CLAIMED IS:
1. A compound of Formula (I):
Figure imgf000076_0001
or a stereoisomer, a geometric isomer, a tautomer, an N-oxide, a solvate, a hydrate, a metabolite, a pharmaceutically acceptable salt or a prodrug thereof, wherein:
each R1, R2, R3, R4, R5 and R6 is independently H, D or F;
X is C6-ioaryl or 5-10 membered heteroaryl comprising 1, 2, 3 or 4 heteroatoms independently selected from O, S or Ν, wherein each of the C6-ioaryl and 5-10 membered heteroaryl is optionally substituted with 1, 2, 3 or 4 substituents independently selected from D, F, CI, Br, I, CN, N02, N3, ORa, SRa, NRaRb, -C(=0)NRaRb, Ci_6alkyl, Ci_6haloalkyl, C2-6alkenyl, C2-6alkynyl, -(Ci_4alkylene)-CN, -(Ci_4alkylene)-ORa, - (Ci-4alkylene)-NRaRb, C6-ioaryl and 5-10 membered heteroaryl;
Z is Ci_6alkyl, C3_6cycloalkyl, -(Ci_4alkylene)-(C3_6cycloalkyl), C3_6heterocyclyl, - (Ci-4alkylene)-(C3-6heterocyclyl), Cs-^fused bicyclyl, -(Ci-4alkylene)-(C5-i2fused bicyclyl), C5_i2spirobicyclyl or -(Ci_4alkylene)-(C5_i2spirobicyclyl), wherein each of the
Figure imgf000076_0002
C3-6cycloalkyl, -(Ci-4alkylene)-(C3-6cycloalkyl), C3-6heterocyclyl, -(Ci_4alkylene)- (C3_6heterocyclyl), Cs-^fused bicyclyl, -(Ci-4alkylene)-(C5-i2fused bicyclyl), C5_i2spirobicyclyl and -(Ci-4alkylene)-(C5-i2spirobicyclyl) is optionally substituted with 1, 2, 3 or 4 substituents independently selected from D, F, CI, Br, I, Ci_4alkyl,
Figure imgf000076_0003
ORa, -(Ci_4alkylene)-ORa, -(Ci_4alkylene)-NRaRb, NRaRb, -C(=0)NRaRb and -OC(=0)NRaRb; and each of the cyclic structures in the bicyclyl groups is a carbocyclic ring or a heterocyclic ring; and
each Ra and Rb is independently H, C ^aliphatic, C3_6cycloalkyl, -(Ci-4alkylene)-(C3_6cycloalkyl), C3_6heterocyclyl, -(Ci-4alkylene)-(C3-6heterocyclyl), C6-ioaryl, -(Ci_4alkylene)-(C6-ioaryl), 5-10 membered heteroaryl or -(Ci_4alkylene) -(5-10membered heteroaryl), provided that when Z is Ci-6alkyl, NRaRb is not NH2 or Me2, or when Ra and Rb are bonded to the same nitrogen atom, Ra and Rb, together with the nitrogen atom they are attached to, optionally form a 3-8 membered heterocyclyl, wherein each of the Ci_6aliphatic, C3_6cycloalkyl, -(Ci_4alkylene)-(C3_6cycloalkyl), C3_6heterocyclyl, -(Ci-4alkylene)-(C3_6heterocyclyl), C6-ioaryl, -(Ci_4alkylene)-(C6-ioaryl), 5-10 membered heteroaryl, -(C alkylene)-(5-10membered heteroaryl) and 3-8 membered heterocyclyl is optionally substituted with 1, 2, 3 or 4 substituents independently selected from D, F, CI, CN, N3, OH, NH2, alkoxy and alkylamino.
2. The compound of claim 1, wherein each R1, R2, R3, R4, R5 and R6 is independently H or D.
3. The compound of claim 1, wherein X is phenyl optionally substituted with 1, 2, 3 or 4 substituents independently selected from D, F, CI, Br, Ci_3alkyl and Ci_3haloalkyl.
4. The compound of claim 1, wherein Z is
Figure imgf000077_0001
C3_6cycloalkyl, C3_6heterocyclyl, -(Ci-4alkylene)-(C3_6heterocyclyl), Cs-^fused bicyclyl, -(Ci_4alkylene)-(C5_i2fused bicyclyl), C5_i2spirobicyclyl or -(Ci_4alkylene)-(C5_i2spirobicyclyl), wherein the
Figure imgf000077_0002
C3-6cycloalkyl, C3_6heterocyclyl, -(Ci_4alkylene)-(C3_6heterocyclyl), Cs-^fused bicyclyl, -(Ci-4alkylene)-(C5_i2fused bicyclyl), Cs-^spirobicyclyl and -(Ci-4alkylene)-(C5_i2spirobicyclyl), each of which is optionally substituted with 1, 2, 3 or 4 substituents independently selected from D, F, CI, Br, I, Ci_3alkyl,
Figure imgf000077_0003
ORa and NRaRb; each of the cyclic structures in the bicyclyl groups is a carbocyclic ring or a heterocyclic ring.
5. The compound of claim 1, wherein each Ra and Rb is independently H, Ci_3alkyl, C3-6cycloalkyl or -(Ci_3alkylene)-(C3_6cycloalkyl), provided that when Z is C^aUcyl, NRaRb is not NH2 or NMe2, wherein each of the Ci_3alkyl, C3_6cycloalkyl and -(Ci-3alkylene)-(C3_6cycloalkyl) is optionally substituted with 1, 2, 3 or 4 substituents independently selected from D, F, OH, NH2, Ci_3alkoxy and Ci_3alkylamino.
6. The compound of claim 1, wherein Z is selected from the following substructures:
Figure imgf000077_0004
(Z1 ) , (Z2) , (Z3) , (Z4) , (Z5) , (Z6) , (Z7) , (Z8) , (Z9) ,
Figure imgf000078_0001
(Z10) , (Z11) , (Z12) , (Z13) , (Z14) , (Z15) , (Z16) , (Z17) ,
Figure imgf000078_0002
(Ζ18) (Ζ19) , (Ζ20) , (Ζ21) , (Ζ22) , (Ζ23) , (Ζ24) ,
(Ζ25) (Ζ26) , (Ζ27) , (Ζ28) , (Ζ29) , (Ζ30) , (Ζ31) , (Ζ32) , (Ζ33) ,
Figure imgf000078_0004
(Ζ34) , (Ζ35) , (Ζ36) , (Ζ37) , (Ζ38) , (Ζ39) , (Ζ40), (Ζ41) , (Ζ42), or a stereoisomer thereof, wherein n is 0, 1, 2 or 3; each W and W is independently O or NH; each hydrogen in Z is optionally substituted with 1, 2, 3 or 4 substituents independently selected from D, F, CI, Br, I, d_3alkyl, Ci_6haloalkyl, ORa and NRaRb, provided that NRaRb is not NH2 or NMe2 in structures (Z25) to (Z32).
7. The compound of claim 1 having one of the following structures:
Figure imgf000079_0001
Figure imgf000080_0001
8. A pharmaceutical composition comprising the compound of any one of claims 1 to 7 and a pharmaceutically acceptable carrier, excipient, diluent, adjuvant, vehicle, or a combination thereof.
9. The pharmaceutical composition of claim 8 further comprising a therapeutic agent selected from a chemotherapeutic agent, an anti-proliferative agent, an agent for treating atherosclerosis, an agent for treating lung fibrosis, and combinations thereof.
10. The pharmaceutical composition of claim 9, wherein the therapeutic agent is chlorambucil, melphalan, cyclophosphamide, ifosfamide, busulfan, carmustine, lomustine, streptozocin, cisplatin, carboplatin, oxaliplatin, dacarbazine, temozolomide, procarbazine, methotrexate, fluorouracil, cytarabine, gemcitabine, mercaptopurine, fludarabine, vinblastine, vincristine, vinorelbine, paclitaxel, docetaxel, topotecan, irinotecan, etoposide, trabectedin, dactinomycin, doxorubicin, epirubicin, daunorubicin, mitoxantrone, bleomycin, mitomycin, ixabepilone, tamoxifen, flutamide, gonadorelin analogues, megestrol, prednisone, dexamethasone, methylprednisolone, thalidomide, interferon alfa, leucovorin, sirolimus, temsirolimus, everolimus, afatinib, alisertib, amuvatinib, apatinib, axitinib, bortezomib, bosutinib, brivanib, cabozantinib, cediranib, crenolanib, crizotinib, dabrafenib, dacomitinib, danusertib, dasatinib, dovitinib, erlotinib, foretinib, ganetespib, gefitinib, ibrutinib, icotinib, imatinib, iniparib, lapatinib, lenvatinib, linifanib, linsitinib, masitinib, momelotinib, motesanib, neratinib, nilotinib, niraparib, oprozomib, olaparib, pazopanib, pictilisib, ponatinib, quizartinib, regorafenib, rigosertib, rucaparib, ruxolitinib, saracatinib, saridegib, sorafenib, sunitinib, tasocitinib, telatinib, tivantinib, tivozanib, tofacitinib, trametinib, vandetanib, veliparib, vemurafenib, vismodegib, volasertib, alemtuzumab, bevacizumab, brentuximabvedotin, catumaxomab, cetuximab, denosumab, gemtuzumab, ipilimumab, nimotuzumab, ofatumumab, panitumumab, ramucirumab, rituximab, tositumomab, trastuzumab, or a combination thereof.
1 1. A method of preventing, managing, treating or lessening the severity of a proliferative disorder in a patient by administering to the patient with the compound of any one of claims 1 to 7 or the pharmaceutical composition of any one of claims 8 to 10.
12. The compound of any one of claims 1 to 7 or the pharmaceutical composition of any one of claims 8 to 10 for use in preventing, managing, treating or lessening the severity of a proliferative disorder in a patient.
13. The method of claim 1 1, wherein the proliferative disorder is metastatic cancer, colon cancer, gastric adenocarcinoma, bladder cancer, breast cancer, kidney cancer, liver cancer, lung cancer, skin cancer, thyroid cancer, cancer of the head and neck, prostate cancer, pancreatic cancer, cancer of the CNS, glioblastoma, a myeloproliferative disorder, atherosclerosis or lung fibrosis.
14. The compound or pharmaceutical composition of claim 12, wherein the proliferative disorder is metastatic cancer, colon cancer, gastric adenocarcinoma, bladder cancer, breast cancer, kidney cancer, pancreatic cancer, cancer of the CNS, glioblastoma, a myeloproliferative disorder, atherosclerosis or lung fibrosis.
15. A method of inhibiting or modulating the activity of a protein kinase in a biological sample comprising contacting a biological sample with the compound of any one of claims 1 to 7 or the pharmaceutical composition of any one of claims 8 to 10.
16. The method of claim 15, wherein the protein kinase is a receptor tyrosine kinase.
17. The method of claim 16, wherein the receptor tyrosine kinase is ALK or c-Met.
PCT/US2013/041918 2012-05-23 2013-05-21 Substituted alkynyl pyridine compounds and methods of use WO2013177092A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201261650513P 2012-05-23 2012-05-23
US61/650,513 2012-05-23

Publications (1)

Publication Number Publication Date
WO2013177092A1 true WO2013177092A1 (en) 2013-11-28

Family

ID=49624271

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2013/041918 WO2013177092A1 (en) 2012-05-23 2013-05-21 Substituted alkynyl pyridine compounds and methods of use

Country Status (1)

Country Link
WO (1) WO2013177092A1 (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN107879966A (en) * 2017-10-16 2018-04-06 扬子江药业集团有限公司 A kind of preparation method of sitafloxacin key intermediate
US10053477B2 (en) 2014-07-04 2018-08-21 Qilu Pharmaceutical Co., Ltd. Spirocyclic aryl phosphorus oxide and aryl phosphorus sulfide
US11040027B2 (en) 2017-01-17 2021-06-22 Heparegenix Gmbh Protein kinase inhibitors for promoting liver regeneration or reducing or preventing hepatocyte death

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070072874A1 (en) * 2003-02-26 2007-03-29 Sugen, Inc. Aminoheteroaryl compounds as protein kinase inhibitors
US20100160283A1 (en) * 2004-10-18 2010-06-24 Amgen Inc. Heteroaryl-substituted alkyne compounds and method of use
US20110098275A1 (en) * 2009-04-21 2011-04-28 Boehringer Ingelheim International Gmbh 5-alkynyl-pyridines

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070072874A1 (en) * 2003-02-26 2007-03-29 Sugen, Inc. Aminoheteroaryl compounds as protein kinase inhibitors
US20100160283A1 (en) * 2004-10-18 2010-06-24 Amgen Inc. Heteroaryl-substituted alkyne compounds and method of use
US20110098275A1 (en) * 2009-04-21 2011-04-28 Boehringer Ingelheim International Gmbh 5-alkynyl-pyridines

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10053477B2 (en) 2014-07-04 2018-08-21 Qilu Pharmaceutical Co., Ltd. Spirocyclic aryl phosphorus oxide and aryl phosphorus sulfide
US11040027B2 (en) 2017-01-17 2021-06-22 Heparegenix Gmbh Protein kinase inhibitors for promoting liver regeneration or reducing or preventing hepatocyte death
CN107879966A (en) * 2017-10-16 2018-04-06 扬子江药业集团有限公司 A kind of preparation method of sitafloxacin key intermediate
CN107879966B (en) * 2017-10-16 2020-01-10 扬子江药业集团有限公司 Preparation method of sitafloxacin key intermediate

Similar Documents

Publication Publication Date Title
US9598400B2 (en) Substituted quinoline compounds and methods of use
US8969388B1 (en) Substituted pyrazolone compounds and methods of use
AU2012223639A1 (en) Substituted quinoline compounds and methods of use
US9326975B2 (en) Substituted pyrazolone compounds and methods of use
EP2958564B1 (en) Heteroaromatic compounds as pi3 kinase modulators
WO2013180949A1 (en) Substituted quinoline compounds and methods of use
WO2013138210A1 (en) Substituted cyclic compounds and methods of use
WO2014022128A1 (en) Pi3 kinase modulators and methods of use
WO2014089324A1 (en) Substituted cyclic compounds and methods of use
WO2014193647A2 (en) Alkenyl compounds and methods of use
WO2014089280A1 (en) Alkynyl compounds and methods of use
WO2015034729A1 (en) Substituted pyridine compounds and methods of use
WO2013177092A1 (en) Substituted alkynyl pyridine compounds and methods of use
WO2013148537A1 (en) Substituted spirobicyclic compounds and methods of use
CN103319468B (en) The spiral shell dicyclic compound replaced and using method and purposes

Legal Events

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

Ref document number: 13794095

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 13794095

Country of ref document: EP

Kind code of ref document: A1