WO2010144338A1 - Triazine derivatives and their therapeutical applications - Google Patents

Triazine derivatives and their therapeutical applications Download PDF

Info

Publication number
WO2010144338A1
WO2010144338A1 PCT/US2010/037570 US2010037570W WO2010144338A1 WO 2010144338 A1 WO2010144338 A1 WO 2010144338A1 US 2010037570 W US2010037570 W US 2010037570W WO 2010144338 A1 WO2010144338 A1 WO 2010144338A1
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
compound
mmol
pharmaceutically acceptable
mixture
Prior art date
Application number
PCT/US2010/037570
Other languages
French (fr)
Inventor
Chunlin Tao
Qinwei Wang
Laxman Nallan
Tulay Polat
Lukasz Koroniak
Neil Desai
Original Assignee
Abraxis Bioscience, Llc
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 Abraxis Bioscience, Llc filed Critical Abraxis Bioscience, Llc
Priority to JP2012515010A priority Critical patent/JP2012529511A/en
Priority to CN201080034890.8A priority patent/CN102573485B/en
Priority to CA2764785A priority patent/CA2764785C/en
Priority to BRPI1010881A priority patent/BRPI1010881A2/en
Priority to AU2010259002A priority patent/AU2010259002B2/en
Priority to EP10786616A priority patent/EP2440050A4/en
Priority to CN201410571301.XA priority patent/CN105175409A/en
Priority to KR1020127000593A priority patent/KR101460095B1/en
Priority to US13/376,964 priority patent/US20120238576A1/en
Publication of WO2010144338A1 publication Critical patent/WO2010144338A1/en
Priority to IL216825A priority patent/IL216825A/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/53Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with three nitrogens as the only ring hetero atoms, e.g. chlorazanil, melamine
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing three or more hetero rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/425Thiazoles
    • A61K31/427Thiazoles not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/02Nasal agents, e.g. decongestants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/02Drugs for dermatological disorders for treating wounds, ulcers, burns, scars, keloids, or the like
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • A61P19/10Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease for osteoporosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • A61P27/06Antiglaucoma agents or miotics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/14Drugs for disorders of the endocrine system of the thyroid hormones, e.g. T3, T4
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/06Antianaemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/10Antioedematous agents; Diuretics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/04Inotropic agents, i.e. stimulants of cardiac contraction; Drugs for heart failure
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings

Definitions

  • the present invention relates generally to the use of compounds to treat a variety of disorders, diseases and pathologic conditions and more specifically to the use of triazine compounds to modulate protein kinases and for treating protein kinase-mediated diseases.
  • Protein kinases constitute a large family of structurally related enzymes that are responsible for the control of a variety of signal transduction processes within the cell. Protein kinases, containing a similar 250-300 amino acid catalytic domain, catalyze the phosphorylation of target protein substrates.
  • the kinases may be categorized into families by the substrates in the phosphorylate (e.g., protein-tyrosine, protein-serine/threonine, lipids, etc.). Tyrosine phosphorylation is a central event in the regulation of a variety of biological processes such as cell proliferation, migration, differentiation and survival. Several families of receptor and non-receptor tyrosine kinases control these events by catalyzing the transfer of phosphate from ATP to a tyrosine residue of specific cell protein targets.
  • families of receptor and non-receptor tyrosine kinases control these events by catalyzing the transfer of phosphate from ATP to a tyrosine residue of specific cell protein targets.
  • kinases in the protein kinase family include, without limitation, abl, Akt, bcr-abl, BIk, Brk, Btk, c-kit, c-Met, c-src, c-fms, CDKl, CDK2, CDK3, CDK4, CDK5, CDK6, CDK7, CDK8, CDK9, CDKlO, cRafl, CSFlR, CSK, EGFR, ErbB2, ErbB3, ErbB4, Erk, Fak, fes, FGFRl, FGFR2, FGFR3, FGFR4, FGFR5, Fgr, flt-1, Fps, Frk, Fyn, Hck, IGF-IR, INS-R, Jak, KDR, Lck, Lyn, MEK, p38, PDGFR, PIK, PKC, PYK2, ros, Tie, Tie-2, TRK, Yes, and Zap70.
  • kinase activity acts as molecular switches regulating cell proliferation, activation, and/or differentiation. Uncontrolled or excessive kinase activity has been observed in many disease states including benign and malignant proliferation disorders as well as diseases resulting from inappropriate activation of the immune system (autoimmune disorders), allograft rejection, and graft vs host disease.
  • Src kinase is involved in proliferation and migration responses in many cell types, cell activation, adhesion, motility, and survival, growth factor receptor signaling, and osteoclast activation (Biscardi et al, Adv. Cancer Res. (1999), 76, 61-119; Yeatman et al, Nat. Rev. Cancer (2004), 4, 470-480; Owens, D. W.; McLean et al., MoI. Biol. Cell (2000), 11, 51-64).
  • Members of the Src family include the following eight kinases in mammals: Src, Fyn, Yes, Fgr, Lyn, Hck, Lck, and BIk (Bolen et al., Annu. Rev.
  • SH4 domain contains the myristylation signals that guide the Src molecule to the cell membrane.
  • Src proteins This unique domain of Src proteins is responsible for their specific interaction with particular receptors and protein targets (Thomas et al., Annu Rev Cell Dev Biol (1997), 13, 513-609).
  • the modulating regions, SH3 and SH2 control intra- as well as intermolecular interactions with protein substrates which affect Src catalytic activity, localization and association with protein targets (Pawson T., Nature (1995), 373, 573-580).
  • the kinase domain, SHl found in all proteins of the Src family, is responsible for the tyrosine kinase activity and has a central role in binding of substrates.
  • v-Src differs from cellular Src (c-Src) on the basis of the structural differences in C-terminal region responsible for regulation of kinase activity.
  • v-Src transforming protein
  • RSV Rous sarcoma virus
  • c- Src normal cellular protein
  • NSCLCs non-small cell lung cancers
  • bladder cancer bladder cancer
  • oesophageal cancer Jankowski et al., Gut, (1992), 33, 1033-8
  • prostate and ovarian cancer Wiener et al., Clin.
  • Src kinase modulates signal transduction through multiple oncogenic pathways, including EGFR, Her2/neu, PDGFR, FGFR, and VEGFR (Frame et al., Biochim. Biophys. Acta (2002), 1602, 114-130; Sakamoto et al., Jpn J Cancer Res, (2001), 92: 941-946).
  • Src kinase inhibitors may be useful anti-cancer agents (Abram et al., Exp. Cell Res., (2000), 254, 1). It is reported that inhibitors of src kinase had significant antiproliferative activity against cancer cell lines (M.M.
  • Src-family kinases are also important for signaling downstream of other immune cell receptors. Fyn, like Lck, is involved in TCR signaling in T cells (Appleby et al., Cell, (1992), 70, 751). Hck and Fgr are involved in Fc ⁇ receptor signaling leading to neutrophil activation (Vicentini et al., J. Immunol. (2002), 168, 6446). Lyn and Src also participate in Fc ⁇ receptor signaling leading to release of histamine and other allergic mediators (Turner, H. and Kinet, J-P Nature (1999), 402, B24). These-f ⁇ ndings suggest that Src family kinase inhibitors may be useful in treating allergic diseases and asthma.
  • Lck plays a role in T-cell signaling. Mice that lack the Lck gene have a poor ability to develop thymocytes. The function of Lck as a positive activator of T-cell signaling suggests that Lck inhibitors may be useful for treating autoimmune disease such as rheumatoid arthritis (Molina et al., Nature, (1992), 357, 161).
  • Hck is a member of the Src protein-tyrosine kinase family and is expressed strongly in macrophages, an important HIV target cell and its inhibition in HIV-infected macrophages might slow disease progression (Ye et al., Biochemistry, (2004), 43 (50), 15775 -15784).
  • Hck, Fgr and Lyn have been identified as important mediators of integrin signaling in myeloid leukocytes (Lowell et al., J. Leukoc. Biol, (1999), 65, 313). Inhibition of these kinase mediators may therefore be useful for treating inflammation (Boschelli et al., Drugs of the Future (2000), 25(7), 717).
  • BCR-ABL encodes the BCR-AEL protein, a constitutively active cytoplasmic tyrosine kinase present in 90% of all patients with chronic myelogenous leukemia (CML) and in 15-30% of adult patients with acute lymphoblastic leukemia (ALL). Numerous studies have demonstrated that the activity of BCR-ABL is required for the cancer causing ability of this chimeric protein.
  • Src kinases play a role in the replication of hepatitis B virus.
  • the virally encoded transcription factor HBx activates Src in a step required for propagation of the virus (Klein et al., EMBOJ. (1999), 18, 5019; Klein et al., MoI. Cell. Biol. (1997), 17, 6427).
  • Some genetic and biochemical data clearly demonstrate that Src-family tyrosine kinases serve as a critical signal relay, via phosphorylation of c-Cbl, for fat accumulation, and provide potential new strategies for treating obesity (Sun et al., Biochemistry, (2005), 44 (44), 14455 -14462).
  • Src inhibitors are also being pursued for the treatment of other diseases including osteoporosis and stroke (Susva et al., Trends Pharmacol. ScL (2000), 21, 489-495; Paul et al., Nat. Med. (2001), 7, 222-227). [0018] It is also possible that inhibitors of the Src kinase activity are useful in the treatment of osteoporosis (Soriano et al., Cell (1991), 64, 693; Boyce et al. J Clin. Invest (1992), 90, 1622; Owens et al., MoI. Biol.
  • src family kinases participate in signal transduction in several cell types. For example, fyn, like Ick, is involved in T-cell activation. Hck and fgr are involved in Fe gamma receptor mediated oxidative burst of neutrophils.
  • T cells play a pivotal role in the regulation of immune responses and are important for establishing immunity to pathogens.
  • T cells are often activated during inflammatory autoimmune diseases, such as rheumatoid arthritis, inflammatory bowel disease, type I diabetes, multiple sclerosis, Sjogren's disease, myasthenia gravis, psoriasis, and lupus. T cell activation is also an important component of transplant rejection, allergic reactions, and asthma.
  • T cells are activated by specific antigens through the T cell receptor, which is expressed on the cell surface. This activation triggers a series of intracellular signaling cascades mediated by enzymes expressed within the cell (Kane et al. Current Opinion in Immunol. (2000), 12, 242). These cascades lead to gene regulation events that result in the production of cytokines, like interleukin-2 (IL-2). IL-2 is a necessary cytokine in T cell activation, leading to proliferation and amplification of specific immune responses. [0022] Therefore, Src kinase and other kinase have become interesting targets for drug discovery (Parang et al., Expert Opin. Ther. Pat.
  • 6,440,965 disclosed substituted pyrimidine derivatives in the treatment of neurodegenerative or neurological disorders
  • PCT WO 02/08205 reported the pyrimidine derivatives having neurotrophic activity
  • PCT WO 03/014111 disclosed arylpiperazines and arylpiperidines and their use as metalloproteinase inhibiting agents
  • PCT WO 03/024448 described compounds as inhibitors of histone deacetylase enzymatic activity
  • PCT WO 04/058776 disclosed compounds which possess anti-angiogenic activity.
  • PCT WO 01/94341 and WO 02/16352 disclosed Src kinase inhibitors of quinazoline derivatives.
  • PCT WO03/026666Aland WO03/018021A1 disclosed pyrimidinyl derivatives as kinase inhibitors.
  • U.S. Pat. No 6498165 reported Src kinase inhibitor compounds of pyrimidine compounds.
  • Peptides as Src Tyrosine Kinase Inhibitors is reported recently (Kumar et al., J Med. Chem., (2006), 49 (11), 3395 -3401).
  • the quinolinecarbonitriles derivatives was reported to be potent dual Inhibitors of Src and AbI Kinases (Diane et al., J Med. Chem., (2004), 47 (7), 1599 -1601).
  • Many inhibitors of kinases are known , there exists a need for new treatment options for conditions associated with protein kinases.
  • the present invention provides an antitumor agent comprising a triazine derivative as described in formula (I) or formula (II), pharmaceutically-acceptable formulations thereof, methods for making novel compounds and compositions for using the compounds.
  • the compounds and compositions comprising the compounds of formula (I) or formula (II) have utility in treatment of a variety of diseases.
  • the combination therapy described herein may be provided by the preparation of the triazine derivative of formula (I) or formula (II) and the other therapeutic agent as separate pharmaceutical formulations followed by the administration thereof to a patient simultaneously, semi-simultaneously, separately or over regular intervals.
  • the present invention also provides methods for using certain chemical compounds such as kinase inhibitors in the treatment of various diseases, disorders, and pathologies, for example, cancer, and vascular disorders, such as myocardial infarction (MI), stroke, or ischemia.
  • the triazine compounds described in this invention may block the enzymatic activity of some or many of the members of the Src family, in addition to blocking the activity of other receptor and non-receptor kinase.
  • Such compounds may be beneficial for treatment of the diseases where disorders affect cell motility, adhesion, and cell cycle progression, and in addition, diseases with related hypoxic conditions, osteoporosis and conditions, which result from or are related to increases in vascular permeability, inflammation or respiratory distress, tumor growth, invasion, angiogenesis, metastases and apoptosis.
  • the present invention comprises compounds as shown in formula (I)
  • A, B, W is selected from S, O, NR 4 , CR 4 or L-R 3 ; R4 is independently selected from hydrogen or an optionally substituted C 1-4 aliphatic group.
  • R 1 represents hydrogen, halogen, hydroxy, amino, cyano, alkyl, cycloalkyl, alkenyl, alkynyl, alkylthio, aryl, arylalkyl, heterocyclic, heteroaryl, heterocycloalkyl, alkylsulfonyl, alkoxycarbonyl and alkylcarbonyl.
  • R 2 is selected from:
  • R 5 represents hydrogen, C 1 -C 4 alkyl, oxo
  • X is CH, when R 6 is hydrogen; or X-R 6 is O; or X is N, R 6 represents groups of hydrogen, Ci-C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -Ci 0 aryl or heteroaryl, (C 3 -C 7 cycloalkyl)Ci-C 4 alkyl, d- C 6 haloalkyl, Ci-C 6 alkoxy, C 1 - C 6 alkylthio, C 2 -C 6 alkanoyl, Ci-C 6 alkoxycarbonyl, C 2 - C 6 alkanoyloxy, mono- and di-(C 3 -C 8 cycloalkyl)amino — C 0- C 4 alkyl, (4- to 7- membered heterocycle)Co-C 4 alkyl, Ci-C 6 alkylsulfonyl, mono- and di-(Ci-C 6 alkyl) s
  • R 3 is selected from:
  • Ci-C 6 alkyl C 2 -C 6 alkenyl, C 2 -C 6 alkynyl;
  • Ar represents heteroaryl or aryl, each of which is substituted with from 0 to 4 substituents independently chosen from:
  • R 7 represents hydrogen, alkyl, cycloalkyl, alkenyl, alkynyl, alkylthio, aryl, arylalkyl.
  • the present invention also comprises compounds of formula (II)
  • Y is selected from -OR 4 , -NR 4 R 5 , and -Q-R 3 ;
  • Q is selected from cycloalkyl and heterocycloalkyl, each of which is optionally substituted with Ci-C 6 alkyl or oxo;
  • R 3 is selected from H, Cj-C 6 alkyl, Ci-C 6 alkyl-R 6 , aryl, and heteroaryl, each of which is optionally substituted with Ci-C 6 alkyl, halo, trifluoromethyl, or oxo;
  • R 4 and R 5 are each independently selected from H, C 1 -C 6 alkyl-R 6 , aryl, and heteroaryl; R is selected from hydroxy, cyano, aryl, heteroaryl, cycloalkyl, heterocycloalkyl, - NH 2 , HiOnO(C 1 -C 6 )alkylamino, di(Ci-C 6 )alkylamino, and C 1 -C 6 alkoxy;
  • X is -NH-Ar 1 -R 1 ;
  • Ar 1 is selected from aryl and heteroaryl, each of which is optionally substituted with C 1 -C 6 alkyl or halo;
  • W is selected from C 1 -C 6 alkyl, cycloalkyl, and -(CH 2 )Ar 1 ;
  • Z is selected from H, Ci-C 6 alkyl, aryl, and heteroaryl.
  • the invention further comprises compounds of formula (II)
  • Y is selected from -OR 4 , -NR 4 R 5 , and -Q-R 3 ;
  • Q is selected from morpholinyl, piperazinyl and piperidinyl;
  • R 3 is selected from H, C 1 -C 6 alkyl, hydroxy(Ci-C 6 )alkyl, cyano(Ci-C 6 )alkyl, pyridinylmethyl, pyridinyl, phenyl, trifluoromethylphenyl, and oxo;
  • R 4 and R 5 are each independently selected from H, Ci-C 6 alkyl -R , and phenyl;
  • R 6 is selected from hydroxy, morpholinyl, di(Ci-C 6 )alkylamino, imidazolyl, and Q- C 6 alkoxy;
  • X is -NH-Ar 1 -R 1 ;
  • Ar 1 is selected from thiazolyl, oxazolyl, oxadiazolyl, methyl-imidazolyl, pyrazolyl;
  • W is selected from Cj-C 6 alkyl and -(CH 2 ) n Ph optionally substituted with Ci-C 6 alkyl or halo;
  • Z is selected from H, Ci-C 6 alkyl, and phenyl.
  • the invention also comprises compounds of formula (II)
  • Y is selected from -OR 4 , -NR 4 R 5 , and -Q-R 3 ;
  • Q is selected from morpholinyl, piperazinyl and piperidinyl;
  • R 3 is selected from H, Cj-C 6 alkyl, hydroxy(Ci-C 6 )alkyl, cyano(Cj-C 6 )alkyl, pyridinylmethyl, pyridinyl, phenyl, trifluoromethylphenyl, and oxo;
  • R 4 and R 5 are each independently selected from H, Cj-C 6 alkyl-R 6 , and phenyl;
  • R 6 is selected from hydroxy, morpholinyl, di(Cj-C 6 )alkylamino, imidazolyl, and C 1 - C 6 alkoxy;
  • X is -NH-Ar'-R 1 ;
  • Ar 1 is selected from thiazolyl, oxazolyl, oxadiazolyl, methyl-imidazolyl, pyrazolyl;
  • W is selected from Cj-C 6 alkyl, cycloalkyl, and -(CH 2 )Ar 2 ;
  • Ar 2 is phenyl, optionally substituted with Cj-C 6 alkyl or halo;
  • Z is selected from H, Cj-C 6 alkyl, and phenyl.
  • alkyl herein alone or as part of another group refers to a monovalent alkane (hydrocarbon) derived radical containing from 1 to 12 carbon atoms unless otherwise defined. Alkyl groups may be substituted at any available point of attachment. An alkyl group substituted with another alkyl group is also referred to as a "branched alkyl group”.
  • Exemplary alkyl groups include methyl, ethyl, propyl, isopropyl, n-butyl, t-butyl, isobutyl, pentyl, hexyl, isohexyl, heptyl, dimethylpentyl, octyl, 2,2,4-trimethylpentyl, nonyl, decyl, undecyl, dodecyl, and the like.
  • substituents include but are not limited to one or more of the following groups: alkyl, aryl, halo (such as F, Cl, Br, I), haloalkyl (such as CCl 3 or CF 3 ), alkoxy, alkylthio, hydroxy, carboxy (-COOH), alkyloxycarbonyl (-C(O)R), alkylcarbonyloxy (- OCOR), amino (-NH 2 ), carbamoyl (-NHCOOR- or -0C0NHR-), urea (- NHCONHR-) or thiol (-SH).
  • alkyl groups are substituted with, for example, amino, heterocycloalkyl, such as morpholine, piperazine, piperidine, azetidine, hydroxyl, methoxy, or heteroaryl groups such as pyrrolidine.
  • “Alkyl” also includes cycloalkyl.
  • cycloalkyl herein alone or as part of another group refers to fully saturated and partially unsaturated hydrocarbon rings of 3 to 9, preferably 3 to 7 carbon atoms.
  • the examples include cyclopropyl, cyclobutyl, cyclopentyl and cyclohexyl, and like. Further, a cycloalkyl may be substituted.
  • alkenyl herein alone or as part of another group refers to a hydrocarbon radical straight, branched or cyclic containing from 2 to 12 carbon atoms and at least one carbon to carbon double bond.
  • groups include the vinyl, allyl, 1-propenyl, isopropenyl, 2-methyl-l-propenyl, 1-butenyl, 2-butenyl, 3-butenyl, 1-pentenyl, 2-pentenyl, 3-pentenyl, 4-pentenyl, 1-hexenyl, 2-hexenyl, 3-hexenyl, 4-hexenyl, 5-hexenyl, 1 -heptenyl, and like.
  • Alkenyl groups may also be substituted at any available point of attachment.
  • exemplary substituents for alkenyl groups include those listed above for alkyl groups, and especially include C 3 to C 7 cycloalkyl groups such as cyclopropyl, cyclopentyl and cyclohexyl, which may be further substituted with, for example, amino, oxo, hydroxyl, etc.
  • alkynyl refers to straight or branched chain alkyne groups, which have one or more unsaturated carbon-carbon bonds, at least one of which is a triple bond.
  • Alkynyl groups include C 2 -C 8 alkynyl, C 2 -C 6 alkynyl and C 2 -C 4 alkynyl groups, which have from 2 to
  • alkynyl group examples include ethenyl, propenyl, isopropenyl, butenyl, isobutenyl, pentenyl, and hexenyl.
  • Alkynyl groups may also be substituted at any available point of attachment.
  • Exemplary substituents for alkynyl groups include those listed above for alkyl groups such as amino, alkylamino, etc.
  • alkoxy alone or as part of another group denotes an alkyl group as described above bonded through an oxygen linkage (-O-).
  • Preferred alkoxy groups have from
  • Examples of such groups include the methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, isobutoxy, sec-butoxy, tert-butoxy, n-pentyloxy, isopentyloxy, n- hexyloxy, cyclohexyloxy, n-heptyloxy, n-octyloxy and 2-ethylhexyloxy.
  • alkylthio refers to an alkyl group as described above attached via a sulfur bridge.
  • Preferred alkoxy and alkylthio groups are those in which an alkyl group is attached via the heteroatom bridge.
  • Preferred alkylthio groups have from 1 to 8 carbon atoms. Examples of such groups include the methylthio, ethylthio, n-propythiol, n-butylthiol, and like.
  • alkoxycarbonyl herein alone or as part of another group denotes an alkoxy group bonded through a carbonyl group.
  • An alkoxycarbonyl radical is represented by the formula: -C(O)OR, where the R group is a straight or branched C)-C 6 alkyl group, cycloalkyl, aryl, or heteroaryl.
  • alkylcarbonyl herein alone or as part of another group refers to an alkyl group bonded through a carbonyl group or -C(O)R.
  • arylalkyl herein alone or as part of another group denotes an aromatic ring bonded through an alkyl group (such as benzyl) as described above.
  • aryl herein alone or as part of another group refers to monocyclic or bicyclic aromatic rings, e.g. phenyl, substituted phenyl and the like, as well as groups which are fused, e.g., napthyl, phenanthrenyl and the like.
  • An aryl group thus contains at least one ring having at least 6 atoms, with up to five such rings being present, containing up to 20 atoms therein, with alternating (resonating) double bonds between adjacent carbon atoms or suitable heteroatoms.
  • Aryl groups may optionally be substituted with one or more groups including, but not limited to halogen such as I, Br, F, or Cl; alkyl, such as methyl, ethyl, propyl, alkoxy, such as methoxy or ethoxy, hydroxy, carboxy, carbamoyl, alkyloxycarbonyl, nitro, alkenyloxy, trifluoromethyl, amino, cycloalkyl, aryl, heteroaryl, cyano, alkyl S(O) n ,
  • aromatic refers to a cyclically conjugated molecular entity with a stability, due to derealization, significantly greater than that of a hypothetical localized structure, such as the Kekule structure.
  • amino herein alone or as part of another group refers to -NH 2 .
  • amino may optionally be substituted with one or two substituents, which may be the same or different, such as alkyl, aryl, arylalkyl, alkenyl, alkynyl, heteroaryl, heteroarylalkyl, cycloheteroalkyl, cycloheteroalkylalkyl, cycloalkyl, cycloalkylalkyl, haloalkyl, hydroxyalkyl, alkoxyalkyl, thioalkyl, carbonyl or carboxyl.
  • substituents may be further substituted with a carboxylic acid, any of the alkyl or aryl substituents set out herein.
  • the amino groups are substituted with carboxyl or carbonyl to form N-acyl or
  • alkylsulfonyl refers to groups of the formula (SO 2 )-alkyl, in which the sulfur atom is the point of attachment.
  • alkylsulfonyl groups include C 1 - C 6 alkylsulfonyl groups, which have from 1 to 6 carbon atoms.
  • Methylsulfonyl is one representative alkylsulfonyl group.
  • heteroatom refers to any atom other than carbon, for example, N, O, or
  • heteroaryl herein alone or as part of another group refers to substituted and unsubstituted aromatic 5 or 6 membered monocyclic groups, 9 or 10 membered bicyclic groups, and 11 to 14 membered tricyclic groups which have at least one heteroatom (O, S or
  • Each ring of the heteroaryl group containing a heteroatom can contain one or two oxygen or sulfur atoms and/or from one to four nitrogen atoms provided that the total number of heteroatoms in each ring is four or less and each ring has at least one carbon atom.
  • the fused rings completing the bicyclic and tricyclic groups may contain only carbon atoms and may be saturated, partially saturated, or unsaturated.
  • the nitrogen and sulfur atoms may optionally be oxidized and the nitrogen atoms may optionally be quaternized.
  • Heteroaryl groups which are bicyclic or tricyclic must include at least one fully aromatic ring but the other fused ring or rings may be aromatic or non- aromatic.
  • the heteroaryl group may be attached at any available nitrogen or carbon atom of any ring.
  • monocyclic heteroaryl groups include pyrrolyl, pyrazolyl, pyrazolinyl, imidazolyl, oxazolyl, diazolyl, isoxazolyl, thiazolyl, thiadiazolyl, isothiazolyl, furanyl, thienyl, oxadiazolyl, pyridyl, pyrazinyl, pyrimidinyl, pyridazinyl, triazinyl and the like.
  • bicyclic heteroaryl groups include indolyl, benzothiazolyl, benzodioxolyl, benzoxaxolyl, benzothienyl, quinolinyl, tetrahydroisoquinolinyl, isoquinolinyl, benzimidazolyl, benzopyranyl, indolizinyl, benzofuranyl, chromonyl, coumarinyl, benzopyranyl, cinnolinyl, quinoxalinyl, indazolyl, pyrrolopyridyl, dihydroisoindolyl, tetrahydroquinolinyl and the like.
  • tricyclic heteroaryl groups include carbazolyl, benzidolyl, phenanthrollinyl, acridinyl, phenanthridinyl, xanthenyl and the like.
  • heterocycle or “heterocycloalkyl” herein alone or as part of another group refers to a cycloalkyl group (nonaromatic) in which one of the carbon atoms in the ring is replaced by a heteroatom selected from O, S or N.
  • the "heterocycle” has from 1 to 3 fused, pendant or spiro rings, at least one of which is a heterocyclic ring (i.e.
  • one or more ring atoms is a heteroatom, with the remaining ring atoms being carbon).
  • the heterocyclic ring may be optionally substituted which means that the heterocyclic ring may be substituted at one or more substitutable ring positions by one or more groups independently selected from alkyl (preferably lower alkyl), heterocycloalkyl, heteroaryl, alkoxy (preferably lower alkoxy), nitro, monoalkylamino (preferably a lower alkylamino), dialkylamino (preferably a alkylamino), cyano, halo, haloalkyl (preferably trifluoromethyl), alkanoyl, aminocarbonyl, monoalkylaminocarbonyl, dialkylaminocarbonyl, alkyl amido (preferably lower alkyl amido), alkoxyalkyl (preferably a lower alkoxy; lower alkyl), alkoxycarbonyl (preferably a lower alkoxycarbonyl), alkylcarbonyloxy (preferably a lower alkyl
  • a heterocyclic ring comprises 1-4 heteroatoms; within certain embodiments each heterocyclic ring has 1 or 2 heteroatoms per ring.
  • Each heterocyclic ring generally contains from 3 to 8 ring members (rings having from to 7 ring members are recited in certain embodiments), and heterocycles comprising fused, pendant or spiro rings typically contain from 9 to 14 ring members which consists of carbon atoms and contains one, two, or three heteroatoms selected from nitrogen, oxygen and/or sulfur.
  • heterocycle or “heterocycloalkyl groups include piperazine, piperidine, morpholine, thiomorpholine, pyrrolidine, imidazolidine and thiazolide.
  • substituted refers to a molecular moiety that is covalently bonded to an atom within a molecule of interest.
  • a “ring substituent” may be a moiety such as a halogen, alkyl group, haloalkyl group or other group discussed herein that is covalently bonded to an atom (preferably a carbon or nitrogen atom) that is a ring member.
  • aryl or heterocyclyl or other group may be substituted at one or more substitutable positions by one or more groups independently selected from alkyl (preferably lower alkyl), alkoxy (preferably lower alkoxy), nitro, monoalkylamino (preferably with one to six carbons), dialkylamino (preferably with one to six carbons), cyano, halo, haloalkyl (preferably trifluoromethyl), alkanoyl, aminocarbonyl, monoalkylaminocarbonyl, dialkylaminocarbonyl, alkyl amido (preferably lower alkyl amido), alkoxyalkyl (preferably a lower alkoxy and lower alkyl), alkoxycarbonyl (preferably a lower alkoxycarbonyl), alkylcarbonyloxy (preferably a lower alkylcarbonyloxy) and aryl (preferably phenyl), said aryl being optionally substituted by halo, lower alkyl and lower
  • a dash (“-") that is not between two letters or symbols is used to indicate a point oft attachment for a substituent. For example, -CONH2 is attached through the carbon atom.
  • a dashed cycle that locates inside of a heterocyle ring is used to indicate a conjugated system. The bonds between two atomes may be single bond or double bond.
  • anticancer agent includes any known agent that is useful for the treatment of cancer including, but is not limited, Acivicin; Aclarubicin; Acodazole Hydrochloride; AcrQnine; Adozelesin; Aldesleukin; Altretamine; Ambomycin; Ametantrone Acetate; Aminoglutethimide; Amsacrine; Anastrozole; Anthramycin; Asparaginase; Asperlin; Azacitidine; Azetepa; Azotomycin; Batimastat; Benzodepa; Bicalutamide; Bisantrene Hydrochloride; Bisnafide Dimesylate; Bizelesin; Bleomycin Sulfate; Brequinar Sodium; Bropirimine; Busulfan; Cactinomycin; Calusterone; Caracemide; Carbetimer; Carboplatin; Carmustine; Carubicin Hydrochloride; Carzelesin; Cedef ⁇
  • Glucuronate Triptorelin; Tubulozole Hydrochloride; Uracil Mustard; Uredepa; Vapreotide;
  • Verteporfin Vinblastine Sulfate; Vincristine Sulfate; Vindesine; Vindesine Sulfate;
  • Vinepidine Sulfate Vinglycinate Sulfate; Vinleursine Sulfate; Vinorelbine Tartrate;
  • Vinrosidine Sulfate Vinzolidine Sulfate; Vorozole; Zeniplatin; Zinostatin; and Zorubicin
  • kinase refers to any enzyme that catalyzes the addition of phosphate groups to a protein residue; for example, serine and threonine kineses catalyze the addition of phosphate groups to serine and threonine residues.
  • Src kinase refers to the related homologs or analogs belonging to the mammalian family of Src kineses, including, for example, c-Src, Fyn, Yes and Lyn kineses and the hematopoietic-restricted kineses Hck, Fgr,
  • terapéuticaally effective amount refers to the amount of the compound or pharmaceutical composition that will elicit the biological or medical response of a tissue, system, animal or human that is being sought by the researcher, veterinarian, medical doctor or other clinician, e.g., restoration or maintenance of vasculostasis or prevention of the compromise or loss or vasculostasis; reduction of tumor burden; reduction of morbidity and/or mortality.
  • the term 'pharmaceutically acceptable refers to the fact that the carrier, diluent or excipient must be compatible with the other ingredients of the formulation and not deleterious to the recipient thereof.
  • administering a compound refers to the act of providing a compound of the invention or pharmaceutical composition to the subject in need of treatment.
  • protected refers that the group is in modified form to preclude undesired side reactions at the protected site.
  • Suitable protecting groups for the compounds of the present invention will be recognized from the present application taking into account the level of skill in the art, and with reference to standard textbooks, such as Greene, T. W. et al.,
  • salts of a compound recited herein is an acid or base salt that is suitable for use in contact with the tissues of human beings or animals without excessive toxicity or carcinogenicity, and preferably without irritation, allergic response, or other problem or complication.
  • Such salts include mineral and organic acid salts of basic residues such as amines, as well as alkali or organic salts of acidic residues such as carboxylic acids.
  • Specific pharmaceutical salts include, but are not limited to, salts of acids such as hydrochloric, phosphoric, hydrobromic, malic, glycolic, fumaric, sulfuric, sulfamic, sulfanilic, formic, toluenesulfonic, methanesulfonic, benzene sulfonic, ethane disulfonic, 2- hydroxyethylsulfonic, nitric, benzoic, 2-acetoxybenzoic, citric, tartaric, lactic, stearic, salicylic, glutamic, ascorbic, pamoic, succinic, fumaric, maleic, propionic, hydroxymaleic, hydroiodic, phenylacetic, alkanoic such as acetic, HOOC- (CH 2 ) n -C00H where n is 0-4, and the like.
  • acids such as hydrochloric, phosphoric, hydrobromic, malic, glycolic, fumaric,
  • pharmaceutically acceptable cations include, but are not limited to sodium, potassium, calcium, aluminum, lithium and ammonium.
  • a pharmaceutically acceptable acid or base salt can be synthesized from a parent compound that contains a basic or acidic moiety by any conventional chemical method. Briefly, such salts can be prepared by reacting the free acid or base forms of these compounds with a stoichiometric amount of the appropriate base or acid in water or in an organic solvent, or in a mixture of the two; generally, the use of nonaqueous media, such as ether, ethyl acetate, ethanol, isopropanol or acetonitrile, is preferred.
  • each compound of formula (I) or formula (II) may, but need not, be formulated as a hydrate, solvate or non- covalent complex.
  • the various crystal forms and polymorphs are within the scope of the present invention.
  • prodrug refers a compound that may not fully satisfy the structural requirements of the compounds provided herein, but is modified in vivo, following administration to a patient, to produce a compound of formula (I) or formula (II), or other formula provided herein.
  • a prodrug may be an acylated derivative of a compound as provided herein.
  • Prodrugs include compounds wherein hydroxy, amine or thiol groups are bonded to any group that, when administered to a mammalian subject, cleaves to form a free hydroxy, amino, or thiol group, respectively.
  • prodrugs include, but are not limited to, acetate, formate and benzoate derivatives of alcohol and amine functional groups within the compounds provided herein.
  • Prodrugs of the compounds provided herein may be prepared by modifying functional groups present in the compounds in such a way that the modifications are cleaved in vivo to yield the parent compounds.
  • Groups that are "optionally substituted” are unsubstituted or are substituted by other than hydrogen at one or more available positions.
  • Such optional substituents include, for example, hydroxy, halogen, cyano, nitro, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 - C 6 alkynyl, C 1 -
  • Optional substitution is also indicated by the phrase "substituted with from 0 to X substituents," where X is the maximum number of possible substituents. Certain optionally substituted groups are substituted with from 0 to 2, 3 or 4 independently selected substituents.
  • R 1 groups of formula (I) are listed below:
  • R 2 groups of formula (I) are listed below:
  • R 3 groups of formula (I) are listed below, wherein the substitute may be the specific ones as defined here or may be one or multiple substitutes as defined above:
  • R 4 is independently selected from hydrogen or an optionally substituted Ci -4 aliphatic group.
  • the compounds of the invention may be compounds of formula (I) wherein
  • R 2 is selected from: (i)amino, alkyl amino, aryl amino, heteroaryl amino; (ii) C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl; (iii) heterocyclic, herteroaryl; and (iv) groups of the formula (Ia):
  • R 5 represents hydrogen, Cj-C 4 alkyl, oxo
  • X is CH, when R 6 is hydrogen; or X-R 6 is O; or X is N, R 6 represents groups of hydrogen, Cj-C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -Cj O aryl or heteroaryl, (C 3 -C 7 cycloalkyl)Cj-C 4 alkyl, C 1 - C 6 haloalkyl, C 1 -C 6 alkoxy, Cj- C 6 alkylthio, C 2 -C 6 alkanoyl, Cj- C 6 alkoxycarbonyl, C 2 - C 6 alkanoyloxy, mono- and di-(C 3 -C 8 cycloalkyl)aminoC 0 -C 4 alkyl, (4- to 7- membered heterocycle)C 0 -C 4 alkyl, Cj-C 6 alkylsulfonyl, mono- and di-(Cj- C
  • R 4 is independetly selected from hydrogen or an optionally substituted Cj -4 aliphatic group.
  • R 3 is selected from:
  • Ar represents heteroaryl or aryl, each of which is substituted with from O to 4 substituents independently chosen from: (1) halogen, hydroxy, amino, cyano, -COOH, -SO 2 NH 2 , oxo, nitro and alkoxycarbonyl; and
  • -C 6 alkyl sulfonamido and mono- and di-(Ci-C 6 alkyl)aminocarbonyl; phenylC 0 -C 4 alkyl and (4- to 7-membered heterocycle) — C 0 -C 4 alkyl, each of which is substituted with from O to 4 secondary substituents independently chosen from halogen, hydroxy, cyano, oxo, imino, C 1 - C 4 alkyl, C 1 -C 4 alkoxy and Cj-C 4 haloalkyl.
  • A, B, W independently represents S, or O, or NR 4 , or CR 4 ;
  • R 7 represents hydrogen, alkyl, cycloalkyl, alkenyl, alkynyl, alkylthio, aryl, arylalkyl.
  • the compounds of the invention may be compounds of formula (I) wherein
  • R 1 represents -CH 3 , -CH 2 CH 3 , -CH 2 CH(CH 3 ) 2 , Cyclopropanyl, Ph.
  • R 2 is selected from: amino, alkyl amino, aryl amino, heteroaryl amino and groups of the formula (Ia):
  • R 5 represents hydrogen, C 1 -C 4 alkyl, oxo
  • X is CH, when R 6 is hydrogen; or X-R 6 is O; or X is N, R 6 represents groups of hydrogen, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -Ci 0 aryl or heteroaryl, (C 3 -C 7 cycloalkyl)Ci-C 4 alkyl, C 1 - C 6 haloalkyl, C 1 -C 6 alkoxy, C 1 - C 6 alkylthio, C 2 -C 6 alkanoyl, Cr C 6 alkoxycarbonyl, C 2 - C 6 alkanoyloxy, mono- and di-(C 3 -C 8 cycloalkyl)aminoC 0 -C 4 alkyl, (4- to 7- membered heterocycle)C 0 -C 4 alkyl, C 1 -C 6 alkylsulfonyl, mono- and CU-(C
  • R 4 is independetly selected from hydrogen or an optionally substituted C 1-4 aliphatic group.
  • R 3 is selected from heteroaryl or aryl, each of which is substituted with from O to 4 substituents independently chosen from:
  • A, B, W independently represents S, or O, or NR 4 , or CR 4 ;
  • K is selected from i) absence; ii) O, S, SO, SO 2 ;
  • R 7 represents hydrogen, alkyl, cycloalkyl, alkenyl, alkynyl, alkylthio, aryl, arylalkyl.
  • R 1 represents, -CH 3 , -CH 2 CH 3 ;
  • R 2 is selected from: alkyl amino, aryl amino, heteroaryl amino and groups of the formula (Ia): wherein:
  • R 5 represents hydrogen, Ci-C 4 alkyl, oxo
  • R 6 represents groups of hydrogen, Ci-C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -Ci 0 aryl or heteroaryl, (C 3 -C 7 cycloalkyl)Ci-C 4 alkyl, C 1 - C 6 haloalkyl, Cj-C 6 alkoxy, Ci- C 6 alkylthio, C 2 -C 6 alkanoyl, Ci- C 6 alkoxycarbonyl, C 2 - C 6 alkanoyloxy, mono- and di-(C 3 - C 8 cycloalkyl)aminoCo-C 4 alkyl, (4- to 7- membered heterocycle)C 0 -C 4 alkyl, Ci-C 6 alkylsulfonyl, mono- and di-(Ci- C 6 alkyl) sulfonamido, and mono- and CU-(C 1 - C 6 alkyl, C
  • R 4 is independetly selected from hydrogen or an optionally substituted C 1-4 aliphatic group.
  • R 3 is selected from heteroaryl or aryl, each of which is substituted with from O to 4 substituents independently chosen from:
  • A, B, W independently represents S, or O, or NR4, or CR4; K is selected from i) absence; ⁇ ) O, S; iii) NR7; R7 represents hydrogen, alkyl.
  • Preferred heterocyclic groups in compounds of formula (I) include
  • the present invention relates to a compound of formula (I) wherein Ri is methyl.
  • the present invention relates to a compound of formula (I) wherein Rj is ethyl.
  • the present invention relates to a compound of formula (I) wherein R] is isopropyl.
  • the present invention relates to a compound of formula (I) wherein Rj is phenyl.
  • the present invention relates to a compound of formula (I) wherein Rj is cyclopropanyl.
  • the present invention relates to a compound of formula (I) wherein R 2 is methyl-piperazinyl.
  • the present invention relates to a compound of formula (I) wherein R 2 is (2-hydroxylethyl)-piperazinyl.
  • the present invention relates to a compound of formula (I) wherein L is oxygen.
  • the present invention relates to a compound of formula (I) wherein L is CO.
  • the present invention relates to a compound of formula (I) wherein L is NHCO.
  • the present invention relates to a compound of formula (I) wherein L is CONH.
  • the present invention relates to a compound of formula (I) wherein L is NR 4 COC(R 4 ).
  • the present invention relates to a compound of formula (I) wherein L is NH.
  • the present invention relates to a compound of formula (I) wherein L is S.
  • the present invention relates to a compound of formula (I) wherein L is SO.
  • the present invention relates to a compound of formula (I) wherein L is SO 2 .
  • the present invention relates to a compound of formula (I) wherein A is N.
  • Examples of specific compounds of the present invention are those compounds defined in the following:
  • a method of preparing the inventive compounds is provided.
  • the compounds of the present invention can be generally prepared using cyanuric chloride as a starting material.
  • Compounds of formula (I) or formula (II) may contain various stereoisomers, geometric isomers, tautomeric isomers, and the like. All of possible isomers and their mixtures are included in the present invention, and the mixing ratio is not particularly limited.
  • the triazine derivative compounds of formula (I) or formula (II) in this invention can be prepared by known procedure in the prior art. The examples could be found in US Patent Application Publication No. 2005/0250945A1 ; US Patent Application Publication No. 2005/0227983 Al; PCT WO 05/007646A1; PCT WO 05/007648A2; PCT WO 05/003103 A2; PCT WO 05/011703 Al ; and J. Med. Chem. (2004), 47(19), 4649-4652. Starting materials are commercially available from suppliers such as Sigma-Aldrich Corp. (St. Louis, MO), or may be synthesized from commercially available precursors using established protocols.
  • reduction refers to the process of reducing a nitro functionality to an amino functionality, or the process of transforming an ester functionality to an alcohol.
  • the reduction of a nitro group can be carried out in a number of ways well known to those skilled in the art of organic synthesis including, but not limited to, catalytic hydrogenation, reduction with SnCl 2 and reduction with titanium bichloride.
  • the reduction of an ester group is typically performed using metal hydride reagents including, but not limited to, diisobutyl-aluminum hydride (DIBAL), lithium aluminum hydride (LAH), and sodium borohydride.
  • DIBAL diisobutyl-aluminum hydride
  • LAH lithium aluminum hydride
  • sodium borohydride sodium borohydride
  • hydrolyze refers to the reaction of a substrate or reactant with water. More specifically, “hydrolyze” refers to the conversion of an ester or nitrite functionality into a carboxylic acid. This process can be catalyzed by a variety of acids or bases well known to those skilled in the art of organic synthesis.
  • the compounds of formula (I) or formula (II) may be prepared by use of known chemical reactions and procedures. The following general preparative methods are presented to aid one of skill in the art in synthesizing the inhibitors, with more detailed examples being presented in the experimental section describing the working examples.
  • Heterocyclic amines are defined in formula (III). Some of heterocyclic amines are commercially available, others may be prepared by known procedure in the prior art (Katritzky, et al. Comprehensive Heterocyclic Chemistry; Permagon Press: Oxford, UK, 1984, March. Advanced Organic Chemistry, 3rd Ed.; John Wiley: New York, 1985), or by using common knowledge of organic chemistry.
  • heterocyclic amine with an amide link can be prepared from commercial compounds as illustrated in Scheme 1.
  • Route 1 the amine is first protected by Boc or other appropriate protecting group; after hydrolysis, the acid can be converted to corresponding amide; followed by removal of protecting group, the desired amine can be obtained.
  • Route 2 the acid, which is either commercially available, or made from its ester form, can also be converted to the desired compound (Ilia).
  • a lot of heterocyclic amines can be prepared by this way.
  • heterocyclic amines can also be generated using standard methods (March, J. Advanced Organic Chemistry, 4th Ed.; John Wiley, New York (1992); Larock, R.C. Comprehensive Organic Transformations, 2nd Ed., John Wiley, New York (1999); PCT No. WO 99/32106).
  • a metal catalyst such as Ni, Pd, or Pt
  • a hydride transfer agent such as formate, cyclohexadiene, or a borohydride
  • Nitroheteros may also be directly reduced using a strong hydride source, such as LAH, (Seyden-Penne. Reductions by the Alumino- and Borohydrides in Organic Synthesis; VCH Publishers: New York (1991)), or using a zero valent metal, such as Fe, Sn or Ca, often in acidic media.
  • LAH strong hydride source
  • a zero valent metal such as Fe, Sn or Ca
  • Many methods exist for the synthesis of nitroaryls March, J. Advanced Organic Chemistry, 4th Ed.; John Wiley, New York (1992); Larock, R.C. Comprehensive Organic Transformations, 2nd Ed., John Wiley, New York (1999)).
  • Nitroheteroaryls may be further elaborated prior to reduction. Nitroheteros substituted with potential leaving groups (eg. F, Cl, Br, etc.) may undergo substitution reactions on treatment with nucleophiles, such as thiolate (exemplified in Scheme 3) or phenoxide. Nitroaryls may also undergo Ullman-type coupling reactions (Scheme 3).
  • potential leaving groups eg. F, Cl, Br, etc.
  • nucleophiles such as thiolate (exemplified in Scheme 3) or phenoxide. Nitroaryls may also undergo Ullman-type coupling reactions (Scheme 3).
  • Scheme 4 illustrates one of the methods to prepare those heterocyclic amines as in Formula IHb, where L is carbonyl.
  • These heterocyclic amines are readily available from reactions of a heterocyclic amine with a substituted aryl carbonyl chloride. Acetyl protection of the amine, which can be easily removed after the Friedel-Crafts reaction, is preferred.
  • These carbonyl linked heterocyclic amines can be further converted to methylene (IIIc) or hydroxyl methylene (HId) linked ones by appropriate reduction.
  • 2-amino thiazole-5-carboxamide or 2-amino-oxazole- 5-carboxamide (HId) are available by the reaction of thiourea or urea with an appropriate ethoxyacrylamide in the presence of NBS, which can be made from the reaction of 3-ethyoxyacryloyl chloride with a corresponding amino compound R' -NH 2 .
  • the 3-ethyoxyacryloyl chloride can be prepared from the corresponding acid or ester.
  • Scheme 6 illustrated the synthesis method for compounds with alkyl or aryl as R 1 .
  • the 6-alkyl or aryl substituted dichloro-triazine (b) may be synthesized by the methods known in the art (e.g., J Med. Chem. 1999, 42, 805-818 and J Med. Chem. 2004, 47, 600- 611) from cyanuric chloride (a) and Grignard reagents.
  • Triazine derivatives can be formed from the reaction of a 6-alkyl or aryl substituted dichloro-triazine (b) with heterocyclic amine, followed by reaction with HR 2.
  • the monochloro-triazine (c) can be converted to amino triazine (d), which can react with YR 2 , to give a triazine derivative (IV).
  • dichloro-triazine (b) can react with HR 2 , followed by reaction with heterocyclic amine to give triazine derivative (IV).
  • monochloro-triazine (e) can be converted to amino triazine (f), which can react with a leaving-group-attached heterocyclic compound (g), to give a triazine derivative (IV).
  • the triazine derivative can also be synthesized by the reaction of cyanuric chloride with a sequence of heterocyclic amines and HR 2 to give 2,4-disubstituted-6-chloro-l,3,5-triazines.
  • the displacement of the last chlorine by amine, hydrazine, hydroxyl or other nucleophilic group can be achieved by increasing the temperature, affording the trisubstituted-l,3,5-triazines (IV).
  • the triazine derivative can be synthesized by the reaction of tri, di- or mono chloride triazine with a heterocyclic amines then the R 3 -L can be added to the heterocyclic moiety.
  • an amide moiety can be added this way, where is triazines (IV).
  • the reaction is preferably conducted in the presence of an inert solvent.
  • an inert solvent there is no particular restriction on the nature of the solvent to be employed, provided that it has no adverse effect on the reaction or on the reagents involved and that it can dissolve the reagents, at least to some extent.
  • suitable solvents include: aliphatic hydrocarbons, such as hexane, heptane, ligroin and petroleum ether; aromatic hydrocarbons, such as benzene, toluene and xylene; halogenated hydrocarbons, especially aromatic and aliphatic hydrocarbons, such as methylene chloride, chloroform, carbon tetrachloride, dichloroethane, chlorobenzene and the dichlorobenzenes; esters, such as ethyl formate, ethyl acetate, propyl acetate, butyl acetate and diethyl carbonate; ethers, such as diethyl ether, diisopropyl ether, tetrahydrofuran, dioxane.
  • aliphatic hydrocarbons such as hexane, heptane, ligroin and petroleum ether
  • aromatic hydrocarbons such as benzene, toluene and
  • ketones such as acetone, methyl ethyl ketone, methyl isobutyl ketone, isophorone and cyclohexanone
  • nitro compounds which may be nitroalkanes or nitroaranes, such as nitroethane and nitrobenzene
  • nitriles such as acetonitrile and isobutyronitrile
  • amides which may be fatty acid amides, such as formamide, dimethylformamide, dimethylacetamide and hexamethylphosphoric triamide
  • sulphoxides such as dimethyl sulphoxide and sulpholane.
  • reaction can take place over a wide range of temperatures, and the precise reaction temperature is not critical to the invention. In general, we find it convenient to carry out the reaction at a temperature of from -50°C to 100°C.
  • compositions of matter that are formulations of one or more active drugs and a pharmaceutically-acceptable carrier.
  • the invention provides a composition for administration to a mammalian subject , which may include a compound of formula (I) or formula (II), or its pharmaceutically acceptable salts.
  • Pharmaceutically acceptable salts of the compounds of this invention include those derived from pharmaceutically acceptable inorganic and organic acids and bases.
  • Suitable acid salts include acetate, adipate, alginate, aspartate, benzoate, benzenesulfonate, bisulfate, butyrate, citrate, camphorate, camphorsulfonate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, formate, fumarate, glucoheptanoate, glycerophosphate, glycolate, hemisulfate, heptanoate, hexanoate, hydrochloride, hydrobromide, hydroiodide, 2-hydroxyethanesulfonate, lactate, maleate, malonate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oxalate, palmoate, pectinate, persulfate, 3-phenylpropionate, phosphate, picrate, pivalate, prop
  • Salts derived from appropriate bases include alkali metal (e.g., sodium and potassium), alkaline earth metal (e.g., magnesium), ammonium and N + (Ci -4 alkyl) 4 salts.
  • alkali metal e.g., sodium and potassium
  • alkaline earth metal e.g., magnesium
  • ammonium and N + (Ci -4 alkyl) 4 salts e.g., sodium and potassium
  • alkali metal e.g., sodium and potassium
  • alkaline earth metal e.g., magnesium
  • ammonium e.g., sodium and sodium and sodium and potassium
  • N + (Ci -4 alkyl) 4 salts e.g., sodium and potassium
  • ammonium e.g., sodium and potassium
  • N + (Ci -4 alkyl) 4 salts e.g., sodium and potassium
  • ammonium e.g., sodium and potassium
  • N + (Ci -4 alkyl) 4 salts
  • compositions of the present invention may be administered orally, parenterally, by inhalation spray, topically, rectally, nasally, buccally, vaginally or via an implanted reservoir.
  • parenteral as used herein includes subcutaneous, intravenous, intramuscular, intra-articular, intra-synovial, intrasternal, intrathecal, intrahepatic, intralesional and intracranial injection or infusion techniques.
  • the compositions are administered orally, intraperitoneally or intravenously.
  • compositions of this invention may be orally administered in any orally acceptable dosage form including, but not limited to, capsules, tablets, troches, elixirs, suspensions, syrups, wafers, chewing gums, aqueous suspensions or solutions.
  • the oral compositions may contain additional ingredients such as: a binder such as macrocrystalline cellulose, gum tragacanth or gelatin; an excipient such as starch or lactose, a disintegrating agent such as alginic acid, corn starch and the like; a lubricant such as magnesium stearate; a glidant such as colloidal silicon dioxide; and a sweetening agent such as sucrose or saccharin or flavoring agent such as peppermint, methyl salicylate, or orange flavoring.
  • a binder such as macrocrystalline cellulose, gum tragacanth or gelatin
  • an excipient such as starch or lactose, a disintegrating agent such as alginic acid, corn starch and the like
  • a lubricant such as magnesium stearate
  • a glidant such as colloidal silicon dioxide
  • a sweetening agent such as sucrose or saccharin or flavoring agent such as peppermint, methyl salicylate, or orange flavoring.
  • tablets or pills may be coated with sugar, shellac, or other enteric coating agents.
  • a syrup may contain, in addition to the active ingredients, sucrose as a sweetening agent and certain preservatives, dyes and colorings and flavors. Materials used in preparing these various compositions should be pharmaceutically or veterinarally pure and non-toxic in the amounts used.
  • the active ingredient may be incorporated into a solution or suspension.
  • the solutions or suspensions may also include the following components: a sterile diluent such as water for injection, saline solution, fixed oils, polyethylene glycols, glycerine, propylene glycol or other synthetic solvents; antibacterial agents such as benzyl alcohol or methyl parabens; antioxidants such as ascorbic acid or sodium bisulfite; chelating agents such as ethylenediaminetetraacetic acid; buffers such as acetates, citrates or phosphates and agents for the adjustment of tonicity such as sodium chloride or dextrose.
  • a sterile diluent such as water for injection, saline solution, fixed oils, polyethylene glycols, glycerine, propylene glycol or other synthetic solvents
  • antibacterial agents such as benzyl alcohol or methyl parabens
  • antioxidants such as ascorbic acid or sodium bisulfite
  • chelating agents
  • the parenteral preparation can be enclosed in ampoules, disposable syringes or multiple dose vials made of glass or plastic.
  • the pharmaceutical forms suitable for injectable use include sterile solutions, dispersions, emulsions, and sterile powders.
  • the final form should be stable under conditions of manufacture and storage. Furthermore, the final pharmaceutical form should be protected against contamination and should, therefore, be able to inhibit the growth of microorganisms such as bacteria or fungi.
  • a single intravenous or intraperitoneal dose can be administered. Alternatively, a slow long-term infusion or multiple short-term daily infusions may be utilized, typically lasting from 1 to 8 days. Alternate day dosing or dosing once every several days may also be utilized.
  • Sterile, injectable solutions may be prepared by incorporating a compound in the required amount into one or more appropriate solvents to which other ingredients, listed above or known to those skilled in the art, may be added as required.
  • Sterile injectable solutions may be prepared by incorporating the compound in the required amount in the appropriate solvent with various other ingredients as required. Sterilizing procedures, such as filtration, may then follow.
  • dispersions are made by incorporating the compound into a sterile vehicle which also contains the dispersion medium and the required other ingredients as indicated above. In the case of a sterile powder, the preferred methods include vacuum drying or freeze drying to which any required ingredients are added.
  • Suitable pharmaceutical carriers include sterile water; saline, dextrose; dextrose in water or saline; condensation products of castor oil and ethylene oxide combining about 30 to about 35 moles of ethylene oxide per mole of castor oil; liquid acid; lower alkanols; oils such as corn oil; peanut oil, sesame oil and the like, with emulsifiers such as mono- or di-glyceride of a fatty acid, or a phosphatide, e.g., lecithin, and the like; glycols; polyalkylene glycols; aqueous media in the presence of a suspending agent, for example, sodium carboxymethylcellulose; sodium alginate; poly(vinylpyrolidone) ; and the like, alone, or with suitable dispensing agents such as lecithin; polyoxyethylene stearate; and the like.
  • a suspending agent for example, sodium carboxymethylcellulose; sodium alginate; poly(vinylpyrolidone)
  • the carrier may also contain adjuvants such as preserving stabilizing, wetting, emulsifying agents and the like together with the penetration enhancer.
  • adjuvants such as preserving stabilizing, wetting, emulsifying agents and the like together with the penetration enhancer.
  • the final form must be sterile and should also be able to pass readily through an injection device such as a hollow needle.
  • the proper viscosity may be achieved and maintained by the proper choice of solvents or excipients.
  • the use of molecular or particulate coatings such as lecithin, the proper selection of particle size in dispersions, or the use of materials with surfactant properties may be utilized.
  • compositions containing triazine derivatives and methods useful for the in vivo delivery of triazine derivatives in the form of nanoparticles which are suitable for any of the aforesaid routes of administration.
  • United States Patent Nos. 5,916,596, 6,506,405 and 6,537,579 teach the preparation of nanoparticles from the biocompatible polymers, such as albumin.
  • methods for the formation of nanoparticles of the present invention by a solvent evaporation technique from an oil-in- water emulsion prepared under conditions of high shear forces (e.g., sonication, high pressure homogenization, or the like).
  • 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.
  • 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.
  • 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 lower 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.
  • Carriers for topical administration of the compounds of this invention include, but are not limited to, mineral oil, liquid petrolatum, white petrolatum, propylene glycol, polyoxyethylene, 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, but are not limited to, mineral oil, sorbitan monostearate, polysorbate 60, cetyl esters wax, cetearyl alcohol, 2- octyldodecanol, benzyl alcohol and water.
  • the pharmaceutically acceptable compositions may be formulated as micronized suspensions in isotonic, pH adjusted sterile saline, or, preferably, as solutions in isotonic, pH adjusted sterile saline, 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.
  • compositions of this invention are formulated for oral administration.
  • the compounds of the invention may be used to treat diseases associated with cellular proliferation or hyperproliferation, such as cancers which include but are not limited to tumors of the nasal cavity, paranasal sinuses, nasopharynx, oral cavity, oropharynx, larynx, hypopharynx, salivary glands, and paragangliomas.
  • diseases associated with cellular proliferation or hyperproliferation such as cancers which include but are not limited to tumors of the nasal cavity, paranasal sinuses, nasopharynx, oral cavity, oropharynx, larynx, hypopharynx, salivary glands, and paragangliomas.
  • the compounds of the invention may also be used to treat cancers of the liver and biliary tree (particularly hepatocellular carcinoma), intestinal cancers, particularly colorectal cancer, ovarian cancer, small cell and non-small cell lung cancer, breast cancer, sarcomas (including fibrosarcoma, malignant fibrous histiocytoma, embryonal rhabdomysocarcoma, leiomysosarcoma, neuro-fibrosarcoma, osteosarcoma, synovial sarcoma, liposarcoma, and alveolar soft part sarcoma), neoplasms of the central nervous systems (particularly brain cancer), and lymphomas (including Hodgkin's lymphoma, lymphoplasmacytoid lymphoma, follicular lymphoma, mucosa-associated lymphoid tissue lymphoma, mantle cell lymphoma, B-lineage large cell lymphoma, Burkitt's lymphoma, and
  • the compounds and methods of the present invention are also useful in treating a variety of disorders, including but not limited to, for example: stroke, cardiovascular disease, myocardial infarction, congestive heart failure, cardiomyopathy, myocarditis, ischemic heart disease, coronary artery disease, cardiogenic shock, vascular shock, pulmonary hypertension, pulmonary edema (including cardiogenic pulmonary edema), pleural effusions, rheumatoid arthritis, diabetic retinopathy, retinitis pigmentosa, and retinopathies, including diabetic retinopathy and retinopathy of prematurity, inflammatory diseases, restenosis, asthma, acute or adult respiratory distress syndrome (ARDS), lupus, vascular leakage, protection from ischemic or reperfusion injury such as ischemic or reperfusion injury incurred during organ transplantation, transplantation tolerance induction;
  • agents e.g., chemotherapeutic agents or protein therapeutic agents described below
  • T- cell mediated hypersensitivity diseases including contact hypersensitivity, delayed- type hypersensitivity, and gluten-sensitive enteropathy (Celiac disease); Type 1 diabetes; psoriasis; contact dermatitis (including that due to poison ivy); Hashimoto's thyroiditis; Sjogren's syndrome; Autoimmune Hyperthyroidism, such as Graves' disease; Addison's disease (autoimmune disease of the adrenal glands); autoimmune polyglandular disease (also known as autoimmune polyglandular syndrome); autoimmune alopecia; pernicious anemia; vitiligo; autoimmune hypopituatarism; Guillain-Barre syndrome; other autoimmune diseases; cancers, including those where kineses such as Src-family kineses are activated or overexpressed, such as colon carcinoma and thymoma, or cancers where kinase activity facilitates tumor growth or survival; glomerulonephritis, serum sickness; uticaria; allergic diseases such as respiratory allergies
  • the compounds of the invention may be used to treat diseases associated with undesired cellular proliferation or hyperproliferation comprising identifying the mammal afflicted with said disease or condition and administering to said afflicted mammal a composition comprising the compound of formula 1 , wherein the disease or condition is associated with a kinase.
  • the compounds of the invention may be used to treat diseases associated with undesired cellular proliferation or hyperproliferation comprising identifying the mammal afflicted with said disease or condition and administering to said afflicted mammal a composition comprising a compound of formula (I) or formula (II), wherein the disease or condition is associated with a tyrosine kinase.
  • the compounds of the invention may be used to treat diseases associated with undesired cellular proliferation or hyperproliferation comprising identifying the mammal afflicted with said disease or condition and administering to said afflicted mammal a composition comprising a compound of formula (I) or formula (II), wherein the disease or condition is associated with the kinase that is a serine kinase or a threonine kinase.
  • the compounds of the invention may be used to treat diseases associated with undesired cellular proliferation or hyperproliferation comprising identifying the mammal afflicted with said disease or condition and administering to said afflicted mammal a composition comprising the compound of formula (I) or formula (II), wherein the disease or condition is associated with the kinase that is a Src family kinase.
  • the compounds of the invention may be used to treat diseases associated with undesired cellular proliferation or hyperproliferation comprising identifying the mammal afflicted with said disease or condition and administering to said afflicted mammal a composition comprising the compound of formula (I) or formula (II), wherein the disease or condition is associated with the kinase that is a Aurora family kinase.
  • the invention also provides methods of treating a mammal afflicted with the above diseases and conditions.
  • 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.
  • the compositions should be formulated so that a dosage of between 0.01-100 mg/kg body weight/day of the inhibitor can be administered to a patient receiving these compositions.
  • the invention compounds are administered in combination with chemotherapeutic agent, an anti-inflammatory agent, antihistamines, chemotherapeutic agent, immunomodulator, therapeutic antibody or a protein kinase inhibitor, e.g., a tyrosine kinase inhibitor, to a subject in need of such treatment.
  • chemotherapeutic agent an anti-inflammatory agent, antihistamines, chemotherapeutic agent, immunomodulator, therapeutic antibody or a protein kinase inhibitor, e.g., a tyrosine kinase inhibitor
  • the method includes administering one or more of the inventive compounds to the afflicted mammal.
  • the method may further include the administration of a second active agent, such as a cytotoxic agent, including alkylating agents, tumor necrosis factors, intercalators, microtubulin inhibitors, and topoisomerase inhibitors.
  • a second active agent such as a cytotoxic agent, including alkylating agents, tumor necrosis factors, intercalators, microtubulin inhibitors, and topoisomerase inhibitors.
  • the second active agent may be co-administered in the same composition or in a second composition.
  • suitable second active agents include, but are not limited to, a cytotoxic drug such as Acivicin; Aclarubicin; Acodazole Hydrochloride; AcrQnine; Adozelesin; Aldesleukin; Altretamine; Ambomycin; Ametantrone Acetate; Aminoglutethimide; Amsacrine; Anastrozole; Anthramycin; Asparaginase; Asperlin; Azacitidine; Azetepa; Azotomycin; Batimastat; Benzodepa; Bicalutamide; Bisantrene Hydrochloride; Bisnafide Dimesylate; Bizelesin; Bleomycin Sulfate; Brequinar Sodium; Bropirimine; Busulfan; Cactinomycin; Calusterone; Caracemide; Carbetimer; Carboplatin; Carmustine; Carubicin Hydrochloride; Carzelesin; Cedefmgol; Chlorambucil;
  • the compounds and compositions may be used at sub-cytotoxic levels in combination with other agents in order to achieve highly selective activity in the treatment of non-neoplastic disorders, such as heart disease, stroke and neurodegenerative diseases (Whitesell et al., Curr Cancer Drug Targets (2003), 3(5), 349- 58).
  • the exemplary therapeutical agents that may be administered in combination with invention compounds include EGFR inhibitors, such as gefitinib, erlotinib, and cetuximab.
  • Her2 inhibitors include canertinib, EKB-569, and GW-572016.
  • Src inhibitors include Src inhibitors, dasatinib, as well as Casodex (bicalutamide), Tamoxifen, MEK-I kinase inhibitors, MARK kinase inhibitors, PD inhibitors, and PDGF inhibitors, such as imatinib, Hsp90 inhibitors, such as 17-AAG and 17-DMAG.
  • anti-angiogenic and antivascular agents which, by interrupting blood flow to solid tumors, render cancer cells quiescent by depriving them of nutrition. Castration, which also renders androgen dependent carcinomas non-proliferative, may also be utilized.
  • IGFlR inhibitors inhibitors of non- receptor and receptor tyrosine kineses, and inhibitors of integrin.
  • the pharmaceutical composition and method of the present invention may further combine other protein therapeutic agents such as cytokines, immunomodulatory agents and antibodies.
  • cytokine encompasses chemokines, interleukins, lymphokines, monokines, colony stimulating factors, and receptor associated proteins, and functional fragments thereof.
  • the term "functional fragment” refers to a polypeptide or peptide which possesses biological function or activity that is identified through a defined functional assay.
  • the cytokines include endothelial monocyte activating polypeptide II (EMAP- II), granulocyte-macrophage-CSF (GM-CSF), granulocyte-CSF (G- CSF), macrophage- CSF (M-CSF), IL-I, IL-2, IL-3, IL- 4, IL-5, IL-6, IL-12, and IL-13, interferons, and the like and which is associated with a particular biologic, morphologic, or phenotypic alteration in a cell or cell mechanism.
  • EMP- II endothelial monocyte activating polypeptide II
  • GM-CSF granulocyte-macrophage-CSF
  • G- CSF granulocyte-CSF
  • M-CSF macrophage- CSF
  • IL-I IL-2,
  • cyclosporins e.g., cyclosporin A
  • CTLA4-Ig antibodies such as ICAM-3, anti-IL-2 receptor (Anti-Tac), anti- CD45RB, anti-CD2, anti-CD3 (OKT-3), anti-CD4, anti-CD80, anti-CD86
  • agents blocking the interaction between CD40 and gp39 such as antibodies specific for CD40 and for gpn39 (i.e., CDl 54), fusion proteins constructed from CD40 and gp39 (CD40Ig and CD8gp39), inhibitors, such as nuclear translocation inhibitors, of NF-kappa B function, such as deoxyspergualin (DSG), cholesterol biosynthesis inhibitors such as HM:G CoA reductase inhibitors (lovastatin and simvastatin), non-steroidal antiinflammatory drugs (NSAIDs) such as ibuprofen and cyclooxygenase inhibitors such as rofec
  • NSAIDs non-steroidal antiinflammatory
  • This example illustrated Src Kinase Assays.
  • Brifely in a final reaction volume of 25 ⁇ L, c-SRC (h) (5-10 mU) is incubated with 8 mM MOPS pH 7.0, 0.2 mM EDTA, 250 ⁇ M KVEKIGEGTYGVVYK (Cdc2 peptide), 10 mM MgAcetate and [g-33P-ATP] (specific activity approx. 500 cpm/pmol, concentration as required).
  • the reaction is initiated by the addition of the MgATP mix. After incubation for 40 minutes at room temperature, the reaction is stopped by the addition of 5 ⁇ L of a 3% phosphoric acid solution. 10 ⁇ L of the reaction is then spotted onto a P30 filtermat and washed three times for 5 minutes in 75 mM phosphoric acid and once in methanol prior to drying and scintillation counting.
  • Table 1 shows representative data for the inhibition of Src kinase by the compounds of this invention.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Engineering & Computer Science (AREA)
  • Immunology (AREA)
  • Epidemiology (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Diabetes (AREA)
  • Hematology (AREA)
  • Cardiology (AREA)
  • Pulmonology (AREA)
  • Rheumatology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Ophthalmology & Optometry (AREA)
  • Oncology (AREA)
  • Vascular Medicine (AREA)
  • Neurosurgery (AREA)
  • Biomedical Technology (AREA)
  • Pain & Pain Management (AREA)
  • Endocrinology (AREA)
  • Transplantation (AREA)
  • Neurology (AREA)
  • Urology & Nephrology (AREA)
  • Otolaryngology (AREA)
  • Hospice & Palliative Care (AREA)
  • Dermatology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

The present invention comprises inter alia compounds as shown in formula (I) or a pharmaceutically acceptable salt thereof.

Description

TRIAZINE DERIVATIVES AND THEIR THERAPEUTICAL APPLICATIONS
CROSS-REFERENCE TO RELATED APPLICATIONS
[0001] This patent application claims the benefit of U.S. Provisional Patent Application No. 61/185,052 , filed June 8, 2009, which is incorporated by reference.
FIELD OF THE INVENTION
[0002] The present invention relates generally to the use of compounds to treat a variety of disorders, diseases and pathologic conditions and more specifically to the use of triazine compounds to modulate protein kinases and for treating protein kinase-mediated diseases.
BACKGROUND OF THE INVENTION
[0003] Protein kinases constitute a large family of structurally related enzymes that are responsible for the control of a variety of signal transduction processes within the cell. Protein kinases, containing a similar 250-300 amino acid catalytic domain, catalyze the phosphorylation of target protein substrates.
[0004] The kinases may be categorized into families by the substrates in the phosphorylate (e.g., protein-tyrosine, protein-serine/threonine, lipids, etc.). Tyrosine phosphorylation is a central event in the regulation of a variety of biological processes such as cell proliferation, migration, differentiation and survival. Several families of receptor and non-receptor tyrosine kinases control these events by catalyzing the transfer of phosphate from ATP to a tyrosine residue of specific cell protein targets. Sequence motifs have been identified that generally correspond to each of these kinase families [ Hanks et al., FASEB J., (1995), 9, 576-596; Knighton et al., Science, (1991), 253, 407-414; Garcia-Bustos et al., EMBO J., (1994),13:2352-2361). Examples of kinases in the protein kinase family include, without limitation, abl, Akt, bcr-abl, BIk, Brk, Btk, c-kit, c-Met, c-src, c-fms, CDKl, CDK2, CDK3, CDK4, CDK5, CDK6, CDK7, CDK8, CDK9, CDKlO, cRafl, CSFlR, CSK, EGFR, ErbB2, ErbB3, ErbB4, Erk, Fak, fes, FGFRl, FGFR2, FGFR3, FGFR4, FGFR5, Fgr, flt-1, Fps, Frk, Fyn, Hck, IGF-IR, INS-R, Jak, KDR, Lck, Lyn, MEK, p38, PDGFR, PIK, PKC, PYK2, ros, Tie, Tie-2, TRK, Yes, and Zap70.
[0005] Studies indicated that protein kinases play a central role in the regulation and maintenance of a wide variety of cellular processes and cellular function. For example, kinase activity acts as molecular switches regulating cell proliferation, activation, and/or differentiation. Uncontrolled or excessive kinase activity has been observed in many disease states including benign and malignant proliferation disorders as well as diseases resulting from inappropriate activation of the immune system (autoimmune disorders), allograft rejection, and graft vs host disease.
[0006] It is reported that many diseases are associated with abnormal cellular responses triggered by protein kinase-mediated events. These diseases include autoimmune diseases, inflammatory diseases, bone diseases, metabolic diseases, neurological and neurodegenerative diseases, cancer, cardiovascular diseases, allergies and asthma, Alzheimer's disease and hormone-related diseases. In addition, endothelial cell specific receptor PTKs, such as VEGF-2 and Tie-2, mediate the angiogenic process and are involved in supporting the progression of cancers and other diseases involving uncontrolled vascularization. Accordingly, there has been a substantial effort in medicinal chemistry to find protein kinase inhibitors that are effective as therapeutic agents. [0007] One kinase family of particular interest is the Src family of kinases. Src kinase is involved in proliferation and migration responses in many cell types, cell activation, adhesion, motility, and survival, growth factor receptor signaling, and osteoclast activation (Biscardi et al, Adv. Cancer Res. (1999), 76, 61-119; Yeatman et al, Nat. Rev. Cancer (2004), 4, 470-480; Owens, D. W.; McLean et al., MoI. Biol. Cell (2000), 11, 51-64). Members of the Src family include the following eight kinases in mammals: Src, Fyn, Yes, Fgr, Lyn, Hck, Lck, and BIk (Bolen et al., Annu. Rev. Immunol, (1997), 15, 371). These are nonreceptor protein kinases that range in molecular mass from 52 to 62 kD. All are characterized by a common structural organization that is comprised of six distinct functional domains: Src homology domain 4 (SH4), a unique domain, SH3 domain, SH2 domain, a catalytic domain (SHl), and a C-terminal regulatory region (Brown et al., Biochim Biophys Acta (1996), 1287, 121-149; Tatosyan et al. Biochemistry (Moscow) 2000, 65, 49-58). SH4 domain contains the myristylation signals that guide the Src molecule to the cell membrane. This unique domain of Src proteins is responsible for their specific interaction with particular receptors and protein targets (Thomas et al., Annu Rev Cell Dev Biol (1997), 13, 513-609). The modulating regions, SH3 and SH2, control intra- as well as intermolecular interactions with protein substrates which affect Src catalytic activity, localization and association with protein targets (Pawson T., Nature (1995), 373, 573-580). The kinase domain, SHl, found in all proteins of the Src family, is responsible for the tyrosine kinase activity and has a central role in binding of substrates. The N-terminal half of Src kinase contains the site(s) for its tyrosine phosphorylation and regulates the catalytic activity of Src (Thomas et al., Annu Rev Cell Dev Biol (1997), 13: 513-609). v-Src differs from cellular Src (c-Src) on the basis of the structural differences in C-terminal region responsible for regulation of kinase activity. [0008] The prototype member of the Src family protein tyrosine kinases was originally identified as the transforming protein (v-Src) of the oncogenic retrovirus, Rous sarcoma virus, RSV (Brugge et al., Nature (1977), 269, 346-348); Hamaguchi et al. (1995), Oncogene 10: 1037-1043). Viral v-Src is a mutated and activated version of a normal cellular protein (c- Src) with intrinsic tyrosine kinase activity (Collett et al., Proc Natl Acad Sci U S A (1978), 75, 2021-2024). This kinase phosphorylates its protein substrates exclusively on tyrosyl residues (Hunter et al., Proc Natl Acad Sci U S A (1980), 77, 1311-1315). [0009] Investigations indicated that Src is a cytoplasmic protein tyrosine kinase, whose activation and recruitment to perimembranal signaling complexes has important implications for cellular fate. It has well-documented that Src protein levels and Src kinase activity are significantly elevated in human breast cancers (Muthuswamy et al., Oncogene, (1995), 11, 1801-1810); Wang et al., Oncogene (1999), 18, 1227-1237; Warmuth et al., Curr. Pharm. Des. (2003), 9, 2043-2059], colon cancers (Irby et al., Nat Genet (1999), 21, 187-190), pancreatic cancers (Lutz et al., Biochem Biophys Res Commun (1998), 243, 503-508], certain B-cell leukemias and lymphomas (Talamonti et al., J. Clin. Invest. (1993), 91, 53; Lutz et al., Biochem. Biophys. Res. (1998), 243, 503; Biscardi et al., Adv. Cancer Res. (1999), 76, 61; Lynch et al., Leukemia (1993), 7, 1416; Boschelli et al., Drugs of the Future (2000), 25(7), 717), gastrointestinal cancer (Cartwright et al., Proc. Natl. Acad. Sci. USA, (1990), 87, 558- 562 and Mao et al., Oncogene, (1997), 15, 3083-3090), non-small cell lung cancers (NSCLCs) (Mazurenko et al., European Journal of Cancer, (1992), 28, 372-7), bladder cancer (Fanning et al., Cancer Research, (1992), 52, 1457-62), oesophageal cancer (Jankowski et al., Gut, (1992), 33, 1033-8), prostate and ovarian cancer (Wiener et al., Clin. Cancer Research, (1999), 5, 2164-70), melanoma and sarcoma (Bohlen et al., Oncogene, (1993), 8, 2025-2031; tatosyan et al., Biochemistry (Moscow) (2000), 65, 49-58). Furthermore, Src kinase modulates signal transduction through multiple oncogenic pathways, including EGFR, Her2/neu, PDGFR, FGFR, and VEGFR (Frame et al., Biochim. Biophys. Acta (2002), 1602, 114-130; Sakamoto et al., Jpn J Cancer Res, (2001), 92: 941-946). [0010] Thus, it is anticipated that blocking signaling through the inhibition of the kinase activity of Src will be an effective means of modulating aberrant pathways that drive oncologic transformation of cells. Src kinase inhibitors may be useful anti-cancer agents (Abram et al., Exp. Cell Res., (2000), 254, 1). It is reported that inhibitors of src kinase had significant antiproliferative activity against cancer cell lines (M.M. Moasser et al., Cancer Res., (1999), 59, 6145; Tatosyan et al., Biochemistry (Moscow) (2000), 65, 49-58).) and inhibited the transformation of cells to an oncogenic phenotype (R. Kami et al., Oncogene (1999), 18, 4654). Furthermore, antisense Src expressed in ovarian and colon tumor cells has been shown to inhibit tumor growth (Wiener et al., Clin. Cancer Res., (1999), 5, 2164; Staley et al., Cell Growth Diff. (1997), 8, 269). Src kinase inhibitors have also been reported to be effective in an animal model of cerebral ischemia (Paul et al. Nature Medicine, (2001), 7, 222), suggesting that Src kinase inhibitors may be effective at limiting brain damage following stroke. Suppression of arthritic bone destruction has been achieved by the overexpression of CSK in rheumatoid synoviocytes and osteoclasts (Takayanagi et al., J Clin. Invest. (1999), 104, 137). CSK, or C-terminal Src kinase, phosphorylates and thereby inhibits Src catalytic activity. This implies that Src inhibition may prevent joint destruction that is characteristic in patients suffering from rheumatoid arthritis (Boschelli et al., Drugs of the Future (2000), 25(7), 717).
[0011] It is well documented that Src-family kinases are also important for signaling downstream of other immune cell receptors. Fyn, like Lck, is involved in TCR signaling in T cells (Appleby et al., Cell, (1992), 70, 751). Hck and Fgr are involved in Fcγ receptor signaling leading to neutrophil activation (Vicentini et al., J. Immunol. (2002), 168, 6446). Lyn and Src also participate in Fcγ receptor signaling leading to release of histamine and other allergic mediators (Turner, H. and Kinet, J-P Nature (1999), 402, B24). These-fϊndings suggest that Src family kinase inhibitors may be useful in treating allergic diseases and asthma.
[0012] Other Src family kinases are also potential therapeutic targets. Lck plays a role in T-cell signaling. Mice that lack the Lck gene have a poor ability to develop thymocytes. The function of Lck as a positive activator of T-cell signaling suggests that Lck inhibitors may be useful for treating autoimmune disease such as rheumatoid arthritis (Molina et al., Nature, (1992), 357, 161).
[0013] Hck is a member of the Src protein-tyrosine kinase family and is expressed strongly in macrophages, an important HIV target cell and its inhibition in HIV-infected macrophages might slow disease progression (Ye et al., Biochemistry, (2004), 43 (50), 15775 -15784).
[0014] Hck, Fgr and Lyn have been identified as important mediators of integrin signaling in myeloid leukocytes (Lowell et al., J. Leukoc. Biol, (1999), 65, 313). Inhibition of these kinase mediators may therefore be useful for treating inflammation (Boschelli et al., Drugs of the Future (2000), 25(7), 717).
[0015] It is reported that Syk is a tyrosine kinase that plays a critical role in the cell degranulation and eosinophil activation and Syk kinase is implicated in various allergic disorders, in particular asthma (Taylor et al., MoI. Cell. Biol. (1995), 15, 4149). [0016] BCR-ABL encodes the BCR-AEL protein, a constitutively active cytoplasmic tyrosine kinase present in 90% of all patients with chronic myelogenous leukemia (CML) and in 15-30% of adult patients with acute lymphoblastic leukemia (ALL). Numerous studies have demonstrated that the activity of BCR-ABL is required for the cancer causing ability of this chimeric protein.
[0017] Src kinases play a role in the replication of hepatitis B virus. The virally encoded transcription factor HBx activates Src in a step required for propagation of the virus (Klein et al., EMBOJ. (1999), 18, 5019; Klein et al., MoI. Cell. Biol. (1997), 17, 6427). Some genetic and biochemical data clearly demonstrate that Src-family tyrosine kinases serve as a critical signal relay, via phosphorylation of c-Cbl, for fat accumulation, and provide potential new strategies for treating obesity (Sun et al., Biochemistry, (2005), 44 (44), 14455 -14462). Since Src plays a role in additional signaling pathways, Src inhibitors are also being pursued for the treatment of other diseases including osteoporosis and stroke (Susva et al., Trends Pharmacol. ScL (2000), 21, 489-495; Paul et al., Nat. Med. (2001), 7, 222-227). [0018] It is also possible that inhibitors of the Src kinase activity are useful in the treatment of osteoporosis (Soriano et al., Cell (1991), 64, 693; Boyce et al. J Clin. Invest (1992), 90, 1622; Owens et al., MoI. Biol. Cell (2000), 11, 51-64), T cell mediated inflammation (Anderson et al., Adv. Immunol. (1994), 56, 151; Goldman, F D et al. J. Clin. Invest. (1998), 102, 421), and cerebral ischemia (Paul et al. Nature Medicine (2001), 7, 222). [0019] In addition, src family kinases participate in signal transduction in several cell types. For example, fyn, like Ick, is involved in T-cell activation. Hck and fgr are involved in Fe gamma receptor mediated oxidative burst of neutrophils. Src and lyn are believed to be important in Fc epsilon induced degranulation of mast cells, and so may play a role in asthma and other allergic diseases. The kinase lyn is known to be involved in the cellular response to DNA damage induced by UV light (Hiwasa et al., FEBS Lett. (1999), 444, 173) or ionizing radiation ( Kumar et al., J Biol Chein, (1998), 273, 25654). Inhibitors of lyn kinase may thus be useful as potentiators in radiation therapy. [0020] T cells play a pivotal role in the regulation of immune responses and are important for establishing immunity to pathogens. In addition, T cells are often activated during inflammatory autoimmune diseases, such as rheumatoid arthritis, inflammatory bowel disease, type I diabetes, multiple sclerosis, Sjogren's disease, myasthenia gravis, psoriasis, and lupus. T cell activation is also an important component of transplant rejection, allergic reactions, and asthma.
[0021] T cells are activated by specific antigens through the T cell receptor, which is expressed on the cell surface. This activation triggers a series of intracellular signaling cascades mediated by enzymes expressed within the cell (Kane et al. Current Opinion in Immunol. (2000), 12, 242). These cascades lead to gene regulation events that result in the production of cytokines, like interleukin-2 (IL-2). IL-2 is a necessary cytokine in T cell activation, leading to proliferation and amplification of specific immune responses. [0022] Therefore, Src kinase and other kinase have become intriguing targets for drug discovery (Parang et al., Expert Opin. Ther. Pat. (2005), 15, 1183-1207; Parang et al., Curr. Opin. Drug Discovery Dev. (2004), 7, 630-638). Many classes of compounds have been disclosed to modulate or, more specifically, inhibit kinase activity for use to treat kinase- related conditions or other disorders. For example, U.S. Pat. No. US Pat. No. 6,596,746 and the PCT WO 05/096784 A2 disclosed benzotrianes as inhibitors of kinases; the PCT WO 01/81311 disclosed substituted benzoic acid amides for the inhibition of angiogenisis; U.S. Pat. No. 6,440,965, disclosed substituted pyrimidine derivatives in the treatment of neurodegenerative or neurological disorders; PCT WO 02/08205 reported the pyrimidine derivatives having neurotrophic activity; PCT WO 03/014111 disclosed arylpiperazines and arylpiperidines and their use as metalloproteinase inhibiting agents; PCT WO 03/024448 described compounds as inhibitors of histone deacetylase enzymatic activity; PCT WO 04/058776 disclosed compounds which possess anti-angiogenic activity. PCT WO 01/94341 and WO 02/16352 disclosed Src kinase inhibitors of quinazoline derivatives. PCT WO03/026666Aland WO03/018021A1 disclosed pyrimidinyl derivatives as kinase inhibitors. U.S. Pat. No 6498165 reported Src kinase inhibitor compounds of pyrimidine compounds. Peptides as Src Tyrosine Kinase Inhibitors is reported recently (Kumar et al., J Med. Chem., (2006), 49 (11), 3395 -3401). The quinolinecarbonitriles derivatives was reported to be potent dual Inhibitors of Src and AbI Kinases (Diane et al., J Med. Chem., (2004), 47 (7), 1599 -1601). [0023] Although many inhibitors of kinases are known , there exists a need for new treatment options for conditions associated with protein kinases.
BRIEF SUMMARY OF THE INVENTION
[0024] Accordingly, the present invention provides an antitumor agent comprising a triazine derivative as described in formula (I) or formula (II), pharmaceutically-acceptable formulations thereof, methods for making novel compounds and compositions for using the compounds. The compounds and compositions comprising the compounds of formula (I) or formula (II) have utility in treatment of a variety of diseases.
[0025] The combination therapy described herein may be provided by the preparation of the triazine derivative of formula (I) or formula (II) and the other therapeutic agent as separate pharmaceutical formulations followed by the administration thereof to a patient simultaneously, semi-simultaneously, separately or over regular intervals. [0026] The present invention also provides methods for using certain chemical compounds such as kinase inhibitors in the treatment of various diseases, disorders, and pathologies, for example, cancer, and vascular disorders, such as myocardial infarction (MI), stroke, or ischemia. The triazine compounds described in this invention may block the enzymatic activity of some or many of the members of the Src family, in addition to blocking the activity of other receptor and non-receptor kinase. Such compounds may be beneficial for treatment of the diseases where disorders affect cell motility, adhesion, and cell cycle progression, and in addition, diseases with related hypoxic conditions, osteoporosis and conditions, which result from or are related to increases in vascular permeability, inflammation or respiratory distress, tumor growth, invasion, angiogenesis, metastases and apoptosis.
DETAILED DESCRIPTION OF THE INVENTION
[0027] The present invention comprises compounds as shown in formula (I)
Figure imgf000008_0001
or a pharmaceutically acceptable salt thereof, wherein:
A, B, W is selected from S, O, NR4, CR4 or L-R3; R4 is independently selected from hydrogen or an optionally substituted C1-4 aliphatic group.
R1 represents hydrogen, halogen, hydroxy, amino, cyano, alkyl, cycloalkyl, alkenyl, alkynyl, alkylthio, aryl, arylalkyl, heterocyclic, heteroaryl, heterocycloalkyl, alkylsulfonyl, alkoxycarbonyl and alkylcarbonyl.
R2 is selected from:
(i) amino, alkyl amino, aryl amino, heteroaryl amino;
(ii) C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl;
(iii) heterocyclic, heteroaryl; and
(iv) groups of the formula (Ia):
Figure imgf000009_0001
wherein:
R5 represents hydrogen, C1-C4 alkyl, oxo;
X is CH, when R6 is hydrogen; or X-R6 is O; or X is N, R6 represents groups of hydrogen, Ci-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-Ci0 aryl or heteroaryl, (C3-C7cycloalkyl)Ci-C4alkyl, d- C6 haloalkyl, Ci-C6 alkoxy, C1- C6 alkylthio, C2-C6 alkanoyl, Ci-C6 alkoxycarbonyl, C2- C6 alkanoyloxy, mono- and di-(C3-C8 cycloalkyl)amino — C0-C4alkyl, (4- to 7- membered heterocycle)Co-C4alkyl, Ci-C6 alkylsulfonyl, mono- and di-(Ci-C6 alkyl) sulfonamido, and mono- and di-(Ci- C6alkyl)aminocarbonyl, each of which is substituted with from 0 to 4 substituents independently chosen from halogen, hydroxy, cyano, amino, -COOH and oxo;
L represents O, S, SO, CO, S02, C02, NR4, (CH2)m, m = 0-3, CONR4, NR4CO, NR4SO2, SO2NR4, NR4CO2, NR4COR4, NR4SO2NR4, NR4NR45OCONR4, C(R4^CONR4, NR4COC(R4), C(R4)2SO, C(R4^SO2, C(R4^SO2NR4, C(R4^NR4, C(R4)ZNR4CO, C(R4^NR4CO2, C(R^=NNR4, C(R^=N-O, C(R4)ZNR4NR4, C(R4)ZNR4SO2NR4, C(R4)ZNR4CONR4, O(CH2)P, S(CH2)P, p=l-3,or (CH2)qO, or (CH2)qS, q = 1-3.
R3 is selected from:
(i) Ci-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl;
(ii) heterocyclic, (iii) Ar.
Ar represents heteroaryl or aryl, each of which is substituted with from 0 to 4 substituents independently chosen from:
(1) halogen, hydroxy, amino, cyano, -COOH, -SO2NH2, oxo, nitro and alkoxycarbonyl; and
(2) C1-C6 alkyl, Ci-C6alkoxy, C3-C10 cycloalkyl,C2-C6 alkenyl, C2-C6 alkynyl, C2-C6 alkanoyl, C1-C6 haloalkyl, C1-C6 haloalkoxy, mono- and di- (C1- C6alkyl)amino, C1-C6 alkylsulfonyl, mono- and di-(Ci-C6alkyl) sulfonamido and mono- and di-(C1-C6alkyl)aminocarbonyl; phenylC0-C4alkyl and (4- to 7-membered heterocycle) — C0-C4alkyl, each of which is substituted with from O to 4 secondary substituents independently chosen from halogen, hydroxy, cyano, oxo, imino, C1- C4alkyl, Ci-C4alkoxy and Ci-C4haloalkyl.
K is selected from i) absence; ii) O, S, SO, SO2; iii) (CH2)m, m = 0-3, O(CH2)p, p=l-3, (CH2)qO, q = 1-3. iv) NR7
R7 represents hydrogen, alkyl, cycloalkyl, alkenyl, alkynyl, alkylthio, aryl, arylalkyl.
[0028] The present invention also comprises compounds of formula (II)
Figure imgf000010_0001
or a pharmaceutically acceptable salt thereof, wherein:
Y is selected from -OR4, -NR4R5, and -Q-R3;
Q is selected from cycloalkyl and heterocycloalkyl, each of which is optionally substituted with Ci-C6 alkyl or oxo;
R3 is selected from H, Cj-C6 alkyl, Ci-C6 alkyl-R6, aryl, and heteroaryl, each of which is optionally substituted with Ci-C6 alkyl, halo, trifluoromethyl, or oxo;
R4 and R5 are each independently selected from H, C1-C6 alkyl-R6, aryl, and heteroaryl; R is selected from hydroxy, cyano, aryl, heteroaryl, cycloalkyl, heterocycloalkyl, - NH2, HiOnO(C1 -C6)alkylamino, di(Ci-C6)alkylamino, and C1-C6 alkoxy;
X is -NH-Ar1 -R1;
Ar1 is selected from aryl and heteroaryl, each of which is optionally substituted with C1-C6 alkyl or halo;
R1 is selected from -(CH2)nC(0)NHW, -CH2C(O)NHAr1 , and -NH2; n = 0, l;
W is selected from C1-C6 alkyl, cycloalkyl, and -(CH2)Ar1;
Z is selected from H, Ci-C6 alkyl, aryl, and heteroaryl. [0029] The invention further comprises compounds of formula (II)
Figure imgf000011_0001
or a pharmaceutically-acceptable salt thereof, wherein:
Y is selected from -OR4, -NR4R5, and -Q-R3;
Q is selected from morpholinyl, piperazinyl and piperidinyl;
R3 is selected from H, C1-C6 alkyl, hydroxy(Ci-C6)alkyl, cyano(Ci-C6)alkyl, pyridinylmethyl, pyridinyl, phenyl, trifluoromethylphenyl, and oxo;
R4 and R5 are each independently selected from H, Ci-C6 alkyl -R , and phenyl;
R6 is selected from hydroxy, morpholinyl, di(Ci-C6)alkylamino, imidazolyl, and Q- C6 alkoxy;
X is -NH-Ar1 -R1;
Ar1 is selected from thiazolyl, oxazolyl, oxadiazolyl, methyl-imidazolyl, pyrazolyl;
R1 is selected from -(CH2)nC(0)NHW and -NH2; n = 0, l;
W is selected from Cj-C6 alkyl and -(CH2)nPh optionally substituted with Ci-C6 alkyl or halo;
Z is selected from H, Ci-C6 alkyl, and phenyl. [0030] The invention also comprises compounds of formula (II)
Figure imgf000011_0002
(H) or a pharmaceutically-acceptable salt thereof, wherein:
Y is selected from -OR4, -NR4R5, and -Q-R3;
Q is selected from morpholinyl, piperazinyl and piperidinyl;
R3 is selected from H, Cj-C6 alkyl, hydroxy(Ci-C6)alkyl, cyano(Cj-C6)alkyl, pyridinylmethyl, pyridinyl, phenyl, trifluoromethylphenyl, and oxo;
R4 and R5 are each independently selected from H, Cj-C6 alkyl-R6, and phenyl;
R6 is selected from hydroxy, morpholinyl, di(Cj-C6)alkylamino, imidazolyl, and C1- C6 alkoxy;
X is -NH-Ar'-R1;
Ar1 is selected from thiazolyl, oxazolyl, oxadiazolyl, methyl-imidazolyl, pyrazolyl;
R1 is selected from -(CH2)nC(O)NHW, -CH2C(O)NHAr2, and -NH2; n = O, l;
W is selected from Cj-C6 alkyl, cycloalkyl, and -(CH2)Ar2 ;
Ar2 is phenyl, optionally substituted with Cj-C6 alkyl or halo;
Z is selected from H, Cj-C6 alkyl, and phenyl.
[0031] The following definitions refer to the various terms used above and throughout the disclosure.
[0032] Compounds are generally described herein using standard nomenclature. For compounds having asymmetric centers, it should be understood that (unless otherwise specified) all of the optical isomers and mixtures thereof are encompassed. In addition, compounds with carbon- carbon double bonds may occur in Z- and E- forms, with all isomeric forms of the compounds being included in the present invention unless otherwise specified. Where a compound exists in various tautomeric forms, a recited compound is not limited to any one specific tautomer, but rather is intended to encompass all tautomeric forms. Certain compounds are described herein using a general formula that include, variables (e.g. X, Ar.). Unless otherwise specified, each variable within such a formula is defined independently of any other variable, and any variable that occurs more than one time in a formula is defined independently at each occurrence.
[0033] The term "halo" or "halogen" refers to fluorine, chlorine, bromine or iodine. [0034] The term "alkyl" herein alone or as part of another group refers to a monovalent alkane (hydrocarbon) derived radical containing from 1 to 12 carbon atoms unless otherwise defined. Alkyl groups may be substituted at any available point of attachment. An alkyl group substituted with another alkyl group is also referred to as a "branched alkyl group". Exemplary alkyl groups include methyl, ethyl, propyl, isopropyl, n-butyl, t-butyl, isobutyl, pentyl, hexyl, isohexyl, heptyl, dimethylpentyl, octyl, 2,2,4-trimethylpentyl, nonyl, decyl, undecyl, dodecyl, and the like. Exemplary substituents include but are not limited to one or more of the following groups: alkyl, aryl, halo (such as F, Cl, Br, I), haloalkyl (such as CCl3 or CF3), alkoxy, alkylthio, hydroxy, carboxy (-COOH), alkyloxycarbonyl (-C(O)R), alkylcarbonyloxy (- OCOR), amino (-NH2), carbamoyl (-NHCOOR- or -0C0NHR-), urea (- NHCONHR-) or thiol (-SH). In some preferred embodiments of the present invention, alkyl groups are substituted with, for example, amino, heterocycloalkyl, such as morpholine, piperazine, piperidine, azetidine, hydroxyl, methoxy, or heteroaryl groups such as pyrrolidine. "Alkyl" also includes cycloalkyl.
[0035] The term "cycloalkyl" herein alone or as part of another group refers to fully saturated and partially unsaturated hydrocarbon rings of 3 to 9, preferably 3 to 7 carbon atoms. The examples include cyclopropyl, cyclobutyl, cyclopentyl and cyclohexyl, and like. Further, a cycloalkyl may be substituted. A substituted cycloalkyl refers to such rings having one, two, or three substituents, selected from the group consisting of halo, alkyl, substituted alkyl, alkenyl, alkynyl, nitro, cyano, oxo (=0), hydroxy, alkoxy, thioalkyl, -CO2H, -C(=O)H, CO2-alkyl, -C(=O)alkyl, keto, =N-0H, =N-O-alkyl, aryl, heteroaryl, heterocyclo, -NR1R", - C(=0)NR'R", -CO2NR1R", -C(=O)NR'R", -NR1CO2R", - NR1C(O)R", -SO2NR1R", and - NR1SO2R", wherein each of R1 and R" are independently selected from hydrogen, alkyl, substituted alkyl, and cycloalkyl, or R' and R" together form a heterocyclo or heteroaryl ring. [0036] The term "alkenyl" herein alone or as part of another group refers to a hydrocarbon radical straight, branched or cyclic containing from 2 to 12 carbon atoms and at least one carbon to carbon double bond. Examples of such groups include the vinyl, allyl, 1-propenyl, isopropenyl, 2-methyl-l-propenyl, 1-butenyl, 2-butenyl, 3-butenyl, 1-pentenyl, 2-pentenyl, 3-pentenyl, 4-pentenyl, 1-hexenyl, 2-hexenyl, 3-hexenyl, 4-hexenyl, 5-hexenyl, 1 -heptenyl, and like. Alkenyl groups may also be substituted at any available point of attachment. Exemplary substituents for alkenyl groups include those listed above for alkyl groups, and especially include C3 to C7 cycloalkyl groups such as cyclopropyl, cyclopentyl and cyclohexyl, which may be further substituted with, for example, amino, oxo, hydroxyl, etc.
[0037] The term "alkynyl" refers to straight or branched chain alkyne groups, which have one or more unsaturated carbon-carbon bonds, at least one of which is a triple bond. Alkynyl groups include C2-C8 alkynyl, C2-C6 alkynyl and C2-C4 alkynyl groups, which have from 2 to
8, 2 to 6 or 2 to 4 carbon atoms, respectively. Illustrative of the alkynyl group include ethenyl, propenyl, isopropenyl, butenyl, isobutenyl, pentenyl, and hexenyl. Alkynyl groups may also be substituted at any available point of attachment. Exemplary substituents for alkynyl groups include those listed above for alkyl groups such as amino, alkylamino, etc.
The numbers in the subscript after the symbol "C" define the number of carbon atoms a particular group can contain.
[0038] The term "alkoxy" alone or as part of another group denotes an alkyl group as described above bonded through an oxygen linkage (-O-). Preferred alkoxy groups have from
1 to 8 carbon atoms. Examples of such groups include the methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, isobutoxy, sec-butoxy, tert-butoxy, n-pentyloxy, isopentyloxy, n- hexyloxy, cyclohexyloxy, n-heptyloxy, n-octyloxy and 2-ethylhexyloxy.
[0039] The term "alkylthio" refers to an alkyl group as described above attached via a sulfur bridge. Preferred alkoxy and alkylthio groups are those in which an alkyl group is attached via the heteroatom bridge. Preferred alkylthio groups have from 1 to 8 carbon atoms. Examples of such groups include the methylthio, ethylthio, n-propythiol, n-butylthiol, and like.
[0040] The term "oxo," as used herein, refers to a keto (C=O) group. An oxo group that is a substituent of a nonaromatic carbon atom results in a conversion Of-CH2- to -C(=O)-.
[0041] The term "alkoxycarbonyl" herein alone or as part of another group denotes an alkoxy group bonded through a carbonyl group. An alkoxycarbonyl radical is represented by the formula: -C(O)OR, where the R group is a straight or branched C)-C6 alkyl group, cycloalkyl, aryl, or heteroaryl.
[0042] The term "alkylcarbonyl" herein alone or as part of another group refers to an alkyl group bonded through a carbonyl group or -C(O)R.
[0043] The term "arylalkyl" herein alone or as part of another group denotes an aromatic ring bonded through an alkyl group (such as benzyl) as described above.
[0044] The term "aryl" herein alone or as part of another group refers to monocyclic or bicyclic aromatic rings, e.g. phenyl, substituted phenyl and the like, as well as groups which are fused, e.g., napthyl, phenanthrenyl and the like. An aryl group thus contains at least one ring having at least 6 atoms, with up to five such rings being present, containing up to 20 atoms therein, with alternating (resonating) double bonds between adjacent carbon atoms or suitable heteroatoms. Aryl groups may optionally be substituted with one or more groups including, but not limited to halogen such as I, Br, F, or Cl; alkyl, such as methyl, ethyl, propyl, alkoxy, such as methoxy or ethoxy, hydroxy, carboxy, carbamoyl, alkyloxycarbonyl, nitro, alkenyloxy, trifluoromethyl, amino, cycloalkyl, aryl, heteroaryl, cyano, alkyl S(O)n,
(m=0, 1, 2), or thiol.
[0045] The term "aromatic" refers to a cyclically conjugated molecular entity with a stability, due to derealization, significantly greater than that of a hypothetical localized structure, such as the Kekule structure.
[0046] The term "amino" herein alone or as part of another group refers to -NH2. An
"amino" may optionally be substituted with one or two substituents, which may be the same or different, such as alkyl, aryl, arylalkyl, alkenyl, alkynyl, heteroaryl, heteroarylalkyl, cycloheteroalkyl, cycloheteroalkylalkyl, cycloalkyl, cycloalkylalkyl, haloalkyl, hydroxyalkyl, alkoxyalkyl, thioalkyl, carbonyl or carboxyl. These substituents may be further substituted with a carboxylic acid, any of the alkyl or aryl substituents set out herein. In some embodiments, the amino groups are substituted with carboxyl or carbonyl to form N-acyl or
N-carbamoyl derivatives.
[0047] The term "alkylsulfonyl" refers to groups of the formula (SO2)-alkyl, in which the sulfur atom is the point of attachment. Preferably, alkylsulfonyl groups include C1- C6 alkylsulfonyl groups, which have from 1 to 6 carbon atoms. Methylsulfonyl is one representative alkylsulfonyl group.
[0048] The term "heteroatom" refers to any atom other than carbon, for example, N, O, or
S.
[0049] The term "heteroaryl" herein alone or as part of another group refers to substituted and unsubstituted aromatic 5 or 6 membered monocyclic groups, 9 or 10 membered bicyclic groups, and 11 to 14 membered tricyclic groups which have at least one heteroatom (O, S or
N) in at least one of the rings. Each ring of the heteroaryl group containing a heteroatom can contain one or two oxygen or sulfur atoms and/or from one to four nitrogen atoms provided that the total number of heteroatoms in each ring is four or less and each ring has at least one carbon atom.
[0050] The fused rings completing the bicyclic and tricyclic groups may contain only carbon atoms and may be saturated, partially saturated, or unsaturated. The nitrogen and sulfur atoms may optionally be oxidized and the nitrogen atoms may optionally be quaternized. Heteroaryl groups which are bicyclic or tricyclic must include at least one fully aromatic ring but the other fused ring or rings may be aromatic or non- aromatic. The heteroaryl group may be attached at any available nitrogen or carbon atom of any ring. The heteroaryl ring system may contain zero, one, two or three substituents selected from the group consisting of halo, alkyl, substituted alkyl, alkenyl, alkynyl, aryl, nitro, cyano, hydroxy, alkoxy, thioalkyl, -CO2H, -C(=O)H, -CO2-alkyl, -C(=O)alkyl, phenyl, benzyl, phenylethyl, phenyloxy, phenylthio, cycloalkyl, substituted cycloalkyl, heterocyclo, heteroaryl, -NR1R", -C(=0)NR'R", -CO2NR1R" ,-C(=0)NR'R",- NR'C02R",-NRC(=0)R",- SO2NR1R", and -NR1SO2R", wherein each of R' and R" is independently selected from hydrogen, alkyl, substituted alkyl, and cycloalkyl, or R and R" together form a heterocyclo or heteroaryl ring.
[0051] Preferably monocyclic heteroaryl groups include pyrrolyl, pyrazolyl, pyrazolinyl, imidazolyl, oxazolyl, diazolyl, isoxazolyl, thiazolyl, thiadiazolyl, isothiazolyl, furanyl, thienyl, oxadiazolyl, pyridyl, pyrazinyl, pyrimidinyl, pyridazinyl, triazinyl and the like. [0052] Preferably bicyclic heteroaryl groups include indolyl, benzothiazolyl, benzodioxolyl, benzoxaxolyl, benzothienyl, quinolinyl, tetrahydroisoquinolinyl, isoquinolinyl, benzimidazolyl, benzopyranyl, indolizinyl, benzofuranyl, chromonyl, coumarinyl, benzopyranyl, cinnolinyl, quinoxalinyl, indazolyl, pyrrolopyridyl, dihydroisoindolyl, tetrahydroquinolinyl and the like.
[0053] Preferably tricyclic heteroaryl groups include carbazolyl, benzidolyl, phenanthrollinyl, acridinyl, phenanthridinyl, xanthenyl and the like. [0054] The term "heterocycle" or "heterocycloalkyl" herein alone or as part of another group refers to a cycloalkyl group (nonaromatic) in which one of the carbon atoms in the ring is replaced by a heteroatom selected from O, S or N. The "heterocycle" has from 1 to 3 fused, pendant or spiro rings, at least one of which is a heterocyclic ring (i.e. , one or more ring atoms is a heteroatom, with the remaining ring atoms being carbon). The heterocyclic ring may be optionally substituted which means that the heterocyclic ring may be substituted at one or more substitutable ring positions by one or more groups independently selected from alkyl (preferably lower alkyl), heterocycloalkyl, heteroaryl, alkoxy (preferably lower alkoxy), nitro, monoalkylamino (preferably a lower alkylamino), dialkylamino (preferably a alkylamino), cyano, halo, haloalkyl (preferably trifluoromethyl), alkanoyl, aminocarbonyl, monoalkylaminocarbonyl, dialkylaminocarbonyl, alkyl amido (preferably lower alkyl amido), alkoxyalkyl (preferably a lower alkoxy; lower alkyl), alkoxycarbonyl (preferably a lower alkoxycarbonyl), alkylcarbonyloxy (preferably a lower alkylcarbonyloxy) and aryl (preferably phenyl), said aryl being optionally substituted by halo, lower alkyl and lower alkoxy groups. A heterocyclic group may generally be linked via any ring or substituent atom, provided that a stable compound results. N-linked heterocyclic groups are linked via a component nitrogen atom.
[0055] Typically, a heterocyclic ring comprises 1-4 heteroatoms; within certain embodiments each heterocyclic ring has 1 or 2 heteroatoms per ring. Each heterocyclic ring generally contains from 3 to 8 ring members (rings having from to 7 ring members are recited in certain embodiments), and heterocycles comprising fused, pendant or spiro rings typically contain from 9 to 14 ring members which consists of carbon atoms and contains one, two, or three heteroatoms selected from nitrogen, oxygen and/or sulfur. [0056] Examples of "heterocycle" or "heterocycloalkyl groups include piperazine, piperidine, morpholine, thiomorpholine, pyrrolidine, imidazolidine and thiazolide. [0057] The term "substituent," as used herein, refers to a molecular moiety that is covalently bonded to an atom within a molecule of interest. For example, a "ring substituent" may be a moiety such as a halogen, alkyl group, haloalkyl group or other group discussed herein that is covalently bonded to an atom (preferably a carbon or nitrogen atom) that is a ring member.
[0058] The term "optionally substituted " as it refers that the aryl or heterocyclyl or other group may be substituted at one or more substitutable positions by one or more groups independently selected from alkyl (preferably lower alkyl), alkoxy (preferably lower alkoxy), nitro, monoalkylamino (preferably with one to six carbons), dialkylamino (preferably with one to six carbons), cyano, halo, haloalkyl (preferably trifluoromethyl), alkanoyl, aminocarbonyl, monoalkylaminocarbonyl, dialkylaminocarbonyl, alkyl amido (preferably lower alkyl amido), alkoxyalkyl (preferably a lower alkoxy and lower alkyl), alkoxycarbonyl (preferably a lower alkoxycarbonyl), alkylcarbonyloxy (preferably a lower alkylcarbonyloxy) and aryl (preferably phenyl), said aryl being optionally substituted by halo, lower alkyl and lower alkoxy groups. Optional substitution is also indicated by the phrase "substituted with from 0 to X substituents," where X is the maximum number of possible substituents. Certain optionally substituted groups are substituted with from 0 to 2, 3 or 4 independently selected substituents.
[0059] A dash ("-") that is not between two letters or symbols is used to indicate a point oft attachment for a substituent. For example, -CONH2 is attached through the carbon atom. [0060] A dashed cycle that locates inside of a heterocyle ring is used to indicate a conjugated system. The bonds between two atomes may be single bond or double bond. [0061] The term "anticancer" agent includes any known agent that is useful for the treatment of cancer including, but is not limited, Acivicin; Aclarubicin; Acodazole Hydrochloride; AcrQnine; Adozelesin; Aldesleukin; Altretamine; Ambomycin; Ametantrone Acetate; Aminoglutethimide; Amsacrine; Anastrozole; Anthramycin; Asparaginase; Asperlin; Azacitidine; Azetepa; Azotomycin; Batimastat; Benzodepa; Bicalutamide; Bisantrene Hydrochloride; Bisnafide Dimesylate; Bizelesin; Bleomycin Sulfate; Brequinar Sodium; Bropirimine; Busulfan; Cactinomycin; Calusterone; Caracemide; Carbetimer; Carboplatin; Carmustine; Carubicin Hydrochloride; Carzelesin; Cedefϊngol; Chlorambucil; Cirolemycin; Cisplatin; Cladribine; Crisnatol Mesylate; Cyclophosphamide; Cytarabine; Dacarbazine; Dactinomycin; Daunorubicin Hydrochloride; Decitabine; Dexormaplatin; Dezaguanine; Dezaguanine Mesylate; Diaziquone; Docetaxel; Doxorubicin; Doxorubicin Hydrochloride; Droloxifene; Droloxifene Citrate; Dromostanolone Propionate; Duazomycin; Edatrexate; Eflomithine Hydrochloride; Elsamitrucin; Enloplatin; Enpromate; Epipropidine; Epirubicin Hydrochloride; Erbulozole; Esorubicin Hydrochloride; Estramustine; Estramustine Phosphate Sodium; Etanidazole; Ethiodized Oil 1 131; Etoposide; Etoposide Phosphate; Etoprine; Fadrozole Hydrochloride; Fazarabine; Fenretinide; Floxuridine; Fludarabine Phosphate; Fluorouracil; Flurocitabine; Fosquidone; Fostriecin Sodium; Gemcitabine; Gemcitabine Hydrochloride; Gold Au 198; Hydroxyurea; Idarubicin Hydrochloride; Ifosfamide; Ilmofosine; Interferon Alfa-2a; Interferon Alfa-2b; Interferon Alfa-nl; Interferon Alfa-n3; Interferon Beta- 1 a; Interferon Gamma- 1 b; Iproplatin; Irinotecan Hydrochloride; Lanreotide Acetate; Letrozole; Leuprolide Acetate; Liarozole Hydrochloride; Lometrexol Sodium; Lomustine; Losoxantrone Hydrochloride; Masoprocol; Maytansine; Mechlorethamine Hydrochloride; Megestrol Acetate; Melengestrol Acetate; Melphalan; Menogaril; Mercaptopurine; Methotrexate; Methotrexate Sodium; Metoprine; Meturedepa; Mitindomide; Mitocarcin; Mitocromin; Mitogillin; Mitomalcin; Mitomycin; Mitosper; Mitotane; Mitoxantrone Hydrochloride; Mycophenolic Acid; Nocodazole; Nogalamycin; Ormaplatin; Oxisuran; Paclitaxel; Pegaspargase; Peliomycin; Pentamustine; Peplomycin Sulfate; Perfosfamide; Pipobroman; Piposulfan; Piroxantrone Hydrochloride; Plicamycin; Plomestane; Porfimer Sodium; Porfiromycin; Prednimustine; Procarbazine Hydrochloride; Puromycin; Puromycin Hydrochloride; Pyrazofurin; Riboprine; Rogletimide; Safmgol; Safingol Hydrochloride; Semustine; Simtrazene; Sparfosate Sodium; Sparsomycin; Spirogermanium Hydrochloride; Spiromustine; Spiroplatin; Streptonigrin; Streptozocin; Strontium Chloride Sr 89; Sulofenur; Talisomycin; Taxane; Taxoid; Tecogalan Sodium; Tegafur; Teloxantrone Hydrochloride; Temoporfin; Teniposide; Teroxirone; Testolactone;
Thiamiprine; Thioguanine; Thiotepa; Tiazofurin; Tirapazamine; Topotecan Hydrochloride;
Toremifene Citrate; Trestolone Acetate; Triciribine Phosphate; Trimetrexate; Trimetrexate
Glucuronate; Triptorelin; Tubulozole Hydrochloride; Uracil Mustard; Uredepa; Vapreotide;
Verteporfin; Vinblastine Sulfate; Vincristine Sulfate; Vindesine; Vindesine Sulfate;
Vinepidine Sulfate; Vinglycinate Sulfate; Vinleurosine Sulfate; Vinorelbine Tartrate;
Vinrosidine Sulfate; Vinzolidine Sulfate; Vorozole; Zeniplatin; Zinostatin; and Zorubicin
Hydrochloride.
[0062] The term "kinase" refers to any enzyme that catalyzes the addition of phosphate groups to a protein residue; for example, serine and threonine kineses catalyze the addition of phosphate groups to serine and threonine residues.
[0063] The terms "Src kinase," "Src kinase family," and "Src family" refer to the related homologs or analogs belonging to the mammalian family of Src kineses, including, for example, c-Src, Fyn, Yes and Lyn kineses and the hematopoietic-restricted kineses Hck, Fgr,
Lck and BIk.
[0064] The term "therapeutically effective amount" refers to the amount of the compound or pharmaceutical composition that will elicit the biological or medical response of a tissue, system, animal or human that is being sought by the researcher, veterinarian, medical doctor or other clinician, e.g., restoration or maintenance of vasculostasis or prevention of the compromise or loss or vasculostasis; reduction of tumor burden; reduction of morbidity and/or mortality.
[0065] The term 'pharmaceutically acceptable" refers to the fact that the carrier, diluent or excipient must be compatible with the other ingredients of the formulation and not deleterious to the recipient thereof.
[0066] The terms "administration of a compound" or "administering a compound" refer to the act of providing a compound of the invention or pharmaceutical composition to the subject in need of treatment.
[0067] The term "protected" refers that the group is in modified form to preclude undesired side reactions at the protected site. Suitable protecting groups for the compounds of the present invention will be recognized from the present application taking into account the level of skill in the art, and with reference to standard textbooks, such as Greene, T. W. et al.,
Protective Groups in Organic Synthesis, John Wiley & Sons, New York (1999). [0068] The term "pharmaceutically acceptable salt" of a compound recited herein is an acid or base salt that is suitable for use in contact with the tissues of human beings or animals without excessive toxicity or carcinogenicity, and preferably without irritation, allergic response, or other problem or complication. Such salts include mineral and organic acid salts of basic residues such as amines, as well as alkali or organic salts of acidic residues such as carboxylic acids. Specific pharmaceutical salts include, but are not limited to, salts of acids such as hydrochloric, phosphoric, hydrobromic, malic, glycolic, fumaric, sulfuric, sulfamic, sulfanilic, formic, toluenesulfonic, methanesulfonic, benzene sulfonic, ethane disulfonic, 2- hydroxyethylsulfonic, nitric, benzoic, 2-acetoxybenzoic, citric, tartaric, lactic, stearic, salicylic, glutamic, ascorbic, pamoic, succinic, fumaric, maleic, propionic, hydroxymaleic, hydroiodic, phenylacetic, alkanoic such as acetic, HOOC- (CH2)n-C00H where n is 0-4, and the like. Similarly, pharmaceutically acceptable cations include, but are not limited to sodium, potassium, calcium, aluminum, lithium and ammonium. Those of ordinary skill in the art will recognize further pharmaceutically acceptable salts for the compounds provided herein. In general, a pharmaceutically acceptable acid or base salt can be synthesized from a parent compound that contains a basic or acidic moiety by any conventional chemical method. Briefly, such salts can be prepared by reacting the free acid or base forms of these compounds with a stoichiometric amount of the appropriate base or acid in water or in an organic solvent, or in a mixture of the two; generally, the use of nonaqueous media, such as ether, ethyl acetate, ethanol, isopropanol or acetonitrile, is preferred. It will be apparent that each compound of formula (I) or formula (II) may, but need not, be formulated as a hydrate, solvate or non- covalent complex. In addition, the various crystal forms and polymorphs are within the scope of the present invention. Also provided herein are prodrugs of the compounds of formula (I) or formula (II).
[0069] The term of "prodrug" refers a compound that may not fully satisfy the structural requirements of the compounds provided herein, but is modified in vivo, following administration to a patient, to produce a compound of formula (I) or formula (II), or other formula provided herein. For example, a prodrug may be an acylated derivative of a compound as provided herein. Prodrugs include compounds wherein hydroxy, amine or thiol groups are bonded to any group that, when administered to a mammalian subject, cleaves to form a free hydroxy, amino, or thiol group, respectively. Examples of prodrugs include, but are not limited to, acetate, formate and benzoate derivatives of alcohol and amine functional groups within the compounds provided herein. Prodrugs of the compounds provided herein may be prepared by modifying functional groups present in the compounds in such a way that the modifications are cleaved in vivo to yield the parent compounds.
[0070] Groups that are "optionally substituted" are unsubstituted or are substituted by other than hydrogen at one or more available positions. Such optional substituents include, for example, hydroxy, halogen, cyano, nitro, C1-C6 alkyl, C2-C6 alkenyl, C2- C6 alkynyl, C1-
C6 alkoxy, C2-C6 alkyl ether, C3-C6 alkanone, C2-C6 alkylthio, amino, mono- or di-(Ci-C6 alkyl)amino, C1-C6 haloalkyl, -COOH, -CONH2, mono- or di-(CrC6 alkyl)aminocarbonyl,
-SO2NH2, and/or mono or di(C]-C6 alkyl) sulfonamido, as well as carbocyclic and heterocyclic groups.
[0071] Optional substitution is also indicated by the phrase "substituted with from 0 to X substituents," where X is the maximum number of possible substituents. Certain optionally substituted groups are substituted with from 0 to 2, 3 or 4 independently selected substituents.
[0072] Preferred R1 groups of formula (I) are listed below:
[0073] -CH3, -CH2CH3, -CH2CH(CH3)2, Cyclopropanyl, Ph, -CH2Ph, -CH2PhOMe.
[0074] Preferred R2 groups of formula (I) are listed below:
Figure imgf000021_0001
Figure imgf000022_0001
[0075] Preferred R3 groups of formula (I) are listed below, wherein the substitute may be the specific ones as defined here or may be one or multiple substitutes as defined above:
Figure imgf000023_0001
[0076] Preferred L is selected from O, S, SO, CO, SO2, CO2, NR6, (CH2)m, m = 0-3,
CONR4, NR4CO, NR4SO2, SO2NR4, NR4CO2, NR4SO2NR4, NR4NR45OCONR4,
C(R4^CONR4, NR4COC(R4), C(R4^SO, C(RO2SO2, C(R4^SO2NR4, C(R^)2NR4,
C(R4^NR4CO, C(Rj)2NR4CO2, C(R4)=NNR4, C(Rt)=N-O, C(R4)2NR4NR4,
C(Rt)2NR4SO2NR4, C(R4)^R4CONR4.
[0077] R4 is independently selected from hydrogen or an optionally substituted Ci-4 aliphatic group.
[0078] Preferably, the compounds of the invention may be compounds of formula (I) wherein
Ri groups of formula (I) are listed below:
-CH3, -CH2CH3, -CH2CH(CH3)2, Cyclopropanyl, Ph, -CH2Ph, -CH2PhOMe.
R2 is selected from: (i)amino, alkyl amino, aryl amino, heteroaryl amino; (ii) C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl; (iii) heterocyclic, herteroaryl; and (iv) groups of the formula (Ia):
Figure imgf000024_0001
wherein:
R5 represents hydrogen, Cj-C4 alkyl, oxo;
X is CH, when R6 is hydrogen; or X-R6 is O; or X is N, R6 represents groups of hydrogen, Cj-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-CjO aryl or heteroaryl, (C3-C7cycloalkyl)Cj-C4alkyl, C1- C6 haloalkyl, C1-C6 alkoxy, Cj- C6 alkylthio, C2-C6 alkanoyl, Cj- C6 alkoxycarbonyl, C2- C6 alkanoyloxy, mono- and di-(C3-C8 cycloalkyl)aminoC0-C4alkyl, (4- to 7- membered heterocycle)C0-C4alkyl, Cj-C6 alkylsulfonyl, mono- and di-(Cj- C6 alkyl) sulfonamide, and mono- and di-(C]- C6alkyl)aminocarbonyl, each of which is substituted with from O to 4 substituents independently chosen from halogen, hydroxy, cyano, amino, -COOH and oxo;
L represents O, S, SO, CO, SO2, CO2, NR6, (CH2)m, m = 0-3, CONR4, NR4CO, NR4SO2, SO2NR4, NR4CO2, NR4COR4, NR4SO2NR4, NR4NR43OCONR4, C(Rt)2CONR4, NR4COC(R4), C(Rj)2SO, C(R4^SO2, C(RzO2SO2NR4, C(Re)2NR4, C(R4^NR4CO, C(R4^NR4CO2, C(Rj)=NNR4, C(Rt)=N-O, C(R^2NR4NR4, C(R4^NR4SO2NR4, C(R4^NR4CONR4.
R4 is independetly selected from hydrogen or an optionally substituted Cj-4 aliphatic group.
R3 is selected from:
(i) C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl;
(ii) heterocyclic,
(iii) Ar.
Ar represents heteroaryl or aryl, each of which is substituted with from O to 4 substituents independently chosen from: (1) halogen, hydroxy, amino, cyano, -COOH, -SO2NH2, oxo, nitro and alkoxycarbonyl; and
(2) C1-C6 alkyl, Ci-C6alkoxy, C3-C10 cycloalkyl,C2-C6 alkenyl, C2-C6 alkynyl, C2-C6 alkanoyl, C1-C6 haloalkyl, C1-C6 haloalkoxy, mono- and di- (C i- C6alkyl)amino, Cj-C6 alkylsulfonyl, mono- and di-(C!-C6alkyl) sulfonamido and mono- and di-(Ci-C6alkyl)aminocarbonyl; phenylC0-C4alkyl and (4- to 7-membered heterocycle) — C0-C4alkyl, each of which is substituted with from O to 4 secondary substituents independently chosen from halogen, hydroxy, cyano, oxo, imino, C1- C4alkyl, C1-C4alkoxy and Cj-C4haloalkyl.
A, B, W independently represents S, or O, or NR4, or CR4;
K is selected from i) absence; ii) O, S, SO, SO2; iii) (CH2)m, m = 0-3, O(CH2)P, p=l-3, (CH2)qO, q = 1-3. iv) NR7
R7 represents hydrogen, alkyl, cycloalkyl, alkenyl, alkynyl, alkylthio, aryl, arylalkyl. [0079] More preferably, the compounds of the invention may be compounds of formula (I) wherein
R1 represents -CH3, -CH2CH3, -CH2CH(CH3)2, Cyclopropanyl, Ph.
R2 is selected from: amino, alkyl amino, aryl amino, heteroaryl amino and groups of the formula (Ia):
Figure imgf000025_0001
wherein:
R5 represents hydrogen, C1-C4 alkyl, oxo;
X is CH, when R6 is hydrogen; or X-R6 is O; or X is N, R6 represents groups of hydrogen, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-Ci0 aryl or heteroaryl, (C3-C7cycloalkyl)Ci-C4alkyl, C1- C6 haloalkyl, C1-C6 alkoxy, C1- C6 alkylthio, C2-C6 alkanoyl, Cr C6 alkoxycarbonyl, C2- C6 alkanoyloxy, mono- and di-(C3-C8 cycloalkyl)aminoC0-C4alkyl, (4- to 7- membered heterocycle)C0-C4alkyl, C1-C6 alkylsulfonyl, mono- and CU-(C1- C6 alkyl) sulfonamido, and mono- and di-(Cj- C6alkyl)aminocarbonyl, each of which is substituted with from O to 4 substituents independently chosen from halogen, hydroxy, cyano, amino, -COOH and oxo;
L represents O, S, CO, SO2, CO2, NR4, (CH2)m, m = 0-3, CONR4, NR4CO, NR4SO2, SO2NR4, NR4CO2, NR4COR4, NR4SO2NR4, NR4NR4, OCONR4, C(R4^CONR4, NR4COC(R4), C(R^2SO, C(R^2SO2, C(R^2SO2NR4, C(R4^NR4, C(R^2NR4CO, C(R^2NR4CO2, C(R4)=NNR4.
R4 is independetly selected from hydrogen or an optionally substituted C1-4 aliphatic group.
R3 is selected from heteroaryl or aryl, each of which is substituted with from O to 4 substituents independently chosen from:
(1) halogen, hydroxy, amino, cyano, -COOH, -SO2NH2, oxo, nitro and alkoxycarbonyl; and
(2) C1-C6 alkyl, Q-Qalkoxy, C3-C10 cycloalkyl,C2-C6 alkenyl, C2-C6 alkynyl, C2-C6 alkanoyl, C1-C6 haloalkyl, C1-C6 haloalkoxy, mono- and di- (C1- C6alkyl)amino, C1-C6 alkylsulfonyl, mono- and di-(CrC6alkyl) sulfonamido and mono- and di-(CrC6alkyl)aminocarbonyl; phenylC0-C4alkyl and (4- to 7-membered heterocycle) — C0-C4alkyl, each of which is substituted with from O to 4 secondary substituents independently chosen from halogen, hydroxy, cyano, oxo, imino, Q- C4alkyl, Ci-C4alkoxy and Ci-C4haloalkyl.
A, B, W independently represents S, or O, or NR4, or CR4;
K is selected from i) absence; ii) O, S, SO, SO2;
Hi) (CH2)m, m = 0-3, O(CH2)P, p=l-3, (CH2)qO, q = 1-3. iv) NR7
R7 represents hydrogen, alkyl, cycloalkyl, alkenyl, alkynyl, alkylthio, aryl, arylalkyl.
[0080] Most preferably, R1 represents, -CH3, -CH2CH3; R2 is selected from: alkyl amino, aryl amino, heteroaryl amino and groups of the formula (Ia):
Figure imgf000027_0001
wherein:
R5 represents hydrogen, Ci-C4 alkyl, oxo;
X is N, R6 represents groups of hydrogen, Ci-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-Ci0 aryl or heteroaryl, (C3-C7cycloalkyl)Ci-C4alkyl, C1- C6 haloalkyl, Cj-C6 alkoxy, Ci- C6 alkylthio, C2-C6 alkanoyl, Ci- C6 alkoxycarbonyl, C2- C6 alkanoyloxy, mono- and di-(C3- C8 cycloalkyl)aminoCo-C4alkyl, (4- to 7- membered heterocycle)C0-C4alkyl, Ci-C6 alkylsulfonyl, mono- and di-(Ci- C6 alkyl) sulfonamido, and mono- and CU-(C1- C6alkyl)aminocarbonyl, each of which is substituted with from 0 to 4 substituents independently chosen from halogen, hydroxy, cyano, amino, -COOH and oxo;
L represents O, S, NR4, (CH2)m, m = 0-3, CONR4, NR4CO, NR4SO2, SO2NR4, NR4CO2, NR4COR6, NR4SO2NR4, C(R4)ZCONR4, C(R4)2SO2, C(Rt)2SO2NR4, C(IU)2NR4, C(R4^NR4CO;
R4 is independetly selected from hydrogen or an optionally substituted C1-4 aliphatic group.
R3 is selected from heteroaryl or aryl, each of which is substituted with from O to 4 substituents independently chosen from:
(1) halogen, hydroxy, amino, cyano, -COOH, -SO2NH2, oxo, nitro and alkoxycarbonyl; and
(2) C1-C6 alkyl, Ci-C6alkoxy, C3-C10 cycloalkyl,C2-C6 alkenyl, C2-C6 alkynyl, C2- C6 alkanoyl, Ci-C6 haloalkyl, Ci-C6 haloalkoxy, mono- and di- (Ci- C6alkyl)amino, Ci-C6 alkylsulfonyl, mono- and di-(Ci-C6alkyl) sulfonamido and mono- and di-(Ci-C6alkyl)aminocarbonyl; phenylC0-C4alkyl and (4- to 7-membered heterocycle)Co-C4alkyl, each of which is substituted with from O to 4 secondary substituents independently chosen from halogen, hydroxy, cyano, oxo, imino, C]- Qalkyl, Ci-C4alkoxy and Ci-C4haloalkyl.
A, B, W independently represents S, or O, or NR4, or CR4; K is selected from i) absence; ϋ) O, S; iii) NR7; R7 represents hydrogen, alkyl. [0081] Preferred heterocyclic groups in compounds of formula (I) include
Figure imgf000028_0001
Which optionally may be substituted.
[0082] According to another embodiment, the present invention relates to a compound of formula (I) wherein Ri is methyl.
[0083] According to another embodiment, the present invention relates to a compound of formula (I) wherein Rj is ethyl.
[0084] According to another embodiment, the present invention relates to a compound of formula (I) wherein R] is isopropyl.
[0085] According to another embodiment, the present invention relates to a compound of formula (I) wherein Rj is phenyl.
[0086] According to another embodiment, the present invention relates to a compound of formula (I) wherein Rj is cyclopropanyl. [0087] According to another embodiment, the present invention relates to a compound of formula (I) wherein R2 is methyl-piperazinyl.
[0088] According to another embodiment, the present invention relates to a compound of formula (I) wherein R2 is (2-hydroxylethyl)-piperazinyl.
[0089] According to another embodiment, the present invention relates to a compound of formula (I) wherein L is oxygen.
[0090] According to another embodiment, the present invention relates to a compound of formula (I) wherein L is CO.
[0091] According to another embodiment, the present invention relates to a compound of formula (I) wherein L is NHCO.
[0092] According to another embodiment, the present invention relates to a compound of formula (I) wherein L is CONH.
[0093] According to another embodiment, the present invention relates to a compound of formula (I) wherein L is NR4COC(R4).
[0094] According to another embodiment, the present invention relates to a compound of formula (I) wherein L is NH.
[0095] According to another embodiment, the present invention relates to a compound of formula (I) wherein L is S.
[0096] According to another embodiment, the present invention relates to a compound of formula (I) wherein L is SO.
[0097] According to another embodiment, the present invention relates to a compound of formula (I) wherein L is SO2.
[0098] According to another embodiment, the present invention relates to a compound of formula (I) wherein A is N.
[0099] Examples of specific compounds of the present invention are those compounds defined in the following:
Figure imgf000030_0001
Figure imgf000031_0001
Figure imgf000032_0001
Figure imgf000033_0001
Figure imgf000034_0001
Figure imgf000035_0001
Figure imgf000036_0001
Figure imgf000037_0001
Figure imgf000038_0001
Figure imgf000039_0001
Figure imgf000040_0001
Figure imgf000041_0001
Figure imgf000042_0001
Figure imgf000043_0001
Figure imgf000044_0001
Figure imgf000045_0001
Figure imgf000046_0001
Figure imgf000047_0001
Figure imgf000048_0001
Figure imgf000049_0001
Figure imgf000050_0001
Figure imgf000051_0001
Figure imgf000052_0001
Figure imgf000053_0001
Figure imgf000054_0001
Figure imgf000055_0001
Figure imgf000056_0001
Figure imgf000057_0001
Figure imgf000058_0001
Figure imgf000059_0001
Figure imgf000060_0001
Figure imgf000061_0001
Figure imgf000062_0001
Figure imgf000063_0001
Figure imgf000064_0001
Figure imgf000065_0001
Figure imgf000066_0001
[0100] In another embodiment, a method of preparing the inventive compounds is provided. The compounds of the present invention can be generally prepared using cyanuric chloride as a starting material. Compounds of formula (I) or formula (II) may contain various stereoisomers, geometric isomers, tautomeric isomers, and the like. All of possible isomers and their mixtures are included in the present invention, and the mixing ratio is not particularly limited.
[0101] The triazine derivative compounds of formula (I) or formula (II) in this invention can be prepared by known procedure in the prior art. The examples could be found in US Patent Application Publication No. 2005/0250945A1 ; US Patent Application Publication No. 2005/0227983 Al; PCT WO 05/007646A1; PCT WO 05/007648A2; PCT WO 05/003103 A2; PCT WO 05/011703 Al ; and J. Med. Chem. (2004), 47(19), 4649-4652. Starting materials are commercially available from suppliers such as Sigma-Aldrich Corp. (St. Louis, MO), or may be synthesized from commercially available precursors using established protocols. By way of example, a synthetic route similar to that shown in any of the following Schemes may be used, together with synthetic methods known in the art of synthetic organic chemistry, or variations thereon as appreciated by those skilled in the art. Each variable in the following schemes refers to any group consistent with the description of the compounds provided herein.
[0102] In the Schemes that follow the term "reduction" refers to the process of reducing a nitro functionality to an amino functionality, or the process of transforming an ester functionality to an alcohol. The reduction of a nitro group can be carried out in a number of ways well known to those skilled in the art of organic synthesis including, but not limited to, catalytic hydrogenation, reduction with SnCl2 and reduction with titanium bichloride. The reduction of an ester group is typically performed using metal hydride reagents including, but not limited to, diisobutyl-aluminum hydride (DIBAL), lithium aluminum hydride (LAH), and sodium borohydride. For an overview of reduction methods see: Hudlicky, M. Reductions in Organic Chemistry, ACS Monograph 188, 1996. In the Schemes that follow, the term "hydrolyze" refers to the reaction of a substrate or reactant with water. More specifically, "hydrolyze" refers to the conversion of an ester or nitrite functionality into a carboxylic acid. This process can be catalyzed by a variety of acids or bases well known to those skilled in the art of organic synthesis.
[0103] The compounds of formula (I) or formula (II) may be prepared by use of known chemical reactions and procedures. The following general preparative methods are presented to aid one of skill in the art in synthesizing the inhibitors, with more detailed examples being presented in the experimental section describing the working examples. [0104] Heterocyclic amines are defined in formula (III). Some of heterocyclic amines are commercially available, others may be prepared by known procedure in the prior art (Katritzky, et al. Comprehensive Heterocyclic Chemistry; Permagon Press: Oxford, UK, 1984, March. Advanced Organic Chemistry, 3rd Ed.; John Wiley: New York, 1985), or by using common knowledge of organic chemistry.
Figure imgf000068_0001
[0105] For example, heterocyclic amine with an amide link (Ilia) can be prepared from commercial compounds as illustrated in Scheme 1. By Route 1 , the amine is first protected by Boc or other appropriate protecting group; after hydrolysis, the acid can be converted to corresponding amide; followed by removal of protecting group, the desired amine can be obtained. Alternatively, by Route 2, the acid, which is either commercially available, or made from its ester form, can also be converted to the desired compound (Ilia). A lot of heterocyclic amines can be prepared by this way.
Scheme 1
Figure imgf000068_0002
[0106] Substituted heterocyclic amines can also be generated using standard methods (March, J. Advanced Organic Chemistry, 4th Ed.; John Wiley, New York (1992); Larock, R.C. Comprehensive Organic Transformations, 2nd Ed., John Wiley, New York (1999); PCT No. WO 99/32106). As shown in Scheme 2, heterocyclic amines can be commonly synthesized by reduction of nitroheteros using a metal catalyst, such as Ni, Pd, or Pt, and H2 or a hydride transfer agent, such as formate, cyclohexadiene, or a borohydride (Rylander. Hydrogenation Methods; Academic Press: London, UK (1985)). Nitroheteros may also be directly reduced using a strong hydride source, such as LAH, (Seyden-Penne. Reductions by the Alumino- and Borohydrides in Organic Synthesis; VCH Publishers: New York (1991)), or using a zero valent metal, such as Fe, Sn or Ca, often in acidic media. Many methods exist for the synthesis of nitroaryls (March, J. Advanced Organic Chemistry, 4th Ed.; John Wiley, New York (1992); Larock, R.C. Comprehensive Organic Transformations, 2nd Ed., John Wiley, New York (1999))).
Scheme 2
Figure imgf000069_0001
[0107] Nitroheteroaryls may be further elaborated prior to reduction. Nitroheteros substituted with potential leaving groups (eg. F, Cl, Br, etc.) may undergo substitution reactions on treatment with nucleophiles, such as thiolate (exemplified in Scheme 3) or phenoxide. Nitroaryls may also undergo Ullman-type coupling reactions (Scheme 3).
Scheme 3
Figure imgf000069_0002
[0108] Scheme 4 illustrates one of the methods to prepare those heterocyclic amines as in Formula IHb, where L is carbonyl. These heterocyclic amines are readily available from reactions of a heterocyclic amine with a substituted aryl carbonyl chloride. Acetyl protection of the amine, which can be easily removed after the Friedel-Crafts reaction, is preferred. These carbonyl linked heterocyclic amines can be further converted to methylene (IIIc) or hydroxyl methylene (HId) linked ones by appropriate reduction.
Scheme 4
Figure imgf000070_0001
[0109] As illustrated in Scheme 5, 2-amino thiazole-5-carboxamide or 2-amino-oxazole- 5-carboxamide (HId) are available by the reaction of thiourea or urea with an appropriate ethoxyacrylamide in the presence of NBS, which can be made from the reaction of 3-ethyoxyacryloyl chloride with a corresponding amino compound R' -NH2. The 3-ethyoxyacryloyl chloride can be prepared from the corresponding acid or ester.
Scheme 5
Figure imgf000070_0002
[0110] The preparation of the compound of formula (IV) in this invention can be carried out by methods known in the art (e.g., J Med. Chem. 1996, 39, 4354-4357; J. Med. Chem. 2004, 47, 600-611; J Med. Chem. 2004, 47, 6283-6291; J Med. Chem. 2005, 48, 1717-1720; J Med. Chem. 2005, 48, 5570-5579; US Patent No. 6340683 Bl; JOC, 2004, 29, 7809-7815).
Figure imgf000071_0001
[0111] Scheme 6 illustrated the synthesis method for compounds with alkyl or aryl as R1. The 6-alkyl or aryl substituted dichloro-triazine (b) may be synthesized by the methods known in the art (e.g., J Med. Chem. 1999, 42, 805-818 and J Med. Chem. 2004, 47, 600- 611) from cyanuric chloride (a) and Grignard reagents. Triazine derivatives can be formed from the reaction of a 6-alkyl or aryl substituted dichloro-triazine (b) with heterocyclic amine, followed by reaction with HR2. Alternatively, the monochloro-triazine (c) can be converted to amino triazine (d), which can react with YR2, to give a triazine derivative (IV). Also, dichloro-triazine (b) can react with HR2, followed by reaction with heterocyclic amine to give triazine derivative (IV). Further more, monochloro-triazine (e) can be converted to amino triazine (f), which can react with a leaving-group-attached heterocyclic compound (g), to give a triazine derivative (IV).
Scheme 6
[0112] As shown in Scheme 7, the triazine derivative can also be synthesized by the reaction of cyanuric chloride with a sequence of heterocyclic amines and HR2 to give 2,4-disubstituted-6-chloro-l,3,5-triazines. The displacement of the last chlorine by amine, hydrazine, hydroxyl or other nucleophilic group can be achieved by increasing the temperature, affording the trisubstituted-l,3,5-triazines (IV).
Scheme 7
Figure imgf000073_0001
[0113] Furthermore, as shown in Scheme 7, the triazine derivative can be synthesized by the reaction of tri, di- or mono chloride triazine with a heterocyclic amines then the R3-L can be added to the heterocyclic moiety. For example, an amide moiety can be added this way, where is triazines (IV).
[0114] Other triazine derivatives of formula (I), where K is not NH, can be prepared in a similar way.
Figure imgf000073_0002
[0115] The reaction is preferably conducted in the presence of an inert solvent. There is no particular restriction on the nature of the solvent to be employed, provided that it has no adverse effect on the reaction or on the reagents involved and that it can dissolve the reagents, at least to some extent. Examples of suitable solvents include: aliphatic hydrocarbons, such as hexane, heptane, ligroin and petroleum ether; aromatic hydrocarbons, such as benzene, toluene and xylene; halogenated hydrocarbons, especially aromatic and aliphatic hydrocarbons, such as methylene chloride, chloroform, carbon tetrachloride, dichloroethane, chlorobenzene and the dichlorobenzenes; esters, such as ethyl formate, ethyl acetate, propyl acetate, butyl acetate and diethyl carbonate; ethers, such as diethyl ether, diisopropyl ether, tetrahydrofuran, dioxane. dimethoxyethane and diethylene glycol dimethyl ether; ketones, such as acetone, methyl ethyl ketone, methyl isobutyl ketone, isophorone and cyclohexanone; nitro compounds, which may be nitroalkanes or nitroaranes, such as nitroethane and nitrobenzene; nitriles, such as acetonitrile and isobutyronitrile; amides, which may be fatty acid amides, such as formamide, dimethylformamide, dimethylacetamide and hexamethylphosphoric triamide; and sulphoxides, such as dimethyl sulphoxide and sulpholane.
[0116] The reaction can take place over a wide range of temperatures, and the precise reaction temperature is not critical to the invention. In general, we find it convenient to carry out the reaction at a temperature of from -50°C to 100°C.
[0117] The present invention provides compositions of matter that are formulations of one or more active drugs and a pharmaceutically-acceptable carrier. In this regard, the invention provides a composition for administration to a mammalian subject , which may include a compound of formula (I) or formula (II), or its pharmaceutically acceptable salts. [0118] Pharmaceutically acceptable salts of the compounds of this invention include those derived from pharmaceutically acceptable inorganic and organic acids and bases. Examples of suitable acid salts include acetate, adipate, alginate, aspartate, benzoate, benzenesulfonate, bisulfate, butyrate, citrate, camphorate, camphorsulfonate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, formate, fumarate, glucoheptanoate, glycerophosphate, glycolate, hemisulfate, heptanoate, hexanoate, hydrochloride, hydrobromide, hydroiodide, 2-hydroxyethanesulfonate, lactate, maleate, malonate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oxalate, palmoate, pectinate, persulfate, 3-phenylpropionate, phosphate, picrate, pivalate, propionate, salicylate, succinate, sulfate, tartrate, thiocyanate, tosylate and undecanoate. Other acids, such as oxalic, while not in themselves pharmaceutically acceptable, may be employed in the preparation of salts useful as intermediates in obtaining the compounds of the invention and their pharmaceutically acceptable acid addition salts.
[0119] Salts derived from appropriate bases include alkali metal (e.g., sodium and potassium), alkaline earth metal (e.g., magnesium), 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 dispersible products may be obtained by such quaternization.
[0120] 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" as used herein includes subcutaneous, intravenous, intramuscular, intra-articular, intra-synovial, intrasternal, intrathecal, intrahepatic, intralesional and intracranial injection or infusion techniques. Preferably, the compositions are administered orally, intraperitoneally or intravenously. [0121] The pharmaceutically acceptable compositions of this invention may be orally administered in any orally acceptable dosage form including, but not limited to, capsules, tablets, troches, elixirs, suspensions, syrups, wafers, chewing gums, aqueous suspensions or solutions.
[0122] The oral compositions may contain additional ingredients such as: a binder such as macrocrystalline cellulose, gum tragacanth or gelatin; an excipient such as starch or lactose, a disintegrating agent such as alginic acid, corn starch and the like; a lubricant such as magnesium stearate; a glidant such as colloidal silicon dioxide; and a sweetening agent such as sucrose or saccharin or flavoring agent such as peppermint, methyl salicylate, or orange flavoring. When the dosage unit form is a capsule, it may additionally contain a liquid carrier such as a fatty oil. Other dosage unit forms may contain other various materials which modify the physical form of the dosage unit, such as, for example, a coating. Thus, tablets or pills may be coated with sugar, shellac, or other enteric coating agents. A syrup may contain, in addition to the active ingredients, sucrose as a sweetening agent and certain preservatives, dyes and colorings and flavors. Materials used in preparing these various compositions should be pharmaceutically or veterinarally pure and non-toxic in the amounts used.
[0123] For the purposes of parenteral therapeutic administration, the active ingredient may be incorporated into a solution or suspension. The solutions or suspensions may also include the following components: a sterile diluent such as water for injection, saline solution, fixed oils, polyethylene glycols, glycerine, propylene glycol or other synthetic solvents; antibacterial agents such as benzyl alcohol or methyl parabens; antioxidants such as ascorbic acid or sodium bisulfite; chelating agents such as ethylenediaminetetraacetic acid; buffers such as acetates, citrates or phosphates and agents for the adjustment of tonicity such as sodium chloride or dextrose. The parenteral preparation can be enclosed in ampoules, disposable syringes or multiple dose vials made of glass or plastic. [0124] The pharmaceutical forms suitable for injectable use include sterile solutions, dispersions, emulsions, and sterile powders. The final form should be stable under conditions of manufacture and storage. Furthermore, the final pharmaceutical form should be protected against contamination and should, therefore, be able to inhibit the growth of microorganisms such as bacteria or fungi. A single intravenous or intraperitoneal dose can be administered. Alternatively, a slow long-term infusion or multiple short-term daily infusions may be utilized, typically lasting from 1 to 8 days. Alternate day dosing or dosing once every several days may also be utilized.
[0125] Sterile, injectable solutions may be prepared by incorporating a compound in the required amount into one or more appropriate solvents to which other ingredients, listed above or known to those skilled in the art, may be added as required. Sterile injectable solutions may be prepared by incorporating the compound in the required amount in the appropriate solvent with various other ingredients as required. Sterilizing procedures, such as filtration, may then follow. Typically, dispersions are made by incorporating the compound into a sterile vehicle which also contains the dispersion medium and the required other ingredients as indicated above. In the case of a sterile powder, the preferred methods include vacuum drying or freeze drying to which any required ingredients are added. [0126] Suitable pharmaceutical carriers include sterile water; saline, dextrose; dextrose in water or saline; condensation products of castor oil and ethylene oxide combining about 30 to about 35 moles of ethylene oxide per mole of castor oil; liquid acid; lower alkanols; oils such as corn oil; peanut oil, sesame oil and the like, with emulsifiers such as mono- or di-glyceride of a fatty acid, or a phosphatide, e.g., lecithin, and the like; glycols; polyalkylene glycols; aqueous media in the presence of a suspending agent, for example, sodium carboxymethylcellulose; sodium alginate; poly(vinylpyrolidone) ; and the like, alone, or with suitable dispensing agents such as lecithin; polyoxyethylene stearate; and the like. The carrier may also contain adjuvants such as preserving stabilizing, wetting, emulsifying agents and the like together with the penetration enhancer. In all cases, the final form, as noted, must be sterile and should also be able to pass readily through an injection device such as a hollow needle. The proper viscosity may be achieved and maintained by the proper choice of solvents or excipients. Moreover, the use of molecular or particulate coatings such as lecithin, the proper selection of particle size in dispersions, or the use of materials with surfactant properties may be utilized.
[0127] In accordance with the invention, there are provided compositions containing triazine derivatives and methods useful for the in vivo delivery of triazine derivatives in the form of nanoparticles, which are suitable for any of the aforesaid routes of administration. [0128] United States Patent Nos. 5,916,596, 6,506,405 and 6,537,579 teach the preparation of nanoparticles from the biocompatible polymers, such as albumin. Thus, in accordance with the present invention, there are provided methods for the formation of nanoparticles of the present invention by a solvent evaporation technique from an oil-in- water emulsion prepared under conditions of high shear forces (e.g., sonication, high pressure homogenization, or the like).
[0129] 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. [0130] 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 lower intestinal tract. Suitable topical formulations are readily prepared for each of these areas or organs.
[0131] 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.
[0132] 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. Carriers for topical administration of the compounds of this invention include, but are not limited to, mineral oil, liquid petrolatum, white petrolatum, propylene glycol, polyoxyethylene, 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. Suitable carriers include, but are not limited to, mineral oil, sorbitan monostearate, polysorbate 60, cetyl esters wax, cetearyl alcohol, 2- octyldodecanol, benzyl alcohol and water.
[0133] For ophthalmic use, the pharmaceutically acceptable compositions may be formulated as micronized suspensions in isotonic, pH adjusted sterile saline, or, preferably, as solutions in isotonic, pH adjusted sterile saline, 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. [0134] 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.
[0135] Most preferably, the pharmaceutically acceptable compositions of this invention are formulated for oral administration.
[0136] In accordance with the invention, the compounds of the invention may be used to treat diseases associated with cellular proliferation or hyperproliferation, such as cancers which include but are not limited to tumors of the nasal cavity, paranasal sinuses, nasopharynx, oral cavity, oropharynx, larynx, hypopharynx, salivary glands, and paragangliomas. The compounds of the invention may also be used to treat cancers of the liver and biliary tree (particularly hepatocellular carcinoma), intestinal cancers, particularly colorectal cancer, ovarian cancer, small cell and non-small cell lung cancer, breast cancer, sarcomas (including fibrosarcoma, malignant fibrous histiocytoma, embryonal rhabdomysocarcoma, leiomysosarcoma, neuro-fibrosarcoma, osteosarcoma, synovial sarcoma, liposarcoma, and alveolar soft part sarcoma), neoplasms of the central nervous systems (particularly brain cancer), and lymphomas (including Hodgkin's lymphoma, lymphoplasmacytoid lymphoma, follicular lymphoma, mucosa-associated lymphoid tissue lymphoma, mantle cell lymphoma, B-lineage large cell lymphoma, Burkitt's lymphoma, and T-cell anaplastic large cell lymphoma).
[0137] The compounds and methods of the present invention, either when administered alone or in combination with other agents (e.g., chemotherapeutic agents or protein therapeutic agents described below) are also useful in treating a variety of disorders, including but not limited to, for example: stroke, cardiovascular disease, myocardial infarction, congestive heart failure, cardiomyopathy, myocarditis, ischemic heart disease, coronary artery disease, cardiogenic shock, vascular shock, pulmonary hypertension, pulmonary edema (including cardiogenic pulmonary edema), pleural effusions, rheumatoid arthritis, diabetic retinopathy, retinitis pigmentosa, and retinopathies, including diabetic retinopathy and retinopathy of prematurity, inflammatory diseases, restenosis, asthma, acute or adult respiratory distress syndrome (ARDS), lupus, vascular leakage, protection from ischemic or reperfusion injury such as ischemic or reperfusion injury incurred during organ transplantation, transplantation tolerance induction; ischemic or reperfusion injury following angioplasty; arthritis (such as rheumatoid arthritis, psoriatic arthritis or osteoarthritis); multiple sclerosis; inflammatory bowel disease, including ulcerative colitis and Crohn's disease; lupus (systemic lupus crythematosis); graft vs. host diseases; T- cell mediated hypersensitivity diseases, including contact hypersensitivity, delayed- type hypersensitivity, and gluten-sensitive enteropathy (Celiac disease); Type 1 diabetes; psoriasis; contact dermatitis (including that due to poison ivy); Hashimoto's thyroiditis; Sjogren's syndrome; Autoimmune Hyperthyroidism, such as Graves' disease; Addison's disease (autoimmune disease of the adrenal glands); autoimmune polyglandular disease (also known as autoimmune polyglandular syndrome); autoimmune alopecia; pernicious anemia; vitiligo; autoimmune hypopituatarism; Guillain-Barre syndrome; other autoimmune diseases; cancers, including those where kineses such as Src-family kineses are activated or overexpressed, such as colon carcinoma and thymoma, or cancers where kinase activity facilitates tumor growth or survival; glomerulonephritis, serum sickness; uticaria; allergic diseases such as respiratory allergies (asthma, hayfever, allergic rhinitis) or skin allergies; mycosis fungoides; acute inflammatory responses (such as acute or adult respiratory distress syndrome and ischemialreperfusion injury); dermatomyositis; alopecia areata; chronic actinic dermatitis; eczema; Behcet's disease; Pustulosis palmoplanteris; Pyoderma gangrenum; Sezary's syndrome; atopic dermatitis; systemic schlerosis; morphea; peripheral limb ischemia and ischemic limb disease; bone disease such as osteoporosis, osteomalacia, hyperparathyroidism, Paget's disease, and renal osteodystrophy; vascular leak syndromes, including vascular leak syndromes induced by chemotherapies or immunomodulators such as IL-2; spinal cord and brain injury or trauma; glaucoma; retinal diseases, including macular degeneration; vitreoretinal disease; pancreatitis; vasculatides, including vasculitis, Kawasaki disease, thromboangiitis obliterans, Wegener s granulomatosis, and Behcet's disease; scleroderma; preeclampsia; thalassemia; Kaposi's sarcoma; von Hippel Lindau disease; and the like. [0138] In accordance with the invention, the compounds of the invention may be used to treat diseases associated with undesired cellular proliferation or hyperproliferation comprising identifying the mammal afflicted with said disease or condition and administering to said afflicted mammal a composition comprising the compound of formula 1 , wherein the disease or condition is associated with a kinase.
[0139] In accordance with the invention, the compounds of the invention may be used to treat diseases associated with undesired cellular proliferation or hyperproliferation comprising identifying the mammal afflicted with said disease or condition and administering to said afflicted mammal a composition comprising a compound of formula (I) or formula (II), wherein the disease or condition is associated with a tyrosine kinase. [0140] In accordance with the invention, the compounds of the invention may be used to treat diseases associated with undesired cellular proliferation or hyperproliferation comprising identifying the mammal afflicted with said disease or condition and administering to said afflicted mammal a composition comprising a compound of formula (I) or formula (II), wherein the disease or condition is associated with the kinase that is a serine kinase or a threonine kinase.
[0141] In accordance with the invention, the compounds of the invention may be used to treat diseases associated with undesired cellular proliferation or hyperproliferation comprising identifying the mammal afflicted with said disease or condition and administering to said afflicted mammal a composition comprising the compound of formula (I) or formula (II), wherein the disease or condition is associated with the kinase that is a Src family kinase. [0142] In accordance with the invention, the compounds of the invention may be used to treat diseases associated with undesired cellular proliferation or hyperproliferation comprising identifying the mammal afflicted with said disease or condition and administering to said afflicted mammal a composition comprising the compound of formula (I) or formula (II), wherein the disease or condition is associated with the kinase that is a Aurora family kinase.
[0143] The invention also provides methods of treating a mammal afflicted with the above diseases and conditions. 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 0.01-100 mg/kg body weight/day of the inhibitor can be administered to a patient receiving these compositions. [0144] In one aspect, the invention compounds are administered in combination with chemotherapeutic agent, an anti-inflammatory agent, antihistamines, chemotherapeutic agent, immunomodulator, therapeutic antibody or a protein kinase inhibitor, e.g., a tyrosine kinase inhibitor, to a subject in need of such treatment.
[0145] The method includes administering one or more of the inventive compounds to the afflicted mammal. The method may further include the administration of a second active agent, such as a cytotoxic agent, including alkylating agents, tumor necrosis factors, intercalators, microtubulin inhibitors, and topoisomerase inhibitors. The second active agent may be co-administered in the same composition or in a second composition. Examples of suitable second active agents include, but are not limited to, a cytotoxic drug such as Acivicin; Aclarubicin; Acodazole Hydrochloride; AcrQnine; Adozelesin; Aldesleukin; Altretamine; Ambomycin; Ametantrone Acetate; Aminoglutethimide; Amsacrine; Anastrozole; Anthramycin; Asparaginase; Asperlin; Azacitidine; Azetepa; Azotomycin; Batimastat; Benzodepa; Bicalutamide; Bisantrene Hydrochloride; Bisnafide Dimesylate; Bizelesin; Bleomycin Sulfate; Brequinar Sodium; Bropirimine; Busulfan; Cactinomycin; Calusterone; Caracemide; Carbetimer; Carboplatin; Carmustine; Carubicin Hydrochloride; Carzelesin; Cedefmgol; Chlorambucil; Cirolemycin; Cisplatin; Cladribine; Crisnatol Mesylate; Cyclophosphamide; Cytarabine; Dacarbazine; Dactinomycin; Daunorubicin Hydrochloride; Decitabine; Dexormaplatin; Dezaguanine; Dezaguanine Mesylate; Diaziquone; Docetaxel; Doxorubicin; Doxorubicin Hydrochloride; Droloxifene; Droloxifene Citrate; Dromostanolone Propionate; Duazomycin; Edatrexate; Eflomithine Hydrochloride; Elsamitrucin; Enloplatin; Enpromate; Epipropidine; Epirubicin Hydrochloride; Erbulozole; Esorubicin Hydrochloride; Estramustine; Estramustine Phosphate Sodium; Etanidazole; Ethiodized Oil 131; Etoposide; Etoposide Phosphate; Etoprine; Fadrozole Hydrochloride; Fazarabine; Fenretinide; Floxuridine; Fludarabine Phosphate; Fluorouracil; Flurocitabine; Fosquidone; Fostriecin Sodium; Gemcitabine; Gemcitabine Hydrochloride; Gold Au 198; Hydroxyurea; Idarubicin Hydrochloride; Ifosfamide; Ilmofosine; Interferon Alfa-2a; Interferon Alfa-2b; Interferon Alfa-nl; Interferon Alfa-n3; Interferon Beta- Da; Interferon Gamma- Ib; Iproplatin; Irinotecan Hydrochloride; Lanreotide Acetate; Letrozole; Leuprolide Acetate; Liarozole Hydrochloride; Lometrexol Sodium; Lomustine; Losoxantrone Hydrochloride; Masoprocol; Maytansine; Mechlorethamine Hydrochloride; Megestrol Acetate; Melengestrol Acetate; Melphalan; Menogaril; Mercaptopurine; Methotrexate; Methotrexate Sodium; Metoprine; Meturedepa; Mitindomide; Mitocarcin; Mitocromin; Mitogillin; Mitomalcin; Mitomycin; Mitosper; Mitotane; Mitoxantrone Hydrochloride; Mycophenolic Acid; Nocodazole; Nogalamycin; Ormaplatin; Oxisuran; Paclitaxel; Pegaspargase; Peliomycin; Pentamustine; Peplomycin Sulfate; Perfosfamide; Pipobroman; Piposulfan; Piroxantrone Hydrochloride; Plicamycin; Plomestane; Porfimer Sodium; Porfϊromycin; Prednimustine; Procarbazine Hydrochloride; Puromycin; Puromycin Hydrochloride; Pyrazofurin; Riboprine; Rogletimide; Safingol; Safingol Hydrochloride; Semustine; Simtrazene; Sparfosate Sodium; Sparsomycin; Spirogermanium Hydrochloride; Spiromustine; Spiroplatin; Streptonigrin; Streptozocin; Strontium Chloride Sr 89; Sulofenur; Talisomycin; Taxane; Taxoid; Tecogalan Sodium; Tegafur; Teloxantrone Hydrochloride; Temoporfin; Teniposide; Teroxirone; Testolactone; Thiamiprine; Thioguanine; Thiotepa; Tiazofurin; Tirapazamine; Topotecan Hydrochloride; Toremifene Citrate; Trestolone Acetate; Triciribine Phosphate; Trimetrexate; Trimetrexate Glucuronate; Triptorelin; Tubulozole Hydrochloride; Uracil Mustard; Uredepa; Vapreotide; Verteporfin; Vinblastine Sulfate; Vincristine Sulfate; Vindesine; Vindesine Sulfate; Vinepidine Sulfate; Vinglycinate Sulfate; Vinleurosine Sulfate; Vinorelbine Tartrate; Vinrosidine Sulfate; Vinzolidine Sulfate; Vorozole; Zeniplatin; Zinostatin; and Zorubicin Hydrochloride.
[0146] In accordance with the invention, the compounds and compositions may be used at sub-cytotoxic levels in combination with other agents in order to achieve highly selective activity in the treatment of non-neoplastic disorders, such as heart disease, stroke and neurodegenerative diseases (Whitesell et al., Curr Cancer Drug Targets (2003), 3(5), 349- 58).
[0147] The exemplary therapeutical agents that may be administered in combination with invention compounds include EGFR inhibitors, such as gefitinib, erlotinib, and cetuximab. Her2 inhibitors include canertinib, EKB-569, and GW-572016. Also included are Src inhibitors, dasatinib, as well as Casodex (bicalutamide), Tamoxifen, MEK-I kinase inhibitors, MARK kinase inhibitors, PD inhibitors, and PDGF inhibitors, such as imatinib, Hsp90 inhibitors, such as 17-AAG and 17-DMAG. Also included are anti-angiogenic and antivascular agents which, by interrupting blood flow to solid tumors, render cancer cells quiescent by depriving them of nutrition. Castration, which also renders androgen dependent carcinomas non-proliferative, may also be utilized. Also included are IGFlR inhibitors, inhibitors of non- receptor and receptor tyrosine kineses, and inhibitors of integrin. [0148] The pharmaceutical composition and method of the present invention may further combine other protein therapeutic agents such as cytokines, immunomodulatory agents and antibodies. As used herein the term "cytokine" encompasses chemokines, interleukins, lymphokines, monokines, colony stimulating factors, and receptor associated proteins, and functional fragments thereof. As used herein, the term "functional fragment" refers to a polypeptide or peptide which possesses biological function or activity that is identified through a defined functional assay. The cytokines include endothelial monocyte activating polypeptide II (EMAP- II), granulocyte-macrophage-CSF (GM-CSF), granulocyte-CSF (G- CSF), macrophage- CSF (M-CSF), IL-I, IL-2, IL-3, IL- 4, IL-5, IL-6, IL-12, and IL-13, interferons, and the like and which is associated with a particular biologic, morphologic, or phenotypic alteration in a cell or cell mechanism.
[0149] Other therapeutic agents for the combinatory therapy include cyclosporins (e.g., cyclosporin A), CTLA4-Ig, antibodies such as ICAM-3, anti-IL-2 receptor (Anti-Tac), anti- CD45RB, anti-CD2, anti-CD3 (OKT-3), anti-CD4, anti-CD80, anti-CD86, agents blocking the interaction between CD40 and gp39, such as antibodies specific for CD40 and for gpn39 (i.e., CDl 54), fusion proteins constructed from CD40 and gp39 (CD40Ig and CD8gp39), inhibitors, such as nuclear translocation inhibitors, of NF-kappa B function, such as deoxyspergualin (DSG), cholesterol biosynthesis inhibitors such as HM:G CoA reductase inhibitors (lovastatin and simvastatin), non-steroidal antiinflammatory drugs (NSAIDs) such as ibuprofen and cyclooxygenase inhibitors such as rofecoxib, steroids such as prednisone or dexamethasone, gold compounds, antiproliferative agents such as methotrexate, FK506 (tacrolimus, Prograf), mycophenolate mofetil, cytotoxic drugs such as azathioprine and cyclophosphamide, TNF-a inhibitors such as tenidap, anti-TNF antibodies or soluble TNF receptor, and rapamycin (sirolimus or Rapamune) or derivatives thereof. [0150] When other therapeutic agents are employed in combination with the compounds of the present invention they may be used for example in amounts as noted in the Physician Desk Reference (PDR) or as otherwise determined by one having ordinary skill in the art. [0151] The following examples are provided to further illustrate the present invention but, of course, should not be construed as in any way limiting its scope. [0152] All experiments were performed under anhydrous conditions (i.e. dry solvents) in an atmosphere of argon, except where stated, using oven-dried apparatus and employing standard techniques in handling air-sensitive materials. Aqueous solutions of sodium bicarbonate (NaHCCβ) and sodium chloride (brine) were saturated. [0153] Analytical thin layer chromatography (TLC) was carried out on Merck Kiesel gel 60 F254 plates with visualization by ultraviolet and/or anisaldehyde, potassium permanganate or phosphomolybdic acid dips.
[0154] NMR spectra: IH Nuclear magnetic resonance spectra were recorded at 400 MHz. Data are presented as follows: chemical shift, multiplicity (s = singlet, d = doublet, t = triplet, q = quartet, qn = quintet, dd = doublet of doublets, m = multiplet, bs = broad singlet), coupling constant (J/Hz) and integration. Coupling constants were taken and calculated directly from the spectra and are uncorrected.
[0155] Low resolution mass spectra: Electrospray (ES+) ionization was used. The protonated parent ion (M+H) or parent sodium ion (M+Na) or fragment of highest mass is quoted. Analytical gradient consisted of 10% ACN in water ramping up to 100% ACN over 5 minutes unless otherwise stated.
Example 1
Figure imgf000084_0001
[0156] A mixture of ethyl β-ethoxyacrylate (26.50 g, 183 mmol) and 2 N sodium hydroxide (110 mL, 220 mmol) was refluxed for 2 h and cooled to 0° C water was removed under vacc, and the yellow solids were triturated with toluene and evaporated to give the sodium β-ethoxyacrylate (25 g, 97%). The mixture of sodium β-thoxyacrylate (10.26 g, 74.29 mmol) and thionyl chloride (25 mL, 343 mmol) was refluxed for 2 h, and evaporated, to give the β-ethoxyacryloyl chloride crude product, which was used without purification. To a cold stirring solution of 3-ethoxyacryloyl chloride in THF (100 mL) was added 2-chloro-6- methylaniline (6.2 mL, 50.35 mmol) and pyridine (9 ml, 111 mmol). The mixture was then warmed and stirred overnight at room temperature. Water was added at 0-10° C, extracted with EtOAc. The organic layer was washed with CuSO4 (3x50 mL) and the resulting solution was passed a pad of silica gel, concentrated under vacuum to give solids. The solids was diluted with toluene and kept, at O0C. The solid was collected by vacuum filtration, washed with water and dried to give 5.2 g (43% yield) of compound 1, (E)-N-(2-chloro-6- methylphenyl)-3-ethoxyacrylamide). 1H NMR (500 Hz, CDCl3) δ 1.26 (t, 3H, J=7 Hz), 2.15 (s, 3H), 3.94 (q, 2H, J=7 Hz), 5.58 (d, IH, J=12.4 Hz), 7.10-7.27 (m, 2H, J=7.5 Hz), 7.27- 7.37 (d, IH, J=7.5 Hz), 7.45(d, IH, J=12.4 Hz); ESI-MS: calcd for (C12Hi4ClNO2) 239, found 240 MH+). Example 2
Figure imgf000085_0001
[0157] To a mixture of compound 1 (5.30 g, 22.11 mmol) in 1,4-dioxane (100 mL) and water (70 mL) was added NBS (4.40 g, 24.72 mmol) at -10 to 0° C. The slurry was warmed and stirred at 20-22° C for 3 h. Thiourea (1.85 g, 26.16 mmol) was added and the mixture heated to 100° C. After 2 h, the resulting solution was cooled to 20-22° C and cone, ammonium hydroxide (6 mL) was added drop wise. The resulting slurry was concentrated under vacuum to about half volume and cooled to 0-5° C. The solid was collected by vacuum filtration, washed with cold water, and dried to give 5.4 g (90% yield) of compound 2 as deep-yellow solids. 1H NMR (500 MHz, DMSO-d6) δ 2.19 (s, 3H), 7.09-7.29 (m, 2H, J=7.5), 7.29-7.43 (d, IH, J=7.5), 7.61 (s, 2H), 7.85 (s, IH), 9.63 (s, IH); ESI-MS: calcd for (CπHioClN3OS) 267, found 268 MH+).
Example 3
Figure imgf000085_0002
[0158] A solution of methylmagnesium bromide in ether (3M, 30 ml, 90 mmole) was added dropwise to a stirred solution of cyanuric chloride (3.91 g, 21.20 mmole) in anhydrous dichloromethane at -10° C. After the addition was complete, the reaction mixture was stirred at -5° C for 4 h, after which time water was added dropwise at a rate such that the temperature of the reaction stayed below 10° C. After warming to room temperature, the reaction mixture was diluted with additional water and methylene chloride and passed through a pad of cilite. The organic layer was dried and evaporated to give 2,4-dichloro-6-methyl-l,3,5-triazine of 4 as yellow solids (3.02 g, 87%). 1H NMR (CDCl3) δ 2.70 (s, 3H).
Example 4
Figure imgf000085_0003
[0159] A solution of Compound 3 (560 mg, 3.41 mmole), diisopropylamine (1.00ml, 5.74 mmole) and Compound 2 (700 mg, 2.65 mmole) in THF (40 mL) was stirred at 0° C for 30 min, then at room temperature for 2 hours. Water was added to the reaction mixture, and the aqueous mixture was extracted twice with EtOAc. The combined extracts were washed with brine, dried, and evaporated in vacuo. Column chromatography provided Compound 4 as light yellow solids (350 mg, 33%). 1H NMR (500 MHz, DMSOd6) 5 2.19 (s, 3H), 2.49 (s, 3H), 7.36-7.58 (m, 3H), 8.23 (br, IH), 9.61 (br, IH), 11.63 (br, IH); ESI-MS: calcd for (C15H12Cl2N6OS) 394, found 395 (MH+).
Example 5
Figure imgf000086_0001
[0160] A mixture of 4 (100 mg, 0.25 mmol), diisopropylethylamine (0.08 mL, 0.50 mmol), and l-(2-hydroxyethyl)piperazine (100 mg, 0.77 mmol) in 1,4-dioxane (15 mL) was refluxed for 12 h. The mixture was concentrated under vacuum, and water was added. The solid was collected by filtration, triturated successively with H2O, aqueous MeOH, and Et2O (2χ) and dried in vacuoto give 5 as light yellow solids (55 g, 45%). 1H NMR (500 MHz, DMSCW6) Sl 1.97 (br s, IH), 10.00 (s, IH), 8.28 (s, IH), 7.40 (d, J= 7.6 Hz, IH), 7.29-7.24 (m, 2H), 4.45 (t, J= 5.4 Hz, IH), 3.87-3.81 (m, 4H), 3.52 (q, J= 6.0 Hz, 2H), 2.46 (m, 4H), 2.42 (t, J= 6.0Hz, 2H), 2.30 (s, 3H), 2.23 (s, 3H). ESI-MS: calcd for (C21H25ClN8O2S) 488, found 489 (MH+). Example 6
Figure imgf000086_0002
[0161] Compound 6 was prepared by the same procedure as was used in the preparation of Compound 5. Light yellow solids were obtained (42% yield). ESI-MS: calcd for (C24H24ClN9OS) 521, found 522 (MH+). Example 7
Figure imgf000087_0001
[0162] A mixture of 4 (200 mg, 0.51 mmol), diisopropylethylamine (0.35 mL, 2.03 mmol), and 1 -methyl piperazine (304 mg, 3.04 mmol) in DMSO (10 mL) was heated at 70° C for 13h. The mixture was extracted by ethyl acetate and the combined organic layers were washed with water and brine. The crude product was recrystallized by MeOH/CHCl3 (23 mg, 9.9%). 1H NMR (500 MHz, DMSOd6) 5 12.01 (br s, IH), 10.0 (br s, IH), 8.29 (s, IH), 7.42(d, J = 7.4Hz, IH), 7.30-7.24 (m, 2H), 3.89 (m, 4H), 2.54-2.43 (m, 4H), 2.34-2.23 (m, 9H), ESI-MS: calcd for (C20H23C1N8OS) 458, found 459 (M+H+). HPLC: retention time: 9.8 min; purity 93%.
Example 8
Figure imgf000087_0002
[0163] Compound 8 was prepared by the same procedure as was used in the preparation of Compound 5. Light yellow solids were obtained (94% yield). ESI-MS: calcd for (Ci9H2OClN7O2S) 445, found 446 (MH+).
Example 9
Figure imgf000087_0003
[0164] A mixture of 4 (100 mg, 0.25 mmol), diisopropylethylamine (0.07 mL, 0.391 mmol), and 4-(2-(piperazin-l-yl) ethyl) morpholine (152 mg, 0.76 mmol) in 1,4-dioxane (15 mL) was refluxed for 12h. The mixture was concentrated under vacuum, and water was added. The mixture was extracted by ethyl acetate and the combined organic was concentrated. The crude product was passed on a pad of silica gel by using 2 to 10% MeOH- NH3/CH2C12 (10 mg, 7%). 1H NMR (500 MHz, DMSO-J6) 811.97 (br s, IH), 9.99 (s, IH), 8.28 (s, IH), 7.40 (d, J= 7.0 Hz, IH), 7.30-7.24 (m, 2H), 3.87-3.80 (m, 4H), 3.54 (m, 4H), 2.58-2.41 (m, 12H), 2.34 (s, 3H), 2.23 (s, 3H), ESI-MS: calcd for (C25H32ClN9O2S) 557, found 558 (MH+). HPLC: retention time: 9.92 min.; purity: 97%.
Example 10
Figure imgf000088_0001
[0165] A mixture of 4 (125 mg, 0.32 mmol), diisopropylethylamine (0.085 mL, 0.48 mmol), and l-(pyridin-4-ylmethyl)piperazine (168 mg, 0.95 mmol) in 1,4-dioxane (15 mL) was refluxed for 12h. The mixture was concentrated under vacuum, and water was added. The mixture was extracted by ethyl acetate and the combined organic layer was concentrated. The crude product was purified by slica gel chromatographyl by using 5% to 10% MeOH/EtOAc (15 mg, 9%). 1H NMR (500 MHz, DMSO-J6) 611.97 (br s, IH), 9.97 (s, IH), 8.52-8.50 (d, J= 5.0 Hz, 2H), 8.27 (s, IH), 7.40-7.35 (m, 4H), 7.29-7.24 (m, 2H), 3.86-3.80 (m, 4H), 3.57 (s, 2H), 2.53-2.41 (m, 4H), 2.33 (s, 3H), 2.23 (s, 3H). ESI-MS: calcd for (C25H26ClN9OS) 535, found 536(MH+). HPLC: retention time: 11.55 min.; purity: 90%.
Example 11
Figure imgf000088_0002
[0166] A mixture of 4 (125 mg, 0.32 mmol), diisopropylethylamine (0.085 mL, 0.48 mmol), and piperidin-4-yl-methanol (109 mg, 0.95 mmol) in 1,4-dioxane (15 mL) was refluxed for 12h. The mixture was concentrated under vacuum, and water was added. The mixture was extracted by ethyl acetate and the combined organic layer was concentrated. The crude product was purified by slica gel chromatographyl by using 10% MeOH/EtOAc (30 mg, 20%). 1H NMR (500 MHz, DMSO-J6) 511.97 (br s, IH), 9.98 (s, IH), 8.28 (s, IH), 7.40 (d, J = 7.5 Hz, IH), 7.30-7.24 (m, 2H), 4.79-4.72 (m, 2H), 4.51 (t, J= 5.5 Hz, IH), 3.26 (m, 2H), 3.10-2.90 (m, 2H), 2.33 (s, 3H), 2.23 (s, 3H), 1.80-1.61 (m, 2H), 1.20-1.0 (m, 2H); ESI-MS: calcd for (C21H24ClN7O2S) 473, found 474(MH+). HPLC: retention time: 8.45 min.; purity: 98%.
Example 12
Figure imgf000089_0001
[0167] A mixture of 4 (125 mg, 0.32 mmol), diisopropylethylamine (0.085 mL, 0.48 mmol), and 2-(piperazin-l-yl)pyrazine (156 mg, 0.95 mmol) in 1,4-dioxane (15 mL) was refluxed for 12h. The mixture was concentrated under vacuum, and water was added. The mixture was extracted by ethyl acetate and the combined organic layer was concentrated. The crude product was purified by slica gel chromatographyl by using 10% MeOH/EtOAc (30 mg, 18%). 1H NMR (500 MHz, DMSO-c/6) 512.02 (br s, IH), 10.0 (s, IH), 8.38 (d, J= 1.2Hz, IH), 8.29 (s, IH), 8.10 (s, IH), 7.86 (d, J = 2.5 Hz, IH), 7.40 (d, J = 7.5 Hz, IH), 7.32-7.24 (m, 2H), 4.10-3.90 (m, 4H), 3.70-3.58 (m, 4H), 2.34 (s, 3H), 2.23 (s, 3H), ESI-MS: calcd for (C23H23C1NI0OS) 522, found 523 (MH+). HPLC: retention time: 24 min.; purity: 92%.
Example 13
Figure imgf000089_0002
[0168] A mixture of 4 (200 mg, 0.51 mmol), diisopropylethylamine (0.35 mL, 2.03 mmol), and piperazine (436 mg, 5.07 mmol) in 1,4-dioxane (25 mL) was refluxed for 12h. The mixture was concentrated under vacuum, and water was added. The mixture was extracted by ethyl acetate and the combined organic layer was concentrated. The crude product was purified by column chromatography by using 10%MeOH/CH2C12 and then recrystallized with MeOH/Chloroform (1 lmg, 7%). 1H NMR (500 MHz, DMSO-J6) 511.95 (br s, IH), 10.0 (s, IH), 8.29 (s, IH), 7.40 (d, J = 7.5 Hz, IH), 7.29-7.24 (m, 2H), 3.90-3.70 (m, 4H), 2.90-2.69 (m, 4H), 2.30 (s, 3H), 2.23 (s, 3H), ESI-MS: calcd for (Ci9H2iClN8OS) 444, found 445 (MH+). HPLC: retention time: 9.24 min.; purity: 93%. Example 14
Figure imgf000090_0001
[0169] A mixture of 4 (125 mg, 0.32 mmol), diisopropylethylamine (0.19 mL, 1.1 mmol), and 3-(piperazin-l-yl)propanenitrile (220 mg, 1.59mmol) in 1,4-dioxane (15 mL) was refluxed for 12h. The mixture was concentrated under vacuum, and water was added. The mixture was extracted by ethyl acetate and the combined organic layer was concentrated. The crude product was recrystallized by MeOH/Chloroform (15mg, 12%). 1H NMR (500 MHz, DMSOd6) 512.00 (br s, IH), 9.99 (s, IH), 8.28 (s, IH), 7.41 (d, J = 7.4Hz, IH), 7.29-7.24 (m, 2H), 3.92-3.79 (m, 4H), 2.71 (t, J= 7Hz, 2H), 2.61 (t, J = 6.5 Hz, 2H), 2.55-2.45 (m, 4H), 2.31 (s, 3H), 2.24 (s, 3H), ESI-MS: calcd for (C22H24ClN9OS) 497, found 498 (MH+). HPLC: retention time: 12.16 min; purity: 88%.
Example 15
Figure imgf000090_0002
[0170] A mixture of 4 (100 mg, 0.25 mmol), diisopropylethylamine (0.07 mL, 1.5 mmol), and l-(pyridin-2-yl)piperazine (83 mg, 0.508 mmol) in 1,4-dioxane (15 mL) was refluxed for 12h. The mixture was concentrated under vacuum, and water was added. The mixture was extracted by ethyl acetate and the combined organic layer was concentrated. The crude product was recrystallized by MeOH/CH2Cl2 (8mg, 6%). 1H NMR (500 MHz, DMSO-^6) 5 12.01 (br s, IH), 10.0 (r s, IH), 8.29 (s, IH), 8.12 (d, J= 3.5 Hz, IH), 7.52-7.60 (m, IH), 7.41 (d, J = 7.4Hz, IH), 7.30-7.24 (m, 2H), 6.91 (d, J= 9 Hz, IH), 6.68-6.64 (m, IH), 4.02-3.92 (m, 4H), 3.68-3.58 (m, 4H), 2.34 (s, 3H), 2.25 (s, 3H), ESI-MS: calcd for (C24H24ClN9OS) 521, found 522 (MH+). HPLC: retention time: 15 min; purity: 89%.
Example 16
Figure imgf000091_0001
[0171] A mixture of 4 (100 mg, 0.25 mmol), diisopropylethylamine (0.07 mL, 1.5 mmol), and 2-(piperazin-l-yl)pyrimidine (83 mg, 0.508 mmol) in 1,4-dioxane (10 mL) was refluxed for 12h. The mixture was concentrated under vacuum, and water was added. The mixture was extracted by ethyl acetate and the combined organic layer was concentrated. The crude product was recrystallized by MeOH/CHCl3 (2 mg, 1.5%). 1H NMR (500 MHz, DMSOd6) 6 12.01 (br s, IH), 9.98(s, IH), 8.38 (d, J= 4.5 Hz, 2H), 8.28 (s, IH), 7.40 (d, J = 7.4Hz, IH), 7.30-7.24 (m, 2H), 6.67 (t J= 3 Hz, IH), 4.12-3.85 (m, 8H), 2.34 (s, 3H), 2.25 (s, 3H), ESI-MS: calcd for (C23H23ClNi0OS) 522, found 523 (MH+). HPLC: retention time: 25 min; purity 97%.
Example 17
Figure imgf000091_0002
[0172] A mixture of 4 (100 mg, 0.25 mmol), diisopropylethylamine (0.07 mL, 1.5 mmol), and 1-phenylpiperazine (82 mg, 0.508 mmol) in 1,4-dioxane (10 mL) was refluxed for 12h. The mixture was concentrated under vacuum, and water was added. The mixture was extracted by ethyl acetate and the combined organic layer was concentrated. The crude product was recrystallized by MeOH/CHCl3 (12 mg, 9%). 1H NMR (500 MHz, DMSO-J6) 6 12.01 (br s, IH), 10.0 (s, IH), 8.30 (s, IH), 7.41 (d, J = 7.4Hz, IH), 7.35-7.24 (m, 4H), 6.99 (d, J= 8 Hz, 2H), 6.81 (t, J = 7Hz, IH), 4.12-3.90 (m, 4H), 3.30-3.10 (m, 4H), 2.34 (s, 3H), 2.24 (s, 3H), ESI-MS: calcd for (C25H25ClN8OS) 520, found 521 (MH+). HPLC: retention time: 34 min; purity 89%.
Example 18
Figure imgf000092_0001
[0173] A mixture of 4 (100 mg, 0.25 mmol), diisopropylethylamine (0.07 mL, 1.5 mmol), and l-(3-(trifluoromethyl)phenyl)piperazine (117 mg, 0.508 mmol) in 1,4-dioxane (10 mL) was refluxed for 12h. The mixture was concentrated under vacuum, and water was added. The mixture was extracted by ethyl acetate and the combined organic layer was concentrated. The crude product was recrystallized by MeOH/CHCl3 (1 lmg, 7%). 1H NMR (500 MHz, DMSO-^6) 512.01 (br s, IH), 10.0 (s, IH), 8.30 (s, IH), 7.48-7.40 (d, J = 7.4 Hz, 2H), 7.34- 7.24 (m, 4H), 7.09 (d, J= 8 Hz, IH), 4.12-3.90 (m, 4H), 3.45-3.30 (m, 4H), 2.34 (s, 3H), 2.24 (s, 3H), ESI-MS: calcd for (C26H24ClF3N8OS) 588, found 589 (MH+). HPLC: retention time: 39 min; purity 93%.
Example 19
Figure imgf000092_0002
[0174] A mixture of 4 (300 mg, 0.25 mmol), diisopropylethylamine (0.66 mL, 3.8 mmol), and piperazin-2-one (761 mg, 7.61 mmol) in DMSO (10 mL) was heated at 65° C for 13h. The mixture was extracted by ethyl acetate and the combined organic layers were washed with water and brine. The crude product was recrystallized by MeOH/CHCl3 (30 mg, 8.6%). 1H NMR (500 MHz, DMSO-^6) 512.01 (br s, IH), 10.0 (br s, IH), 8.30 (s, IH), 8.15 (br s, IH), 7.41(d, J = 7.4Hz, IH), 7.30-7.24 (m, 2H), 4.45-4.32 (m,lH), 4.10-3.92 (m, IH), 2.62- 2.49 (m, 4H), 3.45-3.30 (m, 4H), 2.34 (s, 3H), 2.24 (s, 3H), ESI-MS: calcd for (Ci9H19ClN8O2S) 458, found 481 (M+Na+). HPLC: retention time: 12.7 min; purity 90%.
Example 20
Figure imgf000092_0003
[0175] A mixture of 4 (300 mg, 0.76 mmol) and 3-(lH-imidazol-l-yl)propan-l-amine (821 mg, 4.6 mmol) in 2-propanol (10 mL) was heated at 85° C for 5h. The mixture was extracted by ethyl acetate and the combined organic layers were washed with sodium bicarbonate, water and brine. The crude product was recrystallized by MeOH/CHCl3 (30 mg, 8.1%). 1U NMR (500 MHz, OMSO-d6) 512.01 (br s, IH), 10.05 (br s, IH), 8.30 (s, IH), 7.62 (s, IH), 7.41 (d, J = 7.4Hz, IH), 7.30-7.24 (m, 2H), 7.17(s,lH), 6.83 (s, IH), 4.06 (t, J = 7 Hz, 2H), 2.32-2.24 (m, 8H), 2.05-1.95 (m, 2H) ESI-MS: calcd for C21H22ClN9OS) 483 found 484 (M+H+). HPLC: retention time: 7.9 min; purity 90.5%.
Example 21
Figure imgf000093_0001
[0176] A solution of ethylmagnesium bromide in ether (3 M, 15 ml, 45 mmole) was added dropwise to a stirred solution of cyanuric chloride (5.64 g, 30.58 mmole) in anhydrous dichloromethane at -10° C. After the addition was complete, the reaction mixture was stirred at -5° C for 1 h, after which time water was added dropwise at a rate such that the temperature of the reaction stayed below 10° C. After warming to room temperature, the reaction mixture was diluted with additional water and methylene chloride and passed through a pad of cilite, washed by saturated ammonium chloride, dried and concentrated to give 2,4-dichloro-6- ethyl-l,3,5-triazine 21 as yellow liquid, which solidified after storied in the refrigerator (5.20 g, 96%). 1H NMR (CDCl3) δ 2.95 (q, J = 7.5 Hz. 2H), 1.38 (t, J = 7.5 Hz. 3H).
Example 22
Figure imgf000093_0002
[0177] A solution of Compound 2 (1.07 g, 4.11 mmole), diisopropylamine (10.78 ml, 4.47 mmole) and Compound 21 (1.10 g, 6.18 mmole) in THF (70 mL) was stirred at 0 0C for 8 hours, then cold 5% NaHCO3 was added to the reaction mixture, and the aqueous mixture was extracted twice with EtOAc. The combined extracts were washed with brine, dried, and concentrated in vacuo until a lot of precipitate formed. After filtration, the solids were washed by ethyl acetate dried to give 22 (800 mg, 48%), which was used without purification for the next step reactions.
Example 23
Figure imgf000094_0001
[0178] A mixture of 22 (258 mg, 0.63 mmol), diisopropylethylamine (0.32 mL, 1...83 mmol), and l-(2-hydroxyethyl)piperazine (280 mg, 2...15 mmol) in 1,4-dioxane (50 mL) was stirred at 70° C overnight. The mixture was concentrated under vacuum, and water was added. The mixture was extracted by ethyl acetate and the combined organic was concentrated, passed a pad of silica gel and concentrated to give light yellow solids, which was crystallized from methanol-THF to give white solids 23 (125 mg, 39%). 1H NMR (500 MHz, DMSOd6) 511.97 (br s, IH), 10.00 (s, IH), 8.28 (s, IH), 7.40 (d, J= 7.6 Hz, IH), 7.29- 7.24 (m, 2H), 4.46 (t, J = 5.0 Hz, IH), 3.87-3.81 (m, 4H), 3.52 (q, J= 6.0 Hz, 2H), 2.58-2.41 (m, 8H), 2.23 (s, 3H), 1.20 (t, J = 7.0 Hz, 3H). ESI-MS: calcd for (C22H27C1N8O2S) 502, found 503 (MH+). HPLC: retention time: 12.35 min.; purity: 99%.
Example 24
Figure imgf000094_0002
[0179] Compound 24 was prepared by the same procedure as was used in the preparation of Compound 23. White solids were obtained (29% yield). 1H NMR (500 MHz, DMSO-J6) 8 11.97 (br s, IH), 10.00 (s, IH), 8.31 (s, IH), 8.23 (d, J = 5.0 Hz, 2H), 7.40 (d, J= 8.0 Hz, IH), 7.30-7.25 (m, 2H), 7.00 (d, J= 5.0 Hz, 2H), 4.00 (m, 4H), 3.70-3.65 (m, 4H), 2.61 (br, 2H), 2.24 (s, 3H), 1.25 (br, 3H). ESI-MS: calcd for (C25H26ClN9OS) 535, found 536 (MH+). HPLC: retention time: 16.18 min.; purity: 99%. Example 25
Figure imgf000095_0001
[0180] Compound 25 was prepared by the same procedure as was used in the preparation of Compound 23. White solids were obtained (50% yield). 1H NMR (500 MHz, DMSO-^6) 6 11.97 (br s, IH), 10.00 (s, IH), 8.28 (s, IH), 7.40 (d, J= 7.5 Hz, IH), 7.30-7.25 (m, 2H), 3.84 (m, 4H), 3.70-3.65 (m, 4H), 2.61 (br, 2H), 2.23 (s, 3H), 1.25 (br, 3H). ESI-MS: calcd for (C20H22ClN7O2S) 459, found 460 (MH+). HPLC: retention time: 23.91 min.; purity: 99%.
Example 26
Figure imgf000095_0002
[0181] Compound 26 was prepared by the same procedure as was used in the preparation of Compound 23. White solids were obtained (22% yield). 1H NMR (500 MHz, DMSO-^6) 5 11.97 (br s, IH), 10.00 (s, IH), 8.28 (s, IH), 7.60 (br, IH), 7.40 (d, J= 7.6 Hz, IH), 7.29-7.24 (m, 2H), 3.42 (m, 2H), 2.52 (m, 4H), 2.45-2.17 (m, 9H), 1.20 (m, 3H). ESI-MS: calcd for (C20H25ClN8OS) 460, found 461 (MH+). HPLC: retention time: 10.96 min.; purity: 95%.
Example 27
Figure imgf000095_0003
[0182] A mixture of compound 22 (250 mg, 0.61 mmol) and diisopropylethylamine (0.43 mL, 2.45 mmol), and 2-aminoethanol (373 mg, 6.13 mmol) in DMSO was heated at 70° C for over night. The mixture was extracted by ethyl acetate and the combined organic layers were washed with water and brine. The crude product was recrystallized by MeOH/CHCl3 to give compound 27 (23 mg, 8.6%). 1H NMR (500 MHz, DMSO-^6) 511.83 (br s, IH), 10.02 (br s, IH), 8.31 (s, IH), 7.80 (brs,lH), 7.41 (d, J= 7.4Hz, IH), 7.30-7.24 (m, 2H), 4.80-4.72 (brs, IH), 3.62-3.38 (m, 4H), 2.58-2.50 (m, 2H), 2.24 (s, 3H), 1.20 (m, 3H); ESI-MS: calcd for C18H20ClN7O2S) 433 found 434 (M+H+). HPLC: retention time: 12.2 min; purity 90.6%. Example 28
Figure imgf000096_0001
[0183] A mixture of compound 22 (250 mg, 0.61 mmol) and diisopropylethylamine (0.43 mL, 2.45 mmol), and 3-morpholinopropan-l-amine (882 mg, 6.13 mmol) was heated at 70° C in DMSO for over night. The mixture was extracted by ethyl acetate and the combined organic layers were washed with water and brine. The crude product was recrystallized by MeOH/CHCla to give compound 28 (33 mg, 10%). 1H NMR (500 MHz, DMSO-^6) δl 1.45 (br s, IH), 9.96 (br s, IH), 8.28 (s, IH), 8.10 (brs, IH), 7.41 (d, J= 7.4Hz, IH), 7.30-7.24 (m, 2H), 3.62-3.52 (m, 4H), 2.63-2.50 (m, 6H), 2.42-2.25 (m, 5H), 2.24 (s, 3H), 1.68-1.75 (m, IH), 1.22 (m, 3H); ESI-MS: calcd for C23H29ClN8O2S) 516 found 517 (M+H+). HPLC: retention time: 12.7 min; purity 86%.
Example 29
Figure imgf000096_0002
[0184] A solution of phenylmagnesium bromide in ether (3M, 16 ml, 48 mmole) was added dropwise to a stirred solution of cyanuric chloride (5.93 g, 32.16 mmole) in anhydrous dichloromethane at 5° C. After the addition was complete, the reaction mixture was stirred at 10-20° C for 3 h. The mixture was cooled to 0° C and added water dropwise at a rate such that the temperature of the reaction stayed below 10° C. After warming to room temperature, the reaction mixture was diluted with additional water and methylene chloride and passed through a pad of cilite, washed by saturated ammonium chloride, dried and concentrated to give 2,4-dichloro-6-phenyl-l,3,5-triazine 29 as yellow liquid, which solidified after storage in the refrigerator (1.8 g, 25%). 1H NMR (500 MHz, CDCl3) δ 8.50 (d, J = 8.0 Hz, 2H), 7.70 (t, J = 8.0 Hz, IH), 7.55 (t, J = 8.0 Hz. 2H).
Example 30
Figure imgf000097_0001
[0185] A solution of Compound 2 (500 mg, 1.87 mmole), diisopropylamine (0.33 ml, 1.87 mmole) and Compound 9 (630 mg, 2.81 mmole) in THF (30 mL) was stirred at 0° C for 3 hours, then room temperature for additional 3 hours. Water was added to the reaction mixture, and the aqueous mixture was extracted twice with EtOAc. The combined extracts were washed with brine, dried, and concentrated in vacuo until a lot of precipitate formed. After filtration, the solids were washed by ethyl acetate dried to give compound 30 (250 mg, 29%), which was used without purification for the next step reactions.
Example 31
Figure imgf000097_0002
[0186] A mixture of 30 (220 mg, 0.48 mmol), diisopropylethylamine (0.30 mL, 1.72 mmol), and 1 -(2-hydroxyethyl)piperazine (260 mg, 2.00 mmol) in DMSO (10 mL) was stirred at 60° C overnight. Water was added, followed by ethyl acetate. White solids precipitated from the solution, which was filtered and washed by ethyl acetate to give the desired product 31 (180 mg, 33%). 1H NMR (500 MHz, DMSO-t/6) 5 11.97 (br s, IH), 10.03 (s, IH), 8.45 (br, 2H), 8.32 (s, IH), 7.61 (t, J= 7.0 Hz, IH), 7.55 (t, J= 7.5 Hz, 2H), 7.41(d, J= 8.0 Hz, IH), 7.31-7.24 (m, 2H), 4.48 (t, J= 5.0 Hz, IH), 3.99 (m, 4H), 3.55 (q, J= 6.0 Hz, 2H), 2.54 (br, 4H), 2.45 (t, J = 6.0 Hz, 2 H), 2.25 (s, 3H). ESI-MS: calcd for (C26H27ClN8O2S) 550, found 551 (MH+). HPLC: retention time: 20.02 min.; purity: 98%.
Example 32
Figure imgf000097_0003
[0187] A solution of iso-butylmagnesium bromide in ether (2M, 35 ml, 70.0 mmole) was added dropwise to a stirred solution of cyanuric chloride (5.28 g, 28.63 mmole) in anhydrous dichloromethane at -5° C. After the reaction was completed as indicated by TLC, water was added water dropwise at a rate such that the temperature of the reaction stayed below 10° C. After warming to room temperature, the reaction mixture was diluted with additional water and methylene chloride and passed through a pad of cilite, washed by saturated ammonium chloride, dried and concentrated to give 2,4-dichloro-6-iso-butyl-l,3,5-triazine as yellow slurry liquid residue. The crude product was passed through a pad of silica gel eluted with 10% ethyl acetate in hexanes to give the light yellow liquid of the desired product 32 (3.0 g, 51%). 1H NMR (500 MHz, CDCl3) δ 2.75 (d, J - 7.0 Hz, 2H), 2.29 (m, IH), 0.97 (d, J = 7.0 Hz. 6H).
Example 33
Figure imgf000098_0001
[0188] A solution of Compound 2 (1.17 g, 4.38 mmole), diisopropylamine (2.3 ml, 13.20 mmole) and Compound 32 (1.00 g, 4.85 mmole) in THF (30 mL) was stirred at 0° C for 6 hours. 5% NaHCO3 was added and the reaction mixture was extracted by ethyl acetate (3X). The organic layer was washed by saturated ammonium chloride, brine, dried (Na2SO4), and concentrated in vacuo. The residue was purified by column chromatography on silica gel (0- 2% methanol in DCM) to give the desired product 33 as light-yellow solids (170 mg, 9%). 1H NMR (500 MHz, DMSO-^6) δ 12.98 (br s, IH), 10.11 (s, IH), 8.35 (s, IH), 7.40 (d, J= 7.5 Hz, IH), 7.28 (m, 2H), 2.67 (br, 2H), 2.29 (m, IH), 2.23 (s, 3H), 0.96 (s, 6H). ESI-MS: calcd for (C18H18Cl2N6OS) 436, found 437 (MH+).
Example 34
Figure imgf000098_0002
[0189] A mixture of 33 (120 mg, 0.27 mmol), diisopropylethylamine (0.17 mL, 1.00 mmol), and l-(2-hydroxyethyl)piperazine (130 mg, 1.00 mmol) in 1,4-dioxane (12 mL) was stirred at 60° C overnight. Water was added, followed by ethyl acetate/hexane (5/5). White solids precipitated from the solution, which was recrystalized from methanol/chloroform to give the desired product 34 as white solids (67 mg, 47%). 1H NMR (500 MHz, DMSO-J6) S 11.97 (br s, IH), 9.97 (s, IH), 8.45 (br, 2H), 8.28 (s, IH), 7.40 (d, J= 7.6 Hz, IH), 7.28 (m, 2H), 4.45 (t, J= 5.0 Hz, IH), 3.82 (m, 4H), 3.52 (q, J= 6.0 Hz, 2H), 2.50 (br, 4H), 2.43 (t, J = 6.0 Hz, 2 H), 2.24 (s, 3H), 2.23 (obscured, IH), 0.93 (s, 6H). ESI-MS: calcd for (C24H31C1N8O2S) 530, found 531 (MH+). HPLC: retention time: 17.16 min.; purity: 96%.
Example 35
Figure imgf000099_0001
[0190] To a mixture of compound 1 (180 g, 0.75 mmol) in 1,4-dioxane (3 mL) and water (3 mL) was added NBS (160 mg, 0.90mmol) at 0° C. The slurry was warmed and stirred at 20-22° C for 3 h. Urea (58 mg, 0.97 mmol) was added and the mixture heated to 100° C. After 2 h, the resulting solution was cooled to 20-22° C and cone, ammonium hydroxide (0.2 mL) was added dropwise. The resulting slurry was concentrated under vacuum. The remaining water was removed by co-evaporating with toluene. The residue was purified by column chromatography on silica gel (0-6% 2N ammonia in methanol/100-94% dichloromethane) to give compound 35 as white solids. (120 mg, 63% yield). 1H NMR (500 MHz, DMSO-d6) δ 9.57 (s, IH), 7.60 (s, IH), 7.36 (d, J = 7.5 Hz, IH,), 7.31 (s, 2H), 7.09- 7.29 (m, 2H), 2.19 (s, 3H); ESI-MS: calcd for (Ci !H10ClN3O2) 251, found 252 (MH+), 250 ([M-H]").
Example 36
Figure imgf000099_0002
[0191] To a solution of compound 3 (0.5 g, 3.05 mmol) in DMF (5 mL) was added to a mixture of Boc-piperazine (0.57 g, 3.05 mmol), NaHCO3 (0.51 g, 6.09 mmol) at room temperature. After the competition of the addition, the mixture was stirred at room temperature for 30 minutes. The reaction mixture was extracted with ethyl acetate and the organic layer was further washed with water (2x20 ml), brine (2x20ml). The organic layer was dried (Na2SO4) and concentrated, during which white solid of 36 was formed (450 mg, 47 %). This solid was used without further purification in the next step. 1H NMR (500 MHz, DMSOd6) δ 3.80-3.79 (m, 2H), 3.72-3.70 (m, 2H), 3.42 (br, 4H), 2.34 (s, 3H), 1.42 (s, 9H), ESI-MS: calcd for (C I3H20ClN5O2) 313 found 258 (M-56+H+).
Example 37
Figure imgf000100_0001
[0192] A round bottom flask was flam-dried and flushed with argon, then charged with xantphos (25 mg, 0.05 mmol) and dry 1,4-dioxane (5 mL). After degassing, Pd(OAc)2 ( 5 mg, 0.02 mmol) was added, and the mixture was stirred under an inert atmosphere for 10 min. In another round-bottom flask, compound 36 (70 mg, 0.22 mmol), compound 35 (50 mg, 0.20 mmol)), and K2CO3 (525 mg, 3.8 mmol) were poured into dry 1,5-dioxane (7 mL). Then, the Pd(OAc)2/xantphos solution was added with a syringe. The resulting mixture was subsequently heated to reflux under an inert atmosphere with vigorous stirring until the starting heteroaryl halide has disappeared (overnight). After cooling, the solid material was filtered off and washed with dichloromethane and methanol. The solvent was evaporated, and the resulting crude product was purified by flash column chromatography on cilica gel using EtOAc/DCM/MeOH : 80/20/2 v/v/v as eluent to provide compound 37 as white solids (33 mg, 31%). ESI-MS: calcd for (C24H29ClN8O4) 528, found 529 (MH+), 527 ([M-H]").
Example 38
Figure imgf000100_0002
[0193] Compound 37 (30 mg, 0.06 mmol) was dissolved in dichloromethane (5 mL) and trifluoroacetic acid (1 mL) at 0° C and the mixture was stirred at 0° C for 3 hours. TLC was checked and the starting material was consumed. After concentration, the residue was neutralized by saturated sodium bicarbonate in water and the mixture was extracted by dichloromethane, dried over sodium sulfate and concentrated. The residue was purified by column chromatography on a silica gel (EtOAc/DCM/6% 2M NH3 :: 50/50/6) to give compound 38 as white solids (18 mg, 70%). 1U NMR (500 MHz, DMSOd6) δ 9.93 (s, IH), 7.86 (s, IH), 7.39 (d, J - 7.5 Hz, IH), 7.30-7.25 (m, 2H), 3.71 (br, 4H), 2.68 (br, 4H), 2.26 (s, 3H), 2.21 (s, 3H); ESI-MS: calcd for (C19H21ClN8O2) 428, found 429 (MH+), 427 ([M-H]"). HPLC: retention time: 6.09 min. purity: 96%.
Example 39
Figure imgf000101_0001
[0194] To a solution of compound 21 (1.2 g, 6.74 mmol) in THF (35 mL) was added to a mixture of 1 -hydroxy ethyl piperazine (600 mg, 4.60 mmol), DIPEA (0.80 mL, 4.59 mmol) and THF (35 mL) dropwise at -10° C. After the competition of the addition, the mixture was stirred at -10° C for 30 minutes. Ammonium chloride solution was added and the mixture was extracted by ethyl acetate. The organic layer was dried (Na2SO4) and concentrated, during which yellow precipitate formed. The solids were collected by filtration, washed by ethyl acetate to give compound 39 as yellow solids (350 mg, 28%). 1H NMR (500 MHz, DMSO-d6) δ 5.36 (br, IH), 4.73-4.53 (m, 2H), 3.77 (br, 2H), 3.55 (br, 4H), 3.15 (br, 4H), 2.63 (q, J = 7.5 Hz, 2H), 1.18 (t, J = 7.5 Hz, 3H); ESI-MS: calcd for (C11Hi8ClN5O) 271, found 272 (MH+).
Example 40
Figure imgf000101_0002
[0195] A round bottom flask was flame-dried and flushed with argon, then charged with xantphos (25 mg, 0.05 mmol) and dry 1,4-dioxane (5 mL). After degassing, Pd(OAc)2 ( 5 mg, 0.02 mmol) was added, and the mixture was stirred under an inert atmosphere for 10 min. In another round-bottom flask, compound 39 (58 mg, 0.22 mmol), compound 35 (47 mg, 0.18 mmol)), and K2CO3 (525 mg, 3.8 mmol) were poured into dry 1,5-dioxane (15 mL). Then, the Pd(OAc)2/xantphos solution was added with a syringe. The resulting mixture was subsequently heated to reflux under an inert atmosphere with vigorous stirring until the starting heteroaryl halide has disappeared (overnight). After cooling, the solid material was filtered off and washed with dichloromethane and methanol. The solvent was evaporated, and the resulting crude product was purified by flash column chromatography on silica gel using EtOAc/DCM/MeOH (2N NH3): 50/50/5 v/v/v as eluent to provide compound 40 as white solids (45 mg, 51%). 1H NMR (500 MHz, DMSO-d6) δ 10.25 (br, IH), 9.94 (s, IH), 7.87 (s, IH), 7.39 (d, J = 7.5 Hz, IH), 7.30-7.25 (m, 2H), 4.44 (t, J = 5.5 Hz, IH), 3.77 (br, 4H), 3.50 (q, J = 6.0 Hz, 2H), 2.51 (m, 2H), 2.42-2.40 (m, 6H), 2.21 (s, 3H), 1.18 (t, J = 8.0 Hz, 3H); ESI-MS: calcd for (C22H27ClN8O3) 486, found 487 (MH+), 485 ([M-H]"). HPLC: retention time: 8.16 min. purity: 97%.
Example 41
Figure imgf000102_0001
[0196] To a stirred solution of ethyl 2-aminooxazole-5-carboxylate (0.10 g, 0.64 mmol) in THF (6 mL) was added DIPEA (0.12 mL, 0.70 mmol) at 0° C. After stirring for 10 min. at the same temperature, Compound 21 (0.23 mg, 1.28 mmol) was added. The mixture was stirred at 70° C for 8 h, cooled to room temperature, and diluted with EtOAc and washed with 5% NaHCO3. The organic phase was concentrated and the residue was purified by chromatography on a silica gel column eluted with 1% MeOH in CH2Cl2 to afford Compound 41 (35 mg, 20%) as a yellow solid. 1H NMR (500MHz, DMSO-J6) δ 12.43 (s, IH, NH), 7.95 (s, IH, Ar-H), 4.31 (dd, 2Η, J= 14.3 Hz, CH2), 2.77 (dd, 2Η, J= 14.2 Hz, CH2), 1.30 (t, 3Η, J= 6.7 Hz, CH3), 1.23 (t, 3H, J= 7.5 Hz, CH3). MS (ESI) m/z 296 [M-H]".
Example 42
Figure imgf000102_0002
[0197] To a stirred solution of Compound 41 (0.10 g, 0.34 mmol) in Dioxane (10 mL) were added DIPEA (0.18 mL, 1.02 mmol) and 4-(pyridyl) piperazine (0.06 g, 0.37 mmol) at room temperature. The mixture was heated to 55° C and stirred for 1 hour and cooled to room temperature. The reaction mixture was concentrated. The residue was washed with water, filtered, and dried in vacuo to obtain compound 42 (85 mg, 60%) as a yellow solid. 1H NMR (500MHz, OMSO-d6) δ 11.51 (s, IH, NH), 8.18 (d, 2H, J= 5.8 Hz, Ar-H), 7.87 (s, 1Η, Ar- H), 6.87 (d, 2Η, J= 6.5 Hz, Ar-H), 4.31 (dd, 1Η, J= 14.2 Hz, CH2), 3.92 (bs, 4Η), 3.44 (m, 4H) 2.56 (dd, 2H, J= 15.1 Hz, CH2), 1.29 (t, 3Η, J= 7.0 Hz, CH5). 1.22 (t, 3Η, J= 7.5 Hz, CH3). MS (ESI) m/z 425 [M+Η]+.
Example 43
Figure imgf000103_0001
[0198] A solution of compound 42 (80 mg, 0.18 mmol) in THF-EtOH (1.3 mL, 2:3) and 6 M KOH solution (1.3 mL) was heated to 55° C overnight under inert atmosphere. The solution was cooled to 0° C and acidified with coned HCl to pH 1. The solution was concentrated in vacuo, and the residue was washed with water, ether, and dried in vacuo to obtain Compound 43 as a yellow solid (40 mg, 55%). 1H NMR (500 MHz, DMSO-J6) δ 13.59 (s, IH, NH), 8.29 (d, 2Η, J= 6.6 Hz, Ar-H), 7.81 (s, 1Η, Ar-H), 7.21 (d, 2Η, J= 7.3 Hz, Ar-H), 3.98 (bs, 4Η, Ar-CH2), 3.84-3.82 (m, 4H, Ax-CH2), 2.59 (dd, 2H, J= 15.1 Hz, CH2), 1.22 (t, 3Η, J= 7.5 Hz, CH5). MS (ESI) m/z 397 [M+Η]+.
Example 44
Figure imgf000103_0002
[0199] A mixture of 2-amino oxazole-4-carboxylic acid ethyl ester (0.5 g, 3.2 mmol), Boc anhydride (1.1 g, 4.8 mmol), DIPEA (0.61 mL, 3.5 mmol), DMAP (0.1 g, 0.8 mmol) and DMF (4 mL) was stirred at 40° C overnight. The DMF was removed in vacuo and the residue taken up in ethyl acetate (40 mL). The reaction was washed with brine (2 x 25 mL), saturated sodium bicarbonate (25 mL), 0.0 IM HCl (25 mL), and dried over sodium sulfate. The organic phase was concentrated and the residue was purified by chromatography on a silica gel column eluted with Hexane/EtOAc (2:1) to afford Compound 44 (490 mg, 60%) as a light yellow solid. 1H NMR (400 MHz, DMSO-Jd) δ 10.90 (s, IH, NH), 8.51 (s, 1Η, Ar-H), 4.25 (dd, J= 7.2 Hz, 2H, CH2), 1.44 (s, 9H, C(CHs)3), 1-27 (t, J= 7.2 Hz, 3H, CH5). MS (ESI) m/z 255 [M-H]"
Example 45
Figure imgf000104_0001
[0200] A mixture of Compound 44 (0.25 g, 0.97 mmol), IN NaOH (2 mL) and methanol (1 mL) was stirred at 35° C. The reaction was determined to be complete by TLC after 3 h. The methanol was removed in vacuo and the aqueous layer carefully adjusted to pH 2 with IN HCl at which time a crystalline white solid precipitated out of solution. Filtration of the white solid gave compound 45 (0.2 g, 95%). 1H NMR (400 MHz, OUSO-d6) δ 10.82 (s, IH), 8.41 (s, IH), 1.44 (s, 9H).
Example 46
Figure imgf000104_0002
[0201] A 2 M solution of oxalyl chloride (0.44 mL, 14.5 mmol) was added dropwise to a stirred suspension of Compound 45 (0.1 g, 0.44 mmol) and DMF (2 drops) in CH2Cl2 (4 mL) at 0° C. The solution was warmed to room temperature, stirred at for 4 h and concentrated. The residue was coevaporated with toluene and dried in vacuo to obtain the crude acid chloride. 2-chloro-6-methylaniline (0.15 mL, 1.2 mmol) was added dropwise to a stirred solution of crude acid chloride in CH2Cl2 (2 mL) at 0° C. After 15 min. at the same temperature, pyridine (0.07 mL, 0.9 mmol) was added slowly. The solution was warmed to room temperature and stirred for overnight. Reaction mixture was diluted with EtOAc and washed with H2O and brine. The EtOAc extract was separated, dried (NaSO4), filtered, and concentrated. The residue was chromatographed on a silica gel column eluted with Hexane/EtOAc (1 :1) afforded Compound 46 (30 mg, 19%) as a white solid. 1H NMR (400 MHz, COCh-d6) δ 8.37 (s, IH), 8.01 (s, IH), 7.45 (s, IH), 7.31-7.29 (m, IH), 7.19-7.13 (m, 2H), 2.32 (s, 3H), 1.55 (s, 9H). MS (ESI) m/z 374 [M+Na]+.
Example 47
Figure imgf000105_0001
[0202] A solution of Compound 46 (30 mg, 0.85 mmol) in trifluoroacetic acid (0.2 mL) and CH2Cl2 (1.2 mL) mL) was stirred at 0° C for 5 h and concentrated. The residue was chromatographed on a silica gel column eluted with CH2Cl2/Me0H (40:1) to give ompound 47 as white solid. 1H NMR (400 MHz, CDCl3-Jo) δ 9.32 (s, IH), 7.98 (s, IH), 7.45 (s, IH), 7.37-7.36 (m, IH), 7.35-7.22 (m, IH), 7.08 (s, 2H), 2.19 (s, 3H). MS (ESI) m/z 252 [M+H]+.
Example 48
Figure imgf000105_0002
[0203] A mixture of 5-amino-l,3,4-oxadiazole-2-carboxylic acid ethyl ester (2.0 g, 12.7 mmol), Boc anhydride (4.17 g, 19.1 mmol), DIPEA (1.8 mL, 14.0 mmol), DMAP (413 mg, 3.2 mmol) and DMF (15 mL) was stirred at 40° C overnight. The DMF was removed in vacuo and the residue taken up in ethyl acetate (40 mL). The reaction was washed with brine (2 x 100 mL), saturated sodium bicarbonate (100 mL), 0.01M HCl (100 mL), and dried over sodium sulfate. The organic phase was concentrated and the residue was purified by chromatography on a silica gel column eluted with Hexane/EtOAc (3:1) to afford Compound 48 (2.2 g, 67%) as a white solid. 1U NMR (400 MHz, CDCl3-Jo) δ 8.91 (bs, IH), 4.51 (dd, J = 14.3 Hz, 2H), 1.56 (s, 9H), 1.44 (t, J= 14.3 Hz, 3H). MS (ESI) m/z 280 [M+Na]+.
Example 49
Figure imgf000105_0003
[0204] A mixture of Compound 48 (1.0 g, 3.9 mmol), IN NaOH (10 mL) and methanol (3 mL) was stirred at 35 0C. The reaction was determined to be complete by TLC after 3 h. The methanol was removed in vacuo and the aqueous layer carefully adjusted to pH 2 with IN HCl. Solvent was removed and residue was dried in vacuo to give 49 as a white solid. 1H NMR (400 MHz, DMSO-J<5) δ 10.95 (bs, IH,), 1.44 (s, 9H). MS (ESI) m/z 230 [M+H]+. Example 50
Figure imgf000106_0001
[0205] A 2 M solution of oxalyl chloride (4.4 niL, 8.74 mmol) was added dropwise to a stirred suspension of Compound 49 (1.0 g, 4.37 mmol) and DMF (30 mL) in CH2Cl2 (25 mL) at 0° C. The solution was warmed to room temperature, stirred at for 4 h and concentrated. The residue was coevaporated with toluene and dried in vacuo to obtain the crude acid chloride. 2-chloro-6-methylaniline (1.6 mL, 13.11 mmol) was added dropwise to a stirred solution of crude acid chloride in CH2Cl2 (20 mL) at O0C. After 15 min. at the same temperature, pyridine (0.45 mL, 5.24 mmol) was added slowly. The solution was warmed to room temperature and stirred for overnight. Reaction mixture was concentrated and the residue was chromatographed on a silica gel column eluted with CH2Cl2/Me0H (40:1) afforded Compound 50 as a white solid. 1H NMR (400 MHz, DMSO-Λ5) δ 11.61 (s, IH), 10.83 (s, IH), 7.41-7.99 (m, IH), 7.29-7.28 (m, 2H), 1.24 (s, 3H), 1.49 (s, 9H). MS (ESI) m/z 375 [M+Na]+.
Example 51
Figure imgf000106_0002
[0206] A solution of Compound 50 (0.3 g, 0.85 mmol) dissolved in a mixture Of CH2Cl2 and TFA (7 mL 6:l).The mixture was stirred at 0° C to room temperature for 3 h. Reaction mixture was concentrated and the residue was chromatographed on a silica gel column eluted with CH2Cl2MeOH (40:1) afforded Compound 51 as a white solid. 1H NMR (400 MHz, DMSO-J6) δ 10.51 (s, IH), 7.66 (s, 2H), ), 7.40-7.38 (m, IH), 7.28-7.27 (m, 2H), 3.03 (s, 3H). MS (ESI) m/z 253 [M+Na]+
Example 52
Figure imgf000107_0001
[0207] Compound 51 (0.05 g, 0.2 mmol), Compound 39 (0.07 g, 0.24 mmol), Pd(OAc)2 (6 mg, 0.02 mmol), Xantphos (36 mg, 0.04 mmol) and K2CO3 (0.55 g, 4.0 mmol) were added in 2-5 mL a screw capped microwave vial. Dioxane:DMF (2.5 mL, 1.5:1) was added and vial was sealed with a cap. The mixture was allowed to stir at 180° C for 10 min. under microwave (Biotage, Initiator 2.0) condition. Reaction mixture was filtered and the solid was washed with CH2Cl2 and MeOH, concentrated. The residue was chromatographed on a silica gel column eluted with 4% MeOH in CH2Cl2 afforded Compound 52 as a white solid (21 mg, 22%). 1H NMR (500MHz, DMSO-J6) δ 11.95 (s, IH), 10.70 (s, IH), 7.42-7.29 (m, 3H), 4.42 (t, IH, J= 10.7 Hz), 3-82-3.81 (m, 4H), 3.51 (dd, J= 9.3 Hz, 2H) 2.56 (dd, J= 15.1 Hz, 2H), 2.44-2.40 (m, 2H), 2.24 (s, 3H), 1.20 (t, 3H, J= 15.1 Hz). MS (ESI) m/z 488 [M+H]+.
Example 53
Figure imgf000107_0002
[0208] A 1.6 M solution of n-butyllithium in hexanes (2.27 ml, 3.6 mmol) was added dropwise to a solution of 2-chloro-l -methyl- lH-imidazole* (0.4 g, 3.45 mmol) in anhydrous TΗF (10 ml) at - 78° C. The reaction mixture was maintained below -78° C during the addition. After 15 min, a solution of 2-chloro-6-methylphenyl isocyanate (0.64 g, 3.79 mmol) in TΗF (5 ml) was added and the solution was stirred at -78° C for further 2h. The solution was quenched by the addition of aq NH4Cl and partitioned between EtOAc and water. The organic layer was separated and washed with brine, dried and concentrated. The crude product was purified on silica gel chromatography by using 30% EtOAc/Hexane, to give compound 53 as white solid (353 mg, 36% yield). 1H NMR (500 Hz, DMSO-d6) δ 9.96(s, IH), 7.82 (s, IH), 7.40 (dd, J= 1.7, 5.74 Hz, IH), 7.27 (m, 2H), 3.83 (s, 3H,), 2.23 (s, 3H); ESI-MS: calcd for (C12H11C12N3O) 282, found 282 (M-H+). HPLC: retention time: 17.83 min.; purity: 96%. Example 54
Figure imgf000108_0001
[0209] Sodium hydride (95% dispersion, 0.023 g, 0.85 mmol) was added to a solution of Compound 53 (0.2 g, 0.71 mmol) in DMF (1OmL). After 30 min, the mixture was treated with 4-methoxybenzyl chloride (115 uL, 0.85 mmol) and tetrabutylammonium iodide (0.043 g, 0.12 mmol) and then stirred at room temperature for 13 h. The reaction mixture was partitioned between EtOAc and water. The organic layer was separated and washed with brine, dried and concentrated. The crude product was purified on silica gel chromatography by using 15% EtOAc/Hexane to give compound 54 as white solid (254 mg, 89 % yield). 1H NMR (500 Hz, DMSOd6) δ 7.37 (d, J= 7.80 Hz, IH), 7.29 ( t, J= 7.78 Hz, IH), 7.22 (d, J= 7.21 Hz, IH), 7.14 (d, J= 5.25 Hz, 2H), 6.82 (d, J= 6.72 Hz, 2H), 5.03 (d, J= 14 Hz, IH), 4.59 (d, J= 14 Hz, IH), 3.76 (s, 3H,), 3.71 (s, 3H), 1.84 (s, 3H); ESI-MS: calcd for (C20H19C12N3O2) 403, found 404 (M+H+).
Example 55
Figure imgf000108_0002
[0210] A mixture of Compound 21 (1.0 g, 5.65 mmol), 5% aq NaHCO3 (10 ml), and 1-methyl piperazine (0.51 g, 5.15 mmol) in THF:acetone:water (52:13:13) was was stirred at room temperature for 12 h. The reaction mixture was partitioned between EtOAc and water. The organic layer was separated and washed with brine, dried and concentrated. The crude product 55 was used in the next reaction without further purification.
Example 56
Figure imgf000108_0003
[0211] A mixture of Compound 55 (1.0 g, 4.14 mmol) and NaN3 (0.8Ig, 12.45 mmol) in 20 ml of DMF was stirred at room temperature for 13h. The reaction mixture was partitioned between EtOAc and water. The organic layer was separated and washed with brine, water and concentrated on the rotavapor. The crude product was passed on the pad of silica gel by using 5% MeOH/DCM to give compound 56 as white solid (0.6 g, 59 %). ESI-MS: calcd for (C10H16N8) is 248 and found 249 (M+H+).
Example 57
Figure imgf000109_0002
[0212] A mixture of Compound 56 (1.2 g, 4.83 mmol) and 52 mg of 10 % palladium-on- carbon in 30 ml of absolute ethanol was stirred at 25 0C under 1 atm of hydrogen for 2 h. The catalyst was filtered through the celite and washed ethanol. The filterate was concentrated on the rotavapor to give compound 57 as light yellow solid (1.05 g, 98%). 1H NMR (500 Hz, DMSO-d6) δ 6.70 (brs, 2H), 3.65 (m, 4H,), 2.35 (m, 2H), 2.25 (m, 4H), 2.15 (s, 3H), 1.1 (t, J = 7.5 Hz, 3H) ; ESI-MS: calcd for (Ci0H18N6) 222 found 223 (M+H+).
Example 58
Figure imgf000109_0001
[0213] A mixture of Compound 54 (100 mg, 0.25 mmol), Compound 57 (55.1 mg, 0.25 mmol), Pd(OAc)2 (5.5 mg, 0.025 mmol), Xantphos (29 mg, 0.05 mmol) and NaO1Bu (26 mg, 0.27 mmol) and 1, 4-dioxane (3 ml) were added in 2-5 mL of a screw capped microwave vial. The mixture was subjected to microwave heating using a Biotage, Initiator 2.0 at 180 0C for a period of 7 min. Reaction mixture was filtered and the solid was washed with CH2Cl2 and MeOH, concentrated. The residue was chromatographed on a silica gel column eluted with 5% NH3MeOH in CH2Cl2 to give compound 58. ESI-MS: calcd for (C30H36ClN9O2) 589, found 590 (M+H+). This crude was used in the next reaction without further purification. Example 59
Figure imgf000110_0001
[0214] A solution of Compound 58 (55 mg, 0.09 mmol) dissolved in a mixture Of CH2Cl2 and TFA (2 ml, 1:1) was treated with triflic acid ( 0.03 ml, 0.33 mmol) and stirred at room temperature for 3h. The reaction mixture was adjusted to the pH 9 by using saturated NaHCO3 and extracted with dichloromethane. The organic layer was evaporated and purified by column chromatography using 5% NH3MeOH in dichlromethane to afford yellow solids of compound 59. ESI-MS: calcd for (C22H28ClN9O) 469, found 470 (M+H+). HPLC: retention time: 6.53 min.; purity: 93%.
Example 60
Figure imgf000110_0002
[0215] A mixture of ethyl β-ethoxyacrylate (26.50 g, 183 mmol) and 2 N sodium hydroxide (110 mL, 220 mmol) was refluxed for 2 h and cooled to 0° C. water was removed under vacc, and the yellow solids were triturated with toluene and evaporated to give the sodium β-ethoxyacrylate (25 g, 97%). The mixture of sodium β-thoxyacrylate (10.26 g, 74.29 mmol) and thionyl chloride (25 mL, 343 mmol) was refluxed for 2 h, and evaporated to give the β-ethoxyacryloyl chloride crude product, which was used without purification. To a cold stirring solution of 3-ethoxyacryloyl chloride (7.3 g, 54.2 mmol) in THF (70 mL) was added cyclopropyl amine (8.3 mL, 119.2 mmol) and pyridine (8.8 ml, 108 mmol). The mixture was then warmed and stirred overnight at room temperature. Water was added and extracted with EtOAc. The organic layer was evaporated and the resulting residue was purified by silica gel using 3:1 (hexane-EtoAc), concentrated under vacuum to give 2.7 g (34% yields) of compound 60. 1H NMR (400 MHz, CDCl3) δ 7.50 (d, J = 12 Hz, IH), 5.60 (br s, IH), 5.24 (d, J= 11.5 Hz, IH), 3.94 (q, J - 6.0 Hz, 2H), 2.70 (m, IH), 1.35 (t, J=6.8 Hz, 3H), 0.75 (q, J = 6.0 Hz, 2H), 0.49 (br, 2H); ESI-MS: calcd for (C8Hi3NO2) 155, found 156 (MH+). Example 61
Figure imgf000111_0001
[0216] To a mixture of compound 60 (0.5 g, 3.23 mmol) in 1, 4-dioxane (25 mL) and water (17 mL) was added NBS (0.6 g, 3.55 mmol) at room temperature. The slurry was warmed and stirred at 20-22° C for 3 h. Thiourea (0.26 g, 3.42 mmol) was added and the mixture was refluxed to 100 0C. After 2 h, the resulting solution was cooled to 20-22° C and cone, ammonium hydroxide (6 mL) was added drop- wise. The resulting slurry was concentrated under vacuum to about half volume and cooled to 0-5° C. The solid was collected by vacuum filtration, washed with cold water, and dried to give 0.5 g (85% yields) of compound 61 as deep-yellow solids. 1U NMR (500 MHz, CDCl3) δ 10.61 (br s, IH), 7.62 (d, 3.6 Hz, IH), 7.12 (s, IH), 6.96 (br s, 2H), 2.13 (m, 4H), 2.07 (m, IH); ESI-MS: calcd for (C7H9N3OS) 183, found 184 (MH+).
Example 62
Figure imgf000111_0002
[0217] A solution of Compound 61 (0.5 g, 2.73 mmol), diisopropylamine (0.5 ml, 3.0 mmol) and Compound 21 (1.10 g, 6.18 mmole) in THF (15 mL) was stirred at 0° C for 8 hours, then cold 5% NaHCO3 was added to the reaction mixture, and the aqueous mixture was extracted twice with EtOAc. The combined extracts were washed with brine, dried, and concentrated in vacuo until a lot of precipitate formed. After filtration, the solids were washed by ethyl acetate dried to give 62 (140 mg, 16%), which was used in the next step without purification for the next step reactions.
Example 63
Figure imgf000111_0003
[0218] A mixture of compound 62 (0.25 g, 0.78 mmol) and diisopropylethylamine (0.53 mL, 3.07 mmol), and N, N-dimethylethane-1, 2-diamine (0.27g, 3.07 mmol) in DMSO was heated at 70° C for over night. The mixture was extracted by ethyl acetate and the combined organic layers were washed with water and brine. The crude product was recrystallized by MeOH/CHCl3 (25 mg, 6%). 1H NMR (400 MHz, DMSO-c/6) 6 11.75 (br s, IH), 8.32 (d, J= 3.6 Hz, IH), 7.90 (brs,lH), 7.5 (br s, IH), 3.42 (m, 2H), 2.52 (m, 2H), 2.45 (m, 2H), 2.23 (s, 3H), 2.17 (m, 3H), 1.20 (m, 4H), 0.6 (m, 2H), 0.45 (m, 2H); ESI-MS: calcd for (C16H24N8OS) 376, found 377 (M+H+). HPLC: retention time: 12.2 min; purity 90.6%.
Example 64
Figure imgf000112_0001
[0219] A mixture of ethyl β-ethoxyacrylate (26.50 g, 183 mmol) and 2 N sodium hydroxide (110 mL, 220 mmol) was refiuxed for 2 h and cooled to 0° C. water was removed under vac, and the yellow solids were triturated with toluene and evaporated to give the sodium β-ethoxyacrylate (25 g, 97%). The mixture of sodium β-methoxyacrylate (10.26 g, 74.29 mmol) and thionyl chloride (25 mL, 343 mmol) was refiuxed for 2 h, and evaporated to give the β-ethoxyacryloyl chloride crude product, which was used without further purification. To a cold stirring solution of 3-ethoxyacryloyl chloride (7.3 g, 54.2 mmol) in THF (70 mL) was added isopropylamine (13.8 mL, 162.2 mmol) and pyridine (8.8 ml, 108 mmol). The mixture was then warmed and stirred overnight at room temperature. Water was added and extracted with EtOAc. The organic layer was evaporated and the resulting residue was purified by silica gel using 1 : 1 (hexane-EtoAc), concentrated under vacuum to give 2.3 g of compound 64. 1H NMR (400 MHz, CDCl3) δ 7.50 (d, J = 12.4 Hz, IH), 5.20 (m, 2H), 4.18 (m, IH), 3.94 (q, J= 6.0 Hz, 2H), 1.31 (t, J=5.2 Hz, 3H), 1.16 (s, 3H), 1.14 (s, 3H); ESI-MS: calcd for (C8H15NO2) 157, found 158 (MH+).
Example 65
Figure imgf000112_0002
[0220] To a mixture of compound 64 (1.0 g, 6.37 mmol) in 1, 4-dioxane (50 mL) and water (34 mL) was added NBS (1.19 g, 7.00 mmol) at room temperature. The slurry was warmed and stirred at 20-22° C for 3 h. Thiourea (0.51 g, 6.75 mmol) was added and the mixture was refluxed to 100° C. After 2 h, the resulting solution was cooled to 20-22° C and cone, ammonium hydroxide (6 mL) was added drop-wise. The resulting slurry was concentrated under vacuum to about half volume and cooled to 0-5° C. The solid was collected by vacuum filtration, washed with cold water, and dried to give 0.8 g (85% yields) of compound 65 as deep-yellow solids. 1H NMR (400 MHz, CDCl3) δ 10.61 (br s, IH), 7.62 (d, J = 3.6 Hz, IH), 7.12 (s, IH), 6.96 (br s, 2H), 2.13 (m, 4H), 2.07 (m, IH); ESI-MS: calcd for (C7H9N3OS) 183, found 184 (MH+).
Example 66
Figure imgf000113_0001
[0221] A solution of Compound 65 (0.5 g, 2.7 mmol), diisopropylamine (0.52 ml, 2.97 mmol) and Compound 21 (1.1O g, 6.18 mmol) in THF (15 mL) was stirred at 0° C for 8 hours, then cold 5% NaHCO3 was added to the reaction mixture, and the aqueous mixture was extracted twice with EtOAc. The combined extracts were washed with brine, dried, and concentrated in vacuo until a lot of precipitate formed. After filtration, the solids were washed by ethyl acetate dried to give 66 (371 mg, 42 %), which was used in the next step without purification for the next step reactions.
Example 67
Figure imgf000113_0002
[0222] A mixture of compound 66 (0.25 g, 0.78 mmol) and diisopropylethylamine (0.53 mL, 3.07 mmol), and N, N-dimethylethane-1, 2-diamine (0.27g, 3.07 mmol) in DMSO was heated at 70° C for over night. The mixture was extracted by ethyl acetate and the combined organic layers were washed with water and brine. The crude product was recrystallized by MeOH/CHCl3 (30mg, 10%). 1H NMR (400 MHz, DMSO-J6) 6 11.75 (br s, IH), 8.32 (d, J = 3.6 Hz, IH), 7.90 (brs,lH), 7.5 (br s, IH), 3.42 (m, 2H), 2.52 (m, 2H), 2.45 (m, 2H), 2.23 (s, 3H), 2.17 (m, 3H), 2.15 (m, IH), 1.12 (s, 3H), 1.1 (s, 3H), 1.0 (s, 3H); ESI-MS: calcd for (Ci6H26N8OS) 378 found 379 (M+H+). HPLC: retention time: 6.2 min; purity 84.5%. Example 68
Figure imgf000114_0001
[0223] A mixture of ethyl β-ethoxyacrylate (26.50 g, 183 mmol) and 2 N sodium hydroxide (110 mL, 220 mmol) was refluxed for 2 h and cooled to 0° C. water was removed under vac, and the yellow solids were triturated with toluene and evaporated to give the sodium β-ethoxyacrylate (25 g, 97%). The mixture of sodium β-methoxyacrylate (10.26 g, 74.29 mmol) and thionyl chloride (25 mL, 343 mmol) was refluxed for 2 h, and evaporated to give the β-ethoxyacryloyl chloride crude product, which was used in the next step without further purification. To a cold stirring solution of 3-ethoxyacryloyl chloride (5.0 g, 36.23 mmol) in THF (40 mL) was added 2,6-dimethylaniline (4.3 g, 35.5 mmol) and pyridine (4.4 ml, 54.34 mmol). The mixture was then warmed and stirred overnight at room temperature. Water was added and extracted with EtOAc. The organic layer was evaporated and the resulting residue was purified by silica gel using 1 : 1 (hexane-EtOAc), concentrated under vacuum to give 2.3 g of compound 68. 1H NMR (400 MHz, DMSO-dό) δ 8.95 (s, IH), 7.39 (d, J = 12.4 Hz, IH), 7.01 (s, 3H), 5.53 (d, J = 12.4 Hz, IH), 3.92 (q, J= 5.6 Hz, 2H), 2.08 (s, 6H), 1.24 (t, J=5.2 Hz, 3H), ESI-MS: calcd for (C]3H17NO2) 219, found 220 (MH+).
Example 69
Figure imgf000114_0002
[0224] To a mixture of compound 68 (2.5 g, 11.41 mmol) in 1, 4-dioxane (100 mL) and water (70 mL) was added NBS (2.13 g, 12.55 mmol) at room temperature. The slurry was warmed and stirred at 20-22° C for 3 h. Thiourea (0.92 g, 12.09 mmol) was added and the mixture was refluxed to 100° C. After 2 h, the resulting solution was cooled to 20-22° C and cone, ammonium hydroxide (10 mL) was added drop-wise. The resulting slurry was concentrated under vacuum to about half volume and cooled to 0-5° C. The solid was collected by vacuum filtration, washed with cold water, and dried to give 1.4 g (36% yields) of compound 69 as deep-brown solids. 1H NMR (400 MHz, CDCl3) δ 9.32 (s, IH), 7.80 (s, IH), 7.50 (s, 2H), 7.06 (br s, 3H), 2.12 (s, 6H), calcd for (C]2H13N3OS) 247, found 248 (MH+). Example 70
Figure imgf000115_0001
[0225] A solution of Compound 69 (0.5 g, 2.02 mmol), diisopropylethylamine (0.39 ml, 2.22 mmol) and Compound 21 (0.54 g, 3.02 mmol) in THF (15 mL) was stirred at 0° C for 8 hours, then cold 5% NaHCO3 was added to the reaction mixture, and the aqueous mixture was extracted twice with EtOAc. The combined extracts were washed with brine, dried, and concentrated in vacuo until a lot of precipitate formed. After filtration, the solids were washed by ethyl acetate dried to give 70 (309 mg, 64 %), which was used in the next step without purification for the next step reactions.
Example 71
Figure imgf000115_0002
[0226] A mixture of compound 70 (0.4 g, 1.03 mmol) and diisopropylethylamine (0.72 mL, 4.12 mmol), and N, N-dimethylethane-1, 2-diamine (0.36g, 4.12 mmol) in DMSO was heated at 70° C for over night. The mixture was extracted by ethyl acetate and the combined organic layers were washed with water and brine. The crude product was recrystallized by MeOH/CHCl3 (309 mg, 64%). 1H NMR (400 MHz, DMSO-J6) 811.75 (br s, IH), 9.63 (d, J = 5.2 Hz, IH), 8.22 (s, IH), 7.70 (brs,lH), 7.08 (m, 3H), 3.42 (m, 2H), 2.52 (m, 2H), 2.45 (m, 2H), 2.23 (m, 12H), 1.1 (m, 3H); ESI-MS: calcd for C21H28N8OS) 440 found 441 (M+H+). HPLC: retention time: 16.2 min; purity 98.4%
Example 72
Figure imgf000115_0003
[0227] A mixture of ethyl β-ethoxyacrylate (26.50 g, 183 mmol) and 2 N sodium hydroxide (110 mL, 220 mmol) was refluxed for 2 h and cooled to 0° C water was removed under vac, and the yellow solids were triturated with toluene and evaporated to give the sodium β-ethoxyacrylate (25 g, 97%). The mixture of sodium β-methoxyacrylate (10.26 g, 74.29 mmol) and thionyl chloride (25 mL, 343 mmol) was refluxed for 2 h, and evaporated to give the β-ethoxyacryloyl chloride crude product, which was used in the next step without further purification. To a cold stirring solution of 3-ethoxyacryloyl chloride (5.0 g, 36.23 mmol) in THF (40 mL) was added 2,4,6-trimethylaniline (4.8 g, 35.5 mmol) and DIPEA (9.47 ml, 54.34 mmol). The mixture was then warmed and stirred overnight at room temperature. Water was added and extracted with EtOAc. The organic layer was evaporated and the resulting solids were triturated with EtOAc and filtered the solids to give 1.5 g (18%) of compound 72. 1H NMR (400 MHz, DMSO-d6) δ 8.88 (s, IH), 7.39 (d, 12.4 Hz, IH), 6.89 (s, IH), 6.82 (s, IH), 5.53 (d, J = 12.4 Hz, IH), 3.92 (q, J= 5.6 Hz, 2H), 2.28 (s, 3H), 2.18 (s, 3H), 2.03 (s, 3H), 1.24 (t, J=5.2 Hz, 3H).
Example 73
Figure imgf000116_0001
[0228] To a mixture of compound 72 (1.9 g, 8.15 mmol) in 1, 4-dioxane (50 mL) and water (35 mL) was added NBS (1.52 g, 8.97 mmol) at room temperature. The slurry was warmed and stirred at 20-22° C for 3 h. Thiourea (0.64 g, 8.64 mmol) was added and the mixture was refluxed to 100° C. After 2 h, the resulting solution was cooled to 20-22° C and cone, ammonium hydroxide (10 mL) was added drop- wise. The resulting slurry was concentrated under vacuum to about half volume and cooled to 0-5° C. The solid was collected by vacuum filtration, washed with cold water, and dried to give 0.88 g (44% yields) of compound 35 as deep-brown solids. 1H NMR (400 MHz, CDCl3) δ 9.24 (s, IH), 7.78 (s, IH), 7.48 (s, IH), 6.86 (s, 2H), 6.57 (s, IH), 2.07 (m, 6H), 1.99 (t, J= 5.2 Hz, 3H), calcd for (C13Hi5N3OS) 261, found 262 (MH+).
Example 74
Figure imgf000116_0002
[0229] A solution of Compound 73 (0.5 g, 1.91 mmol), diisopropylethylamine (0.37 ml, 2.1 mmol) and Compound 21 (0.51 g, 2.87 mmol) in THF (15 mL) was stirred at 0° C for 8 hours, then cold 5% NaHCO3 was added to the reaction mixture, and the aqueous mixture was extracted twice with EtOAc. The combined extracts were washed with brine, dried, and concentrated in vacuum. The crude product was triturated with EtOAc and resulting solids were filtered off to give 74 (400 mg, 65 %), which was used in the next step without further purification.
Example 75
Figure imgf000117_0001
[0230] A mixture of compound 74 (0.4 g, 0.99 mmol) and diisopropylethylamine (0.69 mL, 3.98 mmol), and N, N-dimethylethane-1, 2-diamine (0.35g, 3.98 mmol) in DMSO was heated at 600C for over night. The mixture was extracted by ethyl acetate and the combined organic layers were washed with water and brine. The crude product was recrystallized by i- PrOH/CHCl3 to give 37 as white solid, 39 mg, 9% yield. 1H NMR (400 MHz, DMSO-J6) 5 11.75 (br s, IH), 9.52 (d, J= 5.2 Hz, IH), 8.19 (s, IH), 7.70 (brs,lH), 6.88 (s, 2H), 3.48 (m, 2H), 2.52 (m, 2H), 2.45 (m, 2H), 2.10 (m, 15H), 1.19 (m, 3H); ESI-MS: calcd for C22H30N8OS is 454, found 455 (M+H+). HPLC: retention time: 17.1 min; purity 98.6%
Example 76
Figure imgf000117_0002
[0231] A mixture of ethyl β-ethoxyacrylate (26.50 g, 183 mmol) and 2 N sodium hydroxide (110 mL, 220 mmol) was refluxed for 2 h and cooled to 0° C. water was removed under vac, and the yellow solids were triturated with toluene and evaporated to give the sodium β-ethoxyacrylate (25 g, 97%). The mixture of sodium β-methoxyacrylate (10.26 g, 74.29 mmol) and thionyl chloride (25 mL, 343 mmol) was refluxed for 2 h, and evaporated to give the β-ethoxyacryloyl chloride crude product, which was used in the next step without further purification. To a cold stirring solution of 3-ethoxyacryloyl chloride (5.0 g, 36.23 mmol) in THF (40 mL) was added aniline (3.23 ml, 35.5 mmol) and pyridine (4.4 ml, 54.34 mmol). The mixture was then warmed and stirred overnight at room temperature. Water was added and extracted with EtOAc. The organic layer was evaporated and the resulting residue was purified by silica gel using 1 :1 (hexane-EtoAc), concentrated under vacuum to give 2.71 g of compound 76. 1H NMR (400 MHz, DMSO-d6) Example 77
Figure imgf000118_0001
[0232] To a mixture of compound 77 (2.7 g, 14.14 mmol) in 1 , 4-dioxane (100 mL) and water (70 mL) was added NBS (1.14 g, 15.55mmol) at room temperature. The slurry was warmed and stirred at 20-22° C for 3 h. Thiourea (1.14 g, 14.98 mmol) was added and the mixture was refluxed to 100° C. After 2 h, the resulting solution was cooled to 20-22° C and cone, ammonium hydroxide (10 mL) was added drop- wise. The resulting slurry was concentrated under vacuum to about half volume and cooled to 0-5° C. The solid was collected by vacuum filtration, washed with cold water, and dried to give 1.2 g (39% yields) of compound 77 as deep-brown solids. 1H NMR (400 MHz, DMSO-dό) 5 9.8 (s, IH), 7.9 (s, Hz, IH), 7.6 (m, 4H), 7.3 (m, 2H), 7.1 (m, IH),; ESI-MS: calcd for (Ci0H9N3OS): 219 found 220 (M+H+).
Example 78
Figure imgf000118_0002
[0233] A solution of Compound 77 (0.5 g, 2.28 mmol), diisopropylethylamine (0.44 ml, 2.5 mmol) and Compound 21 (0.61 g, 3.42 mmol) in THF (30 mL) was stirred at 0° C for 8 hours, then cold 5% NaHCO3 was added to the reaction mixture, and the aqueous mixture was extracted twice with EtOAc. The combined extracts were washed with brine, dried, and concentrated in vacuo. The crude solids were triturated with EtOAc and after filtration, the solids were washed by ethyl acetate dried to give 78 (400 mg, 49 %), which was used in the next step without further purification.
Example 79
Figure imgf000118_0003
[0234] A mixture of compound 78 (0.4 g, 1.11 mmol) and diisopropylethylamine (0.77 mL, 4.4.44 mmol), and N, N-dimethylethane-1, 2-diamine (0.39g, 4.44 mmol) in DMSO was heated at 60° C for over night. The mixture was extracted by ethyl acetate and the combined organic layers were washed with water and brine. The crude product was recrystallized by MeOH/CHCl3 to give 79 (21 mg, 5%) 1H NMR (400 MHz, DMSO-J6) 811.80 (br s, IH), 10.05 (d, J= 5.2 Hz, IH), 8.32 (d, 7.2Hz, IH), 7.70 (brs,lH), 7.66 (m, 2H), 7.1 (m, 2H), 7.05, IH), 3.42 (m, 2H), 2.52 (m, 2H), 2.45 (m, 2H), 2.23 (m, 6H), 1.1 (m, 3H); ESI-MS: calcd for C19H24N8OS) 412 found 413 (M+H+). HPLC: retention time: 10.2 min; purity 99 %
Example 80
Figure imgf000119_0002
[0235] 2.9 grams of 10% Pd/C was flushed with H2, anhydrous THF was added, and the solution was reflushed with H2. 2,6-Lutidine (21.2g, 198 mmol) and ethyl 4-chloro-4- oxobutanoate (29.8g, 181 mmol) were added, and the solution was stirred at room temperature under H2 for 24 h. The reaction mixture was filtered off through celite and evaporated. The crude residue was dissolved again in CH2Cl2 (500 ml) and washed with Water (200 ml), IN HCl (200 ml) and again with water. The organic layers were dried and evaporated to give 80 (20.2g, 85%). 1H NMR (400 MHz, DMSO-J6) 89.80 (s, IH), 4.14 (q, J = 6.8 Hz, 2H), 2.78 (t, J= 6.0 Hz, 2H), 2.60 (t, J= 6.4 Hz, 2H), 1.24 (t, J= 0.8 Hz, 3H).
Example 81
Figure imgf000119_0003
[0236] Bromine (18.0g, 112 mmol) was added over 0.5 hour to a solution of 80 (12.36g, 107 mmol) in diethyl ether (100 ml) and 1,4-dioxane (91 ml) and the reaction mixture stirred for one hour at room temperature. The reaction mixture was poured into CH2Cl2 (300 ml) and sodium hydrogen carbonate (20.09 g, 237 mmol) was added and stirred for 16 h. The solids were filtered off and the filtrate was concentrated to give 81 (22.9g). The crude brown oil was used in the next step without further purification.
Example 82
Figure imgf000119_0001
[0237] Ethyl 4-bromo-4-oxobutanoate 81 (22.9g, 109.6 mmol) was added to a suspension of thiourea (7.5 g, 98.71 mmol) in Ethanol. The reaction mixture was refluxed for 8h. After cooling, the resulting solids were filtered off and washed with cold EtOH to give 82 (1 Ig,
J CoQ. O Z0 //oΛ).
Example 83
Figure imgf000120_0001
[0238] Sodium hydroxide (8ml of a 1.0 N solution in water) was added to a solution of 82 (2.Ig) in THF (12ml), methanol (4ml), H2O (4ml). The solution was stirred overnight at ambient temperature. The organic solvents were removed under reduced pressure. The residue was acidified using IN HCl to pH 3-4. The solvents were removed and used in the next step without further purification.
Example 84
Figure imgf000120_0002
[0239] Sodiunidicyanoamide (25g, 28ImMoI) in 25mL of H2O was rapidly added to 125mL of cone. HCl at -18° C. Reaction mixture was stirred at -18° C for 15 minutees and at 35° C for another 15 minutes. Cooling at 0° C resulted in formation of white solid that was filtered and washed with ice-cold water to give 12.5g (43%) of intermediate 84 which was used crude for next step.
Example 85
Figure imgf000120_0003
[0240] To 15OmL of CH2Cl2 at room temperature was added DMF ( 11.4mL) followed by POCl3 (11.4mMol). After 5 minutes of stirring the intermediate 84 (12.5g, 120.8mMol) was added in portions. Reaction mixture was stirred overnight at room temperature. Next day, reaction was washed with water (3x), brine (Ix), dried over Na2SO4, filtered and solvent was evaporated to give 7.1g (17%) of off-white solid. 1H NMR (400 MHz, CDCl3) δ 8.91 (s, IH). Example 86
Figure imgf000121_0001
[0241] To the 83 (800mg, 5.06mMol) in 10 niL of DMF at 0° C was added pyridine (0.91 niL, 11.32 mMol) followed by careful, dropwise addition of pentafluorophenyl trifluoroacetate (1.72mL, 10.13 mMol). Reaction mixture was stirred at 0° C for 10 minutes and 90 minutes at room temperature. 3-fluoroaniline (0.97 mL, 10.13 mMol) was added and reaction mixture was stirred overnight at room temperature. Reaction was poured into 5OmL of IN HCl, organic layer was separated. Aqueous layer was extracted with EtOAc. Organic fractions were combined, washed with brine, dried over Na2SO4, filtered and solvent was evaporated to give 86 (0.4 g, 23%) and this was used in the next step without purification.
Example 87
Figure imgf000121_0002
[0242] Crude amide 86 from previous step (0.4g) was heated for 3 hours in 1OmL of methanol and 1OmL of 2N HCl. Reaction mixture was neutralized with Saturated NaHCO3 and methanol was evaporated. Aqueous solution was extracted with EtOAc. Organic fractions were combined, washed with brine, dried over Na2SO4, filtered and solvent was evaporated. Flash column chromatography (silica, CH2Cl2/Me0H 5% to 10%) yielded desired product 87 as yellow oil (360mg, 28% over 2 steps). 1H NMR (400 MHz, DMSO) δ 10.26 (br s, IH), 7.58 (m, 3H), 7.30 (m, 2H), 6.86 (m, 2H), 3.65 (s, 2H). ESI-MS: calcd for Cn Hi 0FN3OS) 251, found 252 (M+H+).
Example 88
Figure imgf000121_0003
[0243] A solution of Compound 87 (0.1 g, 0.39 mmol), diisopropylamine (0.075 ml, 0.43 mmol) and Compound 85 (0.092 g, 0.62 mmol) in THF (10 mL) was stirred at 0° C for 8 hours. DIPEA (128μL, 95mg, and 0.73mMol) was added followed by 1 -methylpiparezine (80 mg, 0.80 mMol) and reaction mixture was stirred overnight at room temperature. Solvent was evaporated and crude material was re-dissolved in EtOAc (3OmL), washed with sat. NaHCO3, brine, dried over Na2SO4, filtered and solvent was evaporated. Flash column chromatography (silica, CH2Cl2/Me0H 5% to 10%) yielded 88 as off-white solid (25mg, 15 %). 1H NMR (400 MHz, DMSO) δ 11.65 (br s, IH), 10.42 (s, IH), 8.30 (s, IH), 7.61 (m, IH), 7.30 (m, 3H), 6.85 (m, IH), 3.67 (m, 6H), 2.35 (m, 4H), 2.16 (s, 3H). ESI-MS: calcd for (C19H21FN8OS) 428, found 429 (M+H+).
Example 89
Figure imgf000122_0001
[0244] A solution of Compound 87 (0.075 g, 0.3 mmol), diisopropylamine (0.075 ml, 0.32 mmol) and Compound 85 (0.067 g, 0.45 mmol) in THF (10 mL) was stirred at 0° C for 8 hours. DIPEA (56μL, 0.32 mmol) was added followed by piperidine (102 mg, 1.20 mmol) and the reaction mixture was stirred overnight at room temperature. Solvent was evaporated and crude material was re-dissolved in EtOAc (3OmL), washed with sat. NaHCO3, brine, dried over Na2SO4, filtered and solvent was evaporated. Flash column chromatography (silica, CH2Cl2MeOH 1% to 5) yielded 89 as off- white solid (10 mg, 8 % over two steps). 1H NMR (400 MHz, DMSO) δ 11.55 (br s, IH), 10.42 (s, IH), 8.30 (s, IH), 7.61 (m, IH), 7.30 (m, 3H), 6.85 (m, IH), 3.80 (m, 6H), 1.65 (m, 2H), 1.52 (m, 4H); ESI-MS: calcd for (Ci9H2OFN7OS) 413, found 414 (M+H+).
Example 90
Figure imgf000122_0002
[0245] To the 83 (800mg, 5.06mmol) in 10 mL of DMF at 0° C was added pyridine (0.91 niL, 11.32 mmol) followed by careful, dropwise addition of pentafluorophenyl trifluoroacetate (1.72mL, 10.13 mmol). Reaction mixture was stirred at 0° C for 10 minutes and 90 minutes at room temperature. 2-chloro-6-methylaniline (1.24mL, 10.13mmol) was added and reaction mixture was stirred overnight at room temperature. Reaction was poured into 5OmL of IN HCl, organic layer was separated. Aqueous layer was extracted with EtOAc. Organic fractions were combined, washed with brine, dried over Na2SO4, filtered and solvent was evaporated to give 90 (0.25 g, 42 %) and this was used in the next step without further purification.
Example 91
Figure imgf000123_0001
[0246] Crude amide 90 from previous step (0.25g) was heated for 8 hours in 1OmL of methanol and 1OmL of 2N HCl. Reaction mixture was neutralized with Saturated NaHCO3 and methanol was evaporated. Aqueous solution was extracted with EtOAc. Organic fractions were combined, washed with brine, dried over Na2SO4, filtered and solvent was evaporated. Flash column chromatography (silica, CH2Cl2/Me0H 5% to 10%) yielded desired product 91 (174mg, 12% over 2 steps). 1H NMR (400 MHz, DMSO) δ 9.68 (s, IH), 7.31 (m, IH), 7.20 (m, 2H), 6.75 (m, 3H), 3.63 (s, 2H), 2.12 (s, 3H); ESI-MS: calcd for Ci2Hi2ClN3OS) 281 found 282 (M+H+).
Example 92
Figure imgf000123_0002
[0247] A solution of Compound 91 (0.07 g, 0.25 mmol), diisopropylamine (47 ul,
0.27 mmol) and Compound 85 (0.056 g, 0.37 mmol) in THF (10 mL) was stirred at 0° C for 8 hours. DIPEA (0.17 mL, 1.0 mmol) was added followed by 1-methylpiparezine (0.11 ml, 1.0 mmol) and reaction mixture was stirred overnight at room temperature. Solvent was evaporated and crude material was re-dissolved in EtOAc (3OmL), washed with sat. NaHCO3, brine, dried over Na2SO4, filtered and solvent was evaporated. Flash column chromatography (silica, CH2Cl2MeOH 5% to 10%) yielded 92 as off-white solid (27mg, 24 %). 1H NMR (400 MHz, DMSO) δ 11.48 (br s, IH), 9.85 (s, IH), 8.30 (s, IH), 7.35 (m, IH), 7.27 (s, IH), 7.21 (m, 2H), 3.85 (m, 6H), 2.35 (m, 4H), 2.19 (s, 3H), 2.13 (s, 3H); ESI-MS: calcd for (C20H23C1N8OS) 458, found 459(M+H+).
Example 93
Figure imgf000124_0001
[0248] Compound 91 (111 mg, 0.38 mmol), 2-(4-(4-chloro-6-ethyl-l, 3, 5-triazin-2- yl)piperazin-l-yl)ethanol (39) (122 mg, 0.45 mmol), Pd(OAc)2 (10 mg, 0.04 mmol), Xantphos (48 mg, 0.08 mmol) and K2CO3 (1.0 g, 7.5 mmol) were added in screw capped microwave vial. THF: DMF (2.5 mL, 1.5:1) was added and vial was sealed with a cap. The mixture was heated at 150° C for 10 min. under microwave (Biotage, Initiator 2.0) conditions. Reaction mixture was filtered and the solid was washed with CH2Cl2 and MeOH, concentrated, (silica, CH2Cl2/Me0H 5% to 10%) yielded 93 as off-white solid (25 mg, 14 %). 1H NMR (400 MHz, DMSO) δ 11.45 (br s, IH), 9.55 (br s, IH), 7.33 (m, 2H), 7.14 (m, 2H), 4.40 (t, IH, J = 5.4 Hz), 3.73 (bs, 2H), 3.64 (bs, 2H), 3.51-3.47 (m, 4H), 2.49-2.35 (m, 8H), 2.11 (s, 3H), 1.15 (t, 3H, J = 7.6 Hz).; ESI-MS: calcd for (C23H29ClN8O2S) 517, found 518 (M+H+).
Example 94
Figure imgf000124_0002
[0249] To the compound 22 (lOOmg, 0.244mMol) in 5mL oh /PrOH was added DIPEA (170μL, 126mg, 0.976mMol) and JV,N-diethylehylenedialine (52μL, 43mg, 0.366mMol). Reaction mixture was microwaved at 120° C for 40 minutes. Disappearance of starting material was confirmed by TLC. Solvent was removed under reduced pressure and flash column chromatography yielded 85mg (44%) of desired product 94. 1H NMR (400 MHz, DMSO) δ 11.78 (bs, IH), 9.92 (s, IH), 8.27 (s, IH), 7.62 (bs, IH), 7.40 (dd, J= 12, 1.6 Hz, IH), 7.27 (m, 2H), 3.60-3.50 (m, 2H), 2.65 - 2.45 (m, 8H), 2.23 (s, 3H), 1.23 (m, 3H), 0.92 (t, J= 7.2 Hz, 6H). ESI-MS: calcd for (C22H29ClN8OS) 488, MS (ESI) m/z 489 [M+H]+.
Example 95
Figure imgf000125_0001
[0250] To 2-amino-l-propene-l,l,3-tricarbonitrile (15.5g, 117.3mMol) in 15OmL OfH2O at room temperature was added 50-60% hydrazine hydrate (7.4mL, 7.6g, 129.03mMol). After solid dissolved reaction mixture was heated at 90° C for 30 minutes and then ice cooled. Formed solid was filtered and dried overnight on high vacuum to give 13g (75%) of product 95 that was used crude for next step.
Example 96
Figure imgf000125_0002
[0251] Compound 95 (13g, 88.4mMo) was added to 12OmL of ION NaOH (aq) and heated at 100° C overnight. Reaction mixture was cooled in ice bath and pH was adjusted to 3 using cone. HCl. After cooling on ice bath for 1 hr formed solid was collected, washed with H2O, dried on air for 1 hr then washed with EtOAc and dried on high vacuum to give 13.2g (81%) of desired product 96 that was used crude for next step.
Example 97
Figure imgf000125_0003
[0252] Compound 96 (13.2g, 71.3mMol) in 25OmL of H2O was refluxed at 1250C for 5 hrs. Reaction mixture was cooled at room temperature and green residue was filtered off. Filtrate was evaporated to give crude product that was dried on high vacuum to yield 1 Og (99%) of desired product 97. 1H NMR (400 MHz, DMSO) δ 6.88 (s, 2H), 5.59 (bs, 2H), 5.25 (s, IH), 3.62 (s, 2H). 1H NMR (400 MHz, DMSO+D2O) δ 5.28(s, IH), 3.62 (s, 2H).
Example 98
Figure imgf000126_0001
[0253] To compound 97 (700mg, 4.96mMol) in 7mL of DMF at 0° C was added pyridine (883μL, 863mg, 10.9ImMoI) followed by careful, dropwise addition of pentafluorophenyl trifluoroacetate (1.68mL, 2.78g, 9.92mMol). Reaction mixture was stirred at 0° C for 10 minutes and 90 minutes at room temperature. 3-fluoroaniline (954μL, l.lg, 9.92mMol) was added and reaction mixture was stirred overnight at room temperature. Reaction was poured into 5OmL of IN HCl, organic layer was separated. Aqueous layer was extracted with EtOAc. Organic fractions were combined, washed with brine, dried over Na2SO4, filtered and solvent was evaporated to give 1.6g (quant.) of crude product 98 that was used in the next step without purification.
Example 99
Figure imgf000126_0002
[0254] Crude amide 98 from previous step (1.6g, 4.96mMol) was heated for 3 hours in 1OmL of methanol and 1OmL of 2N HCl. Reaction mixture was neutralized with cone. NaHCO3 and methanol was evaporated. Aqueous solution was extracted with EtOAc. Organic fractions were combined, washed with brine, dried over Na2SO4, filtered and solvent was evaporated. Flash column chromatography (silica, CH2Cl2/Me0H 5% to 15%) yielded desired product 99 as yellow oil (640mg, 55% over 2 steps). 1H NMR (400 MHz, DMSO) δ 10.26 (s, IH), 7.58 (m, IH), 7.30 (m, 2H), 7.12 (bs, IH), 6.86 (m, IH), 5.30 (s, IH), 3.47 (s, 2H). calcd for (C1 IH11FN4O) 234, MS (ESI) m/z 235 [M+H]+.
Example 100
Figure imgf000126_0003
[0255] To compound 97 (700mg, 4.96mMol) in 7mL of DMF at O0C was added pyridine (883 μL, 863mg, 10.9ImMoI) followed by careful, dropwise addition of pentafluorophenyl trifluoroacetate (1.68mL, 2.78g, 9.92mMol). Reaction mixture was stirred at 0° C for 10 minutes and 90 minutes at room temperature. 2-chloro-6-methylaniline (1.22mL, 1.4g, 9.92mMol) was added and reaction mixture was stirred overnight at room temperature. Reaction was poured into 5OmL of IN HCl, organic layer was separated. Aqueous layer was extracted with EtOAc. Organic fractions were combined, washed with brine, dried over Na2SO4, filtered and solvent was evaporated to give 1.79g (quant.) of crude product 100 that was used in the next step without purification.
Example 101
Figure imgf000127_0001
[0256] Crude amide 100 from previous step (1.79g, 4.96mMol) was heated for 3 hours in 1OmL of methanol and 1OmL of 2N HCl. Reaction mixture was neutralized with cone. NaHCO3 and methanol was evaporated. Aqueous solution was extracted with EtOAc. Organic fractions were combined, washed with brine, dried over Na2SO4, filtered and solvent was evaporated. Flash column chromatography (silica, CH2Cl2/Me0H 5% to 15%) yielded desired product 101 as yellow oil (250mg, 19% over 2 steps). 1H NMR (400 MHz, DMSO) δ 11.20 (bs, IH), 9.62 (s, IH), 7.32 (m, IH), 7.21 (m, 2H), 5.35 (s, IH), 4.55 (bs, 2H), 3.50 (s, 2H), 2.15 (s, 3H). calcd for (C12H13ClN4O) 264, MS (ESI) m/z 265 [M+H]+.
Example 102
Figure imgf000127_0002
[0257] To the 97 (700mg, 4.96mMol) in 7mL of DMF at 0° C was added pyridine (883μL, 863mg, 10.9ImMoI) followed by careful, dropwise addition of pentafluorophenyl trifluoroacetate (1.68mL, 2.78g, 9.92mMol). Reaction mixture was stirred at 0° C for 10 minutes and 90 minutes at room temperature. 4-fluorobenzylamine (1.13mL, 1.12g, 9.92mMol) was added and reaction mixture was stirred overnight at room temperature. Reaction was poured into 5OmL of IN HCl, organic layer was separated. Aqueous layer was extracted with EtOAc. Organic fractions were combined, washed with brine, dried over Na2SO4, filtered and solvent was evaporated to give 1.7Ig (quant.) of crude product 102 that was used in the next step without purification. Example 103
Figure imgf000128_0001
[0258] Crude amide 102 from previous step (1.7Ig, 4.96mMol) was heated for 3 hours in 1OmL of methanol and 1OmL of 2N HCl. Reaction mixture was neutralized with cone. NaHCO3 and methanol was evaporated. Aqueous solution was extracted with EtOAc. Organic fractions were combined, washed with brine, dried over Na2SO4, filtered and solvent was evaporated. Flash column chromatography (silica, CH2Cl2/Me0H 5% to 15%) yielded desired product 103 as yellow oil (520mg, 42% over 2 steps). 1H NMR (400 MHz, DMSO) δ 11.50 (bs, IH), 8.37 (t, J= 6.0 Hz, IH), 7.27 (m, 2H), 7.13 (m, 2H), 5.75 (bs, 2H), 5.25 (s, IH), 4.23 (d, J= 6.0Hz, 2H), 3.17 (s, 2H). calcd for (C12H13FN4O) 248, MS (ESI) m/z 249 [M+H]+.
Example 104
Figure imgf000128_0002
[0259] To compound 85 (lOOmg, 0.67mMol) in 5mL of THF at room temperature was added 99 (157mg, 0.67mMol) and DIPEA (128μL, 95mg, 0.73mMol) in 5mL of THF. Reaction mixture was stirred for 4 hours at room temperature. DIPEA (128μL, 95mg, 0.73mMol) was added followed by 1-methylpiparezine (75μL, 67mg, 0.67mMol) and reaction mixture was stirred overnight at room temperature. Solvent was evaporated and crude material was re-dissolved in EtOAc (3OmL), washed with sat. NaHCO3, brine, dried over Na2SO4, filtered and solvent was evaporated. Flash column chromatography (silica, CH2Cl2MeOH 5% to 10%) yielded 104 as off-white solid (24mg, 9%). ]R NMR (400 MHz, DMSO) δ 12.17 (s, IH), 10.43 (s, IH), 9.75 (s, IH), 8.16 (s, IH), 7.61 (m, IH), 7.32 (m, 2H), 6.89 (m, IH), 6.48 (s, IH), 3.67 (bs, 6H), 2.29 (bs, 4H), 2.16 (s, 3H). calcd for (C19H22FN9O) 411, m/z 412 [M+H]+.
Example 105
Figure imgf000129_0001
[0260] To compound 85 (lOOmg, 0.67mMol) in 5mL of THF at room temperature was added 99 (157mg, 0.67mMol) and DIPEA (128μL, 95mg, 0.73mMol) in 5mL of THF. Reaction mixture was stirred for 4 hours at room temperature. DIPEA (128μL, 95mg, 0.73mMol) was added followed by 3-dimethylamino-l-propanol (78μL, 69mg, 0.67mMol) and reaction mixture was stirred overnight at room temperature. Solvent was evaporated and crude material was re-dissolved in EtOAc (3OmL), washed with sat. NaHCO3, brine, dried over Na2SO4, filtered and solvent was evaporated. Flash column chromatography (silica, CH2Cl2MeOH 5% to 20%) yielded 105 as off-white solid (32mg, 12%). 1H NMR (400 MHz, DMSO) δ 12.29 (s, IH), 10.42 (bs, 2H), 8.41 (s, IH), 7.60 (m, IH), 7.32 (m, 2H), 6.88 (m, IH), 6.54 (s, IH), 4.31 (t, J= 6.0Hz, 2H), 3.72 (s, 2H), 2.39 (bs, 2H), 2.19 (s, 6H), 1.83 (bs, 2H). calcd for (C19H23FN8O2) 414, m/z 415 [M+H]+.
Example 106
Figure imgf000129_0002
[0261] To compound 85 (70mg, 0.47mMol) in 5mL of THF at room temperature was added 101 (124mg, 0.47mMol) and DIPEA (87μL, 64mg, 0.5OmMoI) in 5mL of THF. Reaction mixture was stirred for 4 hours at room temperature. DIPEA (87μL, 64mg, 0.5OmMoI) was added followed by 1 -methylpiparezine (52μL, 47mg, 0.47mMol) and reaction mixture was stirred overnight at room temperature. Solvent was evaporated and crude material was re-dissolved in EtOAc (3OmL), washed with sat. NaHCO3, brine, dried over Na2SO4, filtered and solvent was evaporated. Flash column chromatography (silica, CH2Cl2MeOH 5% to 10%) yielded 106 as off-white solid (70mg, 34%). 1H NMR (400 MHz, DMSO) δ 12.16 (s, IH), 9.77 (bs, 2H), 8.17 (s, IH), 7.34 (m, IH), 7.22 (m, 2H), 6.54 (s, IH), 3.72 (bs, 6H), 2.30 (bs, 4H), 2.18 (s, 3H), 2.16 (s, 3H). calcd for (C20H24ClN9O) 441, m/z 442 [M+H]+. Example 107
Figure imgf000130_0001
[0262] To compound 85 (70mg, 0.47mMol) in 5mL of THF at room temperature was added 101 (124mg, 0.47mMol) and DIPEA (87μL, 64mg, 0.5OmMoI) in 5mL of THF. Reaction mixture was stirred for 4 hours at room temperature. DIPEA (87μL, 64mg, 0.5OmMoI) was added followed by 3-dimethylamino-l-propanol (55μL, 48mg, 0.47mMol) and reaction mixture was stirred overnight at room temperature. Solvent was evaporated and crude material was re-dissolved in EtOAc (3OmL), washed with sat. NaHCO3, brine, dried over Na2SO4, filtered and solvent was evaporated. Flash column chromatography (silica, CH2Cl2MeOH 5% to 15%) yielded 107 as off-white solid (35mg, 170Zo)-1H NMR (400 MHz, DMSO) δ 12.29 (s, IH), 10.42 (bs, IH), 9.78 (s, IH), 8.42 (s, IH), 7.34 (m, IH), 7.22 (m, 2H), 6.61 (s, IH), 4.33 (t, J= 6.4Hz, 2H), 3.72 (s, 2H), 2.36 (bs, 2H), 2.17 (m, 9H), 1.83 (bs, 2H). calcd for (C20H25ClN8O2) 444, m/z 445 [M+H]+.
Example 108
Figure imgf000130_0002
[0263] To compound 85 (lOOmg, 0.67mMol) in 5mL of THF at room temperature was added 103 (166mg, 0.67mMol) and DIPEA (128μL, 95mg, 0.73mMol) in 5mL of THF. Reaction mixture was stirred for 4 hours at room temperature. DIPEA (128μL, 95mg, 0.73mMol) was added followed by 1 -methylpiparezine (75μL, 67mg, 0.67mMol) and reaction mixture was stirred overnight at room temperature. Solvent was evaporated and crude material was re-dissolved in EtOAc (3OmL), washed with sat. NaHCO3, brine, dried over Na2SO4, filtered and solvent was evaporated. Flash column chromatography (silica, CH2Cl2MeOH 5% to 10%) yielded 108 as off-white solid (68mg, 24%). 1H NMR (400 MHz, DMSO) δ 12.08 (s, IH), 9.69 (s, IH), 8.53 (s, IH), 8.17 (s, IH), 7.29 (m, 2H), 7.13 (m, 2H), 6.43 (s, IH), 4.27 (d, J= 6.0Hz, 2H), 3.72 (bs, 4H), 3.51 (s, 2H), 2.31 (bs, 4H), 2.20 (s, 3H). calcd for (C20H24FN9O) 425, m/z 426 [M+H]+. Example 109
Figure imgf000131_0001
[0264] To compound 85 (lOOmg, 0.67mMol) in 5mL of THF at room temperature was added 103 (166mg, 0.67mMol) and DIPEA (128μL, 95mg, 0.73mMol) in 5mL of THF. Reaction mixture was stirred for 4 hours at room temperature. DIPEA (128μL, 95mg, 0.73mMol) was added followed by 3-dimethylarnino-l-propanol (78μL, 69mg, 0.67mMol) and reaction mixture was stirred overnight at room temperature. Solvent was evaporated and crude material was re-dissolved in EtOAc (3OmL), washed with sat. NaHCO3, brine, dried over Na2SO4, filtered and solvent was evaporated. Flash column chromatography (silica, CH2Cl2MeOH 5% to 20%) yielded 109 as off-white solid (30mg, 10%). 1H NMR (400 MHz, DMSO) δ 12.20 (s, IH), 10.38 (bs, IH), 8.54 (s, IH), 8.42 (s, IH), 7.29 (m, 2H), 7.13 (m, 2H), 6.49 (s, IH), 4.32 (t, J= 6.4Hz, 2H), 4.26 (d, J= 6.0Hz, 2H), 3.52 (s, 2H), 2.32 (t, J= 6.8Hz, 2H), 2.14 (s, 6H), 1.83 (m, 2H). calcd for (C20H25FN8O2) 428, m/z 451 [M+Na]+.
Example 110:
Figure imgf000131_0002
[0265] To compound 85 (lOOmg, 0.67mMol) in 5mL of THF at room temperature was added 99 (157mg, 0.67mMol) and DIPEA (128μL, 95mg, 0.73mMol) in 5mL of THF. Reaction mixture was stirred for 4 hours at room temperature. DIPEA (128μL, 95mg, 0.73mMol) was added followed by N,N,N-trimethylethylenediamine (87μL, 68mg, 0.67mMol) and reaction mixture was stirred overnight at room temperature. Solvent was evaporated and crude material was re-dissolved in EtOAc (3OmL), washed with sat. NaHCO3, brine, dried over Na2SO4, filtered and solvent was evaporated. Flash column chromatography (silica, CH2Cl2/Me0H 5% to 20%) yielded 110 as off-white solid (37mg, 13%). 1H NMR (400 MHz, DMSO @ 8O0C) δ 11.95 (bs, IH), 10.13 (s, IH), 9.20 (bs, IH), 8.17 (s, IH), 7.56 (m, IH), 7.33 (m, 2H), 6.85 (m, IH), 6.49 (s, IH), 3.67 (t, J= 6.0Hz, 2H), 3.08 (s, 3H), 2.48 (m, 2H), 2.21 (s, 6H). calcd for (Ci9H24FN9O) 413, m/z 414 [M+H]+. Example 111 :
Figure imgf000132_0001
[0266] To compound 85 (lOOmg, 0.67mMol) in 5mL of THF at room temperature was added 99 (157mg, 0.67mMol) and DIPEA (128μL, 95mg, 0.73mMol) in 5mL of THF. Reaction mixture was stirred for 4 hours at room temperature. DIPEA (128μL, 95mg, 0.73mMol) was added followed by 1 -methoxy-2 -propylamine (71 μL, 60mg, 0.67mMol) and reaction mixture was stirred overnight at room temperature. Solvent was evaporated and crude material was re-dissolved in EtOAc (3OmL), washed with sat. NaHCO3, brine, dried over Na2SO4, filtered and solvent was evaporated. Flash column chromatography (silica, CH2Cl2MeOH 5% to 10%) yielded 111 as off-white solid (40mg, 15%). 1H NMR (400 MHz, DMSO @ 8O0C) δ 11.93 (bs, IH), 10.09 (s, IH), 9.10 (bs, IH), 8.12 (s, IH), 7.56 (m, IH), 7.32 (m, 2H), 6.85 (m, IH), 6.52 (s, IH), 4.18 (m, IH), 3.66 (bs, 2H), 3.39 (m, IH), 3.27 (s, 3H), 1.13 (s, 3H). calcd for (C18H2iFN8O2) 400, m/z 401 [M+H]+.
Example 112:
Figure imgf000132_0002
[0267] To compound 85 (342mg, 2.28mMol) in 2OmL of THF at room temperature was added 99 (535mg, 2.28mMol) and DIPEA (436μL, 323mg, 2.5OmMoI) in 15mL of THF. Reaction mixture was stirred for 4 hours at room temperature. Added 5OmL of H2O, extracted with EtOAc. Organic fractions were combined, washed with sat. NaHCO3, brine, dried over Na2SO4, filtered and solvent was evaporated. Flash column chromatography (silica, CH2Cl2MeOH 20%) yielded 112 as yellow solid (400mg, 51%). 1H NMR (400 MHz, DMSO @ 8O0C) δ 12.25 (bs, IH), 10.68 (s, IH), 10.21 (s, IH), 8.55 (s, IH), 7.56 (m, IH), 7.33 (m, 2H), 6.85 (m, IH), 6.46 (s, IH), 3.74 (s, 2H). calcd for (C14HnClFN7O) 347, m/z 348 [M+H]+.
Example 113:
Figure imgf000133_0001
[0268] To compound 112 (lOOmg, 0.29mMol) in 5mL of THF at room temperature was added DIPEA (55μL, 41mg, 0.32mMol) followed by aniline (26μL, 27mg, 0.29mMol). Reaction mixture was stirred overnight at 60° C. Added 3OmL of EtOAc, washed with sat. NaHCO3, brine, dried over Na2SO4, filtered and solvent was evaporated. Flash column chromatography (silica, CH2Cl2/Me0H 5% to 20%) yielded 113 as light yellow solid (15mg, 13%). 1H NMR (400 MHz, DMSO @ 8O0C) δ 12.03 (bs, IH), 10.10 (s, IH), 9.43 (bs, 2H), 8.31 (s, IH), 7.72 (m, 2H), 7.57 (m, IH), 7.34 (m, 4H), 6.97 (bs, IH), 6.86 (m, IH), 6.49 (bs, IH), 3.70 (s, 2H). calcd for (C20Hi7FN8O) 404, m/z 405 [M+H]+.
Examplel l4
[0269] This example illustrated Src Kinase Assays. Brifely, in a final reaction volume of 25 μL, c-SRC (h) (5-10 mU) is incubated with 8 mM MOPS pH 7.0, 0.2 mM EDTA, 250 μM KVEKIGEGTYGVVYK (Cdc2 peptide), 10 mM MgAcetate and [g-33P-ATP] (specific activity approx. 500 cpm/pmol, concentration as required). The reaction is initiated by the addition of the MgATP mix. After incubation for 40 minutes at room temperature, the reaction is stopped by the addition of 5 μL of a 3% phosphoric acid solution. 10 μL of the reaction is then spotted onto a P30 filtermat and washed three times for 5 minutes in 75 mM phosphoric acid and once in methanol prior to drying and scintillation counting.
Table 1 shows representative data for the inhibition of Src kinase by the compounds of this invention.
Table 1
Figure imgf000133_0002
Figure imgf000134_0001
Figure imgf000135_0001
[0270] All references, including publications, patent applications, and patents, cited herein are hereby incorporated by reference to the same extent as if each reference were individually and specifically indicated to be incorporated by reference and were set forth in its entirety herein.
[0271] The use of the terms "a" and "an" and "the" and similar referents in the context of describing the invention (especially in the context of the following claims) are to be construed to cover both the singular and the plural, unless otherwise indicated herein or clearly contradicted by context. The terms "comprising," "having," "including," and "containing" are to be construed as open-ended terms (i.e., meaning "including, but not limited to,") unless otherwise noted. Recitation of ranges of values herein are merely intended to serve as a shorthand method of referring individually to each separate value falling within the range, unless otherwise indicated herein, and each separate value is incorporated into the specification as if it were individually recited herein. All methods described herein can be performed in any suitable order unless otherwise indicated herein or otherwise clearly contradicted by context. The use of any and all examples, or exemplary language (e.g., "such as") provided herein, is intended merely to better illuminate the invention and does not pose a limitation on the scope of the invention unless otherwise claimed. No language in the specification should be construed as indicating any non-claimed element as essential to the practice of the invention.
[0272] Preferred embodiments of this invention are described herein, including the best mode known to the inventors for carrying out the invention. Variations of those preferred embodiments may become apparent to those of ordinary skill in the art upon reading the foregoing description. The inventors expect skilled artisans to employ such variations as appropriate, and the inventors intend for the invention to be practiced otherwise than as specifically described herein. Accordingly, this invention includes all modifications and equivalents of the subject matter recited in the claims appended hereto as permitted by applicable law. Moreover, any combination of the above-described elements in all possible variations thereof is encompassed by the invention unless otherwise indicated herein or otherwise clearly contradicted by context. 

Claims

What is claimed is:
1. A compound of the formula
Figure imgf000137_0001
or a pharmaceutically acceptable salt thereof, wherein:
A, B, W is selected from S, O, NR4, CR4 or L-R3;
R4 is independently selected from hydrogen or an optionally substituted Cj-4 aliphatic group.
Ri represents hydrogen, halogen, hydroxy, amino, cyano, alkyl, cycloalkyl, alkenyl, alkynyl, alkylthio, aryl, arylalkyl, heterocyclic, heteroaryl, heterocycloalkyl, alkylsulfonyl, alkoxycarbonyl and alkylcarbonyl. R2 is selected from:
(i) amino, alkyl amino, aryl amino, heteroaryl amino; (ii) Ci-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl; (iii) heterocyclic, heteroaryl; and (iv) groups of the formula (Ia):
Figure imgf000137_0002
wherein:
R5 represents hydrogen, Ci-C4 alkyl, oxo;
X is CH, when R6 is hydrogen; or X-R6 is O; or X is N, R6 represents groups of hydrogen, Cj-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-Ci0 aryl or heteroaryl, (C3-C7cycloalkyl)Ci-C4alkyl, Ci- C6 haloalkyl, Ci-C6 alkoxy, C,- C6 alkylthio, C2-C6 alkanoyl, Ci- C6 alkoxycarbonyl, C2- C6 alkanoyloxy, mono- and di-(C3-C8 cycloalkyl)aminoCo-C4alkyl, (4- to 7-membered heterocycle)Co-C4alkyl, Ci-C6 alkylsulfonyl, mono- and CU-(C1- C6 alkyl) sulfonamido, and mono- and di-(CpC6alkyl)aminocarbonyl, each of which is substituted with from 0 to 4 substituents independently chosen from halogen, hydroxy, cyano, amino, -COOH and oxo;
L represents O, S, SO, CO, SO2, CO2, NR4, (CH2V, m = 0"3 > CONR4, NR4CO, NR4SO2, SO2NR4, NR4CO2, NR4COR4, NR4SO2NR4, NR4NR45OCONR4, C(R4)ZCONR4, NR4COC(R4), C(IU)2SO, C(IU)2SO2, C(R4^SO2NR4, C(IU)2NIU, C(R^2NR4CO, C(R4)ZNR4CO2, C(IU)=NNR4, C(R4)=N-0, C(IU)2NR4NR4, C(R4)ZNR4SO2NR4, C(R4)ZNR4CONR4, O(CH2)P, S(CH2)P, p=l-3,or (CH2)qO, or (CH2)qS, q = 1-3;
R3 is selected from:
(i) Ci-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl;
(ii) heterocyclic,
(iii) Ar,
Ar represents heteroaryl or aryl, each of which is substituted with from O to 4 substituents independently chosen from:
(1) halogen, hydroxy, amino, cyano, -COOH, -SOzNH2, oxo, nitro and alkoxycarbonyl; and
(2) Ci-C6 alkyl, Ci-C6alkoxy, C3-Ci0 cycloalkyl,C2-C6 alkenyl, C2-C6 alkynyl, C2-C6 alkanoyl, Cj-C6 haloalkyl, Ci-C6 haloalkoxy, mono- and di- (Ci- C6alkyl)amino, CpC6 alkylsulfonyl, mono- and di-(Ci-C6alkyl) sulfonamido and mono- and di-(Ci-C6alkyl)aminocarbonyl; phenylC0-C4alkyl and (4- to 7-membered heterocycle) — C0-C4alkyl, each of which is substituted with from O to 4 secondary substituents independently chosen from halogen, hydroxy, cyano, oxo, imino, Ci- C4alkyl, Ci-C4alkoxy and Ci-C4haloalkyl;
K is selected from: i) absence; ii) O, S, SO, SO2; iii) (CH2)m, m = 0-3, O(CH2)P, p=l-3, (CH2)qO, q = 1-3, iv) NR7;and
R7 represents hydrogen, alkyl, cycloalkyl, alkenyl, alkynyl, alkylthio, aryl, arylalkyl.
2. A process for making compound of claim 1 or its pharmaceutically acceptable salts, hydrates, solvates, crystal forms salts and individual diastereomers thereof.
3. A pharmaceutical composition comprising at least one compound of claim 1 or its pharmaceutically acceptable salts, hydrates, solvates, crystal forms salts and individual diastereomers thereof, and a pharmaceutically acceptable carrier.
4. A compound selected from the group consisting of:
Figure imgf000139_0001
Figure imgf000140_0001
Figure imgf000141_0001
Figure imgf000142_0001
Figure imgf000143_0001
Figure imgf000144_0001
Figure imgf000145_0001
Figure imgf000146_0001
Figure imgf000147_0001
Figure imgf000148_0001
Figure imgf000149_0001
Figure imgf000150_0001
Figure imgf000151_0001
Figure imgf000152_0001
Figure imgf000153_0001
Figure imgf000154_0001
Figure imgf000155_0001
Figure imgf000156_0001
Figure imgf000157_0001
Figure imgf000158_0001
Figure imgf000159_0001
Figure imgf000160_0001
Figure imgf000161_0001
Figure imgf000162_0001
Figure imgf000163_0001
Figure imgf000164_0001
Figure imgf000165_0001
Figure imgf000166_0001
Figure imgf000167_0001
Figure imgf000168_0001
Figure imgf000169_0001
Figure imgf000170_0001
Figure imgf000171_0001
Figure imgf000172_0001
Figure imgf000173_0001
Figure imgf000174_0001
Figure imgf000175_0001
5. The composition according to claim 3, further comprising an additional therapeutic agent.
6. A method for treating a disease or condition in a mammal characterized by undesired cellular proliferation or hyperproliferation comprising identifying the mammal afflicted with said disease or condition and administering to said afflicted mammal a composition comprising the compound of claim 1.
7. The method of claim 6, wherein the disease or condition is cancer, stroke, congestive heart failure, an ischemia or reperfusion injury, arthritis or other arthropathy, retinopathy or vitreoretinal disease, macular degeneration, autoimmune disease, vascular leakage syndrome, inflammatory disease, edema, transplant rejection, burn, or acute or adult respiratory distress syndrome.
8. The method of claim 7, wherein the disease or condition is cancer.
9. The method of claim 7, wherein the disease or condition is autoimmune disease.
10. The method of claim 7, wherein the disease or condition is stroke.
11. The method of claim 7, wherein the disease or condition is arthritis.
12. The method of claim 7, wherein the disease or condition is inflammatory disease.
13. The method of claim 7, wherein the disease or condition is associated with a kinase.
14. The method according to claim 7, wherein said method further comprises administering an additional therapeutic agent.
15. The method according to claim 7, wherein said additional therapeutic agent is a chemotherapeutic agent.
16. The method of claim 13, wherein the kinase is a tyrosine kinase.
17. The method of claim 13, wherein the kinase is a serine kinase or a threonine kinase.
18. The method of claim 16, wherein the kinase is a Src family kinase.
19. The method of claim 16, wherein the kinase is a AbI family kinase.
20. The method of claim 8, wherein said cancer is selected from the group consisting of cancers of the liver and biliary tree, intestinal cancers, colorectal cancer, ovarian cancer, small cell and non-small cell lung cancer, breast cancer, sarcomas, fibrosarcoma, malignant fibrous histiocytoma, embryonal rhabdomysocarcoma, leiomysosarcoma, neuro-fibrosarcoma, osteosarcoma, synovial sarcoma, liposarcoma, alveolar soft part sarcoma, neoplasms of the central nervous systems, brain cancer, and lymphomas, including Hodgkin's lymphoma, lymphoplasmacytoid lymphoma, follicular lymphoma, mucosa-associated lymphoid tissue lymphoma, mantle cell lymphoma, B-lineage large cell lymphoma, Burkitt's lymphoma, and T-cell anaplastic large cell lymphoma, and combinations thereof.
21. A compound of the formula
Figure imgf000177_0002
or a pharmaceutically acceptable salt thereof, wherein:
Y is selected from -OR4, -NR4R5, and -Q-R3;
Q is selected from cycloalkyl and heterocycloalkyl, each of which is optionally substituted with CpC6 alkyl or oxo;
R3 is selected from H, Ci-C6 alkyl, Q-C6 alkyl-R6, aryl, and heteroaryl, each of which is optionally substituted with Ci-C6 alkyl, halo, trifluoromethyl, or oxo;
R4 and R5 are each independently selected from H, Ci-C6 alkyl-R6, aryl, and heteroaryl;
R6 is selected from hydroxy, cyano, aryl, heteroaryl, cycloalkyl, heterocycloalkyl, - NH2, mono(Ci-C6)alkylamino, di(Ci-C6)alkylamino, and C]-C6 alkoxy;
X is -NH-Ar '-R1;
Ar1 is selected from aryl and heteroaryl, each of which is optionally substituted with Cj-C6 alkyl or halo;
R1 is selected from -(CH2)nC(O)NHW, -CH2C(O)NHAr1 , and -NH2; n = 0, l ;
W is selected from Ci-C6 alkyl, cycloalkyl, and -(CH2)Ar1; Z is selected from H, Ci-C6 alkyl, aryl, and heteroaryl.
22. A compound of the formula
Figure imgf000177_0001
or a pharmaceutically acceptable salt thereof, wherein: Y is selected from -OR4, -NR4R5, and -Q-R3; Q is selected from morpholinyl, piperazinyl and piperidinyl; R3 is selected from H, Cj-C6 alkyl, hydroxy(Ci-C6)alkyl, cyano(Ci-C6)alkyl, pyridinylmethyl, pyridinyl, phenyl, trifluoromethylphenyl, and oxo;
R4 and R5 are each independently selected from H, C1-C6 alkyl-R6, and phenyl;
R6 is selected from hydroxy, morpholinyl, di(Ci-C6)alkylamino, imidazolyl, and C1- C6 alkoxy;
X is -NH- Ar '-R1;
Ar1 is selected from thiazolyl, oxazolyl, oxadiazolyl, methyl-imidazolyl, pyrazolyl;
R1 is selected from -(CH2)nC(0)NHW and -NH2; n = 0, l;
W is selected from Cj-C6 alkyl and -(CH2)nPh optionally substituted with Ci-C6 alkyl or halo;
Z is selected from H, C]-C6 alkyl, and phenyl.
23. A compound of the formula
Figure imgf000178_0001
or a pharmaceutically acceptable salt thereof, wherein:
Y is selected from -OR4, -NR4R5, and -Q-R3;
Q is selected from morpholinyl, piperazinyl and piperidinyl;
R3 is selected from H, Ci-C6 alkyl, hydroxy(Ci-C6)alkyl, cyano(Ci-C6)alkyl, pyridinylmethyl, pyridinyl, phenyl, trifluoromethylphenyl, and oxo;
R4 and R5 are each independently selected from H, Cj-C6 alkyl-R6, and phenyl;
R6 is selected from hydroxy, morpholinyl, di(Ci-C6)alkylamino, imidazolyl, and C1- C6 alkoxy;
X is -NH- Ar '-R1;
Ar1 is selected from thiazolyl, oxazolyl, oxadiazolyl, methyl-imidazolyl, pyrazolyl;
R1 is selected from -(CH2)nC(O)NHW, -CH2C(O)NHAr2, and -NH2; n = O, l;
W is selected from Ci-C6 alkyl, cycloalkyl, and -(CH2)Ar2 ;
Ar2 is phenyl, optionally substituted with CpC6 alkyl or halo;
Z is selected from H, Ci-C6 alkyl, and phenyl.
24. A process for making compound of claim 21 or its pharmaceutically acceptable salts, hydrates, solvates, crystal forms salts and individual diastereomers thereof.
25. A pharmaceutical composition comprising at least one compound of claim 21 or its pharmaceutically acceptable salts, hydrates, solvates, crystal forms salts and individual diastereomers thereof, and a pharmaceutically acceptable carrier.
26. A process for making compound of claim 22 or its pharmaceutically acceptable salts, hydrates, solvates, crystal forms salts and individual diastereomers thereof.
27. A pharmaceutical composition comprising at least one compound of claim 22 or its pharmaceutically acceptable salts, hydrates, solvates, crystal forms salts and individual diastereomers thereof, and a pharmaceutically acceptable carrier.
28. A process for making compound of claim 23 or its pharmaceutically acceptable salts, hydrates, solvates, crystal forms salts and individual diastereomers thereof.
29. A pharmaceutical composition comprising at least one compound of claim 23 or its pharmaceutically acceptable salts, hydrates, solvates, crystal forms salts and individual diastereomers thereof, and a pharmaceutically acceptable carrier.
PCT/US2010/037570 2009-06-08 2010-06-07 Triazine derivatives and their therapeutical applications WO2010144338A1 (en)

Priority Applications (10)

Application Number Priority Date Filing Date Title
JP2012515010A JP2012529511A (en) 2009-06-08 2010-06-07 Triazine derivatives and their therapeutic applications
CN201080034890.8A CN102573485B (en) 2009-06-08 2010-06-07 Triazine derivatives and their therapeutical applications
CA2764785A CA2764785C (en) 2009-06-08 2010-06-07 Triazine derivatives and their therapeutical applications
BRPI1010881A BRPI1010881A2 (en) 2009-06-08 2010-06-07 triazine derivatives and their therapeutic applications.
AU2010259002A AU2010259002B2 (en) 2009-06-08 2010-06-07 Triazine derivatives and their therapeutical applications
EP10786616A EP2440050A4 (en) 2009-06-08 2010-06-07 Triazine derivatives and their therapeutical applications
CN201410571301.XA CN105175409A (en) 2009-06-08 2010-06-07 Triazine Derivatives and their Therapeutical Applications
KR1020127000593A KR101460095B1 (en) 2009-06-08 2010-06-07 Triazine derivatives and their therapeutical applications
US13/376,964 US20120238576A1 (en) 2009-06-08 2010-06-07 Triazine Derivatives and their Therapeutical Applications
IL216825A IL216825A (en) 2009-06-08 2011-12-07 Triazine derivatives, processes for producing the same, compositions comprising the same and uses thereof

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US18505209P 2009-06-08 2009-06-08
US61/185,052 2009-06-08

Publications (1)

Publication Number Publication Date
WO2010144338A1 true WO2010144338A1 (en) 2010-12-16

Family

ID=43309171

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2010/037570 WO2010144338A1 (en) 2009-06-08 2010-06-07 Triazine derivatives and their therapeutical applications

Country Status (10)

Country Link
US (1) US20120238576A1 (en)
EP (1) EP2440050A4 (en)
JP (1) JP2012529511A (en)
KR (1) KR101460095B1 (en)
CN (2) CN105175409A (en)
AU (2) AU2010259002B2 (en)
BR (1) BRPI1010881A2 (en)
CA (1) CA2764785C (en)
IL (1) IL216825A (en)
WO (1) WO2010144338A1 (en)

Cited By (49)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012169934A1 (en) * 2011-06-08 2012-12-13 Общество С Ограниченной Ответственностью "Асинэкс Медхим" Novel kinase inhibitors
CN102827156A (en) * 2012-09-11 2012-12-19 湖南欧亚生物有限公司 Novel industrial synthetic method of dasatinib
US20130131035A1 (en) * 2010-02-17 2013-05-23 Amgen Inc. Carboxamides as inhibitors of voltage-gated sodium channels
WO2013102431A1 (en) * 2012-01-06 2013-07-11 Agios Pharmaceuticals, Inc. Therapeutically active compounds and their methods of use
WO2014102759A2 (en) 2012-12-31 2014-07-03 Ranbaxy Laboratories Limited Process for the preparation of dasatinib and its intermediates
WO2014107535A1 (en) 2013-01-04 2014-07-10 Rempex Pharmaceuticals, Inc. Boronic acid derivatives and therapeutic uses thereof
US8883438B2 (en) 2009-10-21 2014-11-11 Agios Pharmaceuticals, Inc. Method for diagnosing cell proliferation disorders having a neoactive mutation at residue 97 of isocitrate dehydrogenase 1
WO2015003640A1 (en) * 2013-07-11 2015-01-15 Agios Pharmaceuticals, Inc. Therapeutically active compounds and their methods of use
WO2015018060A1 (en) * 2013-08-09 2015-02-12 Agios Pharmaceuticals, Inc. Crystalline forms of therapeutically active compounds and use thereof
US9012491B2 (en) 2011-08-31 2015-04-21 Rempex Pharmaceuticals, Inc. Heterocyclic boronic acid ester derivatives and therapeutic uses thereof
US9073875B2 (en) 2012-11-20 2015-07-07 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of indoleamine 2,3-dioxygenase
US9101638B2 (en) 2013-01-04 2015-08-11 Rempex Pharmaceuticals, Inc. Boronic acid derivatives and therapeutic uses thereof
US9132140B2 (en) 2013-01-04 2015-09-15 Rempex Pharmaceuticals, Inc. Boronic acid derivatives and therapeutic uses thereof
US9156858B2 (en) 2012-05-23 2015-10-13 Rempex Pharmaceuticals, Inc. Boronic acid derivatives and therapeutic uses thereof
US9241947B2 (en) 2013-01-04 2016-01-26 Rempex Pharmaceuticals, Inc. Boronic acid derivatives and therapeutic uses thereof
US9296763B2 (en) 2010-08-10 2016-03-29 Rempex Pharmaceuticals, Inc. Cyclic boronic acid ester derivatives and therapeutic uses thereof
US9434979B2 (en) 2009-10-21 2016-09-06 Shin-San Michael Su Methods and compositions for cell-proliferation-related disorders
US9474779B2 (en) 2012-01-19 2016-10-25 Agios Pharmaceuticals, Inc. Therapeutically active compositions and their methods of use
US9579324B2 (en) 2013-07-11 2017-02-28 Agios Pharmaceuticals, Inc Therapeutically active compounds and their methods of use
US9662327B2 (en) 2011-06-17 2017-05-30 Agios Pharmaceuticals, Inc Phenyl and pyridinyl substituted piperidines and piperazines as inhibitors of IDH1 mutants and their use in treating cancer
US9687497B1 (en) 2014-05-05 2017-06-27 Rempex Pharmaceuticals, Inc. Salts and polymorphs of cyclic boronic acid ester derivatives and therapeutic uses thereof
US9738625B2 (en) 2013-08-02 2017-08-22 Agios Pharmaceuticals, Inc. Therapeutically active compounds and their methods of use
US9751863B2 (en) 2015-08-05 2017-09-05 Agios Pharmaceuticals, Inc. Therapeutically active compounds and their methods of use
US9856279B2 (en) 2011-06-17 2018-01-02 Agios Pharmaceuticals, Inc. Therapeutically active compositions and their methods of use
US9963467B2 (en) 2014-05-19 2018-05-08 Rempex Pharmaceuticals, Inc. Boronic acid derivatives and therapeutic uses thereof
US9968595B2 (en) 2014-03-14 2018-05-15 Agios Pharmaceuticals, Inc. Pharmaceutical compositions of therapeutically active compounds
US9980961B2 (en) 2011-05-03 2018-05-29 Agios Pharmaceuticals, Inc. Pyruvate kinase activators for use in therapy
US10017495B2 (en) 2013-07-11 2018-07-10 Agios Pharmaceuticals, Inc. Therapeutically active compounds and their methods of use
US10029987B2 (en) 2009-06-29 2018-07-24 Agios Pharmaceuticals, Inc. Therapeutic compounds and compositions
US10202339B2 (en) 2012-10-15 2019-02-12 Agios Pharmaceuticals, Inc. Therapeutic compounds and compositions
US10206937B2 (en) 2014-07-01 2019-02-19 Qpex Biopharma, Inc. Boronic acid derivatives and therapeutic uses thereof
US10294249B2 (en) 2016-06-30 2019-05-21 Qpex Biopharma, Inc. Boronic acid derivatives and therapeutic uses thereof
US10376510B2 (en) 2013-07-11 2019-08-13 Agios Pharmaceuticals, Inc. 2,4- or 4,6-diaminopyrimidine compounds as IDH2 mutants inhibitors for the treatment of cancer
US10385074B2 (en) 2014-05-05 2019-08-20 Rempex Pharmaceuticals, Inc. Synthesis of boronate salts and uses thereof
US10561675B2 (en) 2012-06-06 2020-02-18 Rempex Pharmaceuticals, Inc. Cyclic boronic acid ester derivatives and therapeutic uses thereof
US10610125B2 (en) 2009-03-13 2020-04-07 Agios Pharmaceuticals, Inc. Methods and compositions for cell-proliferation-related disorders
US10618918B2 (en) 2015-03-17 2020-04-14 Qpex Biopharma, Inc. Substituted boronic acids as antimicrobials
EP3555092A4 (en) * 2016-12-13 2020-05-13 Princeton Drug Discovery Inc Protein kinase inhibitors
US10653710B2 (en) 2015-10-15 2020-05-19 Agios Pharmaceuticals, Inc. Combination therapy for treating malignancies
US10662205B2 (en) 2014-11-18 2020-05-26 Qpex Biopharma, Inc. Cyclic boronic acid ester derivatives and therapeutic uses thereof
US10689414B2 (en) 2013-07-25 2020-06-23 Agios Pharmaceuticals, Inc. Therapeutically active compounds and their methods of use
US10980788B2 (en) 2018-06-08 2021-04-20 Agios Pharmaceuticals, Inc. Therapy for treating malignancies
US11234976B2 (en) 2015-06-11 2022-02-01 Agios Pharmaceuticals, Inc. Methods of using pyruvate kinase activators
US11286270B2 (en) 2017-10-11 2022-03-29 Qpex Biopharma, Inc. Boronic acid derivatives and synthesis thereof
US11419859B2 (en) 2015-10-15 2022-08-23 Servier Pharmaceuticals Llc Combination therapy for treating malignancies
US11497743B2 (en) 2018-05-16 2022-11-15 Forma Therapeutics, Inc. Treating patients harboring an isocitrate dehydrogenase 1 (IDH-1) mutation
US11498913B2 (en) 2014-09-19 2022-11-15 Forma Therapeutics, Inc. Pyridin-2(1H)-one quinolinone derivatives as mutant-isocitrate dehydrogenase inhibitors
US11576906B2 (en) 2018-05-16 2023-02-14 Forma Therapeutics, Inc. Inhibiting mutant IDH-1
US12016868B2 (en) 2018-04-20 2024-06-25 Qpex Biopharma, Inc. Boronic acid derivatives and therapeutic uses thereof

Families Citing this family (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105517996B (en) * 2013-07-11 2019-03-26 安吉奥斯医药品有限公司 Therapeutical active compound and its application method
CN111087353B (en) * 2013-07-11 2023-10-24 法国施维雅药厂 Therapeutically active compounds and methods of use thereof
BR112016000489A8 (en) 2013-07-11 2020-01-07 Agios Pharmaceuticals Inc compound, pharmaceutical composition comprising it, use of the composition and methods for making compounds
CN103483289B (en) * 2013-09-06 2016-01-27 浙江科源化工有限公司 A kind of synthetic method of 2-amino-N-(the chloro-6-aminomethyl phenyl of 2-) thiazole-5-methane amide
MX2016011865A (en) * 2014-03-14 2017-04-13 Agios Pharmaceuticals Inc Pharmaceutical compositions of therapeutically active compounds.
CN104163818A (en) * 2014-07-15 2014-11-26 南昌大学 2-amino oxazole compound and preparation method and application thereof
AU2016263564B2 (en) 2015-05-20 2019-12-05 Amgen Inc. Triazole agonists of the APJ receptor
CN105541823B (en) * 2016-02-03 2018-03-06 三峡大学 A kind of compound in triazine class of Han oxazolines ring, preparation method and its application on cancer therapy drug
JP6892452B2 (en) * 2016-02-11 2021-06-23 バイエル・クロップサイエンス・アクチェンゲゼルシャフト Substitution imidazolyl-carboxamides as pest control agents
EP3452466B1 (en) 2016-05-03 2020-08-12 Amgen Inc. Heterocyclic triazole compounds as agonists of the apj receptor
EP3541810B1 (en) 2016-11-16 2020-12-23 Amgen Inc. Triazole phenyl compounds as agonists of the apj receptor
US11191762B2 (en) 2016-11-16 2021-12-07 Amgen Inc. Alkyl substituted triazole compounds as agonists of the APJ Receptor
US10689367B2 (en) 2016-11-16 2020-06-23 Amgen Inc. Triazole pyridyl compounds as agonists of the APJ receptor
EP3541804A1 (en) 2016-11-16 2019-09-25 Amgen Inc. Cycloalkyl substituted triazole compounds as agonists of the apj receptor
US10736883B2 (en) 2016-11-16 2020-08-11 Amgen Inc. Triazole furan compounds as agonists of the APJ receptor
US11046680B1 (en) 2016-11-16 2021-06-29 Amgen Inc. Heteroaryl-substituted triazoles as APJ receptor agonists
CN106883235B (en) * 2016-12-29 2019-04-30 天津国际生物医药联合研究院 The preparation and application of furodiazole compound
US10800760B2 (en) 2017-05-31 2020-10-13 Nantbio, Inc. Trk inhibition
US10738033B2 (en) 2017-05-31 2020-08-11 Nantbio, Inc. Trk inhibition
US11149040B2 (en) 2017-11-03 2021-10-19 Amgen Inc. Fused triazole agonists of the APJ receptor
EP3788037A1 (en) 2018-05-01 2021-03-10 Amgen Inc. Substituted pyrimidinones as agonists of the apj receptor
CN111574473A (en) * 2020-06-04 2020-08-25 宁夏农林科学院农业资源与环境研究所(宁夏土壤与植物营养重点实验室) Method for synthesizing aminothiazole compound

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20080176853A1 (en) * 2006-12-15 2008-07-24 Abraxis Bioscience, Inc. Triazine derivatives and their therapeutical applications

Family Cites Families (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU770600B2 (en) * 1999-10-07 2004-02-26 Amgen, Inc. Triazine kinase inhibitors
AU2001291013A1 (en) * 2000-09-15 2002-03-26 Vertex Pharmaceuticals Incorporated Pyrazole compounds useful as protein kinase inhibitors
HUP0400908A3 (en) * 2000-12-21 2010-03-29 Vertex Pharma Pyrazole compounds useful as protein kinase inhibitors, their use and pharmaceutical compositions containing them
DE60314603T2 (en) * 2002-03-15 2008-02-28 Vertex Pharmaceuticals Inc., Cambridge COMPOSITIONS NECESSARY AS PROTEIN KINASE INHIBITORS
EP1485380B1 (en) 2002-03-15 2010-05-19 Vertex Pharmaceuticals Incorporated Azolylaminoazines as inhibitors of protein kinases
US7179826B2 (en) * 2002-03-15 2007-02-20 Vertex Pharmaceuticals Incorporated Compositions useful as inhibitors of protein kinases
AR061185A1 (en) * 2006-05-26 2008-08-13 Chugai Pharmaceutical Co Ltd HETEROCICLICAL COMPOUNDS AS INHIBITORS OF HSP90. PHARMACEUTICAL COMPOSITIONS.
TW200906818A (en) * 2007-07-31 2009-02-16 Astrazeneca Ab Chemical compounds
CA2723424A1 (en) * 2008-05-19 2009-11-26 Burnham Institute For Medical Research Intestinal alkaline phosphatase modulators and uses thereof
RU2010154502A (en) * 2008-06-11 2012-07-20 Астразенека Аб (Se) TRICYCLIC 2,4-DIAMINO-L, 3,5-TRIAZINE DERIVATIVES USED FOR THE TREATMENT OF MALIGNANT NEW FORMATION AND MYELOPROLIFERAL DISORDERS
WO2010144359A1 (en) * 2009-06-08 2010-12-16 Abraxis Bioscience, Llc Triazine derivatives and their therapeutical applications
AU2010258825B2 (en) * 2009-06-09 2014-08-21 Nantbio, Inc. Ureidophenyl substituted triazine derivatives and their therapeutical applications
KR20120026612A (en) * 2009-06-09 2012-03-19 아브락시스 바이오사이언스, 엘엘씨 Benzyl substituted triazine derivatives and their therapeutical applications
CA2765030C (en) * 2009-06-09 2015-10-27 California Capital Equity, Llc Triazine derivatives and their therapeutical applications

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20080176853A1 (en) * 2006-12-15 2008-07-24 Abraxis Bioscience, Inc. Triazine derivatives and their therapeutical applications

Cited By (106)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10610125B2 (en) 2009-03-13 2020-04-07 Agios Pharmaceuticals, Inc. Methods and compositions for cell-proliferation-related disorders
USRE49582E1 (en) 2009-06-29 2023-07-18 Agios Pharmaceuticals, Inc. Therapeutic compounds and compositions
US10988448B2 (en) 2009-06-29 2021-04-27 Agios Pharmaceuticals, Inc. Therapeutic compounds and compositions
US11866411B2 (en) 2009-06-29 2024-01-09 Agios Pharmaceutical, Inc. Therapeutic compounds and compositions
US10029987B2 (en) 2009-06-29 2018-07-24 Agios Pharmaceuticals, Inc. Therapeutic compounds and compositions
US9434979B2 (en) 2009-10-21 2016-09-06 Shin-San Michael Su Methods and compositions for cell-proliferation-related disorders
US9982309B2 (en) 2009-10-21 2018-05-29 Agios Pharmaceuticals, Inc. Method for treating cell proliferation related disorders
US10711314B2 (en) 2009-10-21 2020-07-14 Agios Pharmaceuticals, Inc. Methods for diagnosing IDH-mutant cell proliferation disorders
US8883438B2 (en) 2009-10-21 2014-11-11 Agios Pharmaceuticals, Inc. Method for diagnosing cell proliferation disorders having a neoactive mutation at residue 97 of isocitrate dehydrogenase 1
US9334269B2 (en) * 2010-02-17 2016-05-10 Amgen Inc. Carboxamides as inhibitors of voltage-gated sodium channels
US20130131035A1 (en) * 2010-02-17 2013-05-23 Amgen Inc. Carboxamides as inhibitors of voltage-gated sodium channels
US10172874B2 (en) 2010-08-10 2019-01-08 Rempex Pharmaceuticals, Inc. Pharmaceutical compositions comprising cyclic boronic acid ester derivatives
US11684629B2 (en) 2010-08-10 2023-06-27 Melinta Subsidiary Corp. Therapeutic uses of pharmaceutical compositions comprising cyclic boronic acid ester derivatives
US10004758B2 (en) 2010-08-10 2018-06-26 Rempex Pharmaceuticals, Inc. Cyclic boronic acid ester derivatives and methods of making the same
US11090319B2 (en) 2010-08-10 2021-08-17 Melinta Subsidiary Corp. Therapeutic uses of pharmaceutical compositions comprising cyclic boronic acid ester derivatives
US9296763B2 (en) 2010-08-10 2016-03-29 Rempex Pharmaceuticals, Inc. Cyclic boronic acid ester derivatives and therapeutic uses thereof
US10639318B2 (en) 2010-08-10 2020-05-05 Rempex Pharmaceuticals, Inc. Therapeutic uses of pharmaceutical compositions comprising cyclic boronic acid ester derivatives
US10183034B2 (en) 2010-08-10 2019-01-22 Rempex Pharmaceuticals, Inc. Therapeutic uses of pharmaceutical compositions comprising cyclic boronic acid ester derivatives
US9694025B2 (en) 2010-08-10 2017-07-04 Rempex Pharmaceuticals, Inc. Cyclic boronic acid ester derivatives and therapeutic uses thereof
US11793806B2 (en) 2011-05-03 2023-10-24 Agios Pharmaceuticals, Inc. Pyruvate kinase activators for use in therapy
US10632114B2 (en) 2011-05-03 2020-04-28 Agios Pharmaceuticals, Inc. Pyruvate kinase activators for use in therapy
US9980961B2 (en) 2011-05-03 2018-05-29 Agios Pharmaceuticals, Inc. Pyruvate kinase activators for use in therapy
WO2012169934A1 (en) * 2011-06-08 2012-12-13 Общество С Ограниченной Ответственностью "Асинэкс Медхим" Novel kinase inhibitors
US9662327B2 (en) 2011-06-17 2017-05-30 Agios Pharmaceuticals, Inc Phenyl and pyridinyl substituted piperidines and piperazines as inhibitors of IDH1 mutants and their use in treating cancer
US9856279B2 (en) 2011-06-17 2018-01-02 Agios Pharmaceuticals, Inc. Therapeutically active compositions and their methods of use
US9012491B2 (en) 2011-08-31 2015-04-21 Rempex Pharmaceuticals, Inc. Heterocyclic boronic acid ester derivatives and therapeutic uses thereof
EP3984997A1 (en) * 2012-01-06 2022-04-20 Les Laboratoires Servier SAS Therapeutically active compounds and their methods of use
CN107417667B (en) * 2012-01-06 2020-04-10 安吉奥斯医药品有限公司 Therapeutically active compounds and methods of use thereof
JP2017075193A (en) * 2012-01-06 2017-04-20 アジオス ファーマシューティカルズ, インコーポレイテッド Therapeutically active compositions and their methods of use
US9732062B2 (en) 2012-01-06 2017-08-15 Agios Pharmaceuticals, Inc. Therapeutically active compounds and their methods of use
CN108912066B (en) * 2012-01-06 2022-06-24 安吉奥斯医药品有限公司 Therapeutically active compounds and methods of use thereof
WO2013102431A1 (en) * 2012-01-06 2013-07-11 Agios Pharmaceuticals, Inc. Therapeutically active compounds and their methods of use
CN107417667A (en) * 2012-01-06 2017-12-01 安吉奥斯医药品有限公司 Therapeutical active compound and its application method
CN104114543B (en) * 2012-01-06 2019-03-15 安吉奥斯医药品有限公司 Therapeutical active compound and its application method
AU2017265096B2 (en) * 2012-01-06 2019-08-08 Les Laboratoires Servier Therapeutically active compounds and their methods of use
EA030187B1 (en) * 2012-01-06 2018-07-31 Аджиос Фармасьютикалз, Инк. Therapeutically active compounds and methods of use thereof
CN108912066A (en) * 2012-01-06 2018-11-30 安吉奥斯医药品有限公司 Therapeutical active compound and its application method
US10294215B2 (en) 2012-01-06 2019-05-21 Agios Pharmaceuticals, Inc. Therapeutically active compounds and their methods of use
US11505538B1 (en) 2012-01-06 2022-11-22 Servier Pharmaceuticals Llc Therapeutically active compounds and their methods of use
EP3406608A1 (en) * 2012-01-06 2018-11-28 Agios Pharmaceuticals, Inc. Therapeutically active compounds and their methods of use
CN104114543A (en) * 2012-01-06 2014-10-22 安吉奥斯医药品有限公司 Therapeutically active compounds and their methods of use
AU2017265096B9 (en) * 2012-01-06 2019-09-05 Les Laboratoires Servier Therapeutically active compounds and their methods of use
JP2015503571A (en) * 2012-01-06 2015-02-02 アジオス ファーマシューティカルズ, インコーポレイテッド Therapeutically active compounds and methods of use thereof
US9850277B2 (en) 2012-01-19 2017-12-26 Agios Pharmaceuticals, Inc. Therapeutically active compositions and their methods of use
US10640534B2 (en) 2012-01-19 2020-05-05 Agios Pharmaceuticals, Inc. Therapeutically active compositions and their methods of use
US11667673B2 (en) 2012-01-19 2023-06-06 Servier Pharmaceuticals Llc Therapeutically active compounds and their methods of use
US10717764B2 (en) 2012-01-19 2020-07-21 Agios Pharmaceuticals, Inc. Therapeutically active compounds and their methods of use
US9474779B2 (en) 2012-01-19 2016-10-25 Agios Pharmaceuticals, Inc. Therapeutically active compositions and their methods of use
US9156858B2 (en) 2012-05-23 2015-10-13 Rempex Pharmaceuticals, Inc. Boronic acid derivatives and therapeutic uses thereof
US10561675B2 (en) 2012-06-06 2020-02-18 Rempex Pharmaceuticals, Inc. Cyclic boronic acid ester derivatives and therapeutic uses thereof
US11007206B2 (en) 2012-06-06 2021-05-18 Melinta Subsidiary Corp. Cyclic boronic acid ester derivatives and therapeutic uses thereof
CN102827156A (en) * 2012-09-11 2012-12-19 湖南欧亚生物有限公司 Novel industrial synthetic method of dasatinib
US10202339B2 (en) 2012-10-15 2019-02-12 Agios Pharmaceuticals, Inc. Therapeutic compounds and compositions
US9073875B2 (en) 2012-11-20 2015-07-07 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of indoleamine 2,3-dioxygenase
US9499497B2 (en) 2012-11-20 2016-11-22 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of indoleamine 2,3-dioxygenase
WO2014102759A2 (en) 2012-12-31 2014-07-03 Ranbaxy Laboratories Limited Process for the preparation of dasatinib and its intermediates
US9365526B2 (en) 2012-12-31 2016-06-14 Sun Pharmaceutical Industries Limited Process for the preparation of dasatinib and its intermediates
US9642869B2 (en) 2013-01-04 2017-05-09 Rempex Pharmaceuticals, Inc. Boronic acid derivatives and therapeutic uses thereof
US9101638B2 (en) 2013-01-04 2015-08-11 Rempex Pharmaceuticals, Inc. Boronic acid derivatives and therapeutic uses thereof
US9241947B2 (en) 2013-01-04 2016-01-26 Rempex Pharmaceuticals, Inc. Boronic acid derivatives and therapeutic uses thereof
US9132140B2 (en) 2013-01-04 2015-09-15 Rempex Pharmaceuticals, Inc. Boronic acid derivatives and therapeutic uses thereof
WO2014107535A1 (en) 2013-01-04 2014-07-10 Rempex Pharmaceuticals, Inc. Boronic acid derivatives and therapeutic uses thereof
US10028961B2 (en) 2013-07-11 2018-07-24 Agios Pharmaceuticals, Inc. Therapeutically active compounds and their methods of use
US10017495B2 (en) 2013-07-11 2018-07-10 Agios Pharmaceuticals, Inc. Therapeutically active compounds and their methods of use
US10172864B2 (en) 2013-07-11 2019-01-08 Agios Pharmaceuticals, Inc. Therapeutically active compounds and their methods of use
WO2015003640A1 (en) * 2013-07-11 2015-01-15 Agios Pharmaceuticals, Inc. Therapeutically active compounds and their methods of use
US10376510B2 (en) 2013-07-11 2019-08-13 Agios Pharmaceuticals, Inc. 2,4- or 4,6-diaminopyrimidine compounds as IDH2 mutants inhibitors for the treatment of cancer
EA030199B1 (en) * 2013-07-11 2018-07-31 Аджиос Фармасьютикалз, Инк. Therapeutically active compounds and methods of use thereof
US10946023B2 (en) 2013-07-11 2021-03-16 Agios Pharmaceuticals, Inc. Therapeutically active compounds and their methods of use
US11844758B2 (en) 2013-07-11 2023-12-19 Servier Pharmaceuticals Llc Therapeutically active compounds and their methods of use
US9579324B2 (en) 2013-07-11 2017-02-28 Agios Pharmaceuticals, Inc Therapeutically active compounds and their methods of use
EA030199B9 (en) * 2013-07-11 2021-08-26 Аджиос Фармасьютикалз, Инк. Therapeutically active compounds and methods of use thereof
US11021515B2 (en) 2013-07-25 2021-06-01 Agios Pharmaceuticals, Inc. Therapeutically active compounds and their methods of use
US10689414B2 (en) 2013-07-25 2020-06-23 Agios Pharmaceuticals, Inc. Therapeutically active compounds and their methods of use
US9738625B2 (en) 2013-08-02 2017-08-22 Agios Pharmaceuticals, Inc. Therapeutically active compounds and their methods of use
US10730854B2 (en) 2013-08-02 2020-08-04 Agios Pharmaceuticals, Inc. Therapeutically active compounds and their methods of use
US10093654B2 (en) 2013-08-02 2018-10-09 Agios Pharmaceuticals, Inc. Therapeutically active compounds and their methods of use
WO2015018060A1 (en) * 2013-08-09 2015-02-12 Agios Pharmaceuticals, Inc. Crystalline forms of therapeutically active compounds and use thereof
US11504361B2 (en) 2014-03-14 2022-11-22 Servier Pharmaceuticals Llc Pharmaceutical compositions of therapeutically active compounds
US10799490B2 (en) 2014-03-14 2020-10-13 Agios Pharmaceuticals, Inc. Pharmaceutical compositions of therapeutically active compounds
US9968595B2 (en) 2014-03-14 2018-05-15 Agios Pharmaceuticals, Inc. Pharmaceutical compositions of therapeutically active compounds
US10449184B2 (en) 2014-03-14 2019-10-22 Agios Pharmaceuticals, Inc. Pharmaceutical compositions of therapeutically active compounds
US9687497B1 (en) 2014-05-05 2017-06-27 Rempex Pharmaceuticals, Inc. Salts and polymorphs of cyclic boronic acid ester derivatives and therapeutic uses thereof
US10669292B2 (en) 2014-05-05 2020-06-02 Rempex Pharmaceuticals, Inc. Synthesis of boronate salts and uses thereof
US10385074B2 (en) 2014-05-05 2019-08-20 Rempex Pharmaceuticals, Inc. Synthesis of boronate salts and uses thereof
US9963467B2 (en) 2014-05-19 2018-05-08 Rempex Pharmaceuticals, Inc. Boronic acid derivatives and therapeutic uses thereof
US10206937B2 (en) 2014-07-01 2019-02-19 Qpex Biopharma, Inc. Boronic acid derivatives and therapeutic uses thereof
US11498913B2 (en) 2014-09-19 2022-11-15 Forma Therapeutics, Inc. Pyridin-2(1H)-one quinolinone derivatives as mutant-isocitrate dehydrogenase inhibitors
US10662205B2 (en) 2014-11-18 2020-05-26 Qpex Biopharma, Inc. Cyclic boronic acid ester derivatives and therapeutic uses thereof
US10618918B2 (en) 2015-03-17 2020-04-14 Qpex Biopharma, Inc. Substituted boronic acids as antimicrobials
US11234976B2 (en) 2015-06-11 2022-02-01 Agios Pharmaceuticals, Inc. Methods of using pyruvate kinase activators
US9751863B2 (en) 2015-08-05 2017-09-05 Agios Pharmaceuticals, Inc. Therapeutically active compounds and their methods of use
US11419859B2 (en) 2015-10-15 2022-08-23 Servier Pharmaceuticals Llc Combination therapy for treating malignancies
US10653710B2 (en) 2015-10-15 2020-05-19 Agios Pharmaceuticals, Inc. Combination therapy for treating malignancies
US11999759B2 (en) 2016-06-30 2024-06-04 Qpex Biopharma, Inc. Boronic acid derivatives and therapeutic uses thereof
US10294249B2 (en) 2016-06-30 2019-05-21 Qpex Biopharma, Inc. Boronic acid derivatives and therapeutic uses thereof
US11180512B2 (en) 2016-06-30 2021-11-23 Qpex Biopharma, Inc. Boronic acid derivatives and therapeutic uses thereof
US10570159B2 (en) 2016-06-30 2020-02-25 Qpex Biopharma, Inc. Boronic acid derivatives and therapeutic uses thereof
EP3555092A4 (en) * 2016-12-13 2020-05-13 Princeton Drug Discovery Inc Protein kinase inhibitors
AU2017375949B2 (en) * 2016-12-13 2021-09-09 Princeton Drug Discovery Inc Protein kinase inhibitors
US11236080B2 (en) 2016-12-13 2022-02-01 Princeton Drug Discovery, Inc Protein kinase inhibitors
US11286270B2 (en) 2017-10-11 2022-03-29 Qpex Biopharma, Inc. Boronic acid derivatives and synthesis thereof
US12016868B2 (en) 2018-04-20 2024-06-25 Qpex Biopharma, Inc. Boronic acid derivatives and therapeutic uses thereof
US11576906B2 (en) 2018-05-16 2023-02-14 Forma Therapeutics, Inc. Inhibiting mutant IDH-1
US11497743B2 (en) 2018-05-16 2022-11-15 Forma Therapeutics, Inc. Treating patients harboring an isocitrate dehydrogenase 1 (IDH-1) mutation
US10980788B2 (en) 2018-06-08 2021-04-20 Agios Pharmaceuticals, Inc. Therapy for treating malignancies

Also Published As

Publication number Publication date
KR20120016674A (en) 2012-02-24
BRPI1010881A2 (en) 2016-05-31
EP2440050A4 (en) 2013-04-03
KR101460095B1 (en) 2014-11-10
IL216825A0 (en) 2012-02-29
JP2012529511A (en) 2012-11-22
US20120238576A1 (en) 2012-09-20
AU2010259002A1 (en) 2012-01-12
CA2764785A1 (en) 2010-12-16
EP2440050A1 (en) 2012-04-18
CN102573485B (en) 2014-11-26
IL216825A (en) 2016-10-31
CN105175409A (en) 2015-12-23
AU2010259002B2 (en) 2014-03-20
CA2764785C (en) 2015-10-27
AU2014203330A1 (en) 2014-07-10
CN102573485A (en) 2012-07-11

Similar Documents

Publication Publication Date Title
AU2010259002B2 (en) Triazine derivatives and their therapeutical applications
EP2425840B1 (en) Triazine derivatives and their therapeutical applications
US20130023497A1 (en) Triazine Derivatives and their Therapeutical Applications
AU2010258825B2 (en) Ureidophenyl substituted triazine derivatives and their therapeutical applications
US9409903B2 (en) Benzyl substituted triazine derivatives and their therapeutical applications
AU2010258853B2 (en) Triazine derivatives and their therapeutical applications
EP2440055B1 (en) Styryl-triazine derivatives and their therapeutical applications
AU2014200528B2 (en) Triazine derivatives and their therapeutical applications

Legal Events

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

Ref document number: 201080034890.8

Country of ref document: CN

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

Ref document number: 10786616

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 2764785

Country of ref document: CA

Ref document number: 2010259002

Country of ref document: AU

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2012515010

Country of ref document: JP

WWE Wipo information: entry into national phase

Ref document number: 9909/DELNP/2011

Country of ref document: IN

ENP Entry into the national phase

Ref document number: 20127000593

Country of ref document: KR

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 2010786616

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2010259002

Country of ref document: AU

Date of ref document: 20100607

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 13376964

Country of ref document: US

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: PI1010881

Country of ref document: BR

ENP Entry into the national phase

Ref document number: PI1010881

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20111208