WO2011130728A1 - Synthetic metabolites of fluoro substituted omega-carboxyaryl diphenyl urea for the treatment and prevention diseases and conditions - Google Patents

Synthetic metabolites of fluoro substituted omega-carboxyaryl diphenyl urea for the treatment and prevention diseases and conditions Download PDF

Info

Publication number
WO2011130728A1
WO2011130728A1 PCT/US2011/032835 US2011032835W WO2011130728A1 WO 2011130728 A1 WO2011130728 A1 WO 2011130728A1 US 2011032835 W US2011032835 W US 2011032835W WO 2011130728 A1 WO2011130728 A1 WO 2011130728A1
Authority
WO
WIPO (PCT)
Prior art keywords
cancer
synthetic
acid
compound
kinase
Prior art date
Application number
PCT/US2011/032835
Other languages
French (fr)
Inventor
Scott Wilhelm
Original Assignee
Bayer Healthcare 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 Bayer Healthcare Llc filed Critical Bayer Healthcare Llc
Priority to CN2011800298301A priority Critical patent/CN103079567A/en
Priority to CA2796744A priority patent/CA2796744A1/en
Publication of WO2011130728A1 publication Critical patent/WO2011130728A1/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
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4412Non condensed pyridines; Hydrogenated derivatives thereof having oxo groups directly attached to the heterocyclic ring
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/60Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D213/78Carbon atoms having three bonds to hetero atoms, with at the most one bond to halogen, e.g. ester or nitrile radicals
    • C07D213/81Amides; Imides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/89Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members with hetero atoms directly attached to the ring nitrogen atom
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • This invention relates to novel compounds, pharmaceutical compositions containing such compounds and the use of those compounds or compositions for treating diseases and conditions mediated by abnormal VEGFR, PDGFR, raf, p38, and/or flt-3 kinase signaling, either alone or in combination with anti-cancer agents.
  • Raf kinase a downstream effector of ras, is recognized as a key mediator of these signals from cell surface receptors to the cell nucleus (Lowy, D. R.; Willumsen, B. M. Ann. Rev. Biochem. 1993, 62, 851; Bos, J. L. Cancer Res. 1989, 49, 4682).
  • tumor cells require a functional stroma, a support structure consisting of fibroblast, smooth muscle cells, endothelial cells, extracellular matrix proteins, and soluble factors (Folkman, J., Semin. Oncol. 2002. 29(6 Suppl 16), 15-8).
  • Tumors induce the formation of stromal tissues through the secretion of soluble growth factors such as PDGF and transforming growth factor-beta (TGF-beta), which in turn stimulate the secretion of complimentary factors by host cells such as fibroblast growth factor (FGF), epidermal growth factor (EGF), and vascular endothelial growth factor (VEGF).
  • FGF fibroblast growth factor
  • EGF epidermal growth factor
  • VEGF vascular endothelial growth factor
  • an agent targeting a single pathway may have limited efficacy. It is desirable to provide treatment against a number of key signaling pathways utilized by tumors to induce angiogenesis in the host stroma. These include PDGF, a potent stimulator of stroma formation (Ostman, A. and C.H. Heldin, Adv. Cancer Res. 2001, 80, 1- 38), FGF, a chemo-attractant and mitogen for fibroblasts and endothelial cells, and VEGF, a potent regulator of vascularization.
  • PDGF a potent stimulator of stroma formation
  • FGF a chemo-attractant and mitogen for fibroblasts and endothelial cells
  • VEGF a potent regulator of vascularization.
  • PDGF is a key regulator of stromal formation, which is secreted by many tumors in a paracrine fashion and is believed to promote the growth of fibroblasts, smooth muscle and endothelial cells, promoting stroma formation and angiogenesis.
  • PDGF was originally identified as the v-sis oncogene product of the simian sarcoma virus (Heldin, C.H., et al., J. Cell. Sci. Suppl. 1985, 3, 65-76).
  • the growth factor is made up of two peptide chains, referred to as A or B chains which share 60% homology in their primary amino acid sequence.
  • the chains are disulfide cross linked to form the 30 kDa mature protein composed of either AA, BB or AB homo- or heterodimmers.
  • PDGF is found at high levels in platelets, and is expressed by endothelial cells and vascular smooth muscle cells. In addition, the production of PDGF is up regulated under low oxygen conditions such as those found in poorly vascularized tumor tissue (Kourembanas, S., et al., Kidney Int. 1997, 51(2), 438-43). PDGF binds with high affinity to the PDGF receptor, a 1106 amino acid 124 kDa transmembrane tyrosine kinase receptor (Heldin, C.H., A. Ostman, and L.
  • PDGFR is found as homo- or heterodimer chains which have 30% homology overall in their amino acid sequence and 64% homology between their kinase domains (Heldin, C.H., et al.. Embo J. 1988, 7(5), 1387-93).
  • PDGFR is a member of a family of tyrosine kinase receptors with split kinase domains that includes VEGFR-2 (KDR), VEGFR-3 (flt-4), c-kit, and flt-3.
  • the PDGF receptor is expressed primarily on fibroblasts, smooth muscle cells, and pericytes and to a lesser extent on neurons, kidney mesangial, Leydig, and Schwann cells of the central nervous system. Upon binding to the receptor, PDGF induces receptor dimerization and undergoes auto- and trans- phosphorylation of tyrosine residues which increase the receptors' kinase activity and promotes the recruitment of downstream effectors through the activation of SH2 protein binding domains.
  • a number of signaling molecules form complexes with activated PDGFR including PI-3-kinase, phospholipase C-gamma, src and GAP (GTPase activating protein for p21-ras) (Soskic, V., et al. Biochemistry 1999, 38(6), 1757-64).
  • PI- 3-kinase PDGF activates the Rho signaling pathway inducing cell motility and migration, and through the activation of GAP, induces mitogenesis through the activation of p21-ras and the MAPK signaling pathway.
  • PDGF is found at high concentrations in platelets and is a potent chemoattractant for fibroblast, smooth muscle cells, neutrophils and macrophages. In addition to its role in wound healing PDGF is known to help maintain vascular homeostasis.
  • PDGF vascular endothelial growth factor
  • angiogenesis PDGF controls interstitial fluid pressure, regulating the permeability of vessels through its regulation of the interaction between connective tissue cells and the extracellular matrix. Inhibiting PDGFR activity can lower interstitial pressure and facilitate the influx of cytotoxics into tumors improving the anti-tumor efficacy of these agents (Pietras, K., et al. Cancer Res. 2002, 62(19), 5476-84; Pietras, K., et al. Cancer Res. 2001, 61(7), 2929-34).
  • PDGF can promote tumor growth through either the paracrine or autocrine stimulation of PDGFR receptors on stromal cells or tumor cells directly, or through the amplification of the receptor or activation of the receptor by recombination.
  • Over expressed PDGF can transform human melanoma cells and keratinocytes (Forsberg, K., et al. Proc. Natl. Acad. Sci. U S A. 1993, 90(2), 393-7; Skobe, M. and N.E. Fusenig, Proc. Natl. Acad. Sci. U S A. 1998, 95(3), 1050-5), two cell types that do not express PDGF receptors, presumably by the direct effect of PDGF on stroma formation and induction of angiogenesis.
  • VEGF vascular endothelial growth factor
  • VPF vascular permeability factor
  • VEGF expression is induced by hypoxia (Shweiki et al. Nature 1992, 359, 843), as well as by a variety of cytokines and growth factors, such as interleukin-1, interleukin-6, epidermal growth factor and transforming growth factor.
  • cytokines and growth factors such as interleukin-1, interleukin-6, epidermal growth factor and transforming growth factor.
  • VEGF and the VEGF family members have been reported to bind to one or more of three transmembrane receptor tyrosine kinases (Mustonen et al. J. Cell Biol.
  • VEGFR-2 also known as kinase insert domain containing receptor (KDR); the murine analogue of VEGFR-2 is known as fetal liver kinase-1 (flk-1)), and VEGFR-3 (also known as flt-4).
  • VEGFR-2 and flt-l have been shown to have different signal transduction properties (Waltenberger et al. J. Biol. Chem. 1994, 269, 26988); Park et al. Oncogene 1995, 10, 135).
  • VEGFR-2 undergoes strong ligand-dependant tyrosine phosphorylation in intact cells, whereas flt-l displays a weak response.
  • binding to VEGFR-2 is believed to be a critical requirement for induction of the full spectrum of VEGF- mediated biological responses.
  • VEGF plays a central role in vasculogenesis, and induces angiogenesis and permeabilization of blood vessels.
  • Deregulated VEGF expression contributes to the development of a number of diseases that are characterized by abnormal angiogenesis and/or hyperpermeability processes. It is believed that regulation of the VEGF-mediated signal transduction cascade by some agents can provide a useful control of abnormal angiogenesis and/or hyperpermeability processes.
  • Tumorigenic cells within hypoxic regions of tumors respond by stimulation of VEGF production, which triggers activation of quiescent endothelial cells to stimulate new blood vessel formation. (Shweiki et al. Proc. Nat'l. Acad. Sci. 1995, 92, 768).
  • VEGF production in tumor regions where there is no angiogenesis may proceed through the ras signal transduction pathway (Grugel et al. J. Biol. Chem. 1995, 270, 25915; Rak et al. Cancer Res. 1995, 55, 4575).
  • In situ hybridization studies have demonstrated VEGF mRNA is strongly upregulated in a wide variety of human tumors, including lung (Mattern et al. Br. J. Cancer 1996, 73, 931), thyroid (Viglietto et al. Oncogene 1995, 11, 1569), breast (Brown et al. Human Pathol. 1995, 26, 86), gastrointestinal tract (Brown et al. Cancer Res.
  • VEGF intraocular angiogenesis
  • ischemic retinal-vein occlusion a cascade of events
  • retinopathy of prematurity a number of retinopathies, including diabetic retinopathy, ischemic retinal-vein occlusion, and retinopathy of prematurity (Aiello et al. New Engl. J. Med. 1994, 331, 1480; Peer et al. Lab. Invest. 1995, 72, 638), and age-related macular degeneration (AMD; see, Lopez et al. Invest. Opththalmol. Vis. Sci. 1996, 37, 855).
  • AMD age-related macular degeneration
  • rheumatoid arthritis In rheumatoid arthritis (RA), the in-growth of vascular pannus may be mediated by production of angiogenic factors. Levels of immunoreactive VEGF are high in the synovial fluid of RA patients, while VEGF levels were low in the synovial fluid of patients with other forms of arthritis of with degenerative joint disease (Koch et al. J. Immunol. 1994, 152, 4149).
  • the angiogenesis inhibitor AGM-170 has been shown to prevent neovascularization of the joint in the rat collagen arthritis model (Peacock et al. J. Exper. Med. 1992, 175, 1135).
  • VEGF expression has also been shown in psoriatic skin, as well as bullous disorders associated with subepidermal blister formation, such as bullous pemphigoid, erythema multiforme, and dermatitis herpetiformis (Brown et al. J. Invest. Dermatol. 1995, 104, 744).
  • VEGF, VEGF-C, VEGF-D vascular endothelial growth factors
  • VEGFR-2, VEGFR-3 lymphangiogenesis
  • VEGF, VEGF-C and VEGF-D are expressed in most tumors, primarily during periods of tumor growth and, often at substantially increased levels.
  • VEGF expression is stimulated by hypoxia, cytokines, oncogenes such as ras, or by inactivation of tumor suppressor genes (McMahon, G. Oncologist 2000, 5(Suppl. 1), 3-10; McDonald, N.Q.; Hendrickson, W.A. Cell 1993, 73, 421-424).
  • VEGFR-3 (also called flt-4) is predominantly expressed on lymphatic endothelium in normal adult tissues. VEGFR-3 function is needed for new lymphatic vessel formation, but not for maintenance of the pre-existing lymphatics. VEGFR-3 is also upregulated on blood vessel endothelium in tumors. Recently VEGF-C and VEGF-D, ligands for VEGFR-3, have been identified as regulators of lymphangiogenesis in mammals. Lymphangiogenesis induced by tumor-associated lymphangiogenic factors could promote the growth of new vessels into the tumor, providing tumor cells access to systemic circulation.
  • VEGF-C vascular endothelial growth factor
  • VEGF-D vascular endothelial growth factor
  • VEGFR-3 vascular endothelial growth factor-3
  • clinicopathological factors that relate directly to the ability of primary tumors to spread (e.g., lymph node involvement, lymphatic invasion, secondary metastases, and disease-free survival).
  • these studies demonstrate a statistical correlation between the expression of lymphangiogenic factors and the ability of a primary solid tumor to metastasize (Skobe, M. et al. Nature Med. 2001, 7(2), 192-198; Stacker, S.A. et al.. Nature Med. 2001, 7(2), 186-191; Makinen, T.
  • hypoxia appears to be an important stimulus for VEGF production in malignant cells.
  • p38 MAP kinase Activation of p38 MAP kinase is required for VEGF induction by tumor cells in response to hypoxia (Blaschke, F. et al. Biochem. Biophys. Res. Commun. 2002, 296, 890-896;
  • mitogen- activated protein kinase (MAPK) p38 has been shown to inhibit both cytokine production (e.g., TNF, IL-1, IL-6, IL-8) and proteolytic enzyme production (e.g., MMP-1, MMP-3) in vitro and/or in vivo.
  • the mitogen activated protein (MAP) kinase p38 is involved in IL-1 and TNF signaling pathways (Lee, J. C; Laydon, J. T.; McDonnell, P. C; Gallagher, T. E; Kumar, S.; Green, D.; McNulty, D.; Blumenthal, M. J.; Heys, J. R.; Landvatter, S.
  • TNF tumor necrosis factor
  • Plasmodium falciparum malaria Perlmann et al. Infect. Immunit. 1997, 65, 116) and cerebral malaria (Rudin et al. Am. J. Pathol. 1997, 150, 257), non-insulin-dependent diabetes mellitus (NIDDM; Stephens et al. J. Biol. Chem. 1997, 272, 971; Ofei et al. Diabetes 1996, 45, 881), congestive heart failure (Doyama et al. Int. J. Cardiol. 1996, 54, 217; McMurray et al. Br. Heart J. 1991, 66, 356), damage following heart disease (Malkiel et al. Mol. Med.
  • TNF levels have also been related to host-versus-graft reactions
  • TNF has also been linked to infectious diseases (review: Beutler et al. Crit. Care Med. 1993, 21, 5423; Degre. Biotherapy
  • HIV human immunodeficiency virus
  • Poli. Proc. Nat'l. Acad. Sci. USA 1990, 87, 782; Vyakaram et al. AIDS 1990, 4, 21; Badley et al. J. Exp. Med. 1997, 185, 55 human immunodeficiency virus
  • MMP matrix-destroying metalloprotease
  • TMPs tissue inhibitors of metalloproteinases
  • aneurysmal aortic disease (Vine et al. Clin. Sci. 1991, 81, 233), birth control (Woessner et al. Steroids 1989, 54, 491), dystrophobic epidermolysis bullosa (Kronberger et al. J. Invest. Dermatol. 1982, 79, 208), degenerative cartilage loss following traumatic joint injury, osteopenias mediated by MMP activity, tempero mandibular joint disease, and demyelating diseases of the nervous system (Chantry et al. J. Neurochem. 1988, 50, 688).
  • mitogen activated protein (MAP) kinase p38 enzyme can provide an approach to the treatment of the above listed diseases including osteoporosis and inflammatory disorders such as rheumatoid arthritis and COPD (Badger, A. M.; Bradbeer, J. N.; Votta, B.; Lee, J. C; Adams, J. L.; Griswold, D. E. J. Pharm. Exper. Ther. 1996, 279, 1453).
  • MAP mitogen activated protein
  • hypoxia appears to be an important stimulus for VEGF production in malignant cells.
  • Activation of p38 kinase is required for VEGF induction by tumor cells in response to hypoxia (Blaschke, F. et al. Biochem. Biophys. Res. Commun. 2002, 296, 890-896; Shemirani, B. et al. Oral Oncology 2002, 38, 251-257).
  • p38 kinase promotes malignant cell invasion, and migration of different tumor types through regulation of collagenase activity and urokinase plasminogen activator expression (Laferriere, J. et al. J. Biol. Chem.
  • omega-carboxyaryl diphenyl urea of Formula I are potent inhibitors of raf kinase, VEGFR kinase, p38 kinase, and PDGFR kinase, which are all molecular targets of interest for the treatment and prevention of osteoporosis, inflammatory disorders, hyper-proliferatrive disorders, and angiogenesis disorders, including cancer.
  • the present invention provides, e.g.,
  • the synthetic metabolites of the compound of this invention include oxidized derivatives of Formula I wherein one or more of the urea nitrogens are substituted with a hydroxy group.
  • the synthetic metabolites of the compound of this invention also include analogs where the methylamide group of the compound of Formula I is hydroxylated then de-methylated by metabolic degradation.
  • the synthetic metabolites of the compound of this invention further include oxidized derivatives where the pyridine nitrogen atom is in the N-oxide form (e.g. carries a hydroxy substituent) leading to those structures referred to in the art as 1-oxo- pyridine and 1 -hydroxy-pyridine.
  • Fig. 1 The relationship of the aforementioned synthetic metabolites with the parent compound of Formula I is illustrated in Fig. 1. Particularly preferred are the M2 and M5 metabolites of the compounds of Formula I.
  • the biotransformation of the compound of Formula I was investigated in vitro with liver microsomes and hepatocytes, and in vivo in the plasma of several species.
  • the N-oxide (M-2) and the demethylated N-oxide (M-5) are of significance. Both metabolites in synthetic form show systemic exposure (area under the curve [AUC], mg*h/L) at steady state similar to regorafenib (the parent compound) in patients at a dose of 160 mg in a 3 weeks on/1 week off Phase I study.
  • the pharmacologic activities of the synthetic metabolites of the compounds of formula I are also of relevance.
  • In vitro biochemical and cellular kinase phosphorylation assays reveal that the synthetic metabolites of compounds of formula I are broad- spectrum kinase inhibitors.
  • the synthetic metabolites were effective against tumor growth and further inhibited tumor vasculature.
  • the synthetic metabolites also demonstrated acute effects on vascular endothelial growth factor (VEGF)- induced hypotension in pharmacodynamic rat models.
  • VEGF vascular endothelial growth factor
  • isolated means that the referenced material is removed from its native environment, e.g., a cell, or a tissue, or from the body.
  • an isolated metabolite can be free of some or all cellular components, i.e., components of the cells in which the native material occurs naturally (e.g., cytoplasmic or membrane component, such as, for example, microsomes).
  • purified refers to material that has been isolated under conditions that reduce or eliminate the presence of unrelated materials, i.e., contaminants, including precursor materials from which the material is obtained.
  • a purified metabolite is preferably substantially free of other metabolites or the precursor compound from which it is derived.
  • substantially free is used operationally, in the context of analytical testing of the material.
  • purified material is substantially free of contaminants is at least 50% pure; more preferably, at least 90% pure, and more preferably still at least 99% pure. Purity can be evaluated by chromatography, gel electrophoresis, HPLC, NMR or mass-spectroscopy analysis, composition analysis, biological assays, and other methods known in the art.
  • compositions of the synthetic metabolites of Forumla I are also within the scope of this invention.
  • pharmaceutically acceptable salt refers to a relatively non- toxic, inorganic or organic acid addition salt of a compound of the present invention.
  • Representative salts of the compound of this invention include the conventional non-toxic salts, for example, from inorganic or organic acids by means well known in the art.
  • such acid addition salts include acetate, adipate, alginate, ascorbate, aspartate, benzoate, benzenesulfonate, bisulfate, butyrate, citrate, camphorate, camphorsulfonate, cinnamate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, fumarate, glucoheptanoate, glycerophosphate, hemisulfate, heptanoate, hexanoate, hydrochloride, hydrobromide, hydroiodide, 2-hydroxyethanesulfonate, itaconate, lactate, maleate, mandelate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oxalate, pamoate, pectinate, persulfate, 3-phenylpropionate
  • the salts or prodrugs of the compounds of Formula I may contain one or more asymmetric centers.
  • Asymmetric carbon atoms may be present in the (R) or (S) configuration or (R,S) configuration.
  • Substituents on a ring may also be present in either cis or trans form. It is intended that all such configurations (including enantiomers and diastereomers), are included within the scope of the present invention.
  • Preferred isomers are those with the configuration which produces the more desirable biological activity.
  • Separated, pure or partially purified isomers or racemic mixtures of the compounds of this invention are also included within the scope of the present invention. The purification of said isomers and the separation of said isomeric mixtures can be accomplished by standard techniques known in the art.
  • Example 4 The particular process to be utilized in the preparation of the synthetic metabolites used in this embodiment of the invention is described in Example 4. Salt forms of the synthetic metabolites of Formula (I) are described in the Examples.
  • the present invention provides compounds which are capable of modulating one or more signal transduction pathways involving raf, VEGFR, PDGFR, p38, and/or flt-3 kinases.
  • Raf is an important signaling molecule involved in the regulation of a number of key cellular processes, including cell growth, cell survival and invasion. It is a member of the Ras/raf/MEK/ERK pathway. This pathway is present in most tumor cells.
  • VEGFR, PDGFR, and flt-3 are transmembrane receptor molecules which, when stimulated by an appropriate ligand, trigger the Ras/raf/MEK/ERK cell signaling pathway, leading to a cascade of cellular events. Each of these receptor molecules have tyrosine kinase activity.
  • VEGFR receptors are stimulated by vascular endothelial growth factors (VEGF), and are important control points in the regulation of endothelial cell development and function.
  • VEGF vascular endothelial growth factors
  • the PDGF-beta receptor regulates cell proliferation and survival in a number of cell types, including mesenchymal cells.
  • Flt-3 is a receptor for the FL ligand. It is structurally similar to c-kit, and modulates the growth of pluripotent haemopoietic cells, influencing the development of T-cells, B-cells, and dendritic cells.
  • Raf or raf-1 kinase is a family of serine/threonine kinases which comprise at least three family members, a-raf, b-raf, and c-raf or raf-1. See, e.g., Dhillon and Kolch, Arch. Biochem. Biophys. 2002, 404, 3-9. C-raf and b-raf are preferred targets for compounds of the present invention.
  • Activating b-raf mutations e.g., V599E mutant
  • V599E mutant have been identified in various cancers, including melanoma, and the compounds described herein can be utilized to inhibit their activity.
  • module it is meant that the functional activity of the pathway (or a component of it) is changed in comparison to its normal activity in the absence of the compound. This effect includes any quality or degree of modulation, including, increasing, agonizing, augmenting, enhancing, facilitating, stimulating, decreasing, blocking, inhibiting, reducing, diminishing, antagonizing, etc.
  • the compounds of the present invention can also modulate one or more of the following processes, including, but not limited to, e.g., cell growth (including, e.g., differentiation, cell survival, and/or proliferation), tumor cell growth (including, e.g., differentiation, cell survival, and/or proliferation), tumor regression, endothelial cell growth (including, e.g., differentiation, cell survival, and/or proliferation), angiogenesis (blood vessel growth), lymphangiogenesis (lymphatic vessel growth), and/or hematopoiesis (e.g., T- and B-cell development, dendritic cell development, etc.).
  • cell growth including, e.g., differentiation, cell survival, and/or proliferation
  • tumor cell growth including, e.g., differentiation, cell survival, and/or proliferation
  • tumor regression including, e.g., differentiation, cell survival, and/or proliferation
  • endothelial cell growth including, e.g., differentiation, cell survival, and/or proliferation
  • angiogenesis blood vessel growth
  • kinase activity it is meant a catalytic activity in which a gamma-phosphate from adenosine triphosphate (ATP) is transferred to an amino acid residue (e.g., serine, threonine, or tyrosine) in a protein substrate.
  • ATP adenosine triphosphate
  • a compound can modulate kinase activity, e.g., inhibiting it by directly competing with ATP for the ATP-binding pocket of the kinase, by producing a conformational change in the enzyme's structure that affects its activity (e.g., by disrupting the biologically-active three- dimensional structure), etc.
  • Kinase activity can be determined routinely using conventional assay methods.
  • Kinase assays typically comprise the kinase enzyme, substrates, buffers, and components of a detection system.
  • a typical kinase assay involves the reaction of a protein kinase with a peptide substrate and an ATP, such as 32P-ATP, to produce a phosphorylated end-product (for instance, a phosphoprotein when a peptide substrate is used).
  • the resulting end-product can be detected using any suitable method.
  • a radioactively labeled phosphoprotein can be separated from the unreacted gamma-32P-ATP using an affinity membrane or gel electrophoresis, and then visualized on the gel using autoradiography or detected with a scintillation counter.
  • Non-radioactive methods can also be used. Methods can utilize an antibody which recognizes the phosphorylated substrate, e.g., an anti-phosphotyrosine antibody.
  • kinase enzyme can incubated with a substrate in the presence of ATP and kinase buffer under conditions which are effective for the enzyme to phosphorylate the substrate.
  • the reaction mixture can be separated, e.g., electrophoretically, and then phosphorylation of the substrate can be measured, e.g., by Western blotting using an anti-phosphotyrosine antibody.
  • the antibody can be labeled with a detectable label, e.g., an enzyme, such as HRP, avidin or biotin, chemiluminescent reagents, etc.
  • detectable label e.g., an enzyme, such as HRP, avidin or biotin, chemiluminescent reagents, etc.
  • Other methods can utilize ELISA formats, affinity membrane separation, fluorescence polarization assays, luminescent assays, etc.
  • TR-FRET time-resolved fluorescence resonance energy transfer
  • the compounds of the present invention can be used to treat and/or prevent any disease or condition mediated by one or more cellular signal transduction pathways involving raf, VEGFR, PDGFR, p38, and/or flt-3 kinases.
  • the term "treating” is used conventionally, e.g., the management or care of a subject for the purpose of combating, alleviating, reducing, relieving, improving the condition of, etc., of a disease or disorder.
  • the compounds can also be described as being used to prevent and/or treat diseases and/or condition mediated by the signaling molecules.
  • mediated indicates, e.g., that the signaling molecule is part of the pathway which is aberrant or disturbed in the disease and/or condition.
  • Raf associated diseases include, e.g., cell-proliferation disorders, cancer, tumors, etc.;
  • VEGFR-2 associated diseases include, e.g., cancer, tumor growth, inflammatory disease, rheumatoid arthritis, retinopathy, psoriasis, glomerulonephritis, asthma, chronic bronchitis, atherosclerosis, transplant rejection, conditions involving angiogenesis, etc.
  • VEGFR-3 associated diseases include, e.g., cancer, corneal disease, inflamed cornea (e.g., Hamrah, Am. J. Path. 2003, 163, 57-68), corneal transplantation (Cursiefen et al., Cornea 2003, 22, 273-81), lymphatic hyperplasia, conditions involving lymphangiogenesis, etc.;
  • PDGFR-beta associated diseases include, e.g., diseases or conditions characterized by cell proliferation, cell matrix production, cell movement, and/or extracellular matrix production.
  • diseases or conditions include, e.g., tumors, malignancies, cancer, metastasis, chronic myeloid leukemia, inflammation, renal disease, diabetic nephropathy, mesangial proliferative glomerulonephritis, fibrotic conditions, atherosclerosis, restenosis, hypertension-related arteriosclerosis, venous bypass graft arteriosclerosis, scleroderma, interstitial pulmonary diseases, synovial disorders, arthritis, leukemias, lymphomas, etc
  • Flt-3 associated diseases include, e.g., immune-related disorders, blood cell disorders, conditions involving hematopoietic cell development (e.g., T-cells, B-cells, dendritic cells, cancer, anemia, HIV, acquired immune deficiency syndrome, etc.
  • p38 associated diseases include inflammatory disorders, immunomodulatory disorders, and other disorders that have been linked to abnormal cytokine production, especially TNF-alpha, or abnormal MMP activity. These disorders include, but are not limited to, rheumatoid arthritis, COPD, osteoporosis, Crohn's disease and psoriasis.
  • compounds of the present invention can be used to treat conditions and disorders disclosed in U.S. Pat. No.
  • the compounds of this invention also have a broad therapeutic activity to treat or prevent the progression of a broad array of diseases, such as inflammatory conditions, coronary restenosis, tumor-associated angiogenesis, atherosclerosis, autoimmune diseases, inflammation, certain kidney diseases associated with proliferation of glomerular or mesangial cells, and ocular diseases associated with retinal vessel proliferation, psoriasis, hepatic cirrhosis, diabetes, atherosclerosis, restenosis, vascular graft restenosis, in-stent stenosis, angiogenesis, ocurlar diseases, pulmonary fibrosis, obliterative bronchiolitis, glomerular nephritis, rheumatoid arthritis.
  • diseases such as inflammatory conditions, coronary restenosis, tumor-associated angiogenesis, atherosclerosis, autoimmune diseases, inflammation, certain kidney diseases associated with proliferation of glomerular or mesangial cells, and ocular diseases associated with retinal vessel proliferation, psoriasis
  • the present invention also provides for treating, preventing, modulating, etc., one or more of the following conditions in humans and/or other mammals: retinopathy, including diabetic retinopathy, ischemic retinal-vein occlusion, retinopathy of prematurity and age related macular degeneration; rheumatoid arthritis, psoriasis, or bullous disorder associated with subepidermal blister formation, including bullous pemphigoid, erythema multiforme, or dermatitis herpetiformis, rheumatic fever, bone resorption, postmenopausal osteoperosis, sepsis, gram negative sepsis, septic shock, endotoxic shock, toxic shock syndrome, systemic inflammatory response syndrome, inflammatory bowel disease (Crohn's disease and ulcerative colitis), Jarisch-Herxheimer reaction, asthma, adult respiratory distress syndrome, acute pulmonary fibrotic disease, pulmonary sarcoidosis, allergic respiratory disease, silicos
  • coli infection effects of enterotoxin A resulting from Staphylococcus infection, meningococcal infection, and infections from Borrelia burgdorferi, Treponema pallidum, cytomegalovirus, influenza virus, Theiler' s encephalomyelitis virus, and the human immunodeficiency virus (HIV), papilloma, blastoglioma, Kaposi's sarcoma, melanoma, lung cancer, ovarian cancer, prostate cancer, squamous cell carcinoma, astrocytoma, head cancer, neck cancer, bladder cancer, breast cancer, colorectal cancer, thyroid cancer, pancreatic cancer, gastric cancer, hepatocellular carcinoma, leukemia, lymphoma, Hodgkin's disease, Burkitt's disease, arthritis, rheumatoid arthritis, diabetic retinopathy, angiogenesis, restenosis, in-stent restenosis, vascular graft restenosis, pulmonary fibro
  • the compounds of this invention can possess more than one of the mentioned activities, and therefore can target a plurality of signal transduction pathways.
  • these compounds can achieve therapeutic and prophylactic effects which normally are only obtained when using a combination of different compounds.
  • the ability to inhibit both new vessel formation (e.g., associated with VEGFR-2 and VEGFR-3 function) (e.g., blood and/or lymph) and cell-proliferation (e.g., associated with raf and PDGFR-beta function) using a single compound is especially beneficial in the treatment of cancer, and other cell- proliferation disorders that are facilitated by neo-vascularization.
  • the present invention relates specifically to compounds which possess at least anti-cell proliferation and anti- angiogenic (i.e., inhibits angiogenesis) activity. Any disorder or condition that would benefit from inhibiting vessel growth and cell proliferation can be treated in accordance with the present invention. Using a single compound is also advantageous because its range of activities can be more precisely defined.
  • the present invention relates to methods of treating and/or preventing diseases and conditions; and/or modulating one or more of the pathways, polypeptides, genes, diseases, conditions, etc., associated with raf, VEGFR, PDGFR, p38, and/or flt-3. These methods generally involve administering effective amounts of compounds of the present invention, where an effective amount is the quantity of the compound which is useful to achieve the desired result.
  • Compounds can be administered in any effective form by any effective route, as discussed in more detail below.
  • Methods include modulating tumor cell proliferation, including inhibiting cell proliferation.
  • the latter indicates that the growth and/or differentiation of tumor cells is reduced, decreased, diminished, slowed, etc.
  • proliferation includes any process which relates to cell growth and division, and includes differentiation and apoptosis.
  • raf kinases play a key role in the activation of the cytoplasmic signaling cascade involved in cell proliferation, differentiation, and apoptosis. For example, studies have found that inhibiting c-raf by anti-sense oligonucleotides can block cell proliferation (see above). Any amount of inhibition is considered therapeutic.
  • Included in the methods of the present invention is a method for using synthetic metabolites of the compound described above (Compound of Formula I), including salts, prodrugs, and compositions thereof, to treat mammalian hyper-proliferative disorders comprising administering to a mammal, including a human in need thereof, an amount of a synthetic metabolite of a compound of this invention, pharmaceutically acceptable salt, and composition thereof, which is effective to treat the disorder.
  • Hyper-proliferative disorders include but are not limited to solid tumors, such as cancers of the breast, respiratory tract, brain, reproductive organs, digestive tract, urinary tract, eye, liver, skin, head and neck, thyroid, parathyroid and their distant metastases. Those disorders also include lymphomas, sarcomas, and leukemias.
  • Any tumor or cancer can be treated, including, but not limited to, cancers having one or more mutations in raf, ras, and/or flt-3, as well as any upstream or downstream member of the signaling pathways of which they are a part.
  • a cancer can be treated with a compound of the present invention irrespective of the mechanism which is responsible for it.
  • Cancers of any organ can be treated, including cancers of, but are not limited to, e.g., colon, pancreas, breast, prostate, bone, liver, kidney, lung, testes, skin, pancreas, stomach, colorectal cancer, renal cell carcinoma, hepatocellular carcinoma, melanoma, etc.
  • breast cancer examples include, but are not limited to, invasive ductal carcinoma, invasive lobular carcinoma, ductal carcinoma in situ, and lobular carcinoma in situ.
  • cancers of the respiratory tract include, but are not limited to, small-cell and non-small-cell lung carcinoma, as well as bronchial adenoma and pleuropulmonary blastoma.
  • brain cancers include, but are not limited to, brain stem and hypophtalmic glioma, cerebellar and cerebral astrocytoma, medulloblastoma, ependymoma, as well as neuroectodermal and pineal tumor.
  • Tumors of the male reproductive organs include, but are not limited to, prostate and testicular cancer.
  • Tumors of the female reproductive organs include, but are not limited to, endometrial, cervical, ovarian, vaginal, and vulvar cancer, as well as sarcoma of the uterus.
  • Tumors of the digestive tract include, but are not limited to, anal, colon, colorectal, esophageal, gallbladder, gastric, pancreatic, rectal, small-intestine, and salivary gland cancers.
  • Tumors of the urinary tract include, but are not limited to, bladder, penile, kidney, renal pelvis, ureter, and urethral cancers.
  • Eye cancers include, but are not limited to, intraocular melanoma and retinoblastoma.
  • liver cancers include, but are not limited to, hepatocellular carcinoma (liver cell carcinomas with or without fibrolamellar variant), cholangiocarcinoma (intrahepatic bile duct carcinoma), and mixed hepatocellular cholangiocarcinoma.
  • Skin cancers include, but are not limited to, squamous cell carcinoma, Kaposi's sarcoma, malignant melanoma, Merkel cell skin cancer, and non-melanoma skin cancer.
  • Head-and-neck cancers include, but are not limited to, laryngeal, hypopharyngeal, nasopharyngeal, and/or oropharyngeal cancers, and lip and oral cavity cancer.
  • Lymphomas include, but are not limited to, AIDS-related lymphoma, non-Hodgkin's lymphoma, cutaneous T-cell lymphoma, Hodgkin's disease, and lymphoma of the central nervous system.
  • Sarcomas include, but are not limited to, sarcoma of the soft tissue, osteosarcoma, malignant fibrous histiocytoma, lymphosarcoma, and rhabdomyosarcoma.
  • Leukemias include, but are not limited to, acute myeloid leukemia, acute lymphoblastic leukemia, chronic lymphocytic leukemia, chronic myelogenous leukemia, and hairy cell leukemia.
  • compounds of the present invention can also cause tumor regression, e.g., a decrease in the size of a tumor, or in the extent of cancer in the body.
  • the present invention also relates to methods of modulating angiogenesis and/or lymphangiogenesis in a system comprising cells, comprising administering to the system an effective amount of a compound described herein.
  • a system comprising cells can be an in vivo system, such as a tumor in a patient, isolated organs, tissues, or cells, in vitro assays systems (CAM, BCE, etc), animal models (e.g., in vivo, subcutaneous, cancer models), hosts in need of treatment (e.g., hosts suffering from diseases having angiogenic and/or lymphangiogenic component, such as cancer), etc. Inappropriate and ectopic expression of angiogenesis can be deleterious to an organism. A number of pathological conditions are associated with the growth of extraneous blood vessels.
  • neovascularization of tumor explants include, e.g., U.S. Pat. Nos.
  • CAM chicken chorioallantoic membrane
  • BCE bovine capillary endothelial cell assay
  • HUVEC human umbilical cord vascular endothelial cell growth inhibition assay
  • Modulation of angiogenesis can be determined by any other method.
  • the degree of tissue vascularity is typically determined by assessing the number and density of vessels present in a given sample.
  • microvessel density MMD
  • MMD microvessel density
  • CD31 also known as platelet-endothelial cell adhesion molecule or PEC AM.
  • a CD31 antibody can be employed in conventional immunohistological methods to immunostain tissue sections as described by, e.g., U.S. Pat. No. 6,017,949; Delias et al., Gyn. Oncol.
  • Vezfl e.g., Xiang et al., Dev. Bio. 1999, 206, 123-141
  • angiopoietin e.g., Tie-1
  • Tie-2 e.g., Sato et al., Nature 1995, 376, 70-74.
  • the present invention relates to methods of screening patients to determine their sensitivity to compounds of the present invention.
  • the invention relates to methods of determining whether a condition can be modulated by a compound disclosed herein, comprising measuring the expression or activity of raf, VEGFR-2, VEGFR-3, PDGFR-beta, p38, and/or flt-3 in a sample comprising cells or a cell extract, wherein said sample has been obtained from a cell or subject having said condition.
  • the condition identifies the condition as being treatable with a compound of the present invention, i.e., whereby said disorder or condition can be modulated by the compound when said expression or activity is increased in said condition as compared to a normal control.
  • the method can further comprise a step of comparing the expression in a sample with a normal control, or expression in a sample obtained from normal or unaffected tissue. Comparing can be done manually, against a standard, in an electronic form (e.g., against a database), etc.
  • the normal control can be a standard sample that is provided with the assay; it can be obtained from adjacent, but unaffected, tissue from the same patient; or, it can be pre-determined values, etc.
  • Gene expression, protein expression (e.g., abundance in a cell), protein activity (e.g., kinase activity), etc., can be determined.
  • a biopsy from a cancer patient can be assayed for the presence, quantity, and/or activity of raf, VEGFR-2, VEGFR-3, PDGFR-beta, p38, and/or flt-3.
  • Increased expression or activity of one or more of these can indicate that the cancer can be targeted for treatment by a compound of the present invention.
  • raf activity can be monitored by its ability to initiate the cascade leading to ERK phosphorylation (i.e., raf/MEK/ERK), resulting in phospho-ERK.
  • Increased phospho-ERK levels in a cancer specimen shows that its raf activity is elevated, suggesting the use of compounds of the present invention to treat it.
  • Measuring expression includes determining or detecting the amount of the polypeptide present in a cell or shed by it, as well as measuring the underlying mRNA, where the quantity of mRNA present is considered to reflect the quantity of polypeptide manufactured by the cell. Furthermore, the genes for raf, VEGFR-2, VEGFR-3, PDGFR-beta, p38, and/or Flt-3 can be analyzed to determine whether there is a gene defect responsible for aberrant expression or polypeptide activity.
  • Polypeptide detection can be carried out by any available method, e.g., by Western blots, ELISA, dot blot, immunoprecipitation, RIA, immunohistochemistry, etc.
  • a tissue section can be prepared and labeled with a specific antibody (indirect or direct and visualized with a microscope.
  • Amount of a polypeptide can be quantitated without visualization, e.g., by preparing a lysate of a sample of interest, and then determining by ELISA or Western the amount of polypeptide per quantity of tissue.
  • Antibodies and other specific binding agents can be used. There is no limitation on how detection is performed.
  • Assays can be utilized which permit quantification and/or presence/absence detection of a target nucleic acid (e.g., genes, mRNA, etc., for raf, VEGFR, PDGFR, p38, and/or flt-3) in a sample. Assays can be performed at the single-cell level, or in a sample comprising many cells, where the assay is "averaging" expression over the entire collection of cells and tissue present in the sample. Any suitable assay format can be used, including, but not limited to, e.g., Southern blot analysis, Northern blot analysis, polymerase chain reaction ("PCR") (e.g., Saiki et al., Science 1988, 241, 53; U.S. Pat. Nos.
  • PCR polymerase chain reaction
  • PCR Protocols A Guide to Methods and Applications, Innis et al., eds., Academic Press, New York, 1990
  • RT-PCR reverse transcriptase polymerase chain reaction
  • RACE rapid amplification of cDNA ends
  • LCR ligase chain reaction
  • RNA fingerprinting techniques nucleic acid sequence based amplification (“NASBA") and other transcription based amplification systems (e.g., U.S. Pat. Nos. 5,409,818 and 5,554,527; WO 88/10315), polynucleotide arrays (e.g., U.S. Pat. Nos.
  • NASBA nucleic acid sequence based amplification
  • transcription based amplification systems e.g., U.S. Pat. Nos. 5,409,818 and 5,554,527; WO 88/10315
  • polynucleotide arrays e.g., U.S. Pat. Nos.
  • any method suitable for single cell analysis of gene or protein expression can be used, including in situ hybridization, immunocytochemistry, MACS, FACS, flow cytometry, etc.
  • expression products can be measured using antibodies, PCR, or other types of nucleic acid amplification (e.g., Brady et al., Methods Mol. & Cell. Biol. 1990, 2, 17-25; Eberwine et al., Proc. Natl. Acad. Sci. 1992, 89, 3010-3014; U.S. Pat. No. 5,723,290).
  • nucleic acid amplification e.g., Brady et al., Methods Mol. & Cell. Biol. 1990, 2, 17-25; Eberwine et al., Proc. Natl. Acad. Sci. 1992, 89, 3010-3014; U.S. Pat. No. 5,723,290.
  • Activity of raf, VEGFR-2, VEGFR-3, PDGFR-beta, p38, and/or flt-3 can be assessed routinely, e.g., as described in the examples below, or using standard assays for kinase activity.
  • the present invention also provides methods of assessing the efficacy of a compound of the present invention in treating a disorder, comprising one or more of the following steps in any effective order, e.g., administering an amount of a compound, measuring the expression or activity of raf, VEGFR-2, VEGFR-3, PDGFR-beta, p38, and/or flt-3 (see above), determining the effect of said compound on said expression or activity. For instance, biopsy samples can be removed from patients who have been treated with a compound of the present invention, and then assayed for the presence and/or activity of the mentioned signaling molecules.
  • decreases in the levels of phospho-ERK in the cancer tissue indicate that the compound is exerting in vivo efficacy and a therapeutic effect.
  • the method can be used to determine appropriate dosages and dosing regimens, e.g., how much compound to administer and at what frequency to administer it.
  • the clinician can determine the appropriate treatment protocol and whether it is achieving the desired effect, e.g., on modulating or inhibiting the signal transduction pathway.
  • Compounds of the present invention also can be used as markers to determine the presence and quantity of raf, VEGFR-2, VEGFR-3, PDGFR-beta, p38, and/or flt-3, in a sample comprising a biological material. This comprises one or more of the following steps in any effective order: (i) contacting said sample comprising a biological material with a compound of the present invention, and (ii) determining whether said compound binds to said material.
  • the compound can be labeled, or it can be used as a competitor to a labeled compound, such as labeled- ATP.
  • the invention also provides methods for treating, preventing, modulating, etc., diseases and conditions in mammals comprising administering a compound of this invention with another modulator of the signal transduction pathway comprising, but not limited to raf, VEGFR, PDGFR, p38, and/or flt-3. These can be present in the same composition or in separate formulations or dosage units. Administration can be the same or different routes, and can be simultaneous or sequential.
  • VEGFR-3 modulation relate to VEGFR-3 modulation and are incorporated herein for their description of disease states mediated by VEGFR-3 and assays to determine such activity.
  • compositions containing a compound of the present invention and pharmaceutically acceptable salts thereof can be utilized to achieve the desired pharmacological effect by administration to a patient in need thereof.
  • a patient for the purpose of this invention, is a mammal, including a human, in need of treatment for the particular condition or disease. Therefore, the present invention includes pharmaceutical compositions which are comprised of a pharmaceutically acceptable carrier and a pharmaceutically effective amount of a compound, or salt thereof, of the present invention.
  • pharmaceutically acceptable carrier is meant as any carrier which is relatively non-toxic and innocuous to a patient at concentrations consistent with effective activity of the active ingredient so that any side effects ascribable to the carrier do not vitiate the beneficial effects of the active ingredient.
  • a pharmaceutically effective amount of compound is that amount which produces a result or exerts an influence on the particular condition being treated.
  • the compound of the present invention can be administered with pharmaceutically-acceptable carriers well known in the art using any effective conventional dosage unit forms, including immediate, slow and timed release preparations, orally, parenterally, topically, nasally, ophthalmically, optically, sublingually, rectally, vaginally, and the like.
  • the compound can be formulated into solid or liquid preparations such as capsules, pills, tablets, troches, lozenges, melts, powders, solutions, suspensions, or emulsions, and may be prepared according to methods known to the art for the manufacture of pharmaceutical compositions.
  • the solid unit dosage forms can be a capsule which can be of the ordinary hard- or soft-shelled gelatin type containing, for example, surfactants, lubricants, and inert fillers such as lactose, sucrose, calcium phosphate, and corn starch.
  • the compounds of this invention may be tableted with conventional tablet bases such as lactose, sucrose and cornstarch in combination with binders such as acacia, corn starch or gelatin, disintegrating agents intended to assist the break-up and dissolution of the tablet following administration such as potato starch, alginic acid, corn starch, and guar gum, gum tragacanth, acacia, lubricants intended to improve the flow of tablet granulation and to prevent the adhesion of tablet material to the surfaces of the tablet dies and punches, for example talc, stearic acid, or magnesium, calcium or zinc stearate, dyes, coloring agents, and flavoring agents such as peppermint, oil of wintergreen, or cherry flavoring, intended to enhance the aesthetic qualities of the tablets and make them more acceptable to the patient.
  • binders such as acacia, corn starch or gelatin
  • disintegrating agents intended to assist the break-up and dissolution of the tablet following administration such as potato starch, alginic acid, corn star
  • Suitable excipients for use in oral liquid dosage forms include dicalcium phosphate and diluents such as water and alcohols, for example, ethanol, benzyl alcohol, and polyethylene alcohols, either with or without the addition of a pharmaceutically acceptable surfactant, suspending agent or emulsifying agent.
  • Various other materials may be present as coatings or to otherwise modify the physical form of the dosage unit. For instance tablets, pills or capsules may be coated with shellac, sugar or both.
  • Dispersible powders and granules are suitable for the preparation of an aqueous suspension. They provide the active ingredient in admixture with a dispersing or wetting agent, a suspending agent and one or more preservatives. Suitable dispersing or wetting agents and suspending agents are exemplified by those already mentioned above. Additional excipients, for example those sweetening, flavoring and coloring agents described above, may also be present.
  • the pharmaceutical compositions of this invention may also be in the form of oil-in-water emulsions.
  • the oily phase may be a vegetable oil such as liquid paraffin or a mixture of vegetable oils.
  • Suitable emulsifying agents may be (1) naturally occurring gums such as gum acacia and gum tragacanth, (2) naturally occurring phosphatides such as soy bean and lecithin, (3) esters or partial esters derived form fatty acids and hexitol anhydrides, for example, sorbitan monooleate, (4) condensation products of said partial esters with ethylene oxide, for example, polyoxyethylene sorbitan monooleate.
  • the emulsions may also contain sweetening and flavoring agents.
  • Oily suspensions may be formulated by suspending the active ingredient in a vegetable oil such as, for example, arachis oil, olive oil, sesame oil or coconut oil, or in a mineral oil such as liquid paraffin.
  • the oily suspensions may contain a thickening agent such as, for example, beeswax, hard paraffin, or cetyl alcohol.
  • the suspensions may also contain one or more preservatives, for example, ethyl or n-propyl p-hydroxybenzoate; one or more coloring agents; one or more flavoring agents; and one or more sweetening agents such as sucrose or saccharin.
  • Syrups and elixirs may be formulated with sweetening agents such as, for example, glycerol, propylene glycol, sorbitol or sucrose. Such formulations may also contain a demulcent, and preservative, such as methyl and propyl parabens and flavoring and coloring agents.
  • sweetening agents such as, for example, glycerol, propylene glycol, sorbitol or sucrose.
  • Such formulations may also contain a demulcent, and preservative, such as methyl and propyl parabens and flavoring and coloring agents.
  • the compounds of this invention may also be administered parenterally, that is, subcutaneously, intravenously, intraocularly, intrasynovially, intramuscularly, or interperitoneally, as injectable dosages of the compound in a physiologically acceptable diluent with a pharmaceutical carrier which can be a sterile liquid or mixture of liquids such as water, saline, aqueous dextrose and related sugar solutions, an alcohol such as ethanol, isopropanol, or hexadecyl alcohol, glycols such as propylene glycol or polyethylene glycol, glycerol ketals such as 2,2-dimethyl-l,l-dioxolane-4-methanol, ethers such as poly(ethylene glycol) 400, an oil, a fatty acid, a fatty acid ester or, a fatty acid glyceride, or an acetylated fatty acid glyceride, with or without the addition of a pharmaceutically acceptable surfactant such as
  • Suitable fatty acids include oleic acid, stearic acid, isostearic acid and myristic acid.
  • Suitable fatty acid esters are, for example, ethyl oleate and isopropyl myristate.
  • Suitable soaps include fatty acid alkali metal, ammonium, and triethanolamine salts and suitable detergents include cationic detergents, for example dimethyl dialkyl ammonium halides, alkyl pyridinium halides, and alkylamine acetates; anionic detergents, for example, alkyl, aryl, and olefin sulfonates, alkyl, olefin, ether, and monoglyceride sulfates, and sulfosuccinates; non-ionic detergents, for example, fatty amine oxides, fatty acid alkanolamides, and poly(oxyethylene-oxypropylene)s or ethylene oxide or propylene oxide copolymers; and amphoteric detergents, for example, alkyl-beta-aminopropionates, and 2-alkylimidazoline quarternary ammonium salts, as well as mixtures.
  • suitable detergents include cationic detergents, for example di
  • compositions of this invention will typically contain from about 0.5% to about 25% by weight of the active ingredient in solution. Preservatives and buffers may also be used advantageously. In order to minimize or eliminate irritation at the site of injection, such compositions may contain a non-ionic surfactant having a hydrophile-lipophile balance (HLB) of from about 12 to about 17. The quantity of surfactant in such formulation ranges from about 5% to about 15% by weight.
  • the surfactant can be a single component having the above HLB or can be a mixture of two or more components having the desired HLB.
  • surfactants used in parenteral formulations are the class of polyethylene sorbitan fatty acid esters, for example, sorbitan monooleate and the high molecular weight adducts of ethylene oxide with a hydrophobic base, formed by the condensation of propylene oxide with propylene glycol.
  • compositions may be in the form of sterile injectable aqueous suspensions.
  • suspensions may be formulated according to known methods using suitable dispersing or wetting agents and suspending agents such as, for example, sodium carboxymethylcellulose, methylcellulose, hydroxypropylmethyl-cellulose, sodium alginate, gum tragacanth and gum acacia; dispersing or wetting agents which may be a naturally occurring phosphatide such as lecithin, a condensation product of an alkylene oxide with a fatty acid, for example, polyoxyethylene stearate, a condensation product of ethylene oxide with a long chain aliphatic alcohol, for example, heptadeca-ethyleneoxycetanol, a condensation product of ethylene oxide with a partial ester derived form a fatty acid and a hexitol such as polyoxyethylene sorbitol monooleate, or a condensation product of an ethylene oxide with a partial ester derived from a fatty acid and a hexito
  • the sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally acceptable diluent or solvent.
  • Diluents and solvents that may be employed are, for example, water, Ringer's solution, isotonic sodium chloride solutions and isotonic glucose solutions.
  • sterile fixed oils are conventionally employed as solvents or suspending media.
  • any bland, fixed oil may be employed including synthetic mono- or diglycerides.
  • fatty acids such as oleic acid can be used in the preparation of injectables.
  • composition of the invention may also be administered in the form of suppositories for rectal administration of the drug.
  • These compositions can be prepared by mixing the drug with a suitable non-irritation excipient which is solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum to release the drug.
  • a suitable non-irritation excipient which is solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum to release the drug.
  • Such material is, for example, cocoa butter and polyethylene glycol.
  • transdermal delivery devices Such transdermal patches may be used to provide continuous or discontinuous infusion of the compounds of the present invention in controlled amounts.
  • the construction and use of transdermal patches for the delivery of pharmaceutical agents is well known in the art (see, e.g., US Patent No. 5,023,252, issued June 11, 1991, incorporated herein by reference).
  • patches may be constructed for continuous, pulsatile, or on demand delivery of pharmaceutical agents.
  • Controlled release formulations for parenteral administration include liposomal, polymeric microsphere and polymeric gel formulations which are known in the art.
  • a mechanical delivery device It may be desirable or necessary to introduce the pharmaceutical composition to the patient via a mechanical delivery device.
  • the construction and use of mechanical delivery devices for the delivery of pharmaceutical agents is well known in the art.
  • Direct techniques for, for example, administering a drug directly to the brain usually involve placement of a drug delivery catheter into the patient's ventricular system to bypass the blood-brain barrier.
  • One such implantable delivery system, used for the transport of agents to specific anatomical regions of the body is described in US Patent No. 5,011,472, issued April 30, 1991.
  • compositions of the invention can also contain other conventional pharmaceutically acceptable compounding ingredients, generally referred to as carriers or diluents, as necessary or desired.
  • Conventional procedures for preparing such compositions in appropriate dosage forms can be utilized. Such ingredients and procedures include those described in the following references, each of which is incorporated herein by reference: Powell, M.F. et al, "Compendium of Excipients for Parenteral Formulations” PDA Journal of Pharmaceutical Science & Technology 1998, 52(5), 238-311; Strickley, R.G “Parenteral Formulations of Small Molecule Therapeutics Marketed in the United States (1999)-Part-1" PDA Journal of Pharmaceutical Science & Technology 1999, 53(6), 324-349; and Nema, S. et al, "Excipients and Their Use in Injectable Products” PDA Journal of Pharmaceutical Science & Technology 1997, 51(4), 166-171.
  • compositions for its intended route of administration include:
  • acidifying agents include but are not limited to acetic acid, citric acid, fumaric acid, hydrochloric acid, nitric acid);
  • alkalinizing agents examples include but are not limited to ammonia solution, ammonium carbonate, diethanolamine, monoethanolamine, potassium hydroxide, sodium borate, sodium carbonate, sodium hydroxide, triethanolamine, trolamine;
  • adsorbents examples include but are not limited to powdered cellulose and activated charcoal
  • aerosol propellants examples include but are not limited to carbon dioxide, CC12F2, F2C1C- CC1F2 and CC1F3
  • air displacement agents examples include but are not limited to nitrogen and argon
  • antifungal preservatives examples include but are not limited to benzoic acid, butylparaben, ethylparaben, methylparaben, propylparaben, sodium benzoate
  • antimicrobial preservatives examples include but are not limited to benzalkonium chloride, benzethonium chloride, benzyl alcohol, cetylpyridinium chloride, chlorobutanol, phenol, phenylethyl alcohol, phenylmercuric nitrate and thimerosal
  • antioxidants examples include but are not limited to ascorbic acid, ascorbyl palmitate, butylated hydroxyanisole, butylated hydroxytoluene, hypophosphorus acid, monothioglycerol, propyl gallate, sodium ascorbate, sodium bisulfite, sodium formaldehyde sulfoxylate, sodium metabisulfite);
  • binding materials examples include but are not limited to block polymers, natural and synthetic rubber, polyacrylates, polyurethanes, silicones, polysiloxanes and styrene-butadiene copolymers;
  • buffering agents examples include but are not limited to potassium metaphosphate, dipotassium phosphate, sodium acetate, sodium citrate anhydrous and sodium citrate dihydrate
  • chelating agents examples include but are not limited to edetate disodium and edetic acid
  • colorants examples include but are not limited to FD&C Red No. 3, FD&C Red No. 20, FD&C Yellow No. 6, FD&C Blue No. 2, D&C Green No. 5, D&C Orange No. 5, D&C Red No. 8, caramel and ferric oxide red
  • chelating agents examples include but are not limited to edetate disodium and edetic acid
  • colorants examples include but are not limited to FD&C Red No. 3, FD&C Red No. 20, FD&C Yellow No. 6, FD&C Blue No. 2, D&C Green No. 5, D&C Orange No. 5, D&C Red No. 8, caramel and ferric oxide red
  • clarifying agents examples include but are not limited to bentonite
  • emulsifying agents examples include but are not limited to acacia, cetomacrogol, cetyl alcohol, glyceryl monostearate, lecithin, sorbitan monooleate, polyoxyethylene 50 monostearate);
  • encapsulating agents examples include but are not limited to gelatin and cellulose acetate phthalate
  • flavorants examples include but are not limited to anise oil, cinnamon oil, cocoa, menthol, orange oil, peppermint oil and vanillin);
  • humectants examples include but are not limited to glycerol, propylene glycol and sorbitol
  • levigating agents examples include but are not limited to mineral oil and glycerin
  • oils examples include but are not limited to arachis oil, mineral oil, olive oil, peanut oil, sesame oil and vegetable oil
  • ointment bases examples include but are not limited to lanolin, hydrophilic ointment, polyethylene glycol ointment, petrolatum, hydrophilic petrolatum, white ointment, yellow ointment, and rose water ointment
  • penetration enhancers include but are not limited to monohydroxy or polyhydroxy alcohols, mono-or polyvalent alcohols, saturated or unsaturated fatty alcohols, saturated or unsaturated fatty esters, saturated or unsaturated dicarboxylic acids, essential oils, phosphatidyl derivatives, cephalin, terpenes, amides, ethers, ketones and ureas
  • monohydroxy or polyhydroxy alcohols mono-or polyvalent alcohols
  • saturated or unsaturated fatty alcohols saturated or unsaturated fatty esters
  • saturated or unsaturated dicarboxylic acids saturated or unsaturated dicarboxylic acids
  • essential oils phosphatidyl derivatives
  • cephalin cephalin
  • terpenes amides, ethers, ketones and ureas
  • plasticizers examples include but are not limited to diethyl phthalate and glycerol
  • solvents examples include but are not limited to ethanol, corn oil, cottonseed oil, glycerol, isopropanol, mineral oil, oleic acid, peanut oil, purified water, water for injection, sterile water for injection and sterile water for irrigation);
  • stiffening agents examples include but are not limited to cetyl alcohol, cetyl esters wax, microcrystalline wax, paraffin, stearyl alcohol, white wax and yellow wax;
  • suppository bases examples include but are not limited to cocoa butter and polyethylene glycols (mixtures));
  • surfactants examples include but are not limited to benzalkonium chloride, nonoxynol 10, oxtoxynol 9, polysorbate 80, sodium lauryl sulfate and sorbitan mono-palmitate);
  • suspending agents examples include but are not limited to agar, bentonite, carbomers, carboxymethylcellulose sodium, hydroxyethyl cellulose, hydroxypropyl cellulose, hydroxypropyl methylcellulose, kaolin, methylcellulose, tragacanth and veegum);
  • sweetening agents examples include but are not limited to aspartame, dextrose, glycerol, mannitol, propylene glycol, saccharin sodium, sorbitol and sucrose;
  • tablet anti- adherents examples include but are not limited to magnesium stearate and talc
  • tablet binders examples include but are not limited to acacia, alginic acid, carboxymethylcellulose sodium, compressible sugar, ethylcellulose, gelatin, liquid glucose, methylcellulose, and pregelatinized starch
  • tablet and capsule diluents examples include but are not limited to dibasic calcium phosphate, kaolin, lactose, mannitol, microcrystalline cellulose, powdered cellulose, precipitated calcium carbonate, sodium carbonate, sodium phosphate, sorbitol and starch
  • tablet coating agents examples include but are not limited to liquid glucose, hydroxyethyl cellulose, hydroxypropyl cellulose, hydroxypropyl methylcellulose, methylcellulose, ethylcellulose, cellulose acetate phthalate and shellac
  • tablet direct compression excipients examples include but are not limited to dibasic calcium phosphate
  • tablet disintegrants examples include but are not limited to alginic acid, carboxymethylcellulose calcium, microcrystalline cellulose, polacrillin potassium, sodium alginate, sodium starch glycollate and starch;
  • tablet glidants examples include but are not limited to colloidal silica, corn starch and talc
  • tablet lubricants examples include but are not limited to calcium stearate, magnesium stearate, mineral oil, stearic acid and zinc stearate
  • tablet/capsule opaquants examples include but are not limited to titanium dioxide
  • tablet polishing agents examples include but are not limited to carnauba wax and white wax
  • thickening agents examples include but are not limited to beeswax, cetyl alcohol and paraffin
  • tonicity agents examples include but are not limited to dextrose and sodium chloride
  • viscosity increasing agents examples include but are not limited to alginic acid, bentonite, carbomers, carboxymethylcellulose sodium, methylcellulose, sodium alginate and tragacanth); and
  • wetting agents examples include but are not limited to heptadecaethylene oxycetanol, lecithin, sorbitol monooleate, polyoxyethylene sorbitol monooleate, and polyoxyethylene stearate).
  • compositions according to the present invention can be illustrated as follows:
  • Sterile IV Solution a 5 mg/mL solution of the desired compound of this invention is made using sterile, injectable water, and the pH is adjusted if necessary. The solution is diluted for administration to 1 - 2 mg/mL with sterile 5% dextrose and is administered as an IV infusion over 60 minutes.
  • Lyophilized powder for IV administration A sterile preparation can be prepared with (i) 100 - 1000 mg of the desired compound of this invention as a lypholized powder, (ii) 32- 327 mg/mL sodium citrate, and (iii) 300 - 3000 mg Dextran 40.
  • the formulation is reconstituted with sterile, injectable saline or dextrose 5% to a concentration of 10 to 20 mg/mL, which is further diluted with saline or dextrose 5% to 0.2 - 0.4 mg/mL, and is administered either IV bolus or by IV infusion over 15 - 60 minutes.
  • Intramuscular suspension The following solution or suspension can be prepared, for intramuscular injection:
  • a large number of unit capsules are prepared by filling standard two- piece hard galantine capsules each with 100 mg of powdered active ingredient, 150 mg of lactose, 50 mg of cellulose and 6 mg of magnesium stearate.
  • Soft Gelatin Capsules A mixture of active ingredient in a digestible oil such as soybean oil, cottonseed oil or olive oil is prepared and injected by means of a positive displacement pump into molten gelatin to form soft gelatin capsules containing 100 mg of the active ingredient. The capsules are washed and dried. The active ingredient can be dissolved in a mixture of polyethylene glycol, glycerin and sorbitol to prepare a water miscible medicine mix. Tablets: A large number of tablets are prepared by conventional procedures so that the dosage unit was 100 mg of active ingredient, 0.2 mg of colloidal silicon dioxide, 5 mg of magnesium stearate, 275 mg of microcrystalline cellulose, 11 mg of starch, and 98.8 mg of lactose. Appropriate aqueous and non-aqueous coatings may be applied to increase palatability, improve elegance and stability or delay absorption.
  • a digestible oil such as soybean oil, cottonseed oil or olive oil is prepared and injected by means of a positive displacement pump into mol
  • Immediate Release Tablets/Capsules These are solid oral dosage forms made by conventional and novel processes. These units are taken orally without water for immediate dissolution and delivery of the medication.
  • the active ingredient is mixed in a liquid containing ingredient such as sugar, gelatin, pectin and sweeteners. These liquids are solidified into solid tablets or caplets by freeze drying and solid state extraction techniques.
  • the drug compounds may be compressed with viscoelastic and thermoelastic sugars and polymers or effervescent components to produce porous matrices intended for immediate release, without the need of water. Dosage of the pharmaceutical compositions of the present invention
  • the effective dosage of the compounds of this invention can readily be determined for treatment of each desired indication.
  • the amount of the active ingredient to be administered in the treatment of one of these conditions can vary widely according to such considerations as the particular compound and dosage unit employed, the mode of administration, the period of treatment, the age and sex of the patient treated, and the nature and extent of the condition treated.
  • the total amount of the active ingredient to be administered can range from about 0.001 mg/kg to about 200 mg/kg, and preferably from about 0.1 mg/kg to about 50 mg/kg body weight per day.
  • a unit dosage may preferably contain from about 5 mg to about 4000 mg of active ingredient, and can be administered one or more times per day.
  • the daily dosage for oral administration will preferably be from 0.1 to 50 mg/kg of total body weight.
  • the daily dosage for administration by injection, including intravenous, intramuscular, subcutaneous and parenteral injections, and use of infusion techniques will preferably be from 0.1 to 10 mg/kg of total body weight.
  • the daily rectal dosage regimen will preferably be from 0.1 to 50 mg/kg of total body weight.
  • the daily vaginal dosage regimen will preferably be from 0.1 to 50 mg/kg of total body weight.
  • the daily topical dosage regimen will preferably be from 0.1 to 10 mg/kg administered between one to four times daily.
  • the transdermal concentration will preferably be that required to maintain a daily dose of from 0.1 to 10 mg/kg.
  • the daily inhalation dosage regimen will preferably be from 0.1 to 10 mg/kg of total body weight. Other dosages and amounts can be selected routinely.
  • the specific initial and continuing dosage regimen for each patient will vary according to the nature and severity of the condition as determined by the attending diagnostician, the activity of the specific compound employed, the age and general condition of the patient, time of administration, route of administration, rate of excretion of the drug, drug combinations, and the like.
  • the desired mode of treatment and number of doses of a compound of the present invention or a pharmaceutically acceptable salt or ester or composition thereof can be ascertained by those skilled in the art using conventional treatment tests.
  • Combination of the compounds and compositions of the present invention with additional active ingredients Compounds of this invention can be administered as the sole pharmaceutical agent or in combination with one or more other pharmaceutical agents where the combination causes no unacceptable adverse effects. This may be of particular relevance for the treatment of hyper- proliferative diseases such as cancer.
  • the compound of this invention can be combined with known cytotoxic agents, signal transduction inhibitors, or with other anti- cancer agents, as well as with admixtures and combinations thereof.
  • the compounds of the present invention can be combined with cytotoxic anti-cancer agents.
  • cytotoxic anti-cancer agents can be found in the 11th Edition of the Merck Index (1996). These agents include, by no way of limitation, asparaginase, bleomycin, carboplatin, carmustine, chlorambucil, cisplatin, colaspase, cyclophosphamide, cytarabine, dacarbazine, dactinomycin, daunorubicin, doxorubicin (adriamycine), epirubicin, etoposide, 5-fluorouracil, hexamethylmelamine, hydroxyurea, ifosfamide, irinotecan, leucovorin, lomustine, mechlorethamine, 6-mercaptopurine, mesna, methotrexate, mitomycin C, mitoxantrone, prednisolone, prednisone, procarbazin
  • cytotoxic drugs suitable for use with the compounds of the invention include, but are not limited to, those compounds acknowledged to be used in the treatment of neoplastic diseases in Goodman and Oilman's The Pharmacological Basis of Therapeutics (Ninth Edition, 1996, McGraw-Hill).
  • agents include, by no way of limitation, aminoglutethimide, L-asparaginase, azathioprine, 5-azacytidine cladribine, busulfan, diethylstilbestrol, 2', 2'-difluorodeoxycytidine, docetaxel, erythrohydroxynonyladenine, ethinyl estradiol, 5-fluorodeoxyuridine, 5-fluorodeoxyuridine monophosphate, fludarabine phosphate, fluoxymesterone, flutamide, hydroxyprogesterone caproate, idarubicin, interferon, medroxyprogesterone acetate, megestrol acetate, melphalan, mitotane, paclitaxel, pentostatin, N-phosphonoacetyl-L-aspartate (PALA), plicamycin, semustine, teniposide, testosterone propionate, thiotepa, trimethylmelamine
  • cytotoxic anti-cancer agents suitable for use in combination with the compounds of the invention also include newly discovered cytotoxic principles such as oxaliplatin, gemcitabine, capecitabine, epothilone and its natural or synthetic derivatives, temozolomide (Quinn et al., J. Clin. Oncology 2003, 21(4), 646-651), tositumomab (Bexxar), trabedectin (Vidal et al., Proceedings of the American Society for Clinical Oncology 2004, 23, abstract 3181), and the inhibitors of the kinesin spindle protein Eg5 (Wood et al., Curr. Opin. Pharmacol. 2001, 1, 370-377).
  • cytotoxic principles such as oxaliplatin, gemcitabine, capecitabine, epothilone and its natural or synthetic derivatives, temozolomide (Quinn et al., J. Clin. Oncology 2003, 21(4), 646-651), to
  • the compounds of the present invention can be combined with other signal transduction inhibitors.
  • signal transduction inhibitors which target the EGFR family, such as EGFR, HER-2, and HER-4 (Raymond et al., Drugs 2000, 60 (Suppl.l), 15-23; Harari et al., Oncogene 2000, 19 (53), 6102-6114), and their respective ligands.
  • Examples of such agents include, by no way of limitation, antibody therapies such as Herceptin (trastuzumab), Erbitux (cetuximab), and pertuzumab.
  • Examples of such therapies also include, by no way of limitation, small-molecule kinase inhibitors such as ZD- 1839 / Iressa (Baselga et al., Drugs 2000, 60 (Suppl. 1), 33-40), OSI-774 / Tarceva (Pollack et al. J. Pharm. Exp. Ther. 1999, 291(2), 739-748), CI-1033 (Bridges, Curr. Med. Chem.
  • small-molecule kinase inhibitors such as ZD- 1839 / Iressa (Baselga et al., Drugs 2000, 60 (Suppl. 1), 33-40), OSI-774 / Tarceva (Pollack et al. J. Pharm. Exp. Ther. 1999, 291(2), 739-748), CI-1033 (Bridges, Curr. Med. Chem.
  • the compounds of the present invention can be combined with other signal transduction inhibitors targeting receptor kinases of the split-kinase domain families (VEGFR, FGFR, PDGFR, flt-3, c-kit, c-fms, and the like), and their respective ligands.
  • These agents include, by no way of limitation, antibodies such as Avastin (bevacizumab).
  • These agents also include, by no way of limitation, small-molecule inhibitors such as STI-571 / Gleevec (Zvelebil, Curr. Opin. Oncol., Endocr. Metab. Invest. Drugs 2000, 2(1), 74-82), PTK-787 (Wood et al., Cancer Res.
  • the compounds of the present invention can be combined with inhibitors of the Raf/MEK/ERK transduction pathway (Avruch et al., Recent Prog. Horm. Res. 2001, 56, 127-155), or the PKB (akt) pathway (Lawlor et al., J. Cell Sci. 2001, 114, 2903-2910).
  • inhibitors of the Raf/MEK/ERK transduction pathway Avruch et al., Recent Prog. Horm. Res. 2001, 56, 127-155
  • PKB akt pathway
  • the compounds of the present invention can be combined with inhibitors of histone deacetylase.
  • histone deacetylase examples include, by no way of limitation, suberoylanilide hydroxamic acid (SAHA), LAQ-824 (Ottmann et al., Proceedings of the American Society for Clinical Oncology 2004, 23, abstract 3024), LBH-589 (Beck et al., Proceedings of the American Society for Clinical Oncology 2004, 23, abstract 3025), MS- 275 (Ryan et al., Proceedings of the American Association of Cancer Research 2004, 45, abstract 2452), and FR-901228 (Piekarz et al., Proceedings of the American Society for Clinical Oncology 2004, 23, abstract 3028).
  • SAHA suberoylanilide hydroxamic acid
  • LAQ-824 Ottmann et al., Proceedings of the American Society for Clinical Oncology 2004, 23, abstract 3024
  • LBH-589 Beck et al., Proceedings of the American Society for Clinical Oncology 2004, 23, abstract 3025
  • the compounds of the present invention can be combined with other anti-cancer agents such as proteasome inhibitors, and m-TOR inhibitors.
  • anti-cancer agents such as proteasome inhibitors, and m-TOR inhibitors.
  • proteasome inhibitors include, by no way of limitation, bortezomib (Mackay et al., Proceedings of the American Society for Clinical Oncology 2004, 23, Abstract 3109), and CCI-779 (Wu et al., Proceedings of the American Association of Cancer Research 2004, 45, abstract 3849).
  • cytotoxic and/or cytostatic anti-cancer agent in combination with a compound or composition of the present invention for the treatment of cancer will serve to: (1) yield better efficacy in reducing the growth of a tumor or even eliminate the tumor as compared to administration of either agent alone, (2) provide for the administration of lesser amounts of the administered chemotherapeutic agents,
  • the present invention provides for the following aspects
  • a pharmaceutically acceptable salt of synthetic metabolite according to Aspect 2 which is
  • a) a basic salt of an organic acid or inorganic acid which is hydrochloric acid, hydrobromic acid, sulfuric acid, phosphoric acid, methanesulfonic acid, trifluoromethanesulfonic acid, benzenesulfonic acid, p-toluene sulfonic acid (tosylate salt), 1-napthalene sulfonic acid, 2-napthalene sulfonic acid, acetic acid, trifluoroacetic acid, malic acid, tartaric acid, citric acid, lactic acid, oxalic acid, succinic acid, fumaric acid, maleic acid, benzoic acid, salicylic acid, phenylacetic acid, or mandelic acid; or
  • Aspect 4 The synthetic metabolite according to Aspect 2 which is a metabolite of 4 ⁇ 4- [3-(4-chloro-3-trifluoromethylphenyl)-ureido]-3-fluorophenoxy ⁇ -pyridine-2-carboxylic acid methylamide.
  • a pharmaceutically acceptable salt of a synthetic metabolite according to Aspect 4 which is a basic salt of hydrochloric acid, hydrobromic acid, sulfuric acid, phosphoric acid, methanesulfonic acid, trifluoromethanesulfonic acid, benzenesulfonic acid, p-toluene sulfonic acid (tosylate salt), 1-napthalene sulfonic acid, 2-napthalene sulfonic acid, acetic acid, trifluoroacetic acid, malic acid, tartaric acid, citric acid, lactic acid, oxalic acid, succinic acid, fumaric acid, maleic acid, benzoic acid, salicylic acid, phenylacetic acid, or mandelic acid.
  • Aspect 6 A compound which is which is a hydrochloride, benzenesulfonate, or methanesulfonate salt of the synthetic metabolite according to Aspect 2.
  • a pharmaceutical composition comprising a synthetic metabolite according to Aspect 2 and a physiologically acceptable carrier.
  • a pharmaceutical composition comprising a synthetic metabolites according to Aspect 4 and a physiologically acceptable carrier.
  • a pharmaceutical composition for the treatment of a disease in a human or other mammal regulated by a protein kinase, associated with an aberration in the protein kinase signal transduction pathway comprising a synthetic metabolite according to Aspect 2 and a physiologically acceptable carrier.
  • a pharmaceutical composition for the treatment of a hyper-proliferative disorder comprising a synthetic metabolite according to Aspect 4 and a physiologically acceptable carrier.
  • a pharmaceutical composition for the treatment of a cancerous cell growth comprising a synthetic metabolite according to Aspect 2 and a physiologically acceptable carrier.
  • a method for regulating tyrosine kinase signal transduction comprising administering to a human or other mammal a synthetic metabolite according to Aspect 2.
  • a method for treating or preventing a disease in a human or other mammal which is regulated by tyrosine kinase and associated with an aberration in the tyrosine kinase signal transduction pathway comprising administering to a human or other mammal a synthetic metabolite according to Aspect 2.
  • a method for treating or preventing a disease in a human and/or other mammal which is a VEGFR-2 mediated disorder comprising administering to a human or other mammal a synthetic metabolite according to Aspect 2.
  • Aspect 15. A method for treating or preventing a disease in a human and/or other mammal which is a PDGFR mediated disorder, said method comprising administering to a human or other mammal a synthetic metabolite according to Aspect 2.
  • Aspect 16 A method for treating or preventing a disease in a human or other mammal which is a raf-mediated disorder, said method comprising administering to a human or other mammal a synthetic metabolite according to Aspect 2.
  • a method for treating or preventing a disease in a human or other mammal which is a p38-mediated disorder comprising administering to a human or other mammal a synthetic metabolite according to Aspect 2.
  • Aspect 18 A method for treating or preventing a disease in a human or other mammal which is a VEGF-mediated disorder, said method comprising administering to a human or other mammal a synthetic metabolite according to Aspect 2.
  • a method for treating or preventing a disease in a human or other mammal which is a hyper-proliferative, inflammatory and/or angiogenesis disorder which comprises administering to a human or other mammal a synthetic metabolite according to Aspect 2.
  • Aspect 20 A method for treating or preventing a disease in a human or other mammal which is a hyper-proliferative disorder which comprises administering to a human or other mammal a synthetic metabolite according to Aspect 2.
  • Aspect 21 A method as in aspect 20, wherein the hyper-proliferative disorder is cancer.
  • Aspect 22 A method as in aspect 21, wherein said method comprises administering to a human or other mammal a synthetic metabolite according to Aspect 2 in combination with one or several additional anti-cancer agents.
  • Aspect 23 A method for treating or preventing a disease in a human or other mammal characterized by abnormal angiogenesis or hyperpermiability processes comprising administering to a human or other mammal a synthetic metabolite according to Aspect 2.
  • Aspect 24 A method as in aspect 23, for treating or preventing a disease in a human or other mammal characterized by abnormal angiogenesis or hyperpermeability processes, comprising administering to a human or other mammal, a synthetic metabolite according to Aspect 2 simultaneously with another anti-angiogenesis agent, either in the same formulation or in separate formulations.
  • Aspect 25 A method for treating or preventing one or more of the following conditions in humans and/or other mammals:
  • rheumatoid arthritis psoriasis, a bullous disorder associated with subepidermal blister formation, including bullous pemphigoid, erythema multiforme, or dermatitis herpetiformis, rheumatoid arthritis, osteoarthritis, septic arthritis, tumor metastasis, periodontal disease, cornal ulceration, proteinuria and coronary thrombosis from atherosclerotic plaque, aneurismal aortic, birth control, dystrophobic epidermolysis bullosa, degenerative cartilage loss following traumatic joint injury, osteopenias mediated by MMP activity, tempero mandibular joint disease or demyelating disease of the nervous system,
  • said method comprising administering to a human or other mammal, a synthetic metabolite according to Aspect 2.
  • a method for treating or preventing one or more of the following conditions in humans and/or other mammals tumor growth, retinopathy, ischemic retinal-vein occlusion, retinopathy of prematurity, age related macular degeneration; rheumatoid arthritis, psoriasis, a bullous disorder associated with subepidermal blister formation, including bullous pemphigoid, erythema multiforme, or dermatitis herpetiformis;
  • rheumatic fever fever, bone resorption, postmenopausal osteoporosis, sepsis, gram negative sepsis, septic shock, endotoxic shock, toxic shock syndrome, systemic inflammatory response syndrome, inflammatory bowel disease (Krohn's disease and ulcerative colitis), Jarisch- Herxheimer reaction, asthma, adult respiratory distress syndrome, acute pulmonary fibrotic disease, pulmonary sarcoidosis, allergic respiratory disease, silicosis, coal worker's pneumoconiosis, alveolar injury, hepatic failure, liver disease during acute inflammation, severe alcoholic hepatitis, malaria (Plasmodium falciparum malaria and cerebral malaria), non- insulin-dependent diabetes mellitus (NIDDM), congestive heart failure, damage following heart disease, atherosclerosis, Alzheimer's disease, acute encephalitis, brain injury, multiple sclerosis (demy elation and oligiodendrocyte loss in multiple sclerosis), advanced cancer, lymphoid malign
  • said method comprising administering to a human or other mammal a synthetic metabolite according to Aspect 2.
  • a method for treating or preventing one or more of the following conditions in humans and/or other mammals tumor growth, retinopathy, diabetic retinopathy, ischemic retinal-vein occlusion, retinopathy of prematurity, age related macular degeneration; rheumatoid arthritis, psoriasis, bullous disorder associated with subepidermal blister formation, bullous pemphigoid, erythema multiforme, and dermatitis herpetiformis, in combination with an infectious disease selected from the group consisting of :
  • tuberculosis Helicobacter pylori infection during peptic ulcer disease, Chaga's disease resulting from Trypanosoma cruzi infection, effects of Shiga-like toxin resulting from E. coli infection, effects of enterotoxin A resulting from Staphylococcus infection, meningococcal infection, and infections from Borrelia burgdorferi, Treponema pallidum, cytomegalovirus, influenza virus, Theiler's encephalomyelitis virus, and the human immunodeficiency virus (HIV);
  • HSV human immunodeficiency virus
  • said method comprising administering to a human or other mammal a synthetic metabolite according to Aspect 2.
  • a method as in aspect 22 wherein the additional anti-cancer agent is selected from the group consisting of asparaginase, bleomycin, carboplatin, carmustine, chlorambucil, cisplatin, colaspase, cyclophosphamide, cytarabine, dacarbazine, dactinomycin, daunorubicin, doxorubicin (adriamycine), epirubicin, etoposide, 5-fluorouracil, hexamethylmelamine, hydroxyurea, ifosfamide, irinotecan, leucovorin, lomustine, mechlorethamine, 6-mercaptopurine, mesna, methotrexate, mitomycin C, mitoxantrone, prednisolone, prednisone, procarbazine, raloxifen, streptozocin, tamoxifen, thioguanine, to
  • trastuzumab trastuzumab, cetuximab, bevacizumab, pertuzumab, ZD- 1839 (Iressa), OSI-774 (Tarceva), CI-1033, GW-2016, CP-724,714, HKI-272, EKB-569, STI-571 (Gleevec), PTK-787, SU-11248, ZD-6474, AG-13736, KRN-951, CP-547,632, CP-673,451, CHIR-258, MLN-518, AZD-2171, PD-325901, ARRY-142886, suberoylanilide hydroxamic acid (SAHA), LAQ-824, LBH-589, MS-275, FR-901228, bortezomib, and CCI-779.
  • SAHA suberoylanilide hydroxamic acid
  • a method as in aspect 22 wherein the additional anti-cancer agent is a cytotoxic agent selected from the group consisting of DNA topoisomerase I and II inhibitors, DNA intercalators, alkylating agents, anti-metabolites, cell-cycle blockers, microtubule disruptors, and Eg5 inhibitors.
  • Aspect 30. A method as in aspect 22 wherein the additional anti-cancer agent is selected from the group consisting of inhibitors of growth factor receptor signaling, histone deacetylase inhibitors, inhibitors of the PKB pathway, inhibitors of the Raf/MEK/ERK pathway, inhibitors of the mTOR pathway, and proteasome inhibitors.
  • Aspect 31. A method for treating or preventing one or more of the following conditions in humans and/or other mammals:
  • rheumatic fever fever, bone resorption, postmenopausal osteoporosis, sepsis, gram negative sepsis, septic shock, endotoxic shock, toxic shock syndrome, systemic inflammatory response syndrome, inflammatory bowel disease (Krohn's disease and ulcerative colitis), Jarisch- Herxheimer reaction, asthma, adult respiratory distress syndrome, acute pulmonary fibrotic disease, pulmonary sarcoidosis, allergic respiratory disease, silicosis, coal worker's pneumoconiosis, alveolar injury, hepatic failure, liver disease during acute inflammation, severe alcoholic hepatitis, malaria (Plasmodium falciparum malaria and cerebral malaria), non- insulin-dependent diabetes mellitus (NIDDM), congestive heart failure, damage following heart disease, atherosclerosis, Alzheimer's disease, acute encephalitis, brain injury, multiple sclerosis (demy elation and oligiodendrocyte loss in multiple sclerosis), advanced cancer, lymphoid malign
  • said method comprising administering to a human or other mammal, a synthetic metabolite according to Aspect 2.
  • Aspect 32 A method for treating or preventing one or more of the following conditions in humans and/or other mammals:
  • E. coli infection effects of enterotoxin A resulting from Staphylococcus infection, meningococcal infection, and infections from Borrelia burgdorferi, Treponema pallidum, cytomegalovirus, influenza virus, Theiler's encephalomyelitis virus, and the human immunodeficiency virus (HIV) ,
  • said method comprising administering to a human or other mammal, a synthetic metabolite according to Aspect 2.
  • Aspect 33 A method for treating or preventing osteoporosis, inflammation, and angiogenesis disorders, with the exclusion of cancer, in a human and/or other mammal by administering an effective amount of a synthetic metabolite according to Aspect 2 to said mammal.
  • a method for treating or preventing cancer in a human or other mammal which comprises administering to a human or other mammal a single active principle combining inhibition of tumor cell proliferation mediated by the raf / MEK / ERK pathway, and inhibition of angiogenesis mediated by PDGF and VEGF, wherein said active principle comprises a synthetic metabolite according to Aspect 2.
  • Aspect 35 A method of aspect 34 where said inhibition of tumor cell proliferation is caused by inhibition of raf kinase, and said inhibition of angiogenesis is caused by dual inhibition of PDGFR-beta and VEGFR-2 kinases.
  • Aspect 36 Aspect 36.
  • a method for treating or preventing cancer in a human or other mammal which comprises administering to a human or other mammal a single active principle combining inhibition of tumor cell proliferation mediated by the raf pathway, and inhibition of angiogenesis mediated by PDGF or VEGF, wherein said active principle comprises a synthetic metabolite according to Aspect 2.
  • Aspect 37 A method of treating and/or preventing a disease and/or condition in a subject in need thereof, comprising administering an effective amount of a synthetic metabolite according to Aspect 2.
  • Aspect 38. A method of aspect 37, wherein said method comprises causing tumor regression in a subject or cells therefrom.
  • Aspect 39 A method of aspect 37, wherein said method comprises inhibiting lymphangiogenesis .
  • Aspect 40 A method of aspect 37, wherein said method comprises inhibiting angiogenesis.
  • Aspect 41 A method of aspect 37, wherein said method comprises inhibiting lymphangiogenesis and angiogenesis.
  • a method of aspect 37, wherein said method comprises stimulating the proliferation of hematopoietic progenitor cells.
  • a method of aspect 37, wherein said method comprises treating a disorder in a mammalian subject mediated by raf, VEGFR-2, VEGFR-3, PDGFR-beta, p38 and/or fit- 3.
  • a method of aspect 37 comprising determining whether a condition can be modulated by said compound, comprising measuring the expression or activity of raf, VEGFR-2, VEGFR-3, PDGFR-beta, p38 and/or flt-3, in a sample comprising cells or a cell extract, wherein said ample is obtained from a subject or cell having said condition, whereby said condition can be modulated by said compound when said expression or activity is different in said condition as compared to a normal control.
  • a method of aspect 44 further comprising comparing the expression in said sample to said normal control.
  • a method of aspect 37, wherein said method comprises assessing the efficacy of said compound disorder, comprising administering said compound, measuring the expression or activity of raf, VEGFR-2, VEGFR-3, PDGFR-beta, p38, and/or flt-3, and determining the effect of said compound on said expression or activity.
  • a method of aspect 37 wherein said method comprises determining the presence of raf, VEGFR-2, VEGFR-3, PDGFR-beta, p38 and/or flt-3 in a sample of a biological material, contacting said sample with said compound, and determining whether said compound binds to said material.
  • a method of aspect 37, wherein said method comprises treating a tumor in a subject in need thereof, comprising administering an effective amount of said compound wherein said amount is effective to inhibit tumor cell proliferation and neovascularization.
  • a method for treating or preventing a tumor or vascular disease in a subject comprising administering to said subject, a synthetic metabolite according to aspect 49.
  • Claim 51 A method of making the synthetic metabolite according to aspect 2, comprising contacting the compound of formula I or a salt thereof with a liver microsome preparation under conditions sufficient for the metabolism of said compound of Formula I and isolating the metabolites from said preparation.
  • Fig. 1 shows metabolites of regorafenib from in vitro and in vivo studies.
  • Fig. 2 shows kinase selectivity profile of regorafenib and its N-oxide (M-2) and desmethyl-N- oxide (M-5) synthetic metabolites.
  • Fig. 3 shows that M-2 and M-5 synthetic metabolites significantly inhibit tumor growth of human xenografts in mice.
  • Fig. 4 shows that regorafenib and its synthetic metabolites M-2 and M-5 inhibit the acute hypotensive effect of VEGF in anesthetized rats.
  • Fig. 5 shows that the M-2 synthetic metabolite of regorafenib affects vasculature extravasation in a rat GS9L glioblastoma model after a single oral dose of 7.5 mg/kg. Assays were performed using kinetic DCE-MRI analysis.
  • the compound of example 1 as a free base (2.0 g) was dissolved in anhydrous tetrahydrofuran (15 mL) and a 4M HCl/dioxane was added (excess). The solution was then concentrated in vacuo to afford 2.32 grams of off-white solids.
  • the crude salt was dissolved in hot ethanol (125 mL), activated carbon was added and the mixture heated at reflux for 15 minutes. The hot suspension was filtered through a pad of Celite 521 and allowed to cool to room temperature. The flask was placed in a freezer overnight. The crystalline solids were collected by suction filtration, washed with ethanol, then hexane and air-dried.
  • the compound of example 1 as a free base (2.25 g) was dissolved in ethanol (100 mL) and a stock solution of methanesulfonic acid (excess) was added. The solution was then concentrated in vacuo to afford a yellow oil. Ethanol was added and concentration repeated, affording 2.41 g of off-white solids.
  • the crude salt was dissolved in hot ethanol (-125 mL) and then cooled slowly to crystallize. After reaching room temperature, the flask was placed in a freezer overnight. The colorless crystalline material was collected by suction filtration; the filter cake was washed with ethanol, then hexane and air-dried, to afford 2.05 g of material, which was dried in a vacuum oven at 60 °C overnight.
  • the synthetic metabolites of the compounds of Formula I can be obtained by incubating the parent regorafenib compound with liver microsomes.
  • the metabolites of the present application can also be synthetically obtained.
  • the metabolites of the present application can be purified using techniques that are known in the art.
  • N-oxide (M-2) and hydroxymethyl (M-3) metabolites were identified as metabolites of regorafenib in vitro upon incubation with liver enzymes of various mammalian species (man, dog, rat, mouse). Studies reveal that and its metabolites show high protein binding in man and animal species.
  • a kinome-wide selectivity profiling of regorafenib and its metabolites M-2 and M-5 was performed by Ambit Biosciences (San Diego, CA, US) using an active-site competitive binding assay (Fabian et al. Nat Biotechnol 2005; 23: 329-336).
  • a total of 402 kinases were analyzed using a single dose of 1 ⁇ of compound. Binding inhibition activities are displayed as the percentage of the kinase that remained bound compared with the DMSO- treated control. The potency of the compound is reflected by the size of the circle.
  • M-2 and M-5 showed kinase selectivity profiles similar to but distinct from regorafenib. Results are shown in Fig. 2.
  • Example 9 Biochemical characterization of M-2 and M-5 metabolites
  • M-2 and M-5 demonstrated potent pharmacologic activities.
  • M-2 and M-5 showed an inhibition profile similar to but distinct from regorafenib.
  • M-2 and M-5 inhibited key targets such as vascular endothelial growth factor (VEGF) receptor 2, TIE-2, and mutant and wild-type c-KIT and B- RAF, with IC50 values very similar to regorafenibfor M-2 and somewhat higher for M-5.
  • VEGF vascular endothelial growth factor
  • mice bearing xenografts of the human breast cancer cell lineMDA-MB-231 (K-RASG13D, B-RAFG464V) or the human colorectal cancer cell line HT-29 (B- RAFV600E) were treated orally (qd x 27) with 10 mg/kg of regorafenib, M-2 or M-5, starting at day 11 after tumor inoculation.
  • the tumor growth inhibitory assays were performed using DCE-MRI.
  • TGI tumor growth inhibition
  • VEGF vascular endothelial growth factor
  • Results are presented in Fig. 5.
  • the left panel shows results for regorafenib and the right panel shows results for the M-2 synthetic metabolite.
  • the left y-axis indicates tumor AUC values, which were normalized to thigh muscle.
  • the right y-axis shows serum levels of regorafenib, M-2, M-4 and M-5. The contribution of regorafenib, M-2, M-4 and M-5 could therefore be independently assessed.
  • Example 13 In vivo pharmacological studies
  • the c-raf biochemical assay is performed with a c-raf enzyme that was activated (phosphorylated) by Lck kinase.
  • Lck-activated c-raf (Lck/c-raf) is first produced in Sf9 insect cells by co-infecting cells with baculoviruses expressing, under the control of the polyhedrin promoter, GST-c-raf (from amino acid 302 to amino acid 648) and Lck (full-length). Both baculoviruses are used at the multiplicity of infection of 2.5 and the cells are harvested 48 h post infection.
  • MEK-1 protein is produced in Sf9 insect cells by infecting cells with the baculovirus expressing GST-MEK-1 (full-length) fusion protein at the multiplicity of infection of 5 and harvesting the cells 48 hours post infection. Similar purification procedure is used for GST- c-raf 302-648 and GST-MEK-1.
  • Transfected cells are suspended at 100 mg of wet cell biomass per mL in a buffer containing 10 mM sodium phosphate, 140 mM sodium chloride pH 7.3, 0.5% Triton X-100 and the protease inhibitor cocktail.
  • the cells are disrupted with Polytron homogenizer and centrifuged 30,000g for 30 minutes. The 30,000g supernatant is applied onto GSH- Sepharose.
  • the resin is washed with a buffer containing 50 mM Tris, pH 8.0, 150 mM NaCl and 0.01% Triton X-100.
  • the GST-tagged proteins are eluted with a solution containing 100 mM Glutathione, 50 mM Tris, pH 8.0, 150 mM NaCl and 0.01% Triton X-100.
  • the purified proteins are dialyzed into a buffer containing 20 mM Tris, pH 7.5, 150 mM NaCl and 20% Glycerol.
  • Test compounds are serially diluted in DMSO using three-fold dilutions to stock concentrations ranging typically from 50 ⁇ to 20 nM (final concentrations in the assay range from 1 ⁇ to 0.4 nM).
  • the c-Raf biochemical assay is performed as a radioactive filtermat assay in 96-well Costar polypropylene plates (Costar 3365). The plates are loaded with 75 ⁇ L solution containing 50 mM HEPES pH 7.5, 70 mM NaCl, 80 ng of Lck/c-raf and 1 ⁇ g MEK-1. Subsequently, 2 ⁇ of the serially diluted individual compounds are added to the reaction, prior to the addition of ATP.
  • the reaction is initiated with 25 ⁇ ATP solution containing 5 ⁇ ATP and 0.3 ⁇ Ci [33P]-ATP.
  • the plates are sealed and incubated at 32 oC for 1 h.
  • the reaction is quenched with the addition of 50 ⁇ of 4 % Phosphoric Acid and harvested onto P30 filtermats (PerkinElmer) using a Wallac Tomtec Harvester. Filtermats are washed with 1 % Phosphoric Acid first and deinonized H 2 O second.
  • the filters are dried in a microwave, soaked in scintillation fluid and read in a Wallac 1205 Betaplate Counter (Wallac Inc., Atlanta, GA, U.S.A.). The results are expressed as percent inhibition.
  • Purified and His-tagged p38 (expressed in E. Coli) is activated in vitro by MMK-6 to a high specific activity.
  • MMK-6 MMK-6
  • all reactions are conducted in 100 ⁇ volumes with reagents diluted to yield 0.05 ⁇ g/well of activated p38 and 10 ⁇ g/well of myelin basic protein in assay buffer (25 mM HEPES 7.4, 20 mM MgC12, 150 mM NaCl).
  • Test compounds (5 ⁇ of a 10% DMSO solution in water) are prepared and diluted into the assay to cover a final concentration range from 5 nM to 2.5 ⁇ .
  • the kinase assay is initiated by addition of 25 ⁇ of an ATP cocktail to give a final concentration of 10 ⁇ cold ATP and 0.2 ⁇ [gamma-33P] ATP per well (200-400 dpm/pmol of ATP).
  • the plate is incubated at 32 oC for 35 min., and the reaction quenched with 7 of a 1 N aq HCl solution.
  • the samples are harvested onto a P30 Filtermat (Wallac, Inc.) using a TomTec 1295 Harvester (Wallac, Inc.), and counted in a LKB 1205 Betaplate Liquid Scintillation Counter (Wallac, Inc.). Negative controls include substrate plus ATP alone.
  • SW1353 cellular assay SW1353 cells (human chondro-sarcoma) are seeded (1000 cells/100 ⁇ L ⁇ DMEM 10% FCS/well) into 96-well plates and incubated overnight. After medium replacement, cells are exposed to test compounds for 1 h at 37 oC, at which time human IL-1 (1 ng/mL, Endogen, Woburn, WA) and recombinant human TNFalpha (10 ng/mL) are added. Cultures are incubated for 48 h at 37 oC, then supernatant IL-6 values are determined by ELISA. The compounds of this invention show significant inhibition of p38 kinase.
  • Example 16 Bio-Plex Phospho-ERK 1 ⁇ 2 immunoassay.
  • MDA-MB-231 cells are plated at 50,000 cells per well in 96 well microtitre plates in complete growth media. For effects of test compounds on basal pERKl/2 inhibition, the next day after plating, MDA- MB-231 cells are transferred to DMEM with 0.1% BSA and incubated with test compounds diluted 1:3 to a final concentration of 3 ⁇ to 12 nM in 0.1% DMSO. Cells are incubated with test compounds for 2 h, washed, and lysed in Bio-Plex whole cell lysis buffer A.
  • Samples are diluted with buffer B 1:1 (v/v) and directly transferred to assay plate or frozen at -80 C degrees until processed.
  • 50 ⁇ L ⁇ of diluted MDA-MB-231 cell lysates are incubated with about 2000 of 5 micron Bio-Plex beads conjugated with an anti-ERKl/2 antibody overnight on a shaker at room temperature.
  • biotinylated phospho-ERKl/2 sandwich immunoassay is performed, beads are washed 3 times during each incubation and then 50 ⁇ L ⁇ of PE-strepavidin is used as a developing reagent.
  • the relative fluorescence units of pERKl/2 are detected by counting 25 beads with Bio-Plex flow cell (probe) at high sensitivity.
  • the IC50 is calculated by taking untreated cells as maximum and no cells (beads only) as background using in an Excel spreadsheet based program.
  • the compound of this invention shows significant inhibition in this assay.
  • This assay is performed in 96-well opaque plates (Costar 3915) in the TR-FRET format. Reaction conditions are as follows: 10 ⁇ ATP , 25 nM poly GT-biotin , 2 nM Eu-labelled phospho-Tyr Ab, 10 nM APC, 7 nM Flk-1 (kinase domain), 1% DMSO, 50 mM HEPES pH 7.5, 10 mM MgC12, 0.1 mM EDTA, 0.015% BRIJ, 0.1 mg/mL BSA, 0.1% mercapto- ethanol). Reaction is initiated upon addition of enzyme. Final reaction volume in each well is 100 ⁇ .
  • Plates are read at both 615 and 665 nM on a Perkin Elmer Victor V Multilabel counter at about 1.5- 2.0 hours after reaction initiation. Signal is calculated as a ratio: (665 nm / 615 nm) * 10000 for each well.
  • the compounds of this invention show significant inhibition of VEGFR2 kinase.
  • This assay is formatted in a 96-well black plate (Costar 3915).
  • the following reagents are used: Europium-labeled anti-phosphotyrosine antibody pY20 (Perand streptavidin-APC; poly GT-biotin from, and mouse PDGFR.
  • reaction conditions are as follows: 1 nM mouse PDGFR is combined with 20 ⁇ ATP, 7 nM poly GT-biotin, 1 nM pY20 antibody, 5 nM streptavidin-APC, and 1% DMSO in assay buffer (50 mM HEPES pH 7.5, 10 mM MgC12, 0.1 mM EDTA, 0.015% BRIJ 35, 0.1 mg/mL BSA, 0.1% mercaptoethanol). Reaction is initiated upon addition of enzyme. Final reaction volume in each well is 100 ⁇ . After 90 minutes, the reaction is stopped by addition of 10 ⁇ L/well of 5 ⁇ staurosporine.
  • IC50 generation for both PDGFR and Flk-1, compounds are added prior to the enzyme initiation.
  • a 50-fold stock plate was made with compounds serially diluted 1:3 in a 50% DMSO/50% dH20 solution.
  • a 2 ⁇ addition of the stock to the assay gives final compound concentrations ranging from 10 ⁇ - 4.56 nM in 1% DMSO.
  • Human breast carcinoma cells (MDA MB-231, NCI) are cultured in standard growth medium (DMEM) supplemented with 10% heat-inactivated FBS at 37oC in 5% C02 (vol/ vol) in a humidified incubator. Cells are plated at a density of 3000 cells per well in 90 ⁇ growth medium in a 96 well culture dish. In order to determine TOh CTG values, 24 hours after plating, 100 ⁇ L ⁇ of CellTiter-Glo Luminescent Reagent (Promega) is added to each well and incubated at room temperature for 30 minutes. Luminescence is recorded on a Wallac Victor II instrument. The CellTiter-Glo reagent results in cell lysis and generation of a luminescent signal proportional to the amount of ATP present, which, in turn is directly proportional to the number of cells present.
  • DMEM standard growth medium
  • FBS heat-inactivated FBS
  • C02 vol/ vol
  • TOh CTG values 24 hours after plating, 100 ⁇
  • Test compounds are dissolved in 100% DMSO to prepare 10 mM stocks. Stocks are further diluted 1:400 in growth medium to yield working stocks of 25 ⁇ test compound in 0.25% DMSO. Test compounds are serially diluted in growth medium containing 0.25% DMSO to maintain constant DMSO concentrations for all wells. 60 ⁇ L ⁇ of diluted test compound are added to each culture well to give a final volume of 180 ⁇ L ⁇ . The cells with and without individual test compounds are incubated for 72 hours at which time ATP dependent luminescence is measured, as described previously, to yield T72h values. Optionally, the IC50 values can be determined with a least squares analysis program using compound concentration versus percent inhibition.
  • the compound of this invention shows significant inhibition of proliferation using this assay.
  • Example 20 pPDGFR-beta sandwich ELISA in AoSMC cells
  • 100K P3-P6 Aortic SMC are plated in each well of 12-well cluster in 1000 volume/ well of SGM-2 using standard cell culture techniques. Next day, cells are rinsed with 1000 ⁇ L ⁇ D- PBS once, then serum starved in 500 ⁇ L ⁇ SBM (smooth muscle cell basal media) with 0.1% BSA overnight. Compounds are diluted at a dose range from (10 ⁇ to 1 nM in 10-fold dilution steps in DMSO. Final DMSO concentration 0.1%). Remove old media by inversion into the sink quickly then add 100 ⁇ L ⁇ of each dilution to corresponding well of cells for 1 h at 37 0C.
  • Cells are then stimulated with 10 ng/mL PDGF-BB ligand for 7 min at 37 0C.
  • the media is decanted and 150 ⁇ L ⁇ of isotonic lysis buffer with protease inhibitor tablet (Complete; EDTA-free) and 0.2 mM Na vanadate is added.
  • Cells are lysed for 15 min at 4 0C on shaker in cold room. Lysates are put in eppendorf tubes to which 15 ⁇ L ⁇ of agarose- conjugated anti-PDGFR-beta antibody is added and incubated at 4 0C overnight. Next day, beads are rinsed in 50-volumes of PBS three times and boiled in lx LDS sample buffer for 5 minutes.
  • Table A illustrates the results of a representative in vitro kinase biochemical assay for p38 kinase, PDGFR kinase and VEGFR2 kinase. These three kinase targets are all involved in stroma activation and endothelial cell proliferation, leading to angiogenesis, and providing blood supply to the tumor tissue.
  • Table B illustrates representative results of two cellular assays for raf kinase activity, which are (i) inhibition of pERK in MDA-MB231 cells, a mechanistic readout of raf kinase activity, and (ii) a proliferation assay of MDA-MB231 cells, a functional assay of raf kinase activity.
  • Table B illustrates the results of PDGFR driven phosphorylation of PDGFR-beta in aortic smooth muscle cells, which is a mechanistic readout of PDGFR kinase inhibition.
  • compounds of the present invention provide a unique combination of inhibition of angiogenesis and tumor cell proliferation. They also possess an improved inhibition profile against several key kinase targets such as raf, p38, PDGFR, and VEGFR-2, which are all molecular targets of interest for the treatment of osteoporosis, inflammatory diseases, and hyper-proliferative diseases, including cancer.
  • key kinase targets such as raf, p38, PDGFR, and VEGFR-2, which are all molecular targets of interest for the treatment of osteoporosis, inflammatory diseases, and hyper-proliferative diseases, including cancer.

Abstract

Synthetic metabolites of compounds of Formula (I); salts thereof, prodrugs thereof, pharmaceutical compositions containing such synthetic metabolites, and use of such compound and compositions to treat diseases mediated by raf, VEGFR, PDGFR, p38 and flt-3. Described are M2, M3, M4, and M5 synthetic metabolites of compounds of formula I, wherein the structures of said compound of formula I and said M2, M3, M4, and M5 synthetic metabolites are:

Description

Synthetic metabolites of Fluoro substituted Omega- Carboxyaryl Diphenyl Urea for the Treatment and Prevention of Diseases and Conditions
Field of the Invention
This invention relates to novel compounds, pharmaceutical compositions containing such compounds and the use of those compounds or compositions for treating diseases and conditions mediated by abnormal VEGFR, PDGFR, raf, p38, and/or flt-3 kinase signaling, either alone or in combination with anti-cancer agents.
Background of the Invention
Activation of the ras signal transduction pathway indicates a cascade of events that have a profound impact on cellular proliferation, differentiation, and transformation. Raf kinase, a downstream effector of ras, is recognized as a key mediator of these signals from cell surface receptors to the cell nucleus (Lowy, D. R.; Willumsen, B. M. Ann. Rev. Biochem. 1993, 62, 851; Bos, J. L. Cancer Res. 1989, 49, 4682). It has been shown that inhibiting the effect of active ras by inhibiting the raf kinase signaling pathway by administration of deactivating antibodies to raf kinase or by co-expression of dominant negative raf kinase or dominant negative MEK, the substrate of raf kinase, leads to the reversion of transformed cells to the normal growth phenotype (see: Daum et al. Trends Biochem. Sci. 1994, 19, 474-80; Fridman et al. J. Biol. Chem. 1994, 269, 30105-8. Kolch et al. (Nature 1991, 349, 426-28) have further indicated that inhibition of raf expression by antisense RNA blocks cell proliferation in membrane-associated oncogenes. Similarly, inhibition of raf kinase (by antisense oligodeoxynucleotides) has been correlated in vitro and in vivo with inhibition of the growth of a variety of human tumor types (Monia et al., Nat. Med. 1996, 2, 668-75).
To support progressive tumor growth beyond the size of 1-2 mm3, it is recognized that tumor cells require a functional stroma, a support structure consisting of fibroblast, smooth muscle cells, endothelial cells, extracellular matrix proteins, and soluble factors (Folkman, J., Semin. Oncol. 2002. 29(6 Suppl 16), 15-8). Tumors induce the formation of stromal tissues through the secretion of soluble growth factors such as PDGF and transforming growth factor-beta (TGF-beta), which in turn stimulate the secretion of complimentary factors by host cells such as fibroblast growth factor (FGF), epidermal growth factor (EGF), and vascular endothelial growth factor (VEGF). These stimulatory factors induce the formation of new blood vessels, or angiogenesis, which brings oxygen and nutrients to the tumor and allows it to grow and provides a route for metastasis. It is believed some therapies directed at inhibiting stroma formation will inhibit the growth of epithelial tumors from a wide variety of histological types. (George, D. Semin. Oncol. 2001. 28(5 Suppl 17), 27-33; Shaheen, R.M., et al., Cancer Res. 2001, 61(4), 1464-8; Shaheen, R.M., et al. Cancer Res. 1999, 59(21), 5412-6). However, because of the complex nature and the multiple growth factors involved in angiogenesis process and tumor progression, an agent targeting a single pathway may have limited efficacy. It is desirable to provide treatment against a number of key signaling pathways utilized by tumors to induce angiogenesis in the host stroma. These include PDGF, a potent stimulator of stroma formation (Ostman, A. and C.H. Heldin, Adv. Cancer Res. 2001, 80, 1- 38), FGF, a chemo-attractant and mitogen for fibroblasts and endothelial cells, and VEGF, a potent regulator of vascularization.
PDGF is a key regulator of stromal formation, which is secreted by many tumors in a paracrine fashion and is believed to promote the growth of fibroblasts, smooth muscle and endothelial cells, promoting stroma formation and angiogenesis. PDGF was originally identified as the v-sis oncogene product of the simian sarcoma virus (Heldin, C.H., et al., J. Cell. Sci. Suppl. 1985, 3, 65-76). The growth factor is made up of two peptide chains, referred to as A or B chains which share 60% homology in their primary amino acid sequence. The chains are disulfide cross linked to form the 30 kDa mature protein composed of either AA, BB or AB homo- or heterodimmers. PDGF is found at high levels in platelets, and is expressed by endothelial cells and vascular smooth muscle cells. In addition, the production of PDGF is up regulated under low oxygen conditions such as those found in poorly vascularized tumor tissue (Kourembanas, S., et al., Kidney Int. 1997, 51(2), 438-43). PDGF binds with high affinity to the PDGF receptor, a 1106 amino acid 124 kDa transmembrane tyrosine kinase receptor (Heldin, C.H., A. Ostman, and L. Ronnstrand, Biochim. Biophys. Acta 1998, 1378(1), 79-113). PDGFR is found as homo- or heterodimer chains which have 30% homology overall in their amino acid sequence and 64% homology between their kinase domains (Heldin, C.H., et al.. Embo J. 1988, 7(5), 1387-93). PDGFR is a member of a family of tyrosine kinase receptors with split kinase domains that includes VEGFR-2 (KDR), VEGFR-3 (flt-4), c-kit, and flt-3. The PDGF receptor is expressed primarily on fibroblasts, smooth muscle cells, and pericytes and to a lesser extent on neurons, kidney mesangial, Leydig, and Schwann cells of the central nervous system. Upon binding to the receptor, PDGF induces receptor dimerization and undergoes auto- and trans- phosphorylation of tyrosine residues which increase the receptors' kinase activity and promotes the recruitment of downstream effectors through the activation of SH2 protein binding domains. A number of signaling molecules form complexes with activated PDGFR including PI-3-kinase, phospholipase C-gamma, src and GAP (GTPase activating protein for p21-ras) (Soskic, V., et al. Biochemistry 1999, 38(6), 1757-64). Through the activation of PI- 3-kinase, PDGF activates the Rho signaling pathway inducing cell motility and migration, and through the activation of GAP, induces mitogenesis through the activation of p21-ras and the MAPK signaling pathway. In adults, it is believed the major function of PDGF is to facilitate and increase the rate of wound healing and to maintain blood vessel homeostasis (Baker, E.A. and D.J. Leaper, Wound Repair Regen. 2000, 8(5), 392-8, and Yu, J., A. Moon, and H.R. Kim, Biochem. Biophys. Res. Commun. 2001, 282(3), 697-700). PDGF is found at high concentrations in platelets and is a potent chemoattractant for fibroblast, smooth muscle cells, neutrophils and macrophages. In addition to its role in wound healing PDGF is known to help maintain vascular homeostasis. During the development of new blood vessels, PDGF recruits pericytes and smooth muscle cells that are needed for the structural integrity of the vessels. PDGF is thought to play a similar role during tumor neovascularization. As part of its role in angiogenesis PDGF controls interstitial fluid pressure, regulating the permeability of vessels through its regulation of the interaction between connective tissue cells and the extracellular matrix. Inhibiting PDGFR activity can lower interstitial pressure and facilitate the influx of cytotoxics into tumors improving the anti-tumor efficacy of these agents (Pietras, K., et al. Cancer Res. 2002, 62(19), 5476-84; Pietras, K., et al. Cancer Res. 2001, 61(7), 2929-34). PDGF can promote tumor growth through either the paracrine or autocrine stimulation of PDGFR receptors on stromal cells or tumor cells directly, or through the amplification of the receptor or activation of the receptor by recombination. Over expressed PDGF can transform human melanoma cells and keratinocytes (Forsberg, K., et al. Proc. Natl. Acad. Sci. U S A. 1993, 90(2), 393-7; Skobe, M. and N.E. Fusenig, Proc. Natl. Acad. Sci. U S A. 1998, 95(3), 1050-5), two cell types that do not express PDGF receptors, presumably by the direct effect of PDGF on stroma formation and induction of angiogenesis. This paracrine stimulation of tumor stroma is also observed in carcinomas of the colon, lung, breast, and prostate (Bhardwaj, B., et al. Clin. Cancer Res. 1996, 2(4), 773-82; Nakanishi, K., et al. Mod. Pathol. 1997, 10(4), 341-7; Sundberg, C, et al. Am. J. Pathol. 1997, 151(2), 479-92; Lindmark, G., et al. Lab. Invest. 1993, 69(6), 682-9; Vignaud, J.M., et al, Cancer Res. 1994, 54(20), 5455-63) where the tumors express PDGF, but not the receptor. The autocrine stimulation of tumor cell growth, where a large faction of tumors analyzed express both the ligand PDGF and the receptor, has been reported in glioblastomas (Fleming, T.P., et al. Cancer Res. 1992, 52(16), 4550-3), soft tissue sarcomas (Wang, J., M.D. Coltrera, and A.M. Gown, Cancer Res. 1994, 54(2), 560-4) and cancers of the ovary (Henriksen, R., et al. Cancer Res. 1993, 53(19), 4550- 4), prostate (Fudge, K., C.Y. Wang, and M.E. Stearns, Mod. Pathol. 1994, 7(5), 549-54), pancreas (Funa, K., et al. Cancer Res. 1990, 50(3), 748-53) and lung (Antoniades, H.N., et al., Proc. Natl. Acad. Sci. U S A 1992, 89(9), 3942-6). Ligand independent activation of the receptor is found to a lesser extent but has been reported in chronic myelomonocytic leukemia (CMML) where the a chromosomal translocation event forms a fusion protein between the Ets-like transcription factor TEL and the PDGF receptor. In addition, activating mutations in PDGFR have been found in gastrointestinal stromal tumors in which c-kit activation is not involved (Heinrich, M.C., et al., Science 2003, 9, 9).
Another major regulator of angiogenesis and vasculogenesis in both embryonic development and some angiogenic-dependent diseases is vascular endothelial growth factor (VEGF; also called vascular permeability factor, VPF). VEGF represents a family of isoforms of mitogens existing in homodimeric forms due to alternative RNA splicing. The VEGF isoforms are highly specific for vascular endothelial cells (for reviews, see: Farrara et al. Endocr. Rev. 1992, 13, 18; Neufield et al. FASEB J. 1999, 13, 9).
VEGF expression is induced by hypoxia (Shweiki et al. Nature 1992, 359, 843), as well as by a variety of cytokines and growth factors, such as interleukin-1, interleukin-6, epidermal growth factor and transforming growth factor. To date, VEGF and the VEGF family members have been reported to bind to one or more of three transmembrane receptor tyrosine kinases (Mustonen et al. J. Cell Biol. 1995, 129, 895), VEGF receptor-1 (also known as flt-l (fms-like tyrosine kinase-1)), VEGFR-2 (also known as kinase insert domain containing receptor (KDR); the murine analogue of VEGFR-2 is known as fetal liver kinase-1 (flk-1)), and VEGFR-3 (also known as flt-4). VEGFR-2 and flt-l have been shown to have different signal transduction properties (Waltenberger et al. J. Biol. Chem. 1994, 269, 26988); Park et al. Oncogene 1995, 10, 135). Thus, VEGFR-2 undergoes strong ligand-dependant tyrosine phosphorylation in intact cells, whereas flt-l displays a weak response. Thus, binding to VEGFR-2 is believed to be a critical requirement for induction of the full spectrum of VEGF- mediated biological responses.
In vivo, VEGF plays a central role in vasculogenesis, and induces angiogenesis and permeabilization of blood vessels. Deregulated VEGF expression contributes to the development of a number of diseases that are characterized by abnormal angiogenesis and/or hyperpermeability processes. It is believed that regulation of the VEGF-mediated signal transduction cascade by some agents can provide a useful control of abnormal angiogenesis and/or hyperpermeability processes. Tumorigenic cells within hypoxic regions of tumors respond by stimulation of VEGF production, which triggers activation of quiescent endothelial cells to stimulate new blood vessel formation. (Shweiki et al. Proc. Nat'l. Acad. Sci. 1995, 92, 768). In addition, VEGF production in tumor regions where there is no angiogenesis may proceed through the ras signal transduction pathway (Grugel et al. J. Biol. Chem. 1995, 270, 25915; Rak et al. Cancer Res. 1995, 55, 4575). In situ hybridization studies have demonstrated VEGF mRNA is strongly upregulated in a wide variety of human tumors, including lung (Mattern et al. Br. J. Cancer 1996, 73, 931), thyroid (Viglietto et al. Oncogene 1995, 11, 1569), breast (Brown et al. Human Pathol. 1995, 26, 86), gastrointestinal tract (Brown et al. Cancer Res. 1993, 53, 4727; Suzuki et al. Cancer Res. 1996, 56, 3004), kidney and bladder (Brown et al. Am. J. Pathol. 1993, 1431, 1255), ovary (Olson et al. Cancer Res. 1994, 54, 1255), and cervical (Guidi et al. J. Nat'l Cancer Inst. 1995, 87, 12137) carcinomas, as well as angiosarcoma (Hashimoto et al. Lab. Invest. 1995, 73, 859) and several intracranial tumors (Plate et al. Nature 1992, 359, 845; Phillips et al. Int. J. Oncol. 1993, 2, 913; Berkman et al. J. Clin. Invest. 1993, 91, 153). Neutralizing monoclonal antibodies to VEGFR-2 have been shown to be efficacious in blocking tumor angiogenesis (Kim et al. Nature 1993, 362, 841; Rockwell et al. Mol. Cell. Differ. 1995, 3, 315).
Overexpression of VEGF, for example under conditions of extreme hypoxia, can lead to intraocular angiogenesis, resulting in hyperproliferation of blood vessels, leading eventually to blindness. Such a cascade of events has been observed for a number of retinopathies, including diabetic retinopathy, ischemic retinal-vein occlusion, and retinopathy of prematurity (Aiello et al. New Engl. J. Med. 1994, 331, 1480; Peer et al. Lab. Invest. 1995, 72, 638), and age-related macular degeneration (AMD; see, Lopez et al. Invest. Opththalmol. Vis. Sci. 1996, 37, 855). In rheumatoid arthritis (RA), the in-growth of vascular pannus may be mediated by production of angiogenic factors. Levels of immunoreactive VEGF are high in the synovial fluid of RA patients, while VEGF levels were low in the synovial fluid of patients with other forms of arthritis of with degenerative joint disease (Koch et al. J. Immunol. 1994, 152, 4149). The angiogenesis inhibitor AGM-170 has been shown to prevent neovascularization of the joint in the rat collagen arthritis model (Peacock et al. J. Exper. Med. 1992, 175, 1135).
Increased VEGF expression has also been shown in psoriatic skin, as well as bullous disorders associated with subepidermal blister formation, such as bullous pemphigoid, erythema multiforme, and dermatitis herpetiformis (Brown et al. J. Invest. Dermatol. 1995, 104, 744).
The vascular endothelial growth factors (VEGF, VEGF-C, VEGF-D) and their receptors (VEGFR-2, VEGFR-3) are not only key regulators of tumor angiogenesis, but also lymphangiogenesis. VEGF, VEGF-C and VEGF-D are expressed in most tumors, primarily during periods of tumor growth and, often at substantially increased levels. VEGF expression is stimulated by hypoxia, cytokines, oncogenes such as ras, or by inactivation of tumor suppressor genes (McMahon, G. Oncologist 2000, 5(Suppl. 1), 3-10; McDonald, N.Q.; Hendrickson, W.A. Cell 1993, 73, 421-424).
The biological activities of the VEGFs are mediated through binding to their receptors. VEGFR-3 (also called flt-4) is predominantly expressed on lymphatic endothelium in normal adult tissues. VEGFR-3 function is needed for new lymphatic vessel formation, but not for maintenance of the pre-existing lymphatics. VEGFR-3 is also upregulated on blood vessel endothelium in tumors. Recently VEGF-C and VEGF-D, ligands for VEGFR-3, have been identified as regulators of lymphangiogenesis in mammals. Lymphangiogenesis induced by tumor-associated lymphangiogenic factors could promote the growth of new vessels into the tumor, providing tumor cells access to systemic circulation. Cells that invade the lymphatics could find their way into the bloodstream via the thoracic duct. Tumor expression studies have allowed a direct comparison of VEGF-C, VEGF-D and VEGFR-3 expression with clinicopathological factors that relate directly to the ability of primary tumors to spread (e.g., lymph node involvement, lymphatic invasion, secondary metastases, and disease-free survival). In many instances, these studies demonstrate a statistical correlation between the expression of lymphangiogenic factors and the ability of a primary solid tumor to metastasize (Skobe, M. et al. Nature Med. 2001, 7(2), 192-198; Stacker, S.A. et al.. Nature Med. 2001, 7(2), 186-191; Makinen, T. et al. Nature Med. 2001, 7(2), 199-205; Mandriota, S.J. et al. EMBO J. 2001, 20(4), 672-82; Karpanen, T. et al. Cancer Res. 2001, 61(5), 1786-90; Kubo, H. et al. Blood 2000, 96(2), 546-53).
Hypoxia appears to be an important stimulus for VEGF production in malignant cells.
Activation of p38 MAP kinase is required for VEGF induction by tumor cells in response to hypoxia (Blaschke, F. et al. Biochem. Biophys. Res. Commun. 2002, 296, 890-896;
Shemirani, B. et al. Oral Oncology 2002, 38, 251-257). In addition to its involvement in angiogenesis through regulation of VEGF secretion, p38 MAP kinase promotes malignant cell invasion, and migration of different tumor types through regulation of collagenase activity and urokinase plasminogen activator expression (Laferriere, J. et al. J. Biol. Chem.
2001, 276, 33762-33772; Westermarck, J. et al. Cancer Res. 2000, 60, 7156-7162; Huang, S. et al. J. Biol. Chem. 2000, 275, 12266-12272; Simon, C. et al. Exp. Cell Res. 2001, 271, 344-355).
Inhibition of the mitogen- activated protein kinase (MAPK) p38 has been shown to inhibit both cytokine production (e.g., TNF, IL-1, IL-6, IL-8) and proteolytic enzyme production (e.g., MMP-1, MMP-3) in vitro and/or in vivo. The mitogen activated protein (MAP) kinase p38 is involved in IL-1 and TNF signaling pathways (Lee, J. C; Laydon, J. T.; McDonnell, P. C; Gallagher, T. E; Kumar, S.; Green, D.; McNulty, D.; Blumenthal, M. J.; Heys, J. R.; Landvatter, S. W.; Strieker, J. E.; McLaughlin, M. M.; Siemens, I. R.; Fisher, S. M.; Livi, G. P.; White, J. R.; Adams, J. L.; Yound, P. R. Nature 1994, 372, 739). Clinical studies have linked tumor necrosis factor (TNF) production and/or signaling to a number of diseases including rheumatoid arthritis (Maini. J. Royal Coll. Physicians London 1996, 30, 344). In addition, excessive levels of TNF have been implicated in a wide variety of inflammatory and/or immunomodulatory diseases, including acute rheumatic fever (Yegin et al. Lancet 1997, 349, 170), bone resorption (Pacifici et al. J. Clin. Endocrinol. Metabol. 1997 , 82, 29), postmenopausal osteoporosis (Pacifici et al. J. Bone Mineral Res. 1996, 11, 1043), sepsis (Blackwell et al. Br. J. Anaesth. 1996, 77, 110), gram negative sepsis (Debets et al. Prog. Clin. Biol. Res. 1989, 308, 463), septic shock (Tracey et al. Nature 1987, 330, 662; Girardin et al. New England J. Med. 1988, 319, 397), endotoxic shock (Beutler et al. Science 1985, 229, 869; Ashkenasi et al. Proc. Nat'l. Acad. Sci. USA 1991, 88, 10535), toxic shock syndrome, (Saha et al. J. Immunol. 1996, 157, 3869; Lina et al. FEMS Immunol. Med. Microbiol. 1996, 13, 81), systemic inflammatory response syndrome (Anon. Crit. Care Med. 1992, 20, 864), inflammatory bowel diseases (Stokkers et al. J. Inflamm. 1995-6, 47, 97) including Crohn's disease (van Deventer et al. Aliment. Pharmacol. Therapeu. 1996, 10 (Suppl. 2), 107; van Dullemen et al. Gastroenterology 1995, 109, 129) and ulcerative colitis (Masuda et al. J. Clin. Lab. Immunol. 1995, 46, 111), Jarisch-Herxheimer reactions (Fekade et al. New England J. Med. 1996, 335, 311), asthma (Amrani et al. Rev. Malad. Respir. 1996, 13, 539), adult respiratory distress syndrome (Roten et al. Am. Rev. Respir. Dis. 1991, 143, 590; Suter et al. Am. Rev. Respir. Dis. 1992, 145, 1016), acute pulmonary fibrotic diseases (Pan et al. Pathol. Int. 1996, 46, 91), pulmonary sarcoidosis (Ishioka et al. Sarcoidosis Vasculitis Diffuse Lung Dis. 1996, 13, 139), allergic respiratory diseases (Casale et al. Am. J. Respir. Cell Mol. Biol. 1996, 15, 35), silicosis (Gossart et al. J. Immunol. 1996, 156, 1540; Vanhee et al. Eur. Respir. J. 1995, 8, 834), coal worker's pneumoconiosis (Bonn et al. Am. Rev. Respir. Dis. 1988, 138, 1589), alveolar injury (Horinouchi et al. Am. J. Respir. Cell Mol. Biol. 1996, 14, 1044), hepatic failure (Gantner et al. J. Pharmacol. Exp. Therap. 1997, 280, 53), liver disease during acute inflammation (Kim et al. J. Biol. Chem. 1997, 272, 1402), severe alcoholic hepatitis (Bird et al. Ann. Intern. Med. 1990, 112, 917), malaria (Grau et al. Immunol. Rev. 1989, 112, 49; Taverne et al. Parasitol. Today 1996, 12, 290) including Plasmodium falciparum malaria (Perlmann et al. Infect. Immunit. 1997, 65, 116) and cerebral malaria (Rudin et al. Am. J. Pathol. 1997, 150, 257), non-insulin-dependent diabetes mellitus (NIDDM; Stephens et al. J. Biol. Chem. 1997, 272, 971; Ofei et al. Diabetes 1996, 45, 881), congestive heart failure (Doyama et al. Int. J. Cardiol. 1996, 54, 217; McMurray et al. Br. Heart J. 1991, 66, 356), damage following heart disease (Malkiel et al. Mol. Med. Today 1996, 2, 336), atherosclerosis (Parums et al. J. Pathol. 1996, 179, A46), Alzheimer's disease (Fagarasan et al. Brain Res. 1996, 723, 231; Aisen et al. Gerontology 1997, 43, 143), acute encephalitis (Ichiyama et al. J. Neurol. 1996, 243, 457), brain injury (Cannon et al. Crit. Care Med. 1992, 20, 1414; Hansbrough et al. Surg. Clin. N. Am. 1987, 67, 69; Marano et al. Surg. Gynecol. Obstetr. 1990, 170, 32), multiple sclerosis (M.S.; Coyle. Adv. Neuroimmunol. 1996, 6, 143; Matusevicius et al. J. Neuroimmunol. 1996, 66, 115) including demyelation and oligiodendrocyte loss in multiple sclerosis (Brosnan et al. Brain Pathol. 1996, 6, 243), advanced cancer (MucWierzgon et al. J. Biol. Regulators Homeostatic Agents 1996, 10, 25), lymphoid malignancies (Levy et al. Crit. Rev. Immunol. 1996, 16, 31), pancreatitis (Exley et al. Gut 1992, 33, 1126) including systemic complications in acute pancreatitis (McKay et al. Br. J. Surg. 1996, 83, 919), impaired wound healing in infection inflammation and cancer (Buck et al. Am. J. Pathol. 1996, 149, 195), myelodysplastic syndromes (Raza et al. Int. J.
Hematol. 1996, 63, 265), systemic lupus erythematosus (Maury et al. Arthritis Rheum. 1989,
32, 146), biliary cirrhosis (Miller et al. Am. J. Gasteroenterolog. 1992, 87, 465), bowel necrosis (Sun et al. J. Clin. Invest. 1988, 81, 1328), psoriasis (Christophers. Austr. J. Dermatol. 1996, 37, S4), radiation injury (Redlich et al. J. Immunol. 1996, 157, 1705), and toxicity following administration of monoclonal antibodies such as OKT3 (Brod et al.
Neurology 1996, 46, 1633). TNF levels have also been related to host-versus-graft reactions
(Piguet et al. Immunol. Ser. 1992, 56, 409) including ischemia reperfusion injury (Colletti et al. J. Clin. Invest. 1989, 85, 1333) and allograft rejections including those of the kidney (Maury et al. J. Exp. Med. 1987, 166, 1132), liver (Imagawa et al. Transplantation 1990, 50,
219), heart (Boiling et al. Transplantation 1992, 53, 283), and skin (Stevens et al. Transplant.
Proc. 1990, 22, 1924), lung allograft rejection (Grossman et al. Immunol. Allergy Clin. N.
Am. 1989, 9, 153) including chronic lung allograft rejection (obliterative bronchitis;
LoCicero et al. J. Thorac. Cardiovasc. Surg. 1990, 99, 1059), as well as complications due to total hip replacement (Cirino et al. Life Sci. 1996, 59, 86). TNF has also been linked to infectious diseases (review: Beutler et al. Crit. Care Med. 1993, 21, 5423; Degre. Biotherapy
1996, 8, 219) including tuberculosis (Rook et al. Med. Malad. Infect. 1996, 26, 904), Helicobacter pylori infection during peptic ulcer disease (Beales et al. Gastroenterology
1997, 112, 136), Chaga's disease resulting from Trypanosoma cruzi infection (Chandrasekar et al. Biochem. Biophys. Res. Commun. 1996, 223, 365), effects of Shiga-like toxin resulting from E. coli infection (Harel et al. J. Clin. Invest. 1992, 56, 40), the effects of enterotoxin A resulting from Staphylococcus infection (Fischer et al. J. Immunol. 1990, 144, 4663), meningococcal infection (Waage et al. Lancet 1987, 355; Ossege et al. J. Neurolog. Sci. 1996, 144, 1), and infections from Borrelia burgdorferi (Brandt et al. Infect. Immunol. 1990, 58 , 983), Treponema pallidum (Chamberlin et al. Infect. Immunol. 1989, 57, 2872), cytomegalovirus (CMV; Geist et al. Am. J. Respir. Cell Mol. Biol. 1997, 16, 31), influenza virus (Beutler et al. Clin. Res. 1986, 34, 491a), Sendai virus (Goldfield et al. Proc. Nat'l. Acad. Sci. USA 1989, 87, 1490), Theiler's encephalomyelitis virus (Sierra et al. Immunology 1993, 78, 399), and the human immunodeficiency virus (HIV; Poli. Proc. Nat'l. Acad. Sci. USA 1990, 87, 782; Vyakaram et al. AIDS 1990, 4, 21; Badley et al. J. Exp. Med. 1997, 185, 55).
A number of diseases are thought to be mediated by excess or undesired matrix-destroying metalloprotease (MMP) activity or by an imbalance in the ratio of the MMPs to the tissue inhibitors of metalloproteinases (TIMPs). These include osteoarthritis (Woessner et al. J. Biol. Chem. 1984, 259, 3633), rheumatoid arthritis (Mullins et al. Biochim. Biophys. Acta 1983, 695, 117; Woolley et al. Arthritis Rheum. 1977, 20, 1231; Gravallese et al. Arthritis Rheum. 1991, 34, 1076), septic arthritis (Williams et al. Arthritis Rheum. 1990, 33, 533), tumor metastasis (Reich et al. Cancer Res. 1988, 48, 3307; Matrisian et al. Proc. Nat'l. Acad. Sci., USA 1986, 83, 9413), periodontal diseases (Overall et al. J. Periodontal Res. 1987, 22, 81), corneal ulceration (Burns et al. Invest. Opthalmol. Vis. Sci. 1989, 30, 1569), proteinuria (Baricos et al. Biochem. J. 1988, 254, 609), coronary thrombosis from atherosclerotic plaque rupture (Henney et al. Proc. Nat'l. Acad. Sci., USA 1991, 88, 8154), aneurysmal aortic disease (Vine et al. Clin. Sci. 1991, 81, 233), birth control (Woessner et al. Steroids 1989, 54, 491), dystrophobic epidermolysis bullosa (Kronberger et al. J. Invest. Dermatol. 1982, 79, 208), degenerative cartilage loss following traumatic joint injury, osteopenias mediated by MMP activity, tempero mandibular joint disease, and demyelating diseases of the nervous system (Chantry et al. J. Neurochem. 1988, 50, 688).
Because inhibition of p38 leads to inhibition of TNF production and MMP production, it is believed inhibition of mitogen activated protein (MAP) kinase p38 enzyme can provide an approach to the treatment of the above listed diseases including osteoporosis and inflammatory disorders such as rheumatoid arthritis and COPD (Badger, A. M.; Bradbeer, J. N.; Votta, B.; Lee, J. C; Adams, J. L.; Griswold, D. E. J. Pharm. Exper. Ther. 1996, 279, 1453).
Hypoxia appears to be an important stimulus for VEGF production in malignant cells. Activation of p38 kinase is required for VEGF induction by tumor cells in response to hypoxia (Blaschke, F. et al. Biochem. Biophys. Res. Commun. 2002, 296, 890-896; Shemirani, B. et al. Oral Oncology 2002, 38, 251-257). In addition to its involvement in angiogenesis through regulation of VEGF secretion, p38 kinase promotes malignant cell invasion, and migration of different tumor types through regulation of collagenase activity and urokinase plasminogen activator expression (Laferriere, J. et al. J. Biol. Chem. 2001, 276, 33762-33772; Westermarck, J. et al. Cancer Res. 2000, 60, 7156-7162; Huang, S. et al. J. Biol. Chem. 2000, 275, 12266-12272; Simon, C. et al. Exp. Cell Res. 2001, 271, 344-355). Therefore, inhibition of p38 kinase is also expected to impact tumor growth by interfering with signaling cascades associated with both angiogenesis and malignant cell invasion. Certain ureas have been described as having activity as serine-threonine kinase and/or as tyrosine kinase inhibitors. In particular, the utility of certain ureas as an active ingredient in pharmaceutical compositions for the treatment of cancer, angiogenesis disorders, inflammatory disorders, has been demonstrated.
For cancer and angiogenesis, see:
Smith et al., Bioorg. Med. Chem. Lett. 2001, 11, 2775-2778.
Lowinger et al., Clin. Cancer Res. 2000, 6(suppl.), 335.
Lyons et al., Endocr.-Relat. Cancer 2001, 8, 219-225.
Riedl et al., Book of Abstracts, 92nd AACR Meeting, New Orleans, LA, USA, abstract 4956. Khire et al., Book of Abstracts, 93rdAACR Meeting, San Francisco, CA, USA, abstract 4211. Lowinger et al., Curr. Pharm. Design 2002, 8, 99-110.
Carter et al., Book of Abstracts, 92ndAACR Meeting, New Orleans, LA, USA, abstract 4954. Vincent et al., Book of Abstracts, 38th ASCO Meeting, Orlando, FL, USA, abstract 1900. Hilger et al., Book of Abstracts, 38th ASCO Meeting, Orlando, FL, USA, abstract 1916. Moore et al., Book of Abstracts, 38th ASCO Meeting, Orlando, FL, USA, abstract 1816. Strumberg et al., Book of Abstracts, 38th ASCO Meeting, Orlando, FL, USA, abstract 121.
For p38 mediated diseases, including inflammatory disorders, see:
Redman et al., Bioorg. Med. Chem. Lett. 2001, 11, 9-12.
Dumas et al., Bioorg. Med. Chem. Lett. 2000, 10, 2047-2050.
Dumas et al., Bioorg. Med. Chem. Lett. 2000, 10, 2051-2054.
Ranges et al., Book of Abstracts, 220th ACS National Meeting, Washington, DC, USA, MEDI 149.
Dumas et al., Bioorg. Med. Chem. Lett. 2002, 12, 1559-1562.
Regan et al., J. Med. Chem. 2002, 45, 2994-3008.
Pargellis et al., Nature Struct. Biol. 2002, 9(4), 268-272.
Madwed J. B., Book of Abstracts, Protein Kinases: Novel Target Identification and Validation for Therapeutic Development, San Diego, CA, USA, March 2002.
Pargellis C. et al., Curr. Opin. Invest. Drugs 2003, 4, 566-571.
Branger J. et al., J. Immunol. 2002, 168, 4070-4077.
Branger J. et al., Blood 2003, 101, 4446-4448. Omega-Carboxyaryl diphenyl ureas are disclosed in WO00/42012, published: July 20, 2000, WO00/41698, published: July 20, 2000, the following published U.S. applications:
US2002-0165394-A1, published November 7, 2002,
US2001-003447-A1, published October 25, 2001,
US2001 -0016659- A 1 , published August 23 , 2001 ,
US2002-013774-A1, published September 26, 2002,
and copending U.S. applications:
09/758,547, filed January 12, 2001,
09/889,227, filed July 12, 2001,
09/993,647, filed November 27, 2001,
10/042,203, filed January 11, 2002 and
10/071,248, filed February 11, 2002,
Description of the Invention
It has been discovered that synthetic metabolites of omega-carboxyaryl diphenyl urea of Formula I below are potent inhibitors of raf kinase, VEGFR kinase, p38 kinase, and PDGFR kinase, which are all molecular targets of interest for the treatment and prevention of osteoporosis, inflammatory disorders, hyper-proliferatrive disorders, and angiogenesis disorders, including cancer.
The present invention provides, e.g.,
(i) novel synthetic metabolites of the compound of Formula (I), salts, prodrugs, and metabolites thereof,
(ii) pharmaceutical compositions containing such compounds, and
(iii) use of such synthetic metabolites or compositions for treating diseases and conditions mediated by raf, VEGFR, PDGFR, flt-3, and p38, either as a sole agent or in combination with cytotoxic therapies. The compound of the Formula I below, salts, prodrugs and metabolites thereof is collectively referred to as the "compounds of the invention." Formula I is as follows:
Figure imgf000014_0001
The synthetic metabolites of the compound of this invention include oxidized derivatives of Formula I wherein one or more of the urea nitrogens are substituted with a hydroxy group. The synthetic metabolites of the compound of this invention also include analogs where the methylamide group of the compound of Formula I is hydroxylated then de-methylated by metabolic degradation. The synthetic metabolites of the compound of this invention further include oxidized derivatives where the pyridine nitrogen atom is in the N-oxide form (e.g. carries a hydroxy substituent) leading to those structures referred to in the art as 1-oxo- pyridine and 1 -hydroxy-pyridine.
Where the plural form of the word compounds, salts, and the like, is used herein, this is taken to mean also a single compound, salt, or the like. In particular, the present invention relates to synthetic forms of M2, M3, M4 and M5 metabolites of the compounds of Formula I, whose structures appear below:
Figure imgf000014_0002
The relationship of the aforementioned synthetic metabolites with the parent compound of Formula I is illustrated in Fig. 1. Particularly preferred are the M2 and M5 metabolites of the compounds of Formula I.
Figure imgf000015_0001
As an embodiment of the present invention, the biotransformation of the compound of Formula I was investigated in vitro with liver microsomes and hepatocytes, and in vivo in the plasma of several species. In man, the N-oxide (M-2) and the demethylated N-oxide (M-5) are of significance. Both metabolites in synthetic form show systemic exposure (area under the curve [AUC], mg*h/L) at steady state similar to regorafenib (the parent compound) in patients at a dose of 160 mg in a 3 weeks on/1 week off Phase I study.
The pharmacologic activities of the synthetic metabolites of the compounds of formula I, particularly, the M-2 and M-5 metabolites, are also of relevance. In vitro biochemical and cellular kinase phosphorylation assays reveal that the synthetic metabolites of compounds of formula I are broad- spectrum kinase inhibitors. In in vivo preclinical models the metabolites were effective against tumor growth and further inhibited tumor vasculature. The synthetic metabolites also demonstrated acute effects on vascular endothelial growth factor (VEGF)- induced hypotension in pharmacodynamic rat models.
The term "synthetic" is art-recognized and refers to production by in vitro chemical or enzymatic synthesis.
As used herein, the term "isolated" means that the referenced material is removed from its native environment, e.g., a cell, or a tissue, or from the body. Thus, an isolated metabolite can be free of some or all cellular components, i.e., components of the cells in which the native material occurs naturally (e.g., cytoplasmic or membrane component, such as, for example, microsomes).
The term "purified" as used herein refers to material that has been isolated under conditions that reduce or eliminate the presence of unrelated materials, i.e., contaminants, including precursor materials from which the material is obtained. For example, a purified metabolite is preferably substantially free of other metabolites or the precursor compound from which it is derived. As used herein, the term "substantially free" is used operationally, in the context of analytical testing of the material. Preferably, purified material is substantially free of contaminants is at least 50% pure; more preferably, at least 90% pure, and more preferably still at least 99% pure. Purity can be evaluated by chromatography, gel electrophoresis, HPLC, NMR or mass-spectroscopy analysis, composition analysis, biological assays, and other methods known in the art.
Salts
Pharmaceutically acceptable salts of the synthetic metabolites of Forumla I are also within the scope of this invention. The term "pharmaceutically acceptable salt" refers to a relatively non- toxic, inorganic or organic acid addition salt of a compound of the present invention. For example, see S. M. Berge, et al. "Pharmaceutical Salts," J. Pharm. Sci. 1977, 66, 1-19. Representative salts of the compound of this invention include the conventional non-toxic salts, for example, from inorganic or organic acids by means well known in the art. For example, such acid addition salts include acetate, adipate, alginate, ascorbate, aspartate, benzoate, benzenesulfonate, bisulfate, butyrate, citrate, camphorate, camphorsulfonate, cinnamate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, fumarate, glucoheptanoate, glycerophosphate, hemisulfate, heptanoate, hexanoate, hydrochloride, hydrobromide, hydroiodide, 2-hydroxyethanesulfonate, itaconate, lactate, maleate, mandelate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oxalate, pamoate, pectinate, persulfate, 3-phenylpropionate, picrate, pivalate, propionate, succinate, sulfonate, tartrate, thiocyanate, tosylate, and undecanoate.
The salts or prodrugs of the compounds of Formula I may contain one or more asymmetric centers. Asymmetric carbon atoms may be present in the (R) or (S) configuration or (R,S) configuration. Substituents on a ring may also be present in either cis or trans form. It is intended that all such configurations (including enantiomers and diastereomers), are included within the scope of the present invention. Preferred isomers are those with the configuration which produces the more desirable biological activity. Separated, pure or partially purified isomers or racemic mixtures of the compounds of this invention are also included within the scope of the present invention. The purification of said isomers and the separation of said isomeric mixtures can be accomplished by standard techniques known in the art.
The particular process to be utilized in the preparation of the synthetic metabolites used in this embodiment of the invention is described in Example 4. Salt forms of the synthetic metabolites of Formula (I) are described in the Examples.
Methods of use
The present invention provides compounds which are capable of modulating one or more signal transduction pathways involving raf, VEGFR, PDGFR, p38, and/or flt-3 kinases. Raf is an important signaling molecule involved in the regulation of a number of key cellular processes, including cell growth, cell survival and invasion. It is a member of the Ras/raf/MEK/ERK pathway. This pathway is present in most tumor cells. VEGFR, PDGFR, and flt-3 are transmembrane receptor molecules which, when stimulated by an appropriate ligand, trigger the Ras/raf/MEK/ERK cell signaling pathway, leading to a cascade of cellular events. Each of these receptor molecules have tyrosine kinase activity.
The VEGFR receptors are stimulated by vascular endothelial growth factors (VEGF), and are important control points in the regulation of endothelial cell development and function. The PDGF-beta receptor regulates cell proliferation and survival in a number of cell types, including mesenchymal cells. Flt-3 is a receptor for the FL ligand. It is structurally similar to c-kit, and modulates the growth of pluripotent haemopoietic cells, influencing the development of T-cells, B-cells, and dendritic cells.
Any gene or isoform of raf, VEGFR, PDGFR, p38, and/or flt-3 can be modulated in accordance with present invention, including both wild-type and mutant forms. Raf or raf-1 kinase is a family of serine/threonine kinases which comprise at least three family members, a-raf, b-raf, and c-raf or raf-1. See, e.g., Dhillon and Kolch, Arch. Biochem. Biophys. 2002, 404, 3-9. C-raf and b-raf are preferred targets for compounds of the present invention. Activating b-raf mutations (e.g., V599E mutant) have been identified in various cancers, including melanoma, and the compounds described herein can be utilized to inhibit their activity.
By the term "modulate", it is meant that the functional activity of the pathway (or a component of it) is changed in comparison to its normal activity in the absence of the compound. This effect includes any quality or degree of modulation, including, increasing, agonizing, augmenting, enhancing, facilitating, stimulating, decreasing, blocking, inhibiting, reducing, diminishing, antagonizing, etc.
The compounds of the present invention can also modulate one or more of the following processes, including, but not limited to, e.g., cell growth (including, e.g., differentiation, cell survival, and/or proliferation), tumor cell growth (including, e.g., differentiation, cell survival, and/or proliferation), tumor regression, endothelial cell growth (including, e.g., differentiation, cell survival, and/or proliferation), angiogenesis (blood vessel growth), lymphangiogenesis (lymphatic vessel growth), and/or hematopoiesis (e.g., T- and B-cell development, dendritic cell development, etc.).
While not wishing to be bound by any theory or mechanism of action, it has been found that compounds of the present invention possess the ability to modulate kinase activity. The methods of the present invention, however, are not limited to any particular mechanism or how the compounds achieve their therapeutic effect. By the term "kinase activity", it is meant a catalytic activity in which a gamma-phosphate from adenosine triphosphate (ATP) is transferred to an amino acid residue (e.g., serine, threonine, or tyrosine) in a protein substrate. A compound can modulate kinase activity, e.g., inhibiting it by directly competing with ATP for the ATP-binding pocket of the kinase, by producing a conformational change in the enzyme's structure that affects its activity (e.g., by disrupting the biologically-active three- dimensional structure), etc.
Kinase activity can be determined routinely using conventional assay methods. Kinase assays typically comprise the kinase enzyme, substrates, buffers, and components of a detection system. A typical kinase assay involves the reaction of a protein kinase with a peptide substrate and an ATP, such as 32P-ATP, to produce a phosphorylated end-product (for instance, a phosphoprotein when a peptide substrate is used). The resulting end-product can be detected using any suitable method. When radioactive ATP is utilized, a radioactively labeled phosphoprotein can be separated from the unreacted gamma-32P-ATP using an affinity membrane or gel electrophoresis, and then visualized on the gel using autoradiography or detected with a scintillation counter. Non-radioactive methods can also be used. Methods can utilize an antibody which recognizes the phosphorylated substrate, e.g., an anti-phosphotyrosine antibody. For instance, kinase enzyme can incubated with a substrate in the presence of ATP and kinase buffer under conditions which are effective for the enzyme to phosphorylate the substrate. The reaction mixture can be separated, e.g., electrophoretically, and then phosphorylation of the substrate can be measured, e.g., by Western blotting using an anti-phosphotyrosine antibody. The antibody can be labeled with a detectable label, e.g., an enzyme, such as HRP, avidin or biotin, chemiluminescent reagents, etc. Other methods can utilize ELISA formats, affinity membrane separation, fluorescence polarization assays, luminescent assays, etc.
An alternative to a radioactive format is time-resolved fluorescence resonance energy transfer (TR-FRET). This method follows the standard kinase reaction, where a substrate, e.g., biotinylated poly(GluTyr), is phosphorylated by a protein kinase in the presence of ATP. The end-product can then detected with a europium chelate phosphospecific antibody (anti- phosphotyrosine or phosphoserine/threonine), and streptavidin-APC, which binds the biotinylated substrate. These two components are brought together spatially upon binding, and energy transfer from the phosphospecific antibody to the acceptor (SA-APC) produces fluorescent readout in the homogeneous format.
The compounds of the present invention can be used to treat and/or prevent any disease or condition mediated by one or more cellular signal transduction pathways involving raf, VEGFR, PDGFR, p38, and/or flt-3 kinases. The term "treating" is used conventionally, e.g., the management or care of a subject for the purpose of combating, alleviating, reducing, relieving, improving the condition of, etc., of a disease or disorder. The compounds can also be described as being used to prevent and/or treat diseases and/or condition mediated by the signaling molecules. The term "mediated" indicates, e.g., that the signaling molecule is part of the pathway which is aberrant or disturbed in the disease and/or condition.
Diseases and conditions that can be treated include any of those mentioned above and below, as well as: Raf associated diseases include, e.g., cell-proliferation disorders, cancer, tumors, etc.;
VEGFR-2 associated diseases include, e.g., cancer, tumor growth, inflammatory disease, rheumatoid arthritis, retinopathy, psoriasis, glomerulonephritis, asthma, chronic bronchitis, atherosclerosis, transplant rejection, conditions involving angiogenesis, etc.; VEGFR-3 associated diseases include, e.g., cancer, corneal disease, inflamed cornea (e.g., Hamrah, Am. J. Path. 2003, 163, 57-68), corneal transplantation (Cursiefen et al., Cornea 2003, 22, 273-81), lymphatic hyperplasia, conditions involving lymphangiogenesis, etc.;
PDGFR-beta associated diseases include, e.g., diseases or conditions characterized by cell proliferation, cell matrix production, cell movement, and/or extracellular matrix production. Specific examples, include, e.g., tumors, malignancies, cancer, metastasis, chronic myeloid leukemia, inflammation, renal disease, diabetic nephropathy, mesangial proliferative glomerulonephritis, fibrotic conditions, atherosclerosis, restenosis, hypertension-related arteriosclerosis, venous bypass graft arteriosclerosis, scleroderma, interstitial pulmonary diseases, synovial disorders, arthritis, leukemias, lymphomas, etc
Flt-3 associated diseases include, e.g., immune-related disorders, blood cell disorders, conditions involving hematopoietic cell development (e.g., T-cells, B-cells, dendritic cells, cancer, anemia, HIV, acquired immune deficiency syndrome, etc. p38 associated diseases include inflammatory disorders, immunomodulatory disorders, and other disorders that have been linked to abnormal cytokine production, especially TNF-alpha, or abnormal MMP activity. These disorders include, but are not limited to, rheumatoid arthritis, COPD, osteoporosis, Crohn's disease and psoriasis. In addition, compounds of the present invention can be used to treat conditions and disorders disclosed in U.S. Pat. No. 6,316,479, e.g., glomerular sclerosis, interstitial nephritis, interstitial pulmonary fibrosis, atherosclerosis, wound scarring and scleroderma. The compounds of this invention also have a broad therapeutic activity to treat or prevent the progression of a broad array of diseases, such as inflammatory conditions, coronary restenosis, tumor-associated angiogenesis, atherosclerosis, autoimmune diseases, inflammation, certain kidney diseases associated with proliferation of glomerular or mesangial cells, and ocular diseases associated with retinal vessel proliferation, psoriasis, hepatic cirrhosis, diabetes, atherosclerosis, restenosis, vascular graft restenosis, in-stent stenosis, angiogenesis, ocurlar diseases, pulmonary fibrosis, obliterative bronchiolitis, glomerular nephritis, rheumatoid arthritis.
The present invention also provides for treating, preventing, modulating, etc., one or more of the following conditions in humans and/or other mammals: retinopathy, including diabetic retinopathy, ischemic retinal-vein occlusion, retinopathy of prematurity and age related macular degeneration; rheumatoid arthritis, psoriasis, or bullous disorder associated with subepidermal blister formation, including bullous pemphigoid, erythema multiforme, or dermatitis herpetiformis, rheumatic fever, bone resorption, postmenopausal osteoperosis, sepsis, gram negative sepsis, septic shock, endotoxic shock, toxic shock syndrome, systemic inflammatory response syndrome, inflammatory bowel disease (Crohn's disease and ulcerative colitis), Jarisch-Herxheimer reaction, asthma, adult respiratory distress syndrome, acute pulmonary fibrotic disease, pulmonary sarcoidosis, allergic respiratory disease, silicosis, coal worker's pneumoconiosis, alveolar injury, hepatic failure, liver disease during acute inflammation, severe alcoholic hepatitis, malaria (Plasmodium falciparum malaria and cerebral malaria), non-insulin-dependent diabetes mellitus (NIDDM), congestive heart failure, damage following heart disease, atherosclerosis, Alzheimer's disease, acute encephalitis, brain injury, multiple sclerosis (demyelation and oligiodendrocyte loss in multiple sclerosis), advanced cancer, lymphoid malignancy, pancreatitis, impaired wound healing in infection, inflammation and cancer, myelodysplastic syndromes, systemic lupus erythematosus, biliary cirrhosis, bowel necrosis, radiation injury/ toxicity following administration of monoclonal antibodies, host-versus-graft reaction (ischemia reperfusion injury and allograft rejections of kidney, liver, heart, and skin), lung allograft rejection (obliterative bronchitis), or complications due to total hip replacement, ad an infectious disease selected from tuberculosis, Helicobacter pylori infection during peptic ulcer disease, Chaga's disease resulting from Trypanosoma cruzi infection, effects of Shiga-like toxin resulting from E. coli infection, effects of enterotoxin A resulting from Staphylococcus infection, meningococcal infection, and infections from Borrelia burgdorferi, Treponema pallidum, cytomegalovirus, influenza virus, Theiler' s encephalomyelitis virus, and the human immunodeficiency virus (HIV), papilloma, blastoglioma, Kaposi's sarcoma, melanoma, lung cancer, ovarian cancer, prostate cancer, squamous cell carcinoma, astrocytoma, head cancer, neck cancer, bladder cancer, breast cancer, colorectal cancer, thyroid cancer, pancreatic cancer, gastric cancer, hepatocellular carcinoma, leukemia, lymphoma, Hodgkin's disease, Burkitt's disease, arthritis, rheumatoid arthritis, diabetic retinopathy, angiogenesis, restenosis, in-stent restenosis, vascular graft restenosis, pulmonary fibrosis, hepatic cirrhosis, atherosclerosis, glomerulonephritis, diabetic nephropathy, thrombic micoangiopathy syndromes, transplant rejection, psoriasis, diabetes, wound healing, inflammation, and neurodegenerative diseases, hyperimmune disorders, hemangioma, myocardial angiogenesis, coronary and cerebral collateral vascularization, ischemia, corneal disease, rubeosis, neovascular glaucoma, macular degeneration retinopathy of prematurity, wound healing, ulcer Helicobacter related diseases, fractures, endometriosis, a diabetic condition, cat scratch fever, thyroid hyperplasia, asthma or edema following burns, trauma, chronic lung disease, stroke, polyps, cysts, synovitis, chronic and allergic inflammation, ovarian hyperstimulation syndrome, pulmonary and cerebral edema, keloid, fibrosis, cirrhosis, carpal tunnel syndrome, adult respiratory distress syndrome, ascites, an ocular condition, a cardiovascular condition, Crow-Fukase (POEMS) disease, Crohn's disease, glomerulonephritis, osteoarthritis, multiple sclerosis, graft rejection, Lyme disease, sepsis, von Hippel Lindau disease, pemphigoid, Paget's disease, polycystic kidney disease, sarcoidosis, throiditis, hyperviscosity syndrome, Osler-Weber-Rendu disease, chronic occlusive pulmonary disease, radiation, hypoxia, preeclampsia, menometrorrhagia, endometriosis, infection by Herpes simplex, ischemic retinopathy, corneal angiogenesis, Herpes Zoster, human immunodeficiency virus, parapoxvirus, protozoa, toxoplasmosis, and tumor-associated effusions and edema. The compounds of this invention can possess more than one of the mentioned activities, and therefore can target a plurality of signal transduction pathways. Thus, these compounds can achieve therapeutic and prophylactic effects which normally are only obtained when using a combination of different compounds. For instance, the ability to inhibit both new vessel formation (e.g., associated with VEGFR-2 and VEGFR-3 function) (e.g., blood and/or lymph) and cell-proliferation (e.g., associated with raf and PDGFR-beta function) using a single compound is especially beneficial in the treatment of cancer, and other cell- proliferation disorders that are facilitated by neo-vascularization. Thus, the present invention relates specifically to compounds which possess at least anti-cell proliferation and anti- angiogenic (i.e., inhibits angiogenesis) activity. Any disorder or condition that would benefit from inhibiting vessel growth and cell proliferation can be treated in accordance with the present invention. Using a single compound is also advantageous because its range of activities can be more precisely defined. As indicated above, the present invention relates to methods of treating and/or preventing diseases and conditions; and/or modulating one or more of the pathways, polypeptides, genes, diseases, conditions, etc., associated with raf, VEGFR, PDGFR, p38, and/or flt-3. These methods generally involve administering effective amounts of compounds of the present invention, where an effective amount is the quantity of the compound which is useful to achieve the desired result. Compounds can be administered in any effective form by any effective route, as discussed in more detail below.
Methods include modulating tumor cell proliferation, including inhibiting cell proliferation. The latter indicates that the growth and/or differentiation of tumor cells is reduced, decreased, diminished, slowed, etc. The term "proliferation" includes any process which relates to cell growth and division, and includes differentiation and apoptosis. As discussed above, raf kinases play a key role in the activation of the cytoplasmic signaling cascade involved in cell proliferation, differentiation, and apoptosis. For example, studies have found that inhibiting c-raf by anti-sense oligonucleotides can block cell proliferation (see above). Any amount of inhibition is considered therapeutic.
Included in the methods of the present invention is a method for using synthetic metabolites of the compound described above (Compound of Formula I), including salts, prodrugs, and compositions thereof, to treat mammalian hyper-proliferative disorders comprising administering to a mammal, including a human in need thereof, an amount of a synthetic metabolite of a compound of this invention, pharmaceutically acceptable salt, and composition thereof, which is effective to treat the disorder. Hyper-proliferative disorders include but are not limited to solid tumors, such as cancers of the breast, respiratory tract, brain, reproductive organs, digestive tract, urinary tract, eye, liver, skin, head and neck, thyroid, parathyroid and their distant metastases. Those disorders also include lymphomas, sarcomas, and leukemias.
Any tumor or cancer can be treated, including, but not limited to, cancers having one or more mutations in raf, ras, and/or flt-3, as well as any upstream or downstream member of the signaling pathways of which they are a part. As discussed earlier, a cancer can be treated with a compound of the present invention irrespective of the mechanism which is responsible for it. Cancers of any organ can be treated, including cancers of, but are not limited to, e.g., colon, pancreas, breast, prostate, bone, liver, kidney, lung, testes, skin, pancreas, stomach, colorectal cancer, renal cell carcinoma, hepatocellular carcinoma, melanoma, etc.
Examples of breast cancer include, but are not limited to, invasive ductal carcinoma, invasive lobular carcinoma, ductal carcinoma in situ, and lobular carcinoma in situ.
Examples of cancers of the respiratory tract include, but are not limited to, small-cell and non-small-cell lung carcinoma, as well as bronchial adenoma and pleuropulmonary blastoma.
Examples of brain cancers include, but are not limited to, brain stem and hypophtalmic glioma, cerebellar and cerebral astrocytoma, medulloblastoma, ependymoma, as well as neuroectodermal and pineal tumor.
Tumors of the male reproductive organs include, but are not limited to, prostate and testicular cancer. Tumors of the female reproductive organs include, but are not limited to, endometrial, cervical, ovarian, vaginal, and vulvar cancer, as well as sarcoma of the uterus.
Tumors of the digestive tract include, but are not limited to, anal, colon, colorectal, esophageal, gallbladder, gastric, pancreatic, rectal, small-intestine, and salivary gland cancers.
Tumors of the urinary tract include, but are not limited to, bladder, penile, kidney, renal pelvis, ureter, and urethral cancers.
Eye cancers include, but are not limited to, intraocular melanoma and retinoblastoma. Examples of liver cancers include, but are not limited to, hepatocellular carcinoma (liver cell carcinomas with or without fibrolamellar variant), cholangiocarcinoma (intrahepatic bile duct carcinoma), and mixed hepatocellular cholangiocarcinoma. Skin cancers include, but are not limited to, squamous cell carcinoma, Kaposi's sarcoma, malignant melanoma, Merkel cell skin cancer, and non-melanoma skin cancer.
Head-and-neck cancers include, but are not limited to, laryngeal, hypopharyngeal, nasopharyngeal, and/or oropharyngeal cancers, and lip and oral cavity cancer.
Lymphomas include, but are not limited to, AIDS-related lymphoma, non-Hodgkin's lymphoma, cutaneous T-cell lymphoma, Hodgkin's disease, and lymphoma of the central nervous system. Sarcomas include, but are not limited to, sarcoma of the soft tissue, osteosarcoma, malignant fibrous histiocytoma, lymphosarcoma, and rhabdomyosarcoma.
Leukemias include, but are not limited to, acute myeloid leukemia, acute lymphoblastic leukemia, chronic lymphocytic leukemia, chronic myelogenous leukemia, and hairy cell leukemia.
In addition to inhibiting the proliferation of tumor cells, compounds of the present invention can also cause tumor regression, e.g., a decrease in the size of a tumor, or in the extent of cancer in the body.
The present invention also relates to methods of modulating angiogenesis and/or lymphangiogenesis in a system comprising cells, comprising administering to the system an effective amount of a compound described herein. A system comprising cells can be an in vivo system, such as a tumor in a patient, isolated organs, tissues, or cells, in vitro assays systems (CAM, BCE, etc), animal models (e.g., in vivo, subcutaneous, cancer models), hosts in need of treatment (e.g., hosts suffering from diseases having angiogenic and/or lymphangiogenic component, such as cancer), etc. Inappropriate and ectopic expression of angiogenesis can be deleterious to an organism. A number of pathological conditions are associated with the growth of extraneous blood vessels. These include, e.g., diabetic retinopathy, neovascular glaucoma, psoriasis, retrolental fibroplasias, angiofibroma, inflammation, etc. In addition, the increased blood supply associated with cancerous and neoplastic tissue, encourages growth, leading to rapid tumor enlargement and metastasis. Moreover, the growth of new blood and lymph vessels in a tumor provides an escape route for renegade cells, encouraging metastasis and the consequence spread of the cancer. Useful systems for measuring angiogenesis and/or lymphangiogenesis, and inhibition thereof, include, e.g., neovascularization of tumor explants (e.g., U.S. Pat. Nos. 5,192,744; 6,024,688), chicken chorioallantoic membrane (CAM) assay (e.g., Taylor and Folkman, Nature 1982, 297, 307-312; Eliceiri et al., J. Cell Biol. 1998, 140, 1255-1263), bovine capillary endothelial (BCE) cell assay (e.g., U.S. Pat. No. 6,024,688; Polverini, P. J. et al., Methods Enzymol. 1991, 198, 440-450), migration assays, and HUVEC (human umbilical cord vascular endothelial cell) growth inhibition assay (e.g., U.S. Pat. No. 6,060,449), and use of the rabbit ear model (e.g., Szuba et al., FASEB J. 2002, 16(14), 1985-7).
Modulation of angiogenesis can be determined by any other method. For example, the degree of tissue vascularity is typically determined by assessing the number and density of vessels present in a given sample. For example, microvessel density (MVD) can be estimated by counting the number of endothelial clusters in a high-power microscopic field, or detecting a marker specific for microvascular endothelium or other markers of growing or established blood vessels, such as CD31 (also known as platelet-endothelial cell adhesion molecule or PEC AM). A CD31 antibody can be employed in conventional immunohistological methods to immunostain tissue sections as described by, e.g., U.S. Pat. No. 6,017,949; Delias et al., Gyn. Oncol. 1997, 67, 27-33; and others. Other markers for angiogenesis, include, e.g., Vezfl (e.g., Xiang et al., Dev. Bio. 1999, 206, 123-141), angiopoietin, Tie-1, and Tie-2 (e.g., Sato et al., Nature 1995, 376, 70-74).
Additionally, the present invention relates to methods of screening patients to determine their sensitivity to compounds of the present invention. For example, the invention relates to methods of determining whether a condition can be modulated by a compound disclosed herein, comprising measuring the expression or activity of raf, VEGFR-2, VEGFR-3, PDGFR-beta, p38, and/or flt-3 in a sample comprising cells or a cell extract, wherein said sample has been obtained from a cell or subject having said condition. When the results of the determination indicate that one or more of the mentioned genes (and/or polypeptides which they encode) differ from the normal state, this identifies the condition as being treatable with a compound of the present invention, i.e., whereby said disorder or condition can be modulated by the compound when said expression or activity is increased in said condition as compared to a normal control. The method can further comprise a step of comparing the expression in a sample with a normal control, or expression in a sample obtained from normal or unaffected tissue. Comparing can be done manually, against a standard, in an electronic form (e.g., against a database), etc. The normal control can be a standard sample that is provided with the assay; it can be obtained from adjacent, but unaffected, tissue from the same patient; or, it can be pre-determined values, etc. Gene expression, protein expression (e.g., abundance in a cell), protein activity (e.g., kinase activity), etc., can be determined.
For instance, a biopsy from a cancer patient can be assayed for the presence, quantity, and/or activity of raf, VEGFR-2, VEGFR-3, PDGFR-beta, p38, and/or flt-3. Increased expression or activity of one or more of these can indicate that the cancer can be targeted for treatment by a compound of the present invention. For example, as described in the examples below, raf activity can be monitored by its ability to initiate the cascade leading to ERK phosphorylation (i.e., raf/MEK/ERK), resulting in phospho-ERK. Increased phospho-ERK levels in a cancer specimen shows that its raf activity is elevated, suggesting the use of compounds of the present invention to treat it. Measuring expression includes determining or detecting the amount of the polypeptide present in a cell or shed by it, as well as measuring the underlying mRNA, where the quantity of mRNA present is considered to reflect the quantity of polypeptide manufactured by the cell. Furthermore, the genes for raf, VEGFR-2, VEGFR-3, PDGFR-beta, p38, and/or Flt-3 can be analyzed to determine whether there is a gene defect responsible for aberrant expression or polypeptide activity.
Polypeptide detection can be carried out by any available method, e.g., by Western blots, ELISA, dot blot, immunoprecipitation, RIA, immunohistochemistry, etc. For instance, a tissue section can be prepared and labeled with a specific antibody (indirect or direct and visualized with a microscope. Amount of a polypeptide can be quantitated without visualization, e.g., by preparing a lysate of a sample of interest, and then determining by ELISA or Western the amount of polypeptide per quantity of tissue. Antibodies and other specific binding agents can be used. There is no limitation on how detection is performed.
Assays can be utilized which permit quantification and/or presence/absence detection of a target nucleic acid (e.g., genes, mRNA, etc., for raf, VEGFR, PDGFR, p38, and/or flt-3) in a sample. Assays can be performed at the single-cell level, or in a sample comprising many cells, where the assay is "averaging" expression over the entire collection of cells and tissue present in the sample. Any suitable assay format can be used, including, but not limited to, e.g., Southern blot analysis, Northern blot analysis, polymerase chain reaction ("PCR") (e.g., Saiki et al., Science 1988, 241, 53; U.S. Pat. Nos. 4,683,195, 4,683,202, and 6,040,166; PCR Protocols: A Guide to Methods and Applications, Innis et al., eds., Academic Press, New York, 1990), reverse transcriptase polymerase chain reaction ("RT-PCR"), anchored PCR, rapid amplification of cDNA ends ("RACE") (e.g., Schaefer in Gene Cloning and Analysis: Current Innovations, Pages 99-115, 1997), ligase chain reaction ("LCR") (EP 320 308), onesided PCR (Ohara et al., Proc. Natl. Acad. Sci. 1989, 86, 5673-5677), indexing methods (e.g., U.S. Pat. No. 5,508,169), in situ hybridization, differential display (e.g., Liang et al., Nucl. Acid. Res. 1993, 21, 3269 3275; U.S. Pat. Nos. 5,262,311, 5,599,672 and 5,965,409; W097/18454; Prashar and Weissman, Proc. Natl. Acad. Sci., 93:659-663, and U.S. Pat. Nos. 6,010,850 and 5,712,126; Welsh et al., Nucleic Acid Res., 20:4965-4970, 1992, and U.S. Pat. No. 5,487,985) and other RNA fingerprinting techniques, nucleic acid sequence based amplification ("NASBA") and other transcription based amplification systems (e.g., U.S. Pat. Nos. 5,409,818 and 5,554,527; WO 88/10315), polynucleotide arrays (e.g., U.S. Pat. Nos. 5,143,854, 5,424,186; 5,700,637, 5,874,219, and 6,054,270; PCT WO 92/10092; PCT WO 90/15070), Qbeta Replicase (PCT/US 87/00880), Strand Displacement Amplification ("SDA"), Repair Chain Reaction ("RCR"), nuclease protection assays, subtraction-based methods, Rapid-Scan, etc. Additional useful methods include, but are not limited to, e.g., template-based amplification methods, competitive PCR (e.g., U.S. Pat. No. 5,747,251), redox-based assays (e.g., U.S. Pat. No. 5,871,918), Taqman-based assays (e.g., Holland et al., Proc. Natl. Acad, Sci. 1991, 88, 7276-7280; U.S. Pat. Nos. 5,210,015 and 5,994,063), realtime fluorescence-based monitoring (e.g., U.S. Pat. 5,928,907), molecular energy transfer labels (e.g., U.S. Pat. Nos. 5,348,853, 5,532,129, 5,565,322, 6,030,787, and 6,117,635; Tyagi and Kramer, Nature Biotech., 14:303-309, 1996). Any method suitable for single cell analysis of gene or protein expression can be used, including in situ hybridization, immunocytochemistry, MACS, FACS, flow cytometry, etc. For single cell assays, expression products can be measured using antibodies, PCR, or other types of nucleic acid amplification (e.g., Brady et al., Methods Mol. & Cell. Biol. 1990, 2, 17-25; Eberwine et al., Proc. Natl. Acad. Sci. 1992, 89, 3010-3014; U.S. Pat. No. 5,723,290). These and other methods can be carried out conventionally, e.g., as described in the mentioned publications. Activity of raf, VEGFR-2, VEGFR-3, PDGFR-beta, p38, and/or flt-3 can be assessed routinely, e.g., as described in the examples below, or using standard assays for kinase activity.
The present invention also provides methods of assessing the efficacy of a compound of the present invention in treating a disorder, comprising one or more of the following steps in any effective order, e.g., administering an amount of a compound, measuring the expression or activity of raf, VEGFR-2, VEGFR-3, PDGFR-beta, p38, and/or flt-3 (see above), determining the effect of said compound on said expression or activity. For instance, biopsy samples can be removed from patients who have been treated with a compound of the present invention, and then assayed for the presence and/or activity of the mentioned signaling molecules. Similarly, as discussed above, decreases in the levels of phospho-ERK in the cancer tissue (e.g., compared to normal tissue or before treatment) indicate that the compound is exerting in vivo efficacy and a therapeutic effect. The method can be used to determine appropriate dosages and dosing regimens, e.g., how much compound to administer and at what frequency to administer it. By monitoring its effect on the signaling molecules in the tissue, the clinician can determine the appropriate treatment protocol and whether it is achieving the desired effect, e.g., on modulating or inhibiting the signal transduction pathway.
Compounds of the present invention also can be used as markers to determine the presence and quantity of raf, VEGFR-2, VEGFR-3, PDGFR-beta, p38, and/or flt-3, in a sample comprising a biological material. This comprises one or more of the following steps in any effective order: (i) contacting said sample comprising a biological material with a compound of the present invention, and (ii) determining whether said compound binds to said material. The compound can be labeled, or it can be used as a competitor to a labeled compound, such as labeled- ATP. The invention also provides methods for treating, preventing, modulating, etc., diseases and conditions in mammals comprising administering a compound of this invention with another modulator of the signal transduction pathway comprising, but not limited to raf, VEGFR, PDGFR, p38, and/or flt-3. These can be present in the same composition or in separate formulations or dosage units. Administration can be the same or different routes, and can be simultaneous or sequential.
The following publications relate to VEGFR-3 modulation and are incorporated herein for their description of disease states mediated by VEGFR-3 and assays to determine such activity.
W095/33772 Alitalo, et. al.
WO95/33050 Charnock- Jones, et. al..
W096/39421 Hu, et. al.
W098/33917 Alitalo, et. al.
WO02/057299 Alitalo, et. al.
WO02/060950 Alitalo, et. al.
WO02/081520 Boesen, et. al. The following publications relate to VEGFR-2 modulation and are incorporated herein for their description of disease states mediated by VEGFR-2 and assays to determine such activity.
EP0882799 Hanai, et. al.
EP1167384 Ferraram, et, al.
EP1086705 Sato, et. al.
EP11300032 Tesar, et. al.
EP1166798 Haberey, et. al.
EP1166799 Haberey, et. al.
EP1170017 Maini, et. al.
EP 1203827 Smith
WO02/083850 Rosen, et. al. The following publications relate to flt-3 modulation and are incorporated herein for their description of disease states mediated by flt-3 and assays to determine such activity.
2002/0034517 Brasel, et. al.
2002/0107365 Lyman, et. al.
2002/0111475 Graddis, et. al.
EP0627487 Beckermann, et. al.
WO9846750 Bauer, et. al.
W09818923 McWherter, et. al.
W09428391 Beckermann, et al.
W09426891 Birnbaum, et. al.
The following patents and publication relate to PDGF/PDGFR modulation and are incorporated herein for their description of the disease states mediated by PDGFR-beta and assays to determine such activity.
5,094,941 Hart, et. al.
5,371,205 Kelly, et. al.
5,418,135 Pang
5,444,151 Vassbotn, et. al.
5,468,468 LaRochelle, et. al.
5,567,584 Sledziewski, et. al.
5,618,678 Kelly, et. al.
5,620,687 Hart, et. al.
5,648,076 Ross, et. al.
5,668,264 Janjic, et. al.
5,686,572 Wolf, et. al.
5,817,310 Ramakrishnan, et. al.
5,833,986 LaRochelle, et. al.
5,863,739 LaRochelle, et. al.
5,872,218 Wolf, et. al.
5,882,644 Chang, et. al.
5,891,652 Wolf, et. al.
5,976,534 Hart, et. al. 5,990,141 Hirth, et. al.
6,022,854 Shuman
6,043,211 Williams, et. al.
6,110,737 Escobedo, et. al.
6,207,816B1 Gold, et. al.
6,228,600B 1 Matsui, et. al.
6,229 ,002B 1 Janjic, et. al.
6,316,603B1 McTigue, et. al.
6,372,438B 1 Williams, et. al.
6,403,769B 1 La Rochelle, et. al.
6,440,445B1 Nowak, et. al.
6,475,782B 1 Escobedo, et. al.
WO02/083849 Rosen, et. al.
WO02/083704 Rosen, et. al.
WO02/081520 Boesen, et. al.
WO02/079498 Thomas, et. al.
WO02/070008 Rockwell, et. al.
WO09959636 Sato, et. al.
WO09946364 Cao, et. al.
WO09940118 Hanai, et. al.
W09931238 Yabana, et. al.
W09929861 Klagsbrun, et. al.
WO9858053 Kendall, et. al.
W09851344 Maini, et. al.
W09833917 Alitalo, et. al.
W09831794 Matsumoto, et. al.
W09816551 Ferrara, et. al.
WO9813071 Kendall, et al.
W09811223 Martiny-Baron, et. al
W09744453 Chen, et. al.
WO9723510 Plouet, et. al.
W09715662 Stinchcomb, et. al.
WO9708313 Ferrara, et. al.
W09639515 Cao, et. al. WO9623065 Smith, et. al.
WO9606641 Fleurbaaij, et. al.
W09524473 Cao, et. al.
W09822316 Kyowa
W09521868 Rockwell, et. al.
WO02/060489 Xia, et. al.
PDGFR-beta EP0869177 Matsui, et. al.
WO09010013 Matsui, et. al.
WO9737029 Matsui, et. al.
PDGFR-alpha
EP 1000617 Lammers, et. al.
EP0869177 Matsui, et. al.
EP0811685 Escobedo, et. al. Pharmaceutical compositions based on the compounds of the present invention
This invention also relates to pharmaceutical compositions containing a compound of the present invention and pharmaceutically acceptable salts thereof. These compositions can be utilized to achieve the desired pharmacological effect by administration to a patient in need thereof. A patient, for the purpose of this invention, is a mammal, including a human, in need of treatment for the particular condition or disease. Therefore, the present invention includes pharmaceutical compositions which are comprised of a pharmaceutically acceptable carrier and a pharmaceutically effective amount of a compound, or salt thereof, of the present invention. The term "pharmaceutically acceptable carrier" is meant as any carrier which is relatively non-toxic and innocuous to a patient at concentrations consistent with effective activity of the active ingredient so that any side effects ascribable to the carrier do not vitiate the beneficial effects of the active ingredient. A pharmaceutically effective amount of compound is that amount which produces a result or exerts an influence on the particular condition being treated. The compound of the present invention can be administered with pharmaceutically-acceptable carriers well known in the art using any effective conventional dosage unit forms, including immediate, slow and timed release preparations, orally, parenterally, topically, nasally, ophthalmically, optically, sublingually, rectally, vaginally, and the like.
For oral administration, the compound can be formulated into solid or liquid preparations such as capsules, pills, tablets, troches, lozenges, melts, powders, solutions, suspensions, or emulsions, and may be prepared according to methods known to the art for the manufacture of pharmaceutical compositions. The solid unit dosage forms can be a capsule which can be of the ordinary hard- or soft-shelled gelatin type containing, for example, surfactants, lubricants, and inert fillers such as lactose, sucrose, calcium phosphate, and corn starch.
In another embodiment, the compounds of this invention may be tableted with conventional tablet bases such as lactose, sucrose and cornstarch in combination with binders such as acacia, corn starch or gelatin, disintegrating agents intended to assist the break-up and dissolution of the tablet following administration such as potato starch, alginic acid, corn starch, and guar gum, gum tragacanth, acacia, lubricants intended to improve the flow of tablet granulation and to prevent the adhesion of tablet material to the surfaces of the tablet dies and punches, for example talc, stearic acid, or magnesium, calcium or zinc stearate, dyes, coloring agents, and flavoring agents such as peppermint, oil of wintergreen, or cherry flavoring, intended to enhance the aesthetic qualities of the tablets and make them more acceptable to the patient. Suitable excipients for use in oral liquid dosage forms include dicalcium phosphate and diluents such as water and alcohols, for example, ethanol, benzyl alcohol, and polyethylene alcohols, either with or without the addition of a pharmaceutically acceptable surfactant, suspending agent or emulsifying agent. Various other materials may be present as coatings or to otherwise modify the physical form of the dosage unit. For instance tablets, pills or capsules may be coated with shellac, sugar or both.
Dispersible powders and granules are suitable for the preparation of an aqueous suspension. They provide the active ingredient in admixture with a dispersing or wetting agent, a suspending agent and one or more preservatives. Suitable dispersing or wetting agents and suspending agents are exemplified by those already mentioned above. Additional excipients, for example those sweetening, flavoring and coloring agents described above, may also be present. The pharmaceutical compositions of this invention may also be in the form of oil-in-water emulsions. The oily phase may be a vegetable oil such as liquid paraffin or a mixture of vegetable oils. Suitable emulsifying agents may be (1) naturally occurring gums such as gum acacia and gum tragacanth, (2) naturally occurring phosphatides such as soy bean and lecithin, (3) esters or partial esters derived form fatty acids and hexitol anhydrides, for example, sorbitan monooleate, (4) condensation products of said partial esters with ethylene oxide, for example, polyoxyethylene sorbitan monooleate. The emulsions may also contain sweetening and flavoring agents.
Oily suspensions may be formulated by suspending the active ingredient in a vegetable oil such as, for example, arachis oil, olive oil, sesame oil or coconut oil, or in a mineral oil such as liquid paraffin. The oily suspensions may contain a thickening agent such as, for example, beeswax, hard paraffin, or cetyl alcohol. The suspensions may also contain one or more preservatives, for example, ethyl or n-propyl p-hydroxybenzoate; one or more coloring agents; one or more flavoring agents; and one or more sweetening agents such as sucrose or saccharin.
Syrups and elixirs may be formulated with sweetening agents such as, for example, glycerol, propylene glycol, sorbitol or sucrose. Such formulations may also contain a demulcent, and preservative, such as methyl and propyl parabens and flavoring and coloring agents.
The compounds of this invention may also be administered parenterally, that is, subcutaneously, intravenously, intraocularly, intrasynovially, intramuscularly, or interperitoneally, as injectable dosages of the compound in a physiologically acceptable diluent with a pharmaceutical carrier which can be a sterile liquid or mixture of liquids such as water, saline, aqueous dextrose and related sugar solutions, an alcohol such as ethanol, isopropanol, or hexadecyl alcohol, glycols such as propylene glycol or polyethylene glycol, glycerol ketals such as 2,2-dimethyl-l,l-dioxolane-4-methanol, ethers such as poly(ethylene glycol) 400, an oil, a fatty acid, a fatty acid ester or, a fatty acid glyceride, or an acetylated fatty acid glyceride, with or without the addition of a pharmaceutically acceptable surfactant such as a soap or a detergent, suspending agent such as pectin, carbomers, methycellulose, hydroxypropylmethylcellulose, or carboxymethylcellulose, or emulsifying agent and other pharmaceutical adjuvants. Illustrative of oils which can be used in the parenteral formulations of this invention are those of petroleum, animal, vegetable, or synthetic origin, for example, peanut oil, soybean oil, sesame oil, cottonseed oil, corn oil, olive oil, petrolatum and mineral oil. Suitable fatty acids include oleic acid, stearic acid, isostearic acid and myristic acid. Suitable fatty acid esters are, for example, ethyl oleate and isopropyl myristate. Suitable soaps include fatty acid alkali metal, ammonium, and triethanolamine salts and suitable detergents include cationic detergents, for example dimethyl dialkyl ammonium halides, alkyl pyridinium halides, and alkylamine acetates; anionic detergents, for example, alkyl, aryl, and olefin sulfonates, alkyl, olefin, ether, and monoglyceride sulfates, and sulfosuccinates; non-ionic detergents, for example, fatty amine oxides, fatty acid alkanolamides, and poly(oxyethylene-oxypropylene)s or ethylene oxide or propylene oxide copolymers; and amphoteric detergents, for example, alkyl-beta-aminopropionates, and 2-alkylimidazoline quarternary ammonium salts, as well as mixtures.
The parenteral compositions of this invention will typically contain from about 0.5% to about 25% by weight of the active ingredient in solution. Preservatives and buffers may also be used advantageously. In order to minimize or eliminate irritation at the site of injection, such compositions may contain a non-ionic surfactant having a hydrophile-lipophile balance (HLB) of from about 12 to about 17. The quantity of surfactant in such formulation ranges from about 5% to about 15% by weight. The surfactant can be a single component having the above HLB or can be a mixture of two or more components having the desired HLB.
Illustrative of surfactants used in parenteral formulations are the class of polyethylene sorbitan fatty acid esters, for example, sorbitan monooleate and the high molecular weight adducts of ethylene oxide with a hydrophobic base, formed by the condensation of propylene oxide with propylene glycol.
The pharmaceutical compositions may be in the form of sterile injectable aqueous suspensions. Such suspensions may be formulated according to known methods using suitable dispersing or wetting agents and suspending agents such as, for example, sodium carboxymethylcellulose, methylcellulose, hydroxypropylmethyl-cellulose, sodium alginate, gum tragacanth and gum acacia; dispersing or wetting agents which may be a naturally occurring phosphatide such as lecithin, a condensation product of an alkylene oxide with a fatty acid, for example, polyoxyethylene stearate, a condensation product of ethylene oxide with a long chain aliphatic alcohol, for example, heptadeca-ethyleneoxycetanol, a condensation product of ethylene oxide with a partial ester derived form a fatty acid and a hexitol such as polyoxyethylene sorbitol monooleate, or a condensation product of an ethylene oxide with a partial ester derived from a fatty acid and a hexitol anhydride, for example polyoxyethylene sorbitan monooleate.
The sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally acceptable diluent or solvent. Diluents and solvents that may be employed are, for example, water, Ringer's solution, isotonic sodium chloride solutions and isotonic glucose solutions. In addition, sterile fixed oils are conventionally employed as solvents or suspending media. For this purpose, any bland, fixed oil may be employed including synthetic mono- or diglycerides. In addition, fatty acids such as oleic acid can be used in the preparation of injectables.
A composition of the invention may also be administered in the form of suppositories for rectal administration of the drug. These compositions can be prepared by mixing the drug with a suitable non-irritation excipient which is solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum to release the drug. Such material is, for example, cocoa butter and polyethylene glycol.
Another formulation employed in the methods of the present invention employs transdermal delivery devices ("patches"). Such transdermal patches may be used to provide continuous or discontinuous infusion of the compounds of the present invention in controlled amounts. The construction and use of transdermal patches for the delivery of pharmaceutical agents is well known in the art (see, e.g., US Patent No. 5,023,252, issued June 11, 1991, incorporated herein by reference). Such patches may be constructed for continuous, pulsatile, or on demand delivery of pharmaceutical agents. Controlled release formulations for parenteral administration include liposomal, polymeric microsphere and polymeric gel formulations which are known in the art.
It may be desirable or necessary to introduce the pharmaceutical composition to the patient via a mechanical delivery device. The construction and use of mechanical delivery devices for the delivery of pharmaceutical agents is well known in the art. Direct techniques for, for example, administering a drug directly to the brain usually involve placement of a drug delivery catheter into the patient's ventricular system to bypass the blood-brain barrier. One such implantable delivery system, used for the transport of agents to specific anatomical regions of the body, is described in US Patent No. 5,011,472, issued April 30, 1991.
The compositions of the invention can also contain other conventional pharmaceutically acceptable compounding ingredients, generally referred to as carriers or diluents, as necessary or desired. Conventional procedures for preparing such compositions in appropriate dosage forms can be utilized. Such ingredients and procedures include those described in the following references, each of which is incorporated herein by reference: Powell, M.F. et al, "Compendium of Excipients for Parenteral Formulations" PDA Journal of Pharmaceutical Science & Technology 1998, 52(5), 238-311; Strickley, R.G "Parenteral Formulations of Small Molecule Therapeutics Marketed in the United States (1999)-Part-1" PDA Journal of Pharmaceutical Science & Technology 1999, 53(6), 324-349; and Nema, S. et al, "Excipients and Their Use in Injectable Products" PDA Journal of Pharmaceutical Science & Technology 1997, 51(4), 166-171.
Commonly used pharmaceutical ingredients which can be used as appropriate to formulate the composition for its intended route of administration include:
acidifying agents (examples include but are not limited to acetic acid, citric acid, fumaric acid, hydrochloric acid, nitric acid);
alkalinizing agents (examples include but are not limited to ammonia solution, ammonium carbonate, diethanolamine, monoethanolamine, potassium hydroxide, sodium borate, sodium carbonate, sodium hydroxide, triethanolamine, trolamine);
adsorbents (examples include but are not limited to powdered cellulose and activated charcoal);
aerosol propellants (examples include but are not limited to carbon dioxide, CC12F2, F2C1C- CC1F2 and CC1F3)
air displacement agents (examples include but are not limited to nitrogen and argon);
antifungal preservatives (examples include but are not limited to benzoic acid, butylparaben, ethylparaben, methylparaben, propylparaben, sodium benzoate); antimicrobial preservatives (examples include but are not limited to benzalkonium chloride, benzethonium chloride, benzyl alcohol, cetylpyridinium chloride, chlorobutanol, phenol, phenylethyl alcohol, phenylmercuric nitrate and thimerosal);
antioxidants (examples include but are not limited to ascorbic acid, ascorbyl palmitate, butylated hydroxyanisole, butylated hydroxytoluene, hypophosphorus acid, monothioglycerol, propyl gallate, sodium ascorbate, sodium bisulfite, sodium formaldehyde sulfoxylate, sodium metabisulfite);
binding materials (examples include but are not limited to block polymers, natural and synthetic rubber, polyacrylates, polyurethanes, silicones, polysiloxanes and styrene-butadiene copolymers);
buffering agents (examples include but are not limited to potassium metaphosphate, dipotassium phosphate, sodium acetate, sodium citrate anhydrous and sodium citrate dihydrate)
carrying agents (examples include but are not limited to acacia syrup, aromatic syrup, aromatic elixir, cherry syrup, cocoa syrup, orange syrup, syrup, corn oil, mineral oil, peanut oil, sesame oil, bacteriostatic sodium chloride injection and bacteriostatic water for injection) chelating agents (examples include but are not limited to edetate disodium and edetic acid) colorants (examples include but are not limited to FD&C Red No. 3, FD&C Red No. 20, FD&C Yellow No. 6, FD&C Blue No. 2, D&C Green No. 5, D&C Orange No. 5, D&C Red No. 8, caramel and ferric oxide red);
clarifying agents (examples include but are not limited to bentonite);
emulsifying agents (examples include but are not limited to acacia, cetomacrogol, cetyl alcohol, glyceryl monostearate, lecithin, sorbitan monooleate, polyoxyethylene 50 monostearate);
encapsulating agents (examples include but are not limited to gelatin and cellulose acetate phthalate)
flavorants (examples include but are not limited to anise oil, cinnamon oil, cocoa, menthol, orange oil, peppermint oil and vanillin);
humectants (examples include but are not limited to glycerol, propylene glycol and sorbitol); levigating agents (examples include but are not limited to mineral oil and glycerin);
oils (examples include but are not limited to arachis oil, mineral oil, olive oil, peanut oil, sesame oil and vegetable oil); ointment bases (examples include but are not limited to lanolin, hydrophilic ointment, polyethylene glycol ointment, petrolatum, hydrophilic petrolatum, white ointment, yellow ointment, and rose water ointment);
penetration enhancers (transdermal delivery) (examples include but are not limited to monohydroxy or polyhydroxy alcohols, mono-or polyvalent alcohols, saturated or unsaturated fatty alcohols, saturated or unsaturated fatty esters, saturated or unsaturated dicarboxylic acids, essential oils, phosphatidyl derivatives, cephalin, terpenes, amides, ethers, ketones and ureas)
plasticizers (examples include but are not limited to diethyl phthalate and glycerol);
solvents (examples include but are not limited to ethanol, corn oil, cottonseed oil, glycerol, isopropanol, mineral oil, oleic acid, peanut oil, purified water, water for injection, sterile water for injection and sterile water for irrigation);
stiffening agents (examples include but are not limited to cetyl alcohol, cetyl esters wax, microcrystalline wax, paraffin, stearyl alcohol, white wax and yellow wax);
suppository bases (examples include but are not limited to cocoa butter and polyethylene glycols (mixtures));
surfactants (examples include but are not limited to benzalkonium chloride, nonoxynol 10, oxtoxynol 9, polysorbate 80, sodium lauryl sulfate and sorbitan mono-palmitate);
suspending agents (examples include but are not limited to agar, bentonite, carbomers, carboxymethylcellulose sodium, hydroxyethyl cellulose, hydroxypropyl cellulose, hydroxypropyl methylcellulose, kaolin, methylcellulose, tragacanth and veegum);
sweetening agents (examples include but are not limited to aspartame, dextrose, glycerol, mannitol, propylene glycol, saccharin sodium, sorbitol and sucrose);
tablet anti- adherents (examples include but are not limited to magnesium stearate and talc); tablet binders (examples include but are not limited to acacia, alginic acid, carboxymethylcellulose sodium, compressible sugar, ethylcellulose, gelatin, liquid glucose, methylcellulose, and pregelatinized starch);
tablet and capsule diluents (examples include but are not limited to dibasic calcium phosphate, kaolin, lactose, mannitol, microcrystalline cellulose, powdered cellulose, precipitated calcium carbonate, sodium carbonate, sodium phosphate, sorbitol and starch); tablet coating agents (examples include but are not limited to liquid glucose, hydroxyethyl cellulose, hydroxypropyl cellulose, hydroxypropyl methylcellulose, methylcellulose, ethylcellulose, cellulose acetate phthalate and shellac); tablet direct compression excipients (examples include but are not limited to dibasic calcium phosphate);
tablet disintegrants (examples include but are not limited to alginic acid, carboxymethylcellulose calcium, microcrystalline cellulose, polacrillin potassium, sodium alginate, sodium starch glycollate and starch);
tablet glidants (examples include but are not limited to colloidal silica, corn starch and talc); tablet lubricants (examples include but are not limited to calcium stearate, magnesium stearate, mineral oil, stearic acid and zinc stearate);
tablet/capsule opaquants (examples include but are not limited to titanium dioxide);
tablet polishing agents (examples include but are not limited to carnauba wax and white wax);
thickening agents (examples include but are not limited to beeswax, cetyl alcohol and paraffin);
tonicity agents (examples include but are not limited to dextrose and sodium chloride);
viscosity increasing agents (examples include but are not limited to alginic acid, bentonite, carbomers, carboxymethylcellulose sodium, methylcellulose, sodium alginate and tragacanth); and
wetting agents (examples include but are not limited to heptadecaethylene oxycetanol, lecithin, sorbitol monooleate, polyoxyethylene sorbitol monooleate, and polyoxyethylene stearate).
Pharmaceutical compositions according to the present invention can be illustrated as follows:
Sterile IV Solution: a 5 mg/mL solution of the desired compound of this invention is made using sterile, injectable water, and the pH is adjusted if necessary. The solution is diluted for administration to 1 - 2 mg/mL with sterile 5% dextrose and is administered as an IV infusion over 60 minutes.
Lyophilized powder for IV administration: A sterile preparation can be prepared with (i) 100 - 1000 mg of the desired compound of this invention as a lypholized powder, (ii) 32- 327 mg/mL sodium citrate, and (iii) 300 - 3000 mg Dextran 40. The formulation is reconstituted with sterile, injectable saline or dextrose 5% to a concentration of 10 to 20 mg/mL, which is further diluted with saline or dextrose 5% to 0.2 - 0.4 mg/mL, and is administered either IV bolus or by IV infusion over 15 - 60 minutes. Intramuscular suspension: The following solution or suspension can be prepared, for intramuscular injection:
50 mg/mL of the desired, water-insoluble compound of this invention
5 mg/mL sodium carboxymethylcellulose
4 mg/mL Tween 80
9 mg/mL sodium chloride
9 mg/mL benzyl alcohol Hard Shell Capsules: A large number of unit capsules are prepared by filling standard two- piece hard galantine capsules each with 100 mg of powdered active ingredient, 150 mg of lactose, 50 mg of cellulose and 6 mg of magnesium stearate.
Soft Gelatin Capsules: A mixture of active ingredient in a digestible oil such as soybean oil, cottonseed oil or olive oil is prepared and injected by means of a positive displacement pump into molten gelatin to form soft gelatin capsules containing 100 mg of the active ingredient. The capsules are washed and dried. The active ingredient can be dissolved in a mixture of polyethylene glycol, glycerin and sorbitol to prepare a water miscible medicine mix. Tablets: A large number of tablets are prepared by conventional procedures so that the dosage unit was 100 mg of active ingredient, 0.2 mg of colloidal silicon dioxide, 5 mg of magnesium stearate, 275 mg of microcrystalline cellulose, 11 mg of starch, and 98.8 mg of lactose. Appropriate aqueous and non-aqueous coatings may be applied to increase palatability, improve elegance and stability or delay absorption.
Immediate Release Tablets/Capsules: These are solid oral dosage forms made by conventional and novel processes. These units are taken orally without water for immediate dissolution and delivery of the medication. The active ingredient is mixed in a liquid containing ingredient such as sugar, gelatin, pectin and sweeteners. These liquids are solidified into solid tablets or caplets by freeze drying and solid state extraction techniques. The drug compounds may be compressed with viscoelastic and thermoelastic sugars and polymers or effervescent components to produce porous matrices intended for immediate release, without the need of water. Dosage of the pharmaceutical compositions of the present invention
Based upon standard laboratory techniques known to evaluate compounds useful for the treatment of any of the aforementioned disorders, by standard toxicity tests and by standard pharmacological assays for the determination of treatment of the conditions identified above in mammals, and by comparison of these results with the results of known medicaments that are used to treat these conditions, the effective dosage of the compounds of this invention can readily be determined for treatment of each desired indication. The amount of the active ingredient to be administered in the treatment of one of these conditions can vary widely according to such considerations as the particular compound and dosage unit employed, the mode of administration, the period of treatment, the age and sex of the patient treated, and the nature and extent of the condition treated.
The total amount of the active ingredient to be administered can range from about 0.001 mg/kg to about 200 mg/kg, and preferably from about 0.1 mg/kg to about 50 mg/kg body weight per day. A unit dosage may preferably contain from about 5 mg to about 4000 mg of active ingredient, and can be administered one or more times per day. The daily dosage for oral administration will preferably be from 0.1 to 50 mg/kg of total body weight. The daily dosage for administration by injection, including intravenous, intramuscular, subcutaneous and parenteral injections, and use of infusion techniques will preferably be from 0.1 to 10 mg/kg of total body weight. The daily rectal dosage regimen will preferably be from 0.1 to 50 mg/kg of total body weight. The daily vaginal dosage regimen will preferably be from 0.1 to 50 mg/kg of total body weight. The daily topical dosage regimen will preferably be from 0.1 to 10 mg/kg administered between one to four times daily. The transdermal concentration will preferably be that required to maintain a daily dose of from 0.1 to 10 mg/kg. The daily inhalation dosage regimen will preferably be from 0.1 to 10 mg/kg of total body weight. Other dosages and amounts can be selected routinely.
The specific initial and continuing dosage regimen for each patient will vary according to the nature and severity of the condition as determined by the attending diagnostician, the activity of the specific compound employed, the age and general condition of the patient, time of administration, route of administration, rate of excretion of the drug, drug combinations, and the like. The desired mode of treatment and number of doses of a compound of the present invention or a pharmaceutically acceptable salt or ester or composition thereof can be ascertained by those skilled in the art using conventional treatment tests. Combination of the compounds and compositions of the present invention with additional active ingredients Compounds of this invention can be administered as the sole pharmaceutical agent or in combination with one or more other pharmaceutical agents where the combination causes no unacceptable adverse effects. This may be of particular relevance for the treatment of hyper- proliferative diseases such as cancer. In this instance, the compound of this invention can be combined with known cytotoxic agents, signal transduction inhibitors, or with other anti- cancer agents, as well as with admixtures and combinations thereof.
In one embodiment, the compounds of the present invention can be combined with cytotoxic anti-cancer agents. Examples of such agents can be found in the 11th Edition of the Merck Index (1996). These agents include, by no way of limitation, asparaginase, bleomycin, carboplatin, carmustine, chlorambucil, cisplatin, colaspase, cyclophosphamide, cytarabine, dacarbazine, dactinomycin, daunorubicin, doxorubicin (adriamycine), epirubicin, etoposide, 5-fluorouracil, hexamethylmelamine, hydroxyurea, ifosfamide, irinotecan, leucovorin, lomustine, mechlorethamine, 6-mercaptopurine, mesna, methotrexate, mitomycin C, mitoxantrone, prednisolone, prednisone, procarbazine, raloxifen, streptozocin, tamoxifen, thioguanine, topotecan, vinblastine, vincristine, and vindesine.
Other cytotoxic drugs suitable for use with the compounds of the invention include, but are not limited to, those compounds acknowledged to be used in the treatment of neoplastic diseases in Goodman and Oilman's The Pharmacological Basis of Therapeutics (Ninth Edition, 1996, McGraw-Hill). These agents include, by no way of limitation, aminoglutethimide, L-asparaginase, azathioprine, 5-azacytidine cladribine, busulfan, diethylstilbestrol, 2', 2'-difluorodeoxycytidine, docetaxel, erythrohydroxynonyladenine, ethinyl estradiol, 5-fluorodeoxyuridine, 5-fluorodeoxyuridine monophosphate, fludarabine phosphate, fluoxymesterone, flutamide, hydroxyprogesterone caproate, idarubicin, interferon, medroxyprogesterone acetate, megestrol acetate, melphalan, mitotane, paclitaxel, pentostatin, N-phosphonoacetyl-L-aspartate (PALA), plicamycin, semustine, teniposide, testosterone propionate, thiotepa, trimethylmelamine, uridine, and vinorelbine. Other cytotoxic anti-cancer agents suitable for use in combination with the compounds of the invention also include newly discovered cytotoxic principles such as oxaliplatin, gemcitabine, capecitabine, epothilone and its natural or synthetic derivatives, temozolomide (Quinn et al., J. Clin. Oncology 2003, 21(4), 646-651), tositumomab (Bexxar), trabedectin (Vidal et al., Proceedings of the American Society for Clinical Oncology 2004, 23, abstract 3181), and the inhibitors of the kinesin spindle protein Eg5 (Wood et al., Curr. Opin. Pharmacol. 2001, 1, 370-377).
In another embodiment, the compounds of the present invention can be combined with other signal transduction inhibitors. Of particular interest are signal transduction inhibitors which target the EGFR family, such as EGFR, HER-2, and HER-4 (Raymond et al., Drugs 2000, 60 (Suppl.l), 15-23; Harari et al., Oncogene 2000, 19 (53), 6102-6114), and their respective ligands. Examples of such agents include, by no way of limitation, antibody therapies such as Herceptin (trastuzumab), Erbitux (cetuximab), and pertuzumab. Examples of such therapies also include, by no way of limitation, small-molecule kinase inhibitors such as ZD- 1839 / Iressa (Baselga et al., Drugs 2000, 60 (Suppl. 1), 33-40), OSI-774 / Tarceva (Pollack et al. J. Pharm. Exp. Ther. 1999, 291(2), 739-748), CI-1033 (Bridges, Curr. Med. Chem. 1999, 6, 825-843), GW-2016 (Lackey et al., 92nd AACR Meeting, New Orleans, March 24-28, 2001, abstract 4582), CP-724,714 (Jani et al., Proceedings of the American Society for Clinical Oncology 2004, 23, abstract 3122), HKI-272 (Rabindran et al., Cancer Res. 2004, 64, 3958- 3965), and EKB-569 (Greenberger et al., 11th NCI-EORTC-AACR Symposium on New Drugs in Cancer Therapy, Amsterdam, November 7-10, 2000, abstract 388).
In another embodiment, the compounds of the present invention can be combined with other signal transduction inhibitors targeting receptor kinases of the split-kinase domain families (VEGFR, FGFR, PDGFR, flt-3, c-kit, c-fms, and the like), and their respective ligands. These agents include, by no way of limitation, antibodies such as Avastin (bevacizumab). These agents also include, by no way of limitation, small-molecule inhibitors such as STI-571 / Gleevec (Zvelebil, Curr. Opin. Oncol., Endocr. Metab. Invest. Drugs 2000, 2(1), 74-82), PTK-787 (Wood et al., Cancer Res. 2000, 60(8), 2178-2189), SU-11248 (Demetri et al., Proceedings of the American Society for Clinical Oncology 2004, 23, abstract 3001), ZD- 6474 (Hennequin et al., 92nd AACR Meeting, New Orleans, March 24-28, 2001, abstract 3152), AG-13736 (Herbst et al., Clin. Cancer Res. 2003, 9, 16 (suppl 1), abstract C253), KRN-951 (Taguchi et al., 95th AACR Meeting, Orlando, FL, 2004, abstract 2575), CP- 547,632 (Beebe et al., Cancer Res. 2003, 63, 7301-7309), CP-673,451 (Roberts et al., Proceedings of the American Association of Cancer Research 2004, 45, abstract 3989), CHIR-258 (Lee et al., Proceedings of the American Association of Cancer Research 2004, 45, abstract 2130), MLN-518 (Shen et al., Blood 2003, 102, 11, abstract 476), and AZD-2171 (Hennequin et al., Proceedings of the American Association of Cancer Research 2004, 45, abstract 4539).
In another embodiment, the compounds of the present invention can be combined with inhibitors of the Raf/MEK/ERK transduction pathway (Avruch et al., Recent Prog. Horm. Res. 2001, 56, 127-155), or the PKB (akt) pathway (Lawlor et al., J. Cell Sci. 2001, 114, 2903-2910). These include, by no way of limitation, PD-325901 (Sebolt-Leopold et al., Proceedings of the American Association of Cancer Research 2004, 45, abstract 4003), and ARRY-142886 (Wallace et al., Proceedings of the American Association of Cancer Research 2004, 45, abstract 3891).
In another embodiment, the compounds of the present invention can be combined with inhibitors of histone deacetylase. Examples of such agents include, by no way of limitation, suberoylanilide hydroxamic acid (SAHA), LAQ-824 (Ottmann et al., Proceedings of the American Society for Clinical Oncology 2004, 23, abstract 3024), LBH-589 (Beck et al., Proceedings of the American Society for Clinical Oncology 2004, 23, abstract 3025), MS- 275 (Ryan et al., Proceedings of the American Association of Cancer Research 2004, 45, abstract 2452), and FR-901228 (Piekarz et al., Proceedings of the American Society for Clinical Oncology 2004, 23, abstract 3028). In another embodiment, the compounds of the present invention can be combined with other anti-cancer agents such as proteasome inhibitors, and m-TOR inhibitors. These include, by no way of limitation, bortezomib (Mackay et al., Proceedings of the American Society for Clinical Oncology 2004, 23, Abstract 3109), and CCI-779 (Wu et al., Proceedings of the American Association of Cancer Research 2004, 45, abstract 3849).
Generally, the use of cytotoxic and/or cytostatic anti-cancer agent in combination with a compound or composition of the present invention for the treatment of cancer will serve to: (1) yield better efficacy in reducing the growth of a tumor or even eliminate the tumor as compared to administration of either agent alone, (2) provide for the administration of lesser amounts of the administered chemotherapeutic agents,
(3) provide for a chemotherapeutic treatment that is well tolerated in the patient with fewer deleterious pharmacological complications than observed with single agent chemotherapies and certain other combined therapies,
(4) provide for treating a broader spectrum of different cancer types in mammals, especially humans,
(5) provide for a higher response rate among treated patients,
(6) provide for a longer survival time among treated patients compared to standard chemotherapy treatments,
(7) provide a longer time for tumor progression, and/or
(8) yield efficacy and tolerability results at least as good as those of the agents used alone, compared to known instances where other cancer agent combinations produce antagonistic effects.
Aspects of the instant invention include, but are not limited to:
In one embodiment, the present invention provides for the following aspects
Aspect 1. A synthetic metabolite of Formula (I) or a salt, or a prodrug or an isolated stereoisomer thereof
Figure imgf000047_0001
Aspect 2. A synthetic M2, M3, M4, or M5 metabolite of a compound of Formula I or a salt of said metabolite, wherein the structures of said M2, M3, M4, and M5 metabolites are:
Figure imgf000048_0001
Figure imgf000049_0001
Aspect 3. A pharmaceutically acceptable salt of synthetic metabolite according to Aspect 2 which is
a) a basic salt of an organic acid or inorganic acid which is hydrochloric acid, hydrobromic acid, sulfuric acid, phosphoric acid, methanesulfonic acid, trifluoromethanesulfonic acid, benzenesulfonic acid, p-toluene sulfonic acid (tosylate salt), 1-napthalene sulfonic acid, 2-napthalene sulfonic acid, acetic acid, trifluoroacetic acid, malic acid, tartaric acid, citric acid, lactic acid, oxalic acid, succinic acid, fumaric acid, maleic acid, benzoic acid, salicylic acid, phenylacetic acid, or mandelic acid; or
b) an acid salt of an organic or inorganic base containing an alkali metal cation, an alkaline earth metal cation, an ammonium cation, an aliphatic substituted ammonium cation or an aromatic substituted ammonium cation. Aspect 4. The synthetic metabolite according to Aspect 2 which is a metabolite of 4{4- [3-(4-chloro-3-trifluoromethylphenyl)-ureido]-3-fluorophenoxy}-pyridine-2-carboxylic acid methylamide.
Aspect 5. A pharmaceutically acceptable salt of a synthetic metabolite according to Aspect 4 which is a basic salt of hydrochloric acid, hydrobromic acid, sulfuric acid, phosphoric acid, methanesulfonic acid, trifluoromethanesulfonic acid, benzenesulfonic acid, p-toluene sulfonic acid (tosylate salt), 1-napthalene sulfonic acid, 2-napthalene sulfonic acid, acetic acid, trifluoroacetic acid, malic acid, tartaric acid, citric acid, lactic acid, oxalic acid, succinic acid, fumaric acid, maleic acid, benzoic acid, salicylic acid, phenylacetic acid, or mandelic acid. Aspect 6. A compound which is which is a hydrochloride, benzenesulfonate, or methanesulfonate salt of the synthetic metabolite according to Aspect 2.
Aspect 7. A pharmaceutical composition comprising a synthetic metabolite according to Aspect 2 and a physiologically acceptable carrier.
Aspect 8. A pharmaceutical composition comprising a synthetic metabolites according to Aspect 4 and a physiologically acceptable carrier. Aspect 9. A pharmaceutical composition for the treatment of a disease in a human or other mammal regulated by a protein kinase, associated with an aberration in the protein kinase signal transduction pathway comprising a synthetic metabolite according to Aspect 2 and a physiologically acceptable carrier. Aspect 10. A pharmaceutical composition for the treatment of a hyper-proliferative disorder comprising a synthetic metabolite according to Aspect 4 and a physiologically acceptable carrier.
Aspect 11. A pharmaceutical composition for the treatment of a cancerous cell growth comprising a synthetic metabolite according to Aspect 2 and a physiologically acceptable carrier.
Aspect 12. A method for regulating tyrosine kinase signal transduction comprising administering to a human or other mammal a synthetic metabolite according to Aspect 2.
Aspect 13. A method for treating or preventing a disease in a human or other mammal which is regulated by tyrosine kinase and associated with an aberration in the tyrosine kinase signal transduction pathway, said method comprising administering to a human or other mammal a synthetic metabolite according to Aspect 2.
Aspect 14. A method for treating or preventing a disease in a human and/or other mammal which is a VEGFR-2 mediated disorder, said method comprising administering to a human or other mammal a synthetic metabolite according to Aspect 2. Aspect 15. A method for treating or preventing a disease in a human and/or other mammal which is a PDGFR mediated disorder, said method comprising administering to a human or other mammal a synthetic metabolite according to Aspect 2. Aspect 16. A method for treating or preventing a disease in a human or other mammal which is a raf-mediated disorder, said method comprising administering to a human or other mammal a synthetic metabolite according to Aspect 2.
Aspect 17. A method for treating or preventing a disease in a human or other mammal which is a p38-mediated disorder, said method comprising administering to a human or other mammal a synthetic metabolite according to Aspect 2.
Aspect 18. A method for treating or preventing a disease in a human or other mammal which is a VEGF-mediated disorder, said method comprising administering to a human or other mammal a synthetic metabolite according to Aspect 2.
Aspect 19. A method for treating or preventing a disease in a human or other mammal which is a hyper-proliferative, inflammatory and/or angiogenesis disorder which comprises administering to a human or other mammal a synthetic metabolite according to Aspect 2.
Aspect 20. A method for treating or preventing a disease in a human or other mammal which is a hyper-proliferative disorder which comprises administering to a human or other mammal a synthetic metabolite according to Aspect 2. Aspect 21. A method as in aspect 20, wherein the hyper-proliferative disorder is cancer.
Aspect 22. A method as in aspect 21, wherein said method comprises administering to a human or other mammal a synthetic metabolite according to Aspect 2 in combination with one or several additional anti-cancer agents.
Aspect 23. A method for treating or preventing a disease in a human or other mammal characterized by abnormal angiogenesis or hyperpermiability processes comprising administering to a human or other mammal a synthetic metabolite according to Aspect 2. Aspect 24. A method as in aspect 23, for treating or preventing a disease in a human or other mammal characterized by abnormal angiogenesis or hyperpermeability processes, comprising administering to a human or other mammal, a synthetic metabolite according to Aspect 2 simultaneously with another anti-angiogenesis agent, either in the same formulation or in separate formulations.
Aspect 25. A method for treating or preventing one or more of the following conditions in humans and/or other mammals:
tumor growth, retinopathy, ischemic retinal-vein occlusion, retinopathy of prematurity, age related macular degeneration; rheumatoid arthritis, psoriasis, a bullous disorder associated with subepidermal blister formation, including bullous pemphigoid, erythema multiforme, or dermatitis herpetiformis, rheumatoid arthritis, osteoarthritis, septic arthritis, tumor metastasis, periodontal disease, cornal ulceration, proteinuria and coronary thrombosis from atherosclerotic plaque, aneurismal aortic, birth control, dystrophobic epidermolysis bullosa, degenerative cartilage loss following traumatic joint injury, osteopenias mediated by MMP activity, tempero mandibular joint disease or demyelating disease of the nervous system,
said method comprising administering to a human or other mammal, a synthetic metabolite according to Aspect 2.
Aspect 26. A method for treating or preventing one or more of the following conditions in humans and/or other mammals: tumor growth, retinopathy, ischemic retinal-vein occlusion, retinopathy of prematurity, age related macular degeneration; rheumatoid arthritis, psoriasis, a bullous disorder associated with subepidermal blister formation, including bullous pemphigoid, erythema multiforme, or dermatitis herpetiformis;
in combination with another condition selected from the group consisting of:
rheumatic fever, bone resorption, postmenopausal osteoporosis, sepsis, gram negative sepsis, septic shock, endotoxic shock, toxic shock syndrome, systemic inflammatory response syndrome, inflammatory bowel disease (Krohn's disease and ulcerative colitis), Jarisch- Herxheimer reaction, asthma, adult respiratory distress syndrome, acute pulmonary fibrotic disease, pulmonary sarcoidosis, allergic respiratory disease, silicosis, coal worker's pneumoconiosis, alveolar injury, hepatic failure, liver disease during acute inflammation, severe alcoholic hepatitis, malaria (Plasmodium falciparum malaria and cerebral malaria), non- insulin-dependent diabetes mellitus (NIDDM), congestive heart failure, damage following heart disease, atherosclerosis, Alzheimer's disease, acute encephalitis, brain injury, multiple sclerosis (demy elation and oligiodendrocyte loss in multiple sclerosis), advanced cancer, lymphoid malignancy, pancreatitis, impaired wound healing in infection, inflammation and cancer, myelodysplastic syndromes, systemic lupus erythematosus, biliary cirrhosis, bowel necrosis, radiation injury/ toxicity following administration of monoclonal antibodies, host-versus-graft reaction (ischemia reperfusion injury and allograft rejections of kidney, liver, heart, and skin), lung allograft rejection (obliterative bronchitis) and complications due to total hip replacement,
said method comprising administering to a human or other mammal a synthetic metabolite according to Aspect 2.
Aspect 27. A method for treating or preventing one or more of the following conditions in humans and/or other mammals: tumor growth, retinopathy, diabetic retinopathy, ischemic retinal-vein occlusion, retinopathy of prematurity, age related macular degeneration; rheumatoid arthritis, psoriasis, bullous disorder associated with subepidermal blister formation, bullous pemphigoid, erythema multiforme, and dermatitis herpetiformis, in combination with an infectious disease selected from the group consisting of :
tuberculosis, Helicobacter pylori infection during peptic ulcer disease, Chaga's disease resulting from Trypanosoma cruzi infection, effects of Shiga-like toxin resulting from E. coli infection, effects of enterotoxin A resulting from Staphylococcus infection, meningococcal infection, and infections from Borrelia burgdorferi, Treponema pallidum, cytomegalovirus, influenza virus, Theiler's encephalomyelitis virus, and the human immunodeficiency virus (HIV);
said method comprising administering to a human or other mammal a synthetic metabolite according to Aspect 2.
Aspect 28. A method as in aspect 22 wherein the additional anti-cancer agent is selected from the group consisting of asparaginase, bleomycin, carboplatin, carmustine, chlorambucil, cisplatin, colaspase, cyclophosphamide, cytarabine, dacarbazine, dactinomycin, daunorubicin, doxorubicin (adriamycine), epirubicin, etoposide, 5-fluorouracil, hexamethylmelamine, hydroxyurea, ifosfamide, irinotecan, leucovorin, lomustine, mechlorethamine, 6-mercaptopurine, mesna, methotrexate, mitomycin C, mitoxantrone, prednisolone, prednisone, procarbazine, raloxifen, streptozocin, tamoxifen, thioguanine, topotecan, vinblastine, vincristine, vindesine, aminoglutethimide, L-asparaginase, azathioprine, 5-azacytidine cladribine, busulfan, diethylstilbestrol, 2',2'- difluorodeoxycytidine, docetaxel, erythrohydroxynonyl adenine, ethinyl estradiol, 5- fluorodeoxyuridine, 5-fluorodeoxyuridine monophosphate, fludarabine phosphate, fluoxymesterone, flutamide, hydroxyprogesterone caproate, idarubicin, interferon, medroxyprogesterone acetate, megestrol acetate, melphalan, mitotane, paclitaxel, pentostatin, N-phosphonoacetyl-L-aspartate (PALA), plicamycin, semustine, teniposide, testosterone propionate, thiotepa, trimethylmelamine, uridine, and vinorelbine, oxaliplatin, gemcitabine, capecitabine, epothilone and its natural or synthetic derivatives, tositumomab, trabedectin, and temozolomide. trastuzumab, cetuximab, bevacizumab, pertuzumab, ZD- 1839 (Iressa), OSI-774 (Tarceva), CI-1033, GW-2016, CP-724,714, HKI-272, EKB-569, STI-571 (Gleevec), PTK-787, SU-11248, ZD-6474, AG-13736, KRN-951, CP-547,632, CP-673,451, CHIR-258, MLN-518, AZD-2171, PD-325901, ARRY-142886, suberoylanilide hydroxamic acid (SAHA), LAQ-824, LBH-589, MS-275, FR-901228, bortezomib, and CCI-779. Aspect 29. A method as in aspect 22 wherein the additional anti-cancer agent is a cytotoxic agent selected from the group consisting of DNA topoisomerase I and II inhibitors, DNA intercalators, alkylating agents, anti-metabolites, cell-cycle blockers, microtubule disruptors, and Eg5 inhibitors. Aspect 30. A method as in aspect 22 wherein the additional anti-cancer agent is selected from the group consisting of inhibitors of growth factor receptor signaling, histone deacetylase inhibitors, inhibitors of the PKB pathway, inhibitors of the Raf/MEK/ERK pathway, inhibitors of the mTOR pathway, and proteasome inhibitors. Aspect 31. A method for treating or preventing one or more of the following conditions in humans and/or other mammals:
rheumatic fever, bone resorption, postmenopausal osteoporosis, sepsis, gram negative sepsis, septic shock, endotoxic shock, toxic shock syndrome, systemic inflammatory response syndrome, inflammatory bowel disease (Krohn's disease and ulcerative colitis), Jarisch- Herxheimer reaction, asthma, adult respiratory distress syndrome, acute pulmonary fibrotic disease, pulmonary sarcoidosis, allergic respiratory disease, silicosis, coal worker's pneumoconiosis, alveolar injury, hepatic failure, liver disease during acute inflammation, severe alcoholic hepatitis, malaria (Plasmodium falciparum malaria and cerebral malaria), non- insulin-dependent diabetes mellitus (NIDDM), congestive heart failure, damage following heart disease, atherosclerosis, Alzheimer's disease, acute encephalitis, brain injury, multiple sclerosis (demy elation and oligiodendrocyte loss in multiple sclerosis), advanced cancer, lymphoid malignancy, pancreatitis, impaired wound healing in infection, inflammation and cancer, myelodysplastic syndromes, systemic lupus erythematosus, biliary cirrhosis, bowel necrosis, psoriasis, radiation injury/ toxicity following administration of monoclonal antibodies, host-versus-graft reaction (ischemia reperfusion injury and allograft rejections of kidney, liver, heart, and skin), lung allograft rejection (obliterative bronchitis) or complications due to total hip replacement,
said method comprising administering to a human or other mammal, a synthetic metabolite according to Aspect 2.
Aspect 32. A method for treating or preventing one or more of the following conditions in humans and/or other mammals:
tuberculosis, Helicobacter pylori infection during peptic ulcer disease, Chaga's disease resulting from Trypanosoma cruzi infection, effects of Shiga-like toxin resulting from
E. coli infection, effects of enterotoxin A resulting from Staphylococcus infection, meningococcal infection, and infections from Borrelia burgdorferi, Treponema pallidum, cytomegalovirus, influenza virus, Theiler's encephalomyelitis virus, and the human immunodeficiency virus (HIV) ,
said method comprising administering to a human or other mammal, a synthetic metabolite according to Aspect 2.
Aspect 33. A method for treating or preventing osteoporosis, inflammation, and angiogenesis disorders, with the exclusion of cancer, in a human and/or other mammal by administering an effective amount of a synthetic metabolite according to Aspect 2 to said mammal.
Aspect 34. A method for treating or preventing cancer in a human or other mammal which comprises administering to a human or other mammal a single active principle combining inhibition of tumor cell proliferation mediated by the raf / MEK / ERK pathway, and inhibition of angiogenesis mediated by PDGF and VEGF, wherein said active principle comprises a synthetic metabolite according to Aspect 2. Aspect 35. A method of aspect 34 where said inhibition of tumor cell proliferation is caused by inhibition of raf kinase, and said inhibition of angiogenesis is caused by dual inhibition of PDGFR-beta and VEGFR-2 kinases. Aspect 36. A method for treating or preventing cancer in a human or other mammal which comprises administering to a human or other mammal a single active principle combining inhibition of tumor cell proliferation mediated by the raf pathway, and inhibition of angiogenesis mediated by PDGF or VEGF, wherein said active principle comprises a synthetic metabolite according to Aspect 2.
Aspect 37. A method of treating and/or preventing a disease and/or condition in a subject in need thereof, comprising administering an effective amount of a synthetic metabolite according to Aspect 2. Aspect 38. A method of aspect 37, wherein said method comprises causing tumor regression in a subject or cells therefrom.
Aspect 39. A method of aspect 37, wherein said method comprises inhibiting lymphangiogenesis .
Aspect 40. A method of aspect 37, wherein said method comprises inhibiting angiogenesis.
Aspect 41. A method of aspect 37, wherein said method comprises inhibiting lymphangiogenesis and angiogenesis.
Aspect 42. A method of aspect 37, wherein said method comprises stimulating the proliferation of hematopoietic progenitor cells.
Aspect 43. A method of aspect 37, wherein said method comprises treating a disorder in a mammalian subject mediated by raf, VEGFR-2, VEGFR-3, PDGFR-beta, p38 and/or fit- 3.
Aspect 44. A method of aspect 37, wherein said method comprises determining whether a condition can be modulated by said compound, comprising measuring the expression or activity of raf, VEGFR-2, VEGFR-3, PDGFR-beta, p38 and/or flt-3, in a sample comprising cells or a cell extract, wherein said ample is obtained from a subject or cell having said condition, whereby said condition can be modulated by said compound when said expression or activity is different in said condition as compared to a normal control.
Aspect 45. A method of aspect 44, further comprising comparing the expression in said sample to said normal control.
Aspect 46. A method of aspect 37, wherein said method comprises assessing the efficacy of said compound disorder, comprising administering said compound, measuring the expression or activity of raf, VEGFR-2, VEGFR-3, PDGFR-beta, p38, and/or flt-3, and determining the effect of said compound on said expression or activity.
Aspect 47. A method of aspect 37, wherein said method comprises determining the presence of raf, VEGFR-2, VEGFR-3, PDGFR-beta, p38 and/or flt-3 in a sample of a biological material, contacting said sample with said compound, and determining whether said compound binds to said material.
Aspect 48. A method of aspect 37, wherein said method comprises treating a tumor in a subject in need thereof, comprising administering an effective amount of said compound wherein said amount is effective to inhibit tumor cell proliferation and neovascularization.
Aspect 49. The synthetic metabolite according to aspect 2, which is an M-2 or an M-5 metabolite having the following structural formula:
Figure imgf000057_0001
Figure imgf000058_0001
or a salt, or a prodrug or an isolated stereoisomer thereof.
Aspect 50. A method for treating or preventing a tumor or vascular disease in a subject, comprising administering to said subject, a synthetic metabolite according to aspect 49.
Claim 51. A method of making the synthetic metabolite according to aspect 2, comprising contacting the compound of formula I or a salt thereof with a liver microsome preparation under conditions sufficient for the metabolism of said compound of Formula I and isolating the metabolites from said preparation.
BRIEF DESCRIPTION OF THE DRAWINGS
Various features and attendant advantages of the present invention will be more fully appreciated as the same becomes better understood when considered in conjunction with the accompanying drawings, in which like reference characters designate the same or similar parts throughout the several views, and wherein:
Fig. 1 shows metabolites of regorafenib from in vitro and in vivo studies. Fig. 2 shows kinase selectivity profile of regorafenib and its N-oxide (M-2) and desmethyl-N- oxide (M-5) synthetic metabolites.
Fig. 3 shows that M-2 and M-5 synthetic metabolites significantly inhibit tumor growth of human xenografts in mice. Fig. 4 shows that regorafenib and its synthetic metabolites M-2 and M-5 inhibit the acute hypotensive effect of VEGF in anesthetized rats.
Fig. 5 shows that the M-2 synthetic metabolite of regorafenib affects vasculature extravasation in a rat GS9L glioblastoma model after a single oral dose of 7.5 mg/kg. Assays were performed using kinetic DCE-MRI analysis.
Fig. 6 shows the plasma concentrations of regorafenib, M-2 and M-5 synthetic metabolite on day 1 and day 21 following administration of 160 mg regorafenib co-precipitate tablet to patients with colo-rectal cancer (n = 9, preliminary data). Data are derived from the same study as shown in Table 4.
EXAMPLES
The present invention is further illustrated with the following non-limiting examples.
Abbreviations used in this specification are as follows:
HPLC high pressure liquid chromatography
MS mass spectrometry
ES electrospray
DMSO dimethylsulfoxide
MP melting point
NMR nuclear resonance spectroscopy
TLC thin layer chromatography
rt room temperature
Preparation of 4-amino-3-fluorophenol
Figure imgf000059_0001
To a dry flask purged with Argon was added 10% Pd/C (80 mg) followed by 3-fluoro-4- nitrophenol (1.2 g, 7.64 mmol) as a solution in ethyl acetate (40 mL). The mixture was stirred under an H2 atmosphere for 4 h. The mixture was filtered through a pad of Celite and the solvent was evaporated under reduced pressure to afford the desired product as a tan solid (940 mg, 7.39 mmol; 97 % yield); 1H-NMR (DMSO-d6) 4.38 (s, 2H), 6.29-6.35 (m, 1H), 6.41 (dd, J=2.5, 12.7, 1H), 6.52-6.62 (m, 1H), 8.76 (s, 1H).
Preparation of 4-(4-amino-3-fluorophenoxy)pyridine-2-carboxylic acid methylamide
Figure imgf000060_0001
A solution of 4-amino-3-fluorophenol (500 mg, 3.9 mmol) in N,N-dimethylacetamide (6 mL) cooled to 0 °C was treated with potassium tert-butoxide (441 mg, 3.9 mmol), and the brown solution was allowed to stir at 0 °C for 25 min. To the mixture was added 4-chloro-N- methyl-2-pyridinecarboxamide (516 mg, 3.0 mmol) as a solution in dimethylacetamide (4 mL). The reaction was heated at 100 °C for 16 h. The mixture was cooled to room temperature, quenched with H20 (20 mL), and extracted with ehtylacetate (4 x 40 mL). The combined organics were washed with H20 (2 x 30 mL), dried (MgS04), and evaporated to afford a red-brown oil. 1H-NMR indicated the presence of residual dimethylacetamide, thus the oil was taken up in diethylether (50 mL) and was further washed with brine (5 x 30 mL). The organic layer was dried (MgS04) and concentrated to give 950 mg of the desired product as a red-brown solid, which was used in the next step without purification.
A method of preparing 4-chloro-N-methyl-2-pyridinecarboxamide is described in Bankston et al., Org. Proc. Res. Dev. 2002, 6(6), 777-781. Example 1: Preparation of 4{4-[3-(4-chloro-3-trifluoromethylphenyl)-ureido]-3- fluorophenoxy}-pyridine-2-carboxylic acid methylamide
Figure imgf000060_0002
To a solution of 4-(4-amino-3-fluorophenoxy)pyridine-2-carboxylic acid methylamide (177 mg, 0.68 mmol) in toluene (3 mL) was added 4-chloro-3-(trifluoromethyl)phenyl isocyanate (150 mg, 0.68 mmol). The mixture was stirred at rt for 72 h. The reaction was concentrated under reduced pressure and the residue was triturated with diethylether. The resulting solid was collected by filtration and dried in vacuo for 4 h to afford the title compound (155 mg, 0.32 mmol; 47% yield); 1H-NMR (DMSO-d6) 2.78 (d, J=4.9, 3H), 7.03-7.08 (m, 1H), 7.16 (dd, J=2.6, 5.6, 1H), 7.32 (dd, J=2.7, 11.6, 1H), 7.39 (d, J=2.5, 1H), 7.60 (s, 2H), 8.07-8.18 (m, 2H), 8.50 (d, J=5.7, 1H), 8.72 (s, 1H), 8.74-8.80 (m, 1H), 9.50 (s, 1H); MS (HPLC/ES) 483.06 m/z = (M + 1).
Example 2: Preparation of 4{4-[3-(4-chloro-3-trifluoromethylphenyl)-ureido]-3- fluorophenoxy}-pyridine-2-carboxylic acid methylamide hydrochloride
The compound of example 1 as a free base (2.0 g) was dissolved in anhydrous tetrahydrofuran (15 mL) and a 4M HCl/dioxane was added (excess). The solution was then concentrated in vacuo to afford 2.32 grams of off-white solids. The crude salt was dissolved in hot ethanol (125 mL), activated carbon was added and the mixture heated at reflux for 15 minutes. The hot suspension was filtered through a pad of Celite 521 and allowed to cool to room temperature. The flask was placed in a freezer overnight. The crystalline solids were collected by suction filtration, washed with ethanol, then hexane and air-dried. The mother liquors were concentrated down and crystallization (in freezer) allowed taking place overnight. A second crop of solids was collected and combined with the first crop. The colorless salt was dried in a vacuum oven at 60 °C over two days. Yield of hydrochloride salt obtained 1.72 g (79%).
Melting point: 215 °C
Elemental analysis:
Calcd. Found
c 48.57 48.68
H 3.11 2.76
N 10.79 10.60
CI 13.65 13.63
F 14.63 14.88 Example 3: Preparation of 4{4-[3-(4-chloro-3-trifluoromethylphenyl)-ureido]-3- fluorophenoxy}-pyridine-2-carboxylic acid methylamide mesylate
The compound of example 1 as a free base (2.25 g) was dissolved in ethanol (100 mL) and a stock solution of methanesulfonic acid (excess) was added. The solution was then concentrated in vacuo to afford a yellow oil. Ethanol was added and concentration repeated, affording 2.41 g of off-white solids. The crude salt was dissolved in hot ethanol (-125 mL) and then cooled slowly to crystallize. After reaching room temperature, the flask was placed in a freezer overnight. The colorless crystalline material was collected by suction filtration; the filter cake was washed with ethanol, then hexane and air-dried, to afford 2.05 g of material, which was dried in a vacuum oven at 60 °C overnight.
Melting point: 231 °C
Elemental analysis:
Calcd. Found
c 45.64 45.34
H 3.31 3.08
N 9.68 9.44
CI 6.12 6.08
F 13.13 13.42
S 5.54 5.59
Example 4: Preparation of 4{4-[3-(4-chloro-3-trifluoromethylphenyl)-ureido]-3- fluorophenoxy}-pyridine-2-carboxylic acid methylamide phenylsulfonate
The compound of example 1 as a free base (2.25 g) was suspended in ethanol (50 mL) and benzensulfonic acid (0.737 g) in ethanol (50 mL) was added. The mixture was heated with vigorous stirring. All solid material dissolved to give a reddish solution. The solution was allowed to cool to room temperature and the flask scratched. Crystal formation was difficult to achieve, some seeds were found, added to solution and placed in freezer overnight. Grayish-tan solids had formed in the flask; the material was broken up & collected by suction filtration. The solids were washed with ethanol, then hexane and air-dried. Weighed product: 2.05 g, 69% yield.
Melting point: 213 °C
Elemental Analysis: Calcd. Found
c 50.59 50.24
H 3.30 3.50
N 8.74 8.54
F 11.86 11.79
CI 5.53 5.63
s 5.00 5.16
Example 5: Preparation of synthetic metabolites from compounds of Formula I
As a representative example, the synthetic metabolites of the compounds of Formula I can be obtained by incubating the parent regorafenib compound with liver microsomes.
The metabolites of the present application can also be synthetically obtained. The metabolites of the present application can be purified using techniques that are known in the art.
Example 6: Metabolite profiles in animals
N-oxide (M-2) and hydroxymethyl (M-3) metabolites were identified as metabolites of regorafenib in vitro upon incubation with liver enzymes of various mammalian species (man, dog, rat, mouse). Studies reveal that and its metabolites show high protein binding in man and animal species.
The metabolite profiles are presented in Fig. 1 and Table 1 , below.
Table 1. Metabolite profiles in incubations of [14C] regorafenib (20 μΜ) with liver microsomes of different species (protein concentration 0.5 mg/mL, 60 min).
Figure imgf000063_0001
Example 7: Pharmacological profiling
After oral administration of 10 mg/kg regorafenib to mice for 5 days, the N-oxide (M-2) exposure accounted for ~16 of the total AUC (R+M-2+M-5), whereas the contribution of M-5 was -2% relative to total AUC. Data are shown in Table 2, below. Following oral administration of 10 mg/kg M-2 to mice, regorafenib exposure reached -17% of total exposure, indicating reduction of the N-oxide to be a relevant metabolic pathway in vivo, whereas M-5 accounted for 5% of total exposure. Table 2. Pharmacokinetic parameters of regorafenib and its metabolites M-2 and M-5 at steady state after their oral administration to female NMRI-Foxn-1 mice.
Figure imgf000064_0001
Example 8. Kinome-wide selectivity profile
A kinome-wide selectivity profiling of regorafenib and its metabolites M-2 and M-5 was performed by Ambit Biosciences (San Diego, CA, US) using an active-site competitive binding assay (Fabian et al. Nat Biotechnol 2005; 23: 329-336). A total of 402 kinases were analyzed using a single dose of 1 μΜ of compound. Binding inhibition activities are displayed as the percentage of the kinase that remained bound compared with the DMSO- treated control. The potency of the compound is reflected by the size of the circle.
In the competitive binding assay, M-2 and M-5 showed kinase selectivity profiles similar to but distinct from regorafenib. Results are shown in Fig. 2. Example 9. Biochemical characterization of M-2 and M-5 metabolites
Characterization of both M-2 and M-5 demonstrated potent pharmacologic activities. In biochemical kinase assays, M-2 and M-5 showed an inhibition profile similar to but distinct from regorafenib. In cellular assays, M-2 and M-5 inhibited key targets such as vascular endothelial growth factor (VEGF) receptor 2, TIE-2, and mutant and wild-type c-KIT and B- RAF, with IC50 values very similar to regorafenibfor M-2 and somewhat higher for M-5. Data are presented in Table 3.
Table 3. Pharmacologic activity of regorafenib, M-2 and M-5in cellular kinase phosphorylation assays.
Figure imgf000065_0001
Example 10. Tumor growth inhibitory effects of M-2 and M-5 metabolites
Human and rat tumor models were to determine the in vivo effects of M-2 and M-5 synthetic metabolites on tumor growth and on tumor vasculature. In these studies, mice bearing xenografts of the human breast cancer cell lineMDA-MB-231 (K-RASG13D, B-RAFG464V) or the human colorectal cancer cell line HT-29 (B- RAFV600E) were treated orally (qd x 27) with 10 mg/kg of regorafenib, M-2 or M-5, starting at day 11 after tumor inoculation. Tumor growth inhibition is given as relative tumor area (p < 0.05 vs vehicle in all instances; n = 8 mice/group). The tumor growth inhibitory assays were performed using DCE-MRI.
As shown in Fig. 3, both synthetic metabolites dosed orally exhibited potent dose-dependent tumor growth inhibition (TGI) in preclinical murine HT-29 colorectal and MDA-MB-231 breast cancer xenografts, achieving significant TGI of 62/58% and 54/50%, respectively, compared with vehicle controls at 10 mg/kg (Fig. 3).
Example 11. Inhibition of VEGF-induced hypotension
The acute effect of the synthetic metabolites on vascular endothelial growth factor (VEGF)- induced hypotension was studied using a rat pharmacodynamic model.
In these studies, a bolus i.v. of VEGF (9 μg/kg) was initially used to induce a transient hypotension in rats (see inset of Fig. 4). The antagonistic effects of regorafenib and its synthetic metabolites on VEGF-induced hypotension were then studied. To this end, following a 10 minute i.v. pretreatment with vehicle, regorafenib, M-2 or M-5 were administered at the indicated doses (Fig. 4). Reduction of diastolic (blue bars) and systolic (orange bars) blood pressure (difference between pre-injection and minimum value) were calculated. Results are presented in Fig. 4. Example 12. Effect on tumor vasculature
The pharmacodynamic effect of regorafenib and M-2 on the tumor vasculature in vivo was analyzed in GS9L rat glioblastoma tumor-bearing Fischerrats by DCE-MRI analysis using the macromolecular contrast agent Gadomer-17. In these studies, rats carrying glioblastoma tumors in their thighs were treatedat a single oral dose of 7.5 mg/kg of M-2 or regorafenib and inhibition of the extravasation of Gadomer-17 was analyzed 2, 6 and 24 hours aftertreatment.
Results are presented in Fig. 5. The left panel shows results for regorafenib and the right panel shows results for the M-2 synthetic metabolite. The left y-axis indicates tumor AUC values, which were normalized to thigh muscle. The right y-axis shows serum levels of regorafenib, M-2, M-4 and M-5. The contribution of regorafenib, M-2, M-4 and M-5 could therefore be independently assessed. Example 13. In vivo pharmacological studies
In colon cancer patients treated daily with 160 mg coprecipitate tablets for a period of 21 days, M-2 and M-5showed systemic exposures very similar to regorafenib. Results are presented in Table 4 and Figure 6(B). As shown in Table 4, bio-transformation of regorafenib in patientswith colorectal cancer results in significant elevation of demethylated and oxidized M-2 and M-5 synthetic metabolite levels. The synthetic metabolites were observed after single or multiple qd dosing with 160 mg co-precipitate tablets.
Continuous qd dosing in patients with colorectal cancer for 19 days resulted in a 25- and 2- fold increase in the AUC of M-5 and M-2 synthetic metabolites, respectively. Cmax values were similarly increased 42-fold and 5-fold, respectively. Values are reflected in comparison to the first dose.
In order to further characterize the metabolites, plasma concentrations of regorafenib, M-2 and M-5 were observed on a daily basis on day 1 and day 21 following administration of 160 mgregorafenib co-precipitate tablet to patients with colorectal cancer (n = 9, preliminary data). Results are shown in Fig. 6. Multiple plasma peaks of regorafenib, M-2 and M-5 were observed, most likelyas a result of enterohepatic cycling. Combined Cmax level of the parent compound and M2/M5 metabolites was 11 mg/L on day 21. The combined pharmacologically relevant plasma concentration was evaluated to -2.5 mg/L, remaining 3 days after the last dosing.
Table 4. Pharmacokinetic parameters of regorafenib, M-2 and M-5 on day 1 and day 21, following administration of 160 mg regorafenib co-precipitate tablet to patients with colorectal cancer (geometric mean ( CV), preliminary data)
Figure imgf000068_0001
Example 14. c-raf (raf-1) Biochemical Assay
The c-raf biochemical assay is performed with a c-raf enzyme that was activated (phosphorylated) by Lck kinase. Lck-activated c-raf (Lck/c-raf) is first produced in Sf9 insect cells by co-infecting cells with baculoviruses expressing, under the control of the polyhedrin promoter, GST-c-raf (from amino acid 302 to amino acid 648) and Lck (full-length). Both baculoviruses are used at the multiplicity of infection of 2.5 and the cells are harvested 48 h post infection.
MEK-1 protein is produced in Sf9 insect cells by infecting cells with the baculovirus expressing GST-MEK-1 (full-length) fusion protein at the multiplicity of infection of 5 and harvesting the cells 48 hours post infection. Similar purification procedure is used for GST- c-raf 302-648 and GST-MEK-1.
Transfected cells are suspended at 100 mg of wet cell biomass per mL in a buffer containing 10 mM sodium phosphate, 140 mM sodium chloride pH 7.3, 0.5% Triton X-100 and the protease inhibitor cocktail. The cells are disrupted with Polytron homogenizer and centrifuged 30,000g for 30 minutes. The 30,000g supernatant is applied onto GSH- Sepharose. The resin is washed with a buffer containing 50 mM Tris, pH 8.0, 150 mM NaCl and 0.01% Triton X-100. The GST-tagged proteins are eluted with a solution containing 100 mM Glutathione, 50 mM Tris, pH 8.0, 150 mM NaCl and 0.01% Triton X-100. The purified proteins are dialyzed into a buffer containing 20 mM Tris, pH 7.5, 150 mM NaCl and 20% Glycerol.
Test compounds are serially diluted in DMSO using three-fold dilutions to stock concentrations ranging typically from 50 μΜ to 20 nM (final concentrations in the assay range from 1 μΜ to 0.4 nM). The c-Raf biochemical assay is performed as a radioactive filtermat assay in 96-well Costar polypropylene plates (Costar 3365). The plates are loaded with 75 μL solution containing 50 mM HEPES pH 7.5, 70 mM NaCl, 80 ng of Lck/c-raf and 1 μg MEK-1. Subsequently, 2 μΕ of the serially diluted individual compounds are added to the reaction, prior to the addition of ATP. The reaction is initiated with 25 μΕ ATP solution containing 5μΜ ATP and 0.3 μCi [33P]-ATP. The plates are sealed and incubated at 32 oC for 1 h. The reaction is quenched with the addition of 50 μΕ of 4 % Phosphoric Acid and harvested onto P30 filtermats (PerkinElmer) using a Wallac Tomtec Harvester. Filtermats are washed with 1 % Phosphoric Acid first and deinonized H2O second. The filters are dried in a microwave, soaked in scintillation fluid and read in a Wallac 1205 Betaplate Counter (Wallac Inc., Atlanta, GA, U.S.A.). The results are expressed as percent inhibition.
% Inhibition = [100-(Tib/Ti)] x 100 where
Tib = (counts per minute with inhibitor)-(background)
Ti = (counts per minute without inhibitor)-(background) The compound of the present invention shows potent inhibition of raf kinase in this assay.
Example 15: p38 kinase in vitro assay
Purified and His-tagged p38 (expressed in E. Coli) is activated in vitro by MMK-6 to a high specific activity. Using a microtiter format, all reactions are conducted in 100 μΕ volumes with reagents diluted to yield 0.05 μg/well of activated p38 and 10 μg/well of myelin basic protein in assay buffer (25 mM HEPES 7.4, 20 mM MgC12, 150 mM NaCl). Test compounds (5 μΕ of a 10% DMSO solution in water) are prepared and diluted into the assay to cover a final concentration range from 5 nM to 2.5 μΜ. The kinase assay is initiated by addition of 25 μΕ of an ATP cocktail to give a final concentration of 10 μΜ cold ATP and 0.2 μα [gamma-33P] ATP per well (200-400 dpm/pmol of ATP). The plate is incubated at 32 oC for 35 min., and the reaction quenched with 7 of a 1 N aq HCl solution. The samples are harvested onto a P30 Filtermat (Wallac, Inc.) using a TomTec 1295 Harvester (Wallac, Inc.), and counted in a LKB 1205 Betaplate Liquid Scintillation Counter (Wallac, Inc.). Negative controls include substrate plus ATP alone. SW1353 cellular assay: SW1353 cells (human chondro-sarcoma) are seeded (1000 cells/100 μL· DMEM 10% FCS/well) into 96-well plates and incubated overnight. After medium replacement, cells are exposed to test compounds for 1 h at 37 oC, at which time human IL-1 (1 ng/mL, Endogen, Woburn, WA) and recombinant human TNFalpha (10 ng/mL) are added. Cultures are incubated for 48 h at 37 oC, then supernatant IL-6 values are determined by ELISA. The compounds of this invention show significant inhibition of p38 kinase.
Example 16: Bio-Plex Phospho-ERK ½ immunoassay.
A 96 well pERK immunoassay, using laser flow cytometry (Bio-Rad) platform has been established to measure inhibition of basal pERK in breast cancer cell line. MDA-MB-231 cells are plated at 50,000 cells per well in 96 well microtitre plates in complete growth media. For effects of test compounds on basal pERKl/2 inhibition, the next day after plating, MDA- MB-231 cells are transferred to DMEM with 0.1% BSA and incubated with test compounds diluted 1:3 to a final concentration of 3 μΜ to 12 nM in 0.1% DMSO. Cells are incubated with test compounds for 2 h, washed, and lysed in Bio-Plex whole cell lysis buffer A. Samples are diluted with buffer B 1:1 (v/v) and directly transferred to assay plate or frozen at -80 C degrees until processed. 50 μL· of diluted MDA-MB-231 cell lysates are incubated with about 2000 of 5 micron Bio-Plex beads conjugated with an anti-ERKl/2 antibody overnight on a shaker at room temperature. The next day, biotinylated phospho-ERKl/2 sandwich immunoassay is performed, beads are washed 3 times during each incubation and then 50 μL· of PE-strepavidin is used as a developing reagent. The relative fluorescence units of pERKl/2 are detected by counting 25 beads with Bio-Plex flow cell (probe) at high sensitivity. The IC50 is calculated by taking untreated cells as maximum and no cells (beads only) as background using in an Excel spreadsheet based program.
The compound of this invention shows significant inhibition in this assay.
Example 17: Flk-1 (murine VEGFR-2) Biochemical Assay
This assay is performed in 96-well opaque plates (Costar 3915) in the TR-FRET format. Reaction conditions are as follows: 10 μΜ ATP , 25 nM poly GT-biotin , 2 nM Eu-labelled phospho-Tyr Ab, 10 nM APC, 7 nM Flk-1 (kinase domain), 1% DMSO, 50 mM HEPES pH 7.5, 10 mM MgC12, 0.1 mM EDTA, 0.015% BRIJ, 0.1 mg/mL BSA, 0.1% mercapto- ethanol). Reaction is initiated upon addition of enzyme. Final reaction volume in each well is 100 μΕ. Plates are read at both 615 and 665 nM on a Perkin Elmer Victor V Multilabel counter at about 1.5- 2.0 hours after reaction initiation. Signal is calculated as a ratio: (665 nm / 615 nm) * 10000 for each well.
The compounds of this invention show significant inhibition of VEGFR2 kinase.
Example 18: Murine PDGFR FRET biochemical assay
This assay is formatted in a 96-well black plate (Costar 3915). The following reagents are used: Europium-labeled anti-phosphotyrosine antibody pY20 (Perand streptavidin-APC; poly GT-biotin from, and mouse PDGFR. The reaction conditions are as follows: 1 nM mouse PDGFR is combined with 20 μΜ ATP, 7 nM poly GT-biotin, 1 nM pY20 antibody, 5 nM streptavidin-APC, and 1% DMSO in assay buffer (50 mM HEPES pH 7.5, 10 mM MgC12, 0.1 mM EDTA, 0.015% BRIJ 35, 0.1 mg/mL BSA, 0.1% mercaptoethanol). Reaction is initiated upon addition of enzyme. Final reaction volume in each well is 100 μΕ. After 90 minutes, the reaction is stopped by addition of 10 μL/well of 5 μΜ staurosporine. Plates are read at both 615 and 665 nm on a Perkin Elmer VictorV Multilabel counter at about 1 hour after the reaction is stopped. Signal is calculated as a ratio: (665 nm / 615 nm) * 10000 for each well. The compounds of this invention show significant inhibition of PDGFR kinase.
For IC50 generation for both PDGFR and Flk-1, compounds are added prior to the enzyme initiation. A 50-fold stock plate was made with compounds serially diluted 1:3 in a 50% DMSO/50% dH20 solution. A 2 μΕ addition of the stock to the assay gives final compound concentrations ranging from 10 μΜ - 4.56 nM in 1% DMSO. The data are expressed as percent inhibition: % inhibition = 100-((Signal with inhibitor-background)/(Signal without inhibitor - background)) * 100
Example 19: MDA-MB231 proliferation assay
Human breast carcinoma cells (MDA MB-231, NCI) are cultured in standard growth medium (DMEM) supplemented with 10% heat-inactivated FBS at 37oC in 5% C02 (vol/ vol) in a humidified incubator. Cells are plated at a density of 3000 cells per well in 90 μΕ growth medium in a 96 well culture dish. In order to determine TOh CTG values, 24 hours after plating, 100 μL· of CellTiter-Glo Luminescent Reagent (Promega) is added to each well and incubated at room temperature for 30 minutes. Luminescence is recorded on a Wallac Victor II instrument. The CellTiter-Glo reagent results in cell lysis and generation of a luminescent signal proportional to the amount of ATP present, which, in turn is directly proportional to the number of cells present.
Test compounds are dissolved in 100% DMSO to prepare 10 mM stocks. Stocks are further diluted 1:400 in growth medium to yield working stocks of 25 μΜ test compound in 0.25% DMSO. Test compounds are serially diluted in growth medium containing 0.25% DMSO to maintain constant DMSO concentrations for all wells. 60 μL· of diluted test compound are added to each culture well to give a final volume of 180 μL·. The cells with and without individual test compounds are incubated for 72 hours at which time ATP dependent luminescence is measured, as described previously, to yield T72h values. Optionally, the IC50 values can be determined with a least squares analysis program using compound concentration versus percent inhibition.
% Inhibition = [l-(T72h test-T0h)/(T72h ctrl-TOh)] x 100, where
T72h test = ATP dependent luminescence at 72 hours in the presence of test compound T72h Ctrl = ATP dependent luminescence at 72 hours in the absence of test compound TOh = ATP dependent luminescence at Time Zero
The compound of this invention shows significant inhibition of proliferation using this assay.
Example 20: pPDGFR-beta sandwich ELISA in AoSMC cells
100K P3-P6 Aortic SMC are plated in each well of 12-well cluster in 1000 volume/ well of SGM-2 using standard cell culture techniques. Next day, cells are rinsed with 1000 μL· D- PBS once, then serum starved in 500 μL· SBM (smooth muscle cell basal media) with 0.1% BSA overnight. Compounds are diluted at a dose range from (10 μΜ to 1 nM in 10-fold dilution steps in DMSO. Final DMSO concentration 0.1%). Remove old media by inversion into the sink quickly then add 100 μL· of each dilution to corresponding well of cells for 1 h at 37 0C. Cells are then stimulated with 10 ng/mL PDGF-BB ligand for 7 min at 37 0C. The media is decanted and 150 μL· of isotonic lysis buffer with protease inhibitor tablet (Complete; EDTA-free) and 0.2 mM Na vanadate is added. Cells are lysed for 15 min at 4 0C on shaker in cold room. Lysates are put in eppendorf tubes to which 15 μL· of agarose- conjugated anti-PDGFR-beta antibody is added and incubated at 4 0C overnight. Next day, beads are rinsed in 50-volumes of PBS three times and boiled in lx LDS sample buffer for 5 minutes. Samples are run on 3-8% gradient Tris -Acetate gels and transferred onto Nitrocellulose. Membranes are blocked in 1% BSA/TBS-T for 1 hr. before incubation in anti-phospho-PDGFR-b (Tyr-857) antibody in blocking buffer (1 : 1000 dilution) for 1 h. After three washes in TBS-T, membranes are incubated in Goat anti-rabbit HRP IgG (1:25000 dilution) for 1 hr. Three more washes followed before addition of ECL substrate. Membranes are exposed to Hyperfilm-ECL. Subsequently, membranes are stripped and reprobed with anti-PDGFR-beta antibody for total PDGFR-beta.
Table A illustrates the results of a representative in vitro kinase biochemical assay for p38 kinase, PDGFR kinase and VEGFR2 kinase. These three kinase targets are all involved in stroma activation and endothelial cell proliferation, leading to angiogenesis, and providing blood supply to the tumor tissue.
Table A
Table B illustrates representative results of two cellular assays for raf kinase activity, which are (i) inhibition of pERK in MDA-MB231 cells, a mechanistic readout of raf kinase activity, and (ii) a proliferation assay of MDA-MB231 cells, a functional assay of raf kinase activity. In addition, Table B illustrates the results of PDGFR driven phosphorylation of PDGFR-beta in aortic smooth muscle cells, which is a mechanistic readout of PDGFR kinase inhibition.
Table B
Figure imgf000073_0002
Overall, compounds of the present invention provide a unique combination of inhibition of angiogenesis and tumor cell proliferation. They also possess an improved inhibition profile against several key kinase targets such as raf, p38, PDGFR, and VEGFR-2, which are all molecular targets of interest for the treatment of osteoporosis, inflammatory diseases, and hyper-proliferative diseases, including cancer.
It is believed that one skilled in the art, using the preceding information and information available in the art, can utilize the present invention to its fullest extent. It should be apparent to one of ordinary skill in the art that changes and modifications can be made to this invention without departing from the spirit or scope of the invention as it is set forth herein. All publications, applications and patents cited above and below are incorporated herein by reference.
The topic headings set forth above and below are meant as guidance where certain information can be found in the application, but are not intended to be the only source in the application where information on such topic can be found.
Without further elaboration, it is believed that one skilled in the art can, using the preceding description, utilize the present invention to its fullest extent. The preceding preferred specific embodiments are, therefore, to be construed as merely illustrative, and not limitative of the remainder of the disclosure in any way whatsoever.
In the foregoing and in the examples, all temperatures are set forth uncorrected in degrees Celsius and, all parts and percentages are by weight, unless otherwise indicated.
The entire disclosures of all applications, patents and publications, cited herein and the following publications are incorporated by reference herein.
1. Wilhelm S et al. "Regorafenib (BAY 73-4506): identification of clinically relevant metabolites and their preclinical pharmacology." American Society of Clinical Oncology (ASCO) Annual Meeting, 2010. Abstract no. 1666 (enclosed).
2. Wilhelm S et al. Mol Cancer Ther 2009; 8 (suppl): abs B42.
3. Eisen T et al. Eur J Cancer Suppl 2009; 7: 424, abs O-71053.
4. Strumberg D et al. J Clin Oncol (Meeting Abstracts) 2009; 27: 161s, abs 35604.
5. Fabian MA et al. Nat Biotechnol 2005; 23: 329-336 The preceding examples can be repeated with similar success by substituting the generically or specifically described reactants and/or operating conditions of this invention for those used in the preceding examples.
From the foregoing description, one skilled in the art can easily ascertain the essential characteristics of this invention and, without departing from the spirit and scope thereof, can make various changes and modifications of the invention to adapt it to various usages and conditions.

Claims

CLAIMS WHAT WE CLAIM IS
Claim 1. An M2, M3, M4, or M5 synthetic metabolite of a compound of Formula I or a salt or a prodrug or a stereoisomer of said synthetic metabolite, wherein the structures of said compound of formula I and said M2, M3, M4, and M5 synthetic metabolites are:
Figure imgf000076_0001
Figure imgf000077_0001
Claim 2. The synthetic metabolite according to claim 1 or a salt or a prodrug or a stereoisomer of said synthetic metabolite, which is an M2 or an M5 synthetic metabolite having the following strucutural formula:
Figure imgf000077_0002
Figure imgf000078_0001
Claim 3. A method of inhibiting tumor growth in a subject comprising administering to said subject the synthetic metabolite according to claim 1 or a pharmaceutical composition comprising said synthetic metabolite.
Claim 4. A method of making the synthetic metabolite according to claim 1, comprising contacting a compound of formula I or a salt thereof with a liver microsome preparation under conditions sufficient for the metabolism of said compound of Formula I and isolating the metabolites from said preparation.
PCT/US2011/032835 2010-04-17 2011-04-18 Synthetic metabolites of fluoro substituted omega-carboxyaryl diphenyl urea for the treatment and prevention diseases and conditions WO2011130728A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
CN2011800298301A CN103079567A (en) 2010-04-17 2011-04-18 Synthetic metabolites of fluoro substituted omega-carboxyaryl diphenyl urea for the treatment and prevention diseases and conditions
CA2796744A CA2796744A1 (en) 2010-04-17 2011-04-18 Synthetic metabolites of fluoro substituted omega-carboxyaryl diphenyl urea for the treatment and prevention of diseases and conditions

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US32529410P 2010-04-17 2010-04-17
US61/325,294 2010-04-17

Publications (1)

Publication Number Publication Date
WO2011130728A1 true WO2011130728A1 (en) 2011-10-20

Family

ID=44144967

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2011/032835 WO2011130728A1 (en) 2010-04-17 2011-04-18 Synthetic metabolites of fluoro substituted omega-carboxyaryl diphenyl urea for the treatment and prevention diseases and conditions

Country Status (3)

Country Link
CN (1) CN103079567A (en)
CA (1) CA2796744A1 (en)
WO (1) WO2011130728A1 (en)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102643229A (en) * 2012-01-17 2012-08-22 湖南有色凯铂生物药业有限公司 N-((4-chloro-3-trifluoromethyl) phenyl)-N'-((2-fluoro-4-(2-formamyl)-4-pyridyloxy) phenyl) urea and application thereof serving as anticancer medicament
CN102885814A (en) * 2012-01-17 2013-01-23 湖南有色凯铂生物药业有限公司 Compound and use of compound as anti-cancer medicine
CN103058922A (en) * 2012-09-06 2013-04-24 湖南有色凯铂生物药业有限公司 Crystal form of aromatic urea for antitumor drug and preparation method of crystal form
CN103508944A (en) * 2012-06-25 2014-01-15 湖南有色凯铂生物药业有限公司 Crystal form A of N-((4-chloro-3-trifluoromethyl)phenyl)-N'-((2-fluoro-4-(2-formamyl)-4-pyridyloxy)phenyl)urea and preparation method thereof

Families Citing this family (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104250226B (en) * 2013-06-27 2019-04-26 常州方楠医药技术有限公司 A method of preparing Rui Gefeini intermediate
WO2015035531A1 (en) * 2013-09-12 2015-03-19 杭州普晒医药科技有限公司 Crystal form of regorafenib salt and preparation method and application thereof
CN104829523B (en) * 2014-04-30 2017-10-31 药源药物化学(上海)有限公司 Rui Gefeini salt and its crystal formation, preparation method
CN105218439B (en) * 2014-06-06 2018-11-20 连云港润众制药有限公司 A kind of crystal of Rui Gefeini and preparation method thereof
CN105566215B (en) * 2014-10-17 2018-02-16 沈阳药科大学 A kind of Rui Gefeini preparation method
CN104592105B (en) * 2015-02-10 2017-01-18 杭州朱养心药业有限公司 Regorafenib and manufacture method thereof
CN107921173B (en) * 2015-08-03 2021-07-06 富士胶片株式会社 Cell structure, non-human model animal, method for producing non-human model animal, and method for evaluating test substance
CN109796401B (en) * 2019-04-04 2023-10-17 新乡双鹭药业有限公司 Preparation method of regorafenib bulk drug
CN112587485A (en) * 2021-01-08 2021-04-02 湖南南新制药股份有限公司 Medicine solid dispersion and preparation method thereof

Citations (51)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR901228A (en) 1943-01-16 1945-07-20 Deutsche Edelstahlwerke Ag Ring gap magnet system
US4220302A (en) 1978-07-21 1980-09-02 Hampton Diane M Nursing bottle holder
US4683202A (en) 1985-03-28 1987-07-28 Cetus Corporation Process for amplifying nucleic acid sequences
US4683195A (en) 1986-01-30 1987-07-28 Cetus Corporation Process for amplifying, detecting, and/or-cloning nucleic acid sequences
WO1988010315A1 (en) 1987-06-19 1988-12-29 Siska Diagnostics, Inc. Transcription-based nucleic acid amplification/detection systems
EP0320308A2 (en) 1987-12-11 1989-06-14 Abbott Laboratories Method for detecting a target nucleic acid sequence
WO1990015070A1 (en) 1989-06-07 1990-12-13 Affymax Technologies N.V. Very large scale immobilized peptide synthesis
US5011472A (en) 1988-09-06 1991-04-30 Brown University Research Foundation Implantable delivery system for biological factors
US5023252A (en) 1985-12-04 1991-06-11 Conrex Pharmaceutical Corporation Transdermal and trans-membrane delivery of drugs
WO1992010092A1 (en) 1990-12-06 1992-06-25 Affymax Technologies N.V. Very large scale immobilized polymer synthesis
US5192744A (en) 1990-01-12 1993-03-09 Northwestern University Method of inhibiting angiogenesis of tumors
US5210015A (en) 1990-08-06 1993-05-11 Hoffman-La Roche Inc. Homogeneous assay system using the nuclease activity of a nucleic acid polymerase
US5262311A (en) 1992-03-11 1993-11-16 Dana-Farber Cancer Institute, Inc. Methods to clone polyA mRNA
US5348853A (en) 1991-12-16 1994-09-20 Biotronics Corporation Method for reducing non-specific priming in DNA amplification
US5409818A (en) 1988-02-24 1995-04-25 Cangene Corporation Nucleic acid amplification process
US5424186A (en) 1989-06-07 1995-06-13 Affymax Technologies N.V. Very large scale immobilized polymer synthesis
US5487985A (en) 1990-10-15 1996-01-30 Stratagene Arbitrarily primed polymerase chain reaction method for fingerprinting genomes
US5508169A (en) 1990-04-06 1996-04-16 Queen's University At Kingston Indexing linkers
US5532129A (en) 1991-11-07 1996-07-02 Enterprise Partners Ii, L.P. Self-organizing molecular photonic structures based on chromophore- and fluorophore-containing polynucleotides and methods of their use
US5554527A (en) 1989-11-24 1996-09-10 Behringwerke Aktiengesellschaft Agent for the storage and suspension of cells, especially erythrocytes
US5599672A (en) 1992-03-11 1997-02-04 Dana-Farber Cancer Institute, Inc. Method of differential display of exposed mRNA by RT/PCR
WO1997018454A2 (en) 1995-11-16 1997-05-22 Baylor College Of Medicine Method for identifying metastatic sequences
US5700637A (en) 1988-05-03 1997-12-23 Isis Innovation Limited Apparatus and method for analyzing polynucleotide sequences and method of generating oligonucleotide arrays
US5712126A (en) 1995-08-01 1998-01-27 Yale University Analysis of gene expression by display of 3-end restriction fragments of CDNA
US5723290A (en) 1994-11-03 1998-03-03 Trustees Of The University Of Pennsylvania Methods for profiling mRNA expression in neurites
US5747251A (en) 1992-10-08 1998-05-05 The Regents Of The University Of California Polymerase chain reaction assays to determine the presence and concentration of a target nucleic acid in a sample
US5871918A (en) 1996-06-20 1999-02-16 The University Of North Carolina At Chapel Hill Electrochemical detection of nucleic acid hybridization
US5874219A (en) 1995-06-07 1999-02-23 Affymetrix, Inc. Methods for concurrently processing multiple biological chip assays
US5928907A (en) 1994-04-29 1999-07-27 The Perkin-Elmer Corporation., Applied Biosystems Division System for real time detection of nucleic acid amplification products
US5994063A (en) 1995-06-23 1999-11-30 Metzker; Michael L. Substituted 4,4-difluoro-4-bora-3A,4A-diaza-s-indacene compounds for homogenous amplification/detection assays
US6017949A (en) 1996-08-02 2000-01-25 D'amato; Robert J. Method of regulating the female reproductive system through angiogenesis inhibitors
US6024688A (en) 1996-03-08 2000-02-15 The Children's Medical Center Corporation Angiostatin fragments and method of use
US6030787A (en) 1994-11-16 2000-02-29 Pe Corporation Hybridization assay using self-quenching fluorescence probe
US6040166A (en) 1985-03-28 2000-03-21 Roche Molecular Systems, Inc. Kits for amplifying and detecting nucleic acid sequences, including a probe
US6054270A (en) 1988-05-03 2000-04-25 Oxford Gene Technology Limited Analying polynucleotide sequences
US6060449A (en) 1996-03-25 2000-05-09 Juridical Foundation The Chemo-Sero-Therapeutic Research Institute Neovascularization inhibitor containing tissue factor pathway inhibitor
WO2000042012A1 (en) 1999-01-13 2000-07-20 Bayer Corporation φ-CARBOXYARYL SUBSTITUTED DIPHENYL UREAS AS RAF KINASE INHIBITORS
WO2000041698A1 (en) 1999-01-13 2000-07-20 Bayer Corporation φ-CARBOXY ARYL SUBSTITUTED DIPHENYL UREAS AS p38 KINASE INHIBITORS
US6117635A (en) 1996-07-16 2000-09-12 Intergen Company Nucleic acid amplification oligonucleotides with molecular energy transfer labels and methods based thereon
US20010003447A1 (en) 1999-12-08 2001-06-14 Hiroyuki Murai Liquid crystal display device
US6316479B1 (en) 1997-05-19 2001-11-13 Sugen, Inc. Isoxazole-4-carboxamide compounds active against protein tryosine kinase related disorders
US20020013774A1 (en) 2000-07-10 2002-01-31 Colondot.Com System and method for negotiating improved terms for products and services being purchased through the internet
US20020165394A1 (en) 1999-01-13 2002-11-07 Bayer Corporation Inhibition of RAF kinase using quinolyl, isoquinolyl or pyridyl ureas
WO2005009961A2 (en) * 2003-07-23 2005-02-03 Bayer Pharmaceuticals Corporation Fluoro substituted omega-carboxyaryl diphenyl urea for the treatment and prevention of diseases and conditions
US7124802B2 (en) 2003-05-21 2006-10-24 Royal Group Technologies Limited Cascade shade
WO2007054216A1 (en) * 2005-11-10 2007-05-18 Bayer Healthcare Ag Diaryl urea for treating pulmonary hypertension
WO2007068380A1 (en) * 2005-12-15 2007-06-21 Bayer Healthcare Ag Diaryl urea for treating virus infections
WO2008089389A2 (en) * 2007-01-19 2008-07-24 Bayer Healthcare Llc Treatment of cancers with acquired resistance to kit inhibitors
WO2008089388A2 (en) * 2007-01-19 2008-07-24 Bayer Healthcare Llc Treatment of cancers having resistance to chemotherapeutic agents
US8700880B2 (en) 2008-10-10 2014-04-15 Apple Inc. Dynamic trampoline and structured code generation in a signed code environment
US9993647B2 (en) 2014-12-31 2018-06-12 Tsinghua University Variable frequency stimulation therapy method

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB0614498D0 (en) * 2006-07-21 2006-08-30 Univ Belfast Improvements in Hydrogen Trapping

Patent Citations (58)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR901228A (en) 1943-01-16 1945-07-20 Deutsche Edelstahlwerke Ag Ring gap magnet system
US4220302A (en) 1978-07-21 1980-09-02 Hampton Diane M Nursing bottle holder
US4683202A (en) 1985-03-28 1987-07-28 Cetus Corporation Process for amplifying nucleic acid sequences
US6040166A (en) 1985-03-28 2000-03-21 Roche Molecular Systems, Inc. Kits for amplifying and detecting nucleic acid sequences, including a probe
US4683202B1 (en) 1985-03-28 1990-11-27 Cetus Corp
US5023252A (en) 1985-12-04 1991-06-11 Conrex Pharmaceutical Corporation Transdermal and trans-membrane delivery of drugs
US4683195A (en) 1986-01-30 1987-07-28 Cetus Corporation Process for amplifying, detecting, and/or-cloning nucleic acid sequences
US4683195B1 (en) 1986-01-30 1990-11-27 Cetus Corp
WO1988010315A1 (en) 1987-06-19 1988-12-29 Siska Diagnostics, Inc. Transcription-based nucleic acid amplification/detection systems
EP0320308A2 (en) 1987-12-11 1989-06-14 Abbott Laboratories Method for detecting a target nucleic acid sequence
US5409818A (en) 1988-02-24 1995-04-25 Cangene Corporation Nucleic acid amplification process
US5700637A (en) 1988-05-03 1997-12-23 Isis Innovation Limited Apparatus and method for analyzing polynucleotide sequences and method of generating oligonucleotide arrays
US6054270A (en) 1988-05-03 2000-04-25 Oxford Gene Technology Limited Analying polynucleotide sequences
US5011472A (en) 1988-09-06 1991-04-30 Brown University Research Foundation Implantable delivery system for biological factors
WO1990015070A1 (en) 1989-06-07 1990-12-13 Affymax Technologies N.V. Very large scale immobilized peptide synthesis
US5143854A (en) 1989-06-07 1992-09-01 Affymax Technologies N.V. Large scale photolithographic solid phase synthesis of polypeptides and receptor binding screening thereof
US5424186A (en) 1989-06-07 1995-06-13 Affymax Technologies N.V. Very large scale immobilized polymer synthesis
US5554527A (en) 1989-11-24 1996-09-10 Behringwerke Aktiengesellschaft Agent for the storage and suspension of cells, especially erythrocytes
US5192744A (en) 1990-01-12 1993-03-09 Northwestern University Method of inhibiting angiogenesis of tumors
US5508169A (en) 1990-04-06 1996-04-16 Queen's University At Kingston Indexing linkers
US5210015A (en) 1990-08-06 1993-05-11 Hoffman-La Roche Inc. Homogeneous assay system using the nuclease activity of a nucleic acid polymerase
US5487985A (en) 1990-10-15 1996-01-30 Stratagene Arbitrarily primed polymerase chain reaction method for fingerprinting genomes
WO1992010092A1 (en) 1990-12-06 1992-06-25 Affymax Technologies N.V. Very large scale immobilized polymer synthesis
US5532129A (en) 1991-11-07 1996-07-02 Enterprise Partners Ii, L.P. Self-organizing molecular photonic structures based on chromophore- and fluorophore-containing polynucleotides and methods of their use
US5565322A (en) 1991-11-07 1996-10-15 Nanogen, Inc. Hybridization of polynucleotides conjugated with chromophores and fluorophores to generate donor-to donor energy transfer system
US5348853A (en) 1991-12-16 1994-09-20 Biotronics Corporation Method for reducing non-specific priming in DNA amplification
US5599672A (en) 1992-03-11 1997-02-04 Dana-Farber Cancer Institute, Inc. Method of differential display of exposed mRNA by RT/PCR
US5262311A (en) 1992-03-11 1993-11-16 Dana-Farber Cancer Institute, Inc. Methods to clone polyA mRNA
US5965409A (en) 1992-03-11 1999-10-12 Dana-Farber Cancer Institute System for comparing levels or amounts of mRNAs
US5747251A (en) 1992-10-08 1998-05-05 The Regents Of The University Of California Polymerase chain reaction assays to determine the presence and concentration of a target nucleic acid in a sample
US5928907A (en) 1994-04-29 1999-07-27 The Perkin-Elmer Corporation., Applied Biosystems Division System for real time detection of nucleic acid amplification products
US5723290A (en) 1994-11-03 1998-03-03 Trustees Of The University Of Pennsylvania Methods for profiling mRNA expression in neurites
US6030787A (en) 1994-11-16 2000-02-29 Pe Corporation Hybridization assay using self-quenching fluorescence probe
US5874219A (en) 1995-06-07 1999-02-23 Affymetrix, Inc. Methods for concurrently processing multiple biological chip assays
US5994063A (en) 1995-06-23 1999-11-30 Metzker; Michael L. Substituted 4,4-difluoro-4-bora-3A,4A-diaza-s-indacene compounds for homogenous amplification/detection assays
US6010850A (en) 1995-08-01 2000-01-04 Yale University Analysis of gene expression by display of 3'-end restriction fragments of cDNAs
US5712126A (en) 1995-08-01 1998-01-27 Yale University Analysis of gene expression by display of 3-end restriction fragments of CDNA
WO1997018454A2 (en) 1995-11-16 1997-05-22 Baylor College Of Medicine Method for identifying metastatic sequences
US6024688A (en) 1996-03-08 2000-02-15 The Children's Medical Center Corporation Angiostatin fragments and method of use
US6060449A (en) 1996-03-25 2000-05-09 Juridical Foundation The Chemo-Sero-Therapeutic Research Institute Neovascularization inhibitor containing tissue factor pathway inhibitor
US5871918A (en) 1996-06-20 1999-02-16 The University Of North Carolina At Chapel Hill Electrochemical detection of nucleic acid hybridization
US6117635A (en) 1996-07-16 2000-09-12 Intergen Company Nucleic acid amplification oligonucleotides with molecular energy transfer labels and methods based thereon
US6017949A (en) 1996-08-02 2000-01-25 D'amato; Robert J. Method of regulating the female reproductive system through angiogenesis inhibitors
US6316479B1 (en) 1997-05-19 2001-11-13 Sugen, Inc. Isoxazole-4-carboxamide compounds active against protein tryosine kinase related disorders
WO2000041698A1 (en) 1999-01-13 2000-07-20 Bayer Corporation φ-CARBOXY ARYL SUBSTITUTED DIPHENYL UREAS AS p38 KINASE INHIBITORS
US20010016659A1 (en) 1999-01-13 2001-08-23 Bernd Riedl Omega-carboxyaryl substituted diphenyl ureas as raf kinase inhibitors
US20020165394A1 (en) 1999-01-13 2002-11-07 Bayer Corporation Inhibition of RAF kinase using quinolyl, isoquinolyl or pyridyl ureas
WO2000042012A1 (en) 1999-01-13 2000-07-20 Bayer Corporation φ-CARBOXYARYL SUBSTITUTED DIPHENYL UREAS AS RAF KINASE INHIBITORS
US20010003447A1 (en) 1999-12-08 2001-06-14 Hiroyuki Murai Liquid crystal display device
US20020013774A1 (en) 2000-07-10 2002-01-31 Colondot.Com System and method for negotiating improved terms for products and services being purchased through the internet
US7124802B2 (en) 2003-05-21 2006-10-24 Royal Group Technologies Limited Cascade shade
WO2005009961A2 (en) * 2003-07-23 2005-02-03 Bayer Pharmaceuticals Corporation Fluoro substituted omega-carboxyaryl diphenyl urea for the treatment and prevention of diseases and conditions
WO2007054216A1 (en) * 2005-11-10 2007-05-18 Bayer Healthcare Ag Diaryl urea for treating pulmonary hypertension
WO2007068380A1 (en) * 2005-12-15 2007-06-21 Bayer Healthcare Ag Diaryl urea for treating virus infections
WO2008089389A2 (en) * 2007-01-19 2008-07-24 Bayer Healthcare Llc Treatment of cancers with acquired resistance to kit inhibitors
WO2008089388A2 (en) * 2007-01-19 2008-07-24 Bayer Healthcare Llc Treatment of cancers having resistance to chemotherapeutic agents
US8700880B2 (en) 2008-10-10 2014-04-15 Apple Inc. Dynamic trampoline and structured code generation in a signed code environment
US9993647B2 (en) 2014-12-31 2018-06-12 Tsinghua University Variable frequency stimulation therapy method

Non-Patent Citations (268)

* Cited by examiner, † Cited by third party
Title
"PCR Protocols: A Guide to Methods and Applications", 1990, ACADEMIC PRESS
AIELLO ET AL., NEW ENGL. J. MED., vol. 331, 1994, pages 1480
AISEN ET AL., GERONTOLOGY, vol. 43, 1997, pages 143
AMRANI ET AL., REV. MALAD. RESPIR., vol. 13, 1996, pages 539
ANON, CRIT. CARE MED., vol. 20, 1992, pages 864
ANTONIADES, H.N. ET AL., PROC. NATL. ACAD. SCI. USA, vol. 89, no. 9, 1992, pages 3942 - 6
ASHKENASI ET AL., PROC. NAT'L. ACAD. SCI. USA, vol. 88, 1991, pages 10535
AVRUCH ET AL., RECENT PROG. HORM. RES., vol. 56, 2001, pages 127 - 155
BADGER, A. M., BRADBEER, J. N., VOTTA, B., LEE, J. C., ADAMS, J. L., GRISWOLD, D. E., J. PHARM. EXPER. THER., vol. 279, 1996, pages 1453
BADLEY ET AL., J. EXP. MED., vol. 185, 1997, pages 55
BAKER, E.A., D.J. LEAPER, WOUND REPAIR REGEN., vol. 8, no. 5, 2000, pages 392 - 8
BANKSTON ET AL., ORG. PROC. RES. DEV., vol. 6, no. 6, 2002, pages 777 - 781
BARICOS ET AL., BIOCHEM. J., vol. 254, 1988, pages 609
BASELGA ET AL., DRUGS, vol. 60, no. 1, 2000, pages 33 - 40
BEALES ET AL., GASTROENTEROLOGY, vol. 112, 1997, pages 136
BECK ET AL., PROCEEDINGS OF THE AMERICAN SOCIETY FOR CLINICAL ONCOLOGY, 2004, pages 23
BEEBE ET AL., CANCER RES., vol. 63, 2003, pages 7301 - 7309
BERKMAN ET AL., J. CLIN. INVEST., vol. 91, 1993, pages 153
BEUTLER ET AL., CLIN. RES., vol. 34, 1986, pages 491A
BEUTLER ET AL., CRIT. CARE MED., vol. 21, 1993, pages 5423
BEUTLER ET AL., SCIENCE, vol. 229, 1985, pages 869
BHARDWAJ, B. ET AL., CLIN. CANCER RES., vol. 2, no. 4, 1996, pages 773 - 82
BIRD ET AL., ANN. INTERN. MED., vol. 112, 1990, pages 917
BLACKWELL ET AL., BR. J. ANAESTH., vol. 77, 1996, pages 110
BLASCHKE, F. ET AL., BIOCHEM. BIOPHYS. RES. COMMUN., vol. 296, 2002, pages 890 - 896
BOLLING ET AL., TRANSPLANTATION, vol. 53, 1992, pages 283
BORM ET AL., AM. REV. RESPIR. DIS., vol. 138, 1988, pages 1589
BOS, J. L., CANCER RES., vol. 49, 1989, pages 4682
BRADY ET AL., METHODS MOL. & CELL. BIOL., vol. 2, 1990, pages 17 - 25
BRANDT ET AL., INFECT. IMMUNOL., vol. 58, 1990, pages 983
BRANGER J. ET AL., BLOOD, vol. 101, 2003, pages 4446 - 4448
BRANGER J. ET AL., J. IMMUNOL., vol. 168, 2002, pages 4070 - 4077
BRIDGES, CURR. MED. CHEM., vol. 6, 1999, pages 825 - 843
BROD ET AL., NEUROLOGY, vol. 46, 1996, pages 1633
BROSNAN ET AL., BRAIN PATHOL., vol. 6, 1996, pages 243
BROWN ET AL., AM. J. PATHOL., vol. 1431, 1993, pages 1255
BROWN ET AL., CANCER RES., vol. 53, 1993, pages 4727
BROWN ET AL., HUMAN PATHOL., vol. 26, 1995, pages 86
BROWN ET AL., J. INVEST. DERMATOL., vol. 104, 1995, pages 744
BUCK ET AL., AM. J. PATHOL., vol. 149, 1996, pages 195
BURNS ET AL., INVEST. OPTHALMOL. VIS. SCI., vol. 30, 1989, pages 1569
CANNON ET AL., CRIT. CARE MED., vol. 20, 1992, pages 1414
CARTER ET AL.: "Book of Abstracts, 92ndAACR Meeting"
CASALE ET AL., AM. J. RESPIR. CELL MOL. BIOL., vol. 15, 1996, pages 35
CHAMBERLIN ET AL., INFECT. IMMUNOL., vol. 57, 1989, pages 2872
CHANDRASEKAR ET AL., BIOCHEM. BIOPHYS. RES. COMMUN., vol. 223, 1996, pages 365
CHANTRY ET AL., J. NEUROCHEM., vol. 50, 1988, pages 688
CHRISTOPHERS, AUSTR. J. DERMATOL., vol. 37, 1996, pages 4
CIRINO ET AL., LIFE SCI., vol. 59, 1996, pages 86
COLLETTI ET AL., J. CLIN. INVEST., vol. 85, 1989, pages 1333
CURSIEFEN ET AL., CORNEA, vol. 22, 2003, pages 273 - 81
DAUM ET AL., TRENDS BIOCHEM. SCI., vol. 19, 1994, pages 474 - 80
DEBETS ET AL., PROG. CLIN. BIOL. RES., vol. 308, 1989, pages 463
DEGRE, BIOTHERAPY, vol. 8, 1996, pages 219
DELLAS ET AL., GYN. ONCOL., vol. 67, 1997, pages 27 - 33
DEMETRI ET AL., PROCEEDINGS OF THE AMERICAN SOCIETY FOR CLINICAL ONCOLOGY, 2004, pages 23
DHILLON, KOLCH, ARCH. BIOCHEM. BIOPHYS., vol. 404, 2002, pages 3 - 9
DOYAMA ET AL., INT. J. CARDIOL., vol. 54, 1996, pages 217
DUMAS ET AL., BIOORG. MED. CHEM. LETT., vol. 10, 2000, pages 2047 - 2050
DUMAS ET AL., BIOORG. MED. CHEM. LETT., vol. 10, 2000, pages 2051 - 2054
DUMAS ET AL., BIOORG. MED. CHEM. LETT., vol. 12, 2002, pages 1559 - 1562
EBERWINE ET AL., PROC. NATL. ACAD. SCI., vol. 89, 1992, pages 3010 - 3014
EISEN T ET AL., EUR J CANCER SUPPL, vol. 7, 2009, pages 424
ELICEIRI ET AL., J. CELL BIOL., vol. 140, 1998, pages 1255 - 1263
EXLEY ET AL., GUT, vol. 33, 1992, pages 1126
FABIAN ET AL., NAT BIOTECHNOL, vol. 23, 2005, pages 329 - 336
FABIAN MA ET AL., NAT BIOTECHNOL, vol. 23, 2005, pages 329 - 336
FAGARASAN ET AL., BRAIN RES., vol. 723, 1996, pages 231
FARRARA ET AL., ENDOCR. REV., vol. 13, 1992, pages 18
FEKADE ET AL., NEW ENGLAND J. MED., vol. 335, 1996, pages 311
FISCHER ET AL., J. IMMUNOL., vol. 144, 1990, pages 4663
FLEMING, T.P. ET AL., CANCER RES., vol. 52, no. 16, 1992, pages 4550 - 3
FOLKMAN, J., SEMIN. ONCOL., vol. 29, no. 6, 2002, pages 15 - 8
FORSBERG, K. ET AL., PROC. NATL. ACAD. SCI. USA., vol. 90, no. 2, 1993, pages 393 - 7
FRIDMAN ET AL., J. BIOL. CHEM., vol. 269, 1994, pages 30105 - 8
FUDGE, K., C.Y. WANG, M.E. STEARNS, MOD. PATHOL., vol. 7, no. 5, 1994, pages 549 - 54
FUNA, K. ET AL., CANCER RES., vol. 50, no. 3, 1990, pages 748 - 53
GANTNER ET AL., J. PHARMACOL. EXP. THERAP., vol. 280, 1997, pages 53
GEIST ET AL., AM. J. RESPIR. CELL MOL. BIOL., vol. 16, 1997, pages 31
GEORGE, D., SEMIN. ONCOL., vol. 28, no. 5, 2001, pages 27 - 33
GIRARDIN ET AL., NEW ENGLAND J. MED., vol. 319, 1988, pages 397
GOLDFIELD ET AL., PROC. NAT'L. ACAD. SCI. USA, vol. 87, 1989, pages 1490
GOODMAN, GILMAN'S: "The Pharmacological Basis of Therapeutics", 1996, MCGRAW-HILL
GOSSART ET AL., J. IMMUNOL., vol. 156, 1996, pages 1540
GRAU ET AL., IMMUNOL. REV., vol. 112, 1989, pages 49
GRAVALLESE ET AL., ARTHRITIS RHEUM., vol. 34, 1991, pages 1076
GREENBERGER ET AL., LLTH NCI-EORTC-AACR SYMPOSIUM ON NEW DRUGS IN CANCER THERAPY, 7 November 2000 (2000-11-07)
GROSSMAN ET AL., IMMUNOL. ALLERGY CLIN. N. AM., vol. 9, 1989, pages 153
GRUGEL ET AL., J. BIOL. CHEM., vol. 270, 1995, pages 25915
GUIDI ET AL., J. NAT'L CANCER INST., vol. 87, 1995, pages 12137
HAMRAH, AM. J. PATH., vol. 163, 2003, pages 57 - 68
HANSBROUGH ET AL., SURG. CLIN. N. AM., vol. 67, 1987, pages 69
HARARI ET AL., ONCOGENE, vol. 19, no. 53, 2000, pages 6102 - 6114
HAREL ET AL., J. CLIN. INVEST., vol. 56, 1992, pages 40
HASHIMOTO ET AL., LAB. INVEST., vol. 73, 1995, pages 859
HEINRICH, M.C. ET AL., SCIENCE, vol. 9, 2003, pages 9
HENNEQUIN ET AL., 92ND AACR MEETING, 24 March 2001 (2001-03-24)
HENNEQUIN ET AL., PROCEEDINGS OF THE AMERICAN ASSOCIATION OF CANCER RESEARCH, 2004, pages 45
HENNEY ET AL., PROC. NAT'L. ACAD. SCI., USA, vol. 88, 1991, pages 8154
HENRIKSEN, R. ET AL., CANCER RES., vol. 53, no. 19, 1993, pages 4550 - 4
HERBST ET AL., CLIN. CANCER RES., vol. 9, no. 1, 2003, pages 16
HILGER ET AL.: "Book of Abstracts, 38th ASCO Meeting"
HOLLAND ET AL., PROC. NATL. ACAD, SCI., vol. 88, 1991, pages 7276 - 7280
HORINOUCHI ET AL., AM. J. RESPIR. CELL MOL. BIOL., vol. 14, 1996, pages 1044
HUANG, S. ET AL., J. BIOL. CHEM., vol. 275, 2000, pages 12266 - 12272
ICHIYAMA ET AL., J. NEUROL., vol. 243, 1996, pages 457
IMAGAWA ET AL., TRANSPLANTATION, vol. 50, 1990, pages 219
ISHIOKA ET AL., §ARCOIDOSIS VASCULITIS DIFFUSE LUNG DIS., vol. 13, 1996, pages 139
JANI ET AL., PROCEEDINGS OF THE AMERICAN SOCIETY FOR CLINICAL ONCOLOGY, vol. 23, 2004
KARPANEN, T. ET AL., CANCER RES., vol. 61, no. 5, 2001, pages 1786 - 90
KHIRE ET AL.: "Book of Abstracts, 93rdAACR Meeting"
KIM ET AL., J. BIOL. CHEM., vol. 272, 1997, pages 1402
KIM ET AL., NATURE, vol. 362, 1993, pages 841
KOCH ET AL., J. IMMUNOL., vol. 152, 1994, pages 4149
KOLCH ET AL., NATURE, vol. 349, 1991, pages 426 - 28
KRONBERGER ET AL., J. INVEST. DERMATOL., vol. 79, 1982, pages 208
KUBO, H. ET AL., BLOOD, vol. 96, no. 2, 2000, pages 546 - 53
LACKEY ET AL., 92ND AACR MEETING, 24 March 2001 (2001-03-24)
LAFERRIERE, J. ET AL., J. BIOL. CHEM., vol. 276, 2001, pages 33762 - 33772
LAWLOR ET AL., J. CELL SCI., vol. 114, 2001, pages 2903 - 2910
LEE ET AL., PROCEEDINGS OF THE AMERICAN ASSOCIATION OF CANCER RESEARCH, 2004, pages 45
LEE, J. C., LAYDON, J. T., MCDONNELL, P. C., GALLAGHER, T. F., KUMAR, S., GREEN, D., MCNULTY, D., BLUMENTHAL, M. J., HEYS, J. R.,, NATURE, vol. 372, 1994, pages 739
LEVY ET AL., CRIT. REV. IMMUNOL., vol. 16, 1996, pages 31
LIANG ET AL., NUCL. ACID. RES., vol. 21, 1993, pages 3269 - 3275
LINA ET AL., FEMS IMMUNOL. MED. MICROBIOL., vol. 13, 1996, pages 81
LINDMARK, G. ET AL., LAB. INVEST., vol. 69, no. 6, 1993, pages 682 - 9
LOCICERO ET AL., J. THORAC. CARDIOVASC. SURG., vol. 99, 1990, pages 1059
LOPEZ ET AL., INVEST. OPTHTHALMOL. VIS. SCI., vol. 37, 1996, pages 855
LOWINGER ET AL., CLIN. CANCER RES., vol. 6, 2000, pages 335
LOWINGER ET AL., CURR. PHARM. DESIGN, vol. 8, 2002, pages 99 - 110
LOWY, D. R., WILLUMSEN, B. M., ANN. REV. BIOCHEM., vol. 62, 1993, pages 851
LYONS ET AL., ENDOCR.-RELAT. CANCER, vol. 8, 2001, pages 219 - 225
M.S.; COYLE, ADV. NEUROIMMUNOL., vol. 6, 1996, pages 143
MACKAY ET AL., PROCEEDINGS OF THE AMERICAN SOCIETY FOR CLINICAL ONCOLOGY, 2004, pages 23
MADWED J. B.: "Book of Abstracts, Protein Kinases: Novel Target Identification and Validation for Therapeutic Development", March 2002
MAINI. J., ROYAL COLL. PHYSICIANS LONDON, vol. 30, 1996, pages 344
MAKINEN, T. ET AL., NATURE MED., vol. 7, no. 2, 2001, pages 199 - 205
MALKIEL ET AL., MOL. MED. TODAY, vol. 2, 1996, pages 336
MANDRIOTA, S.J. ET AL., EMBO J., vol. 20, no. 4, 2001, pages 672 - 82
MARANO ET AL., SURG. GYNECOL. OBSTETR., vol. 170, 1990, pages 32
MASUDA ET AL., J. CLIN. LAB. IMMUNOL., vol. 46, 1995, pages 111
MATRISIAN ET AL., PROC. NAT'L. ACAD. SCI., USA, vol. 83, 1986, pages 9413
MATTERN ET AL., BR. J. CANCER, vol. 73, 1996, pages 931
MATUSEVICIUS ET AL., J. NEUROIMMUNOL., vol. 66, 1996, pages 115
MAURY ET AL., ARTHRITIS RHEUM., vol. 32, 1989, pages 146
MAURY ET AL., J. EXP. MED., vol. 166, 1987, pages 1132
MCDONALD, N.Q., HENDRICKSON, W.A., CELL, vol. 73, 1993, pages 421 - 424
MCKAY ET AL., BR. J. SURG., vol. 83, 1996, pages 919
MCMAHON, G., ONCOLOGIST, vol. 5, no. 1, 2000, pages 3 - 10
MCMURRAY ET AL., BR. HEART J., vol. 66, 1991, pages 356
MILLER ET AL., AM. J. GASTEROENTEROLOG., vol. 87, 1992, pages 465
MONIA ET AL., NAT. MED., vol. 2, 1996, pages 668 - 75
MOORE ET AL.: "Book of Abstracts, 38th ASCO Meeting"
MUCWIERZGON ET AL., J. BIOL. REGULATORS HOMEOSTATIC AGENTS, vol. 10, 1996, pages 25
MULLINS ET AL., BIOCHIM. BIOPHYS. ACTA, vol. 695, 1983, pages 117
MUSTONEN ET AL., J. CELL BIOL., vol. 129, 1995, pages 895
NAKANISHI, K. ET AL., MOD. PATHOL., vol. 10, no. 4, 1997, pages 341 - 7
NEMA, S. ET AL.: "Excipients and Their Use in Injectable Products", PDA JOURNAL OF PHARMACEUTICAL SCIENCE & TECHNOLOGY, vol. 51, no. 4, 1997, pages 166 - 171
NEUFIELD ET AL., FASEB J., vol. 13, 1999, pages 9
OFEI ET AL., DIABETES, vol. 45, 1996, pages 881
OHARA ET AL., PROC. NATL. ACAD. SCI., vol. 86, 1989, pages 5673 - 5677
OLSON ET AL., CANCER RES., vol. 54, 1994, pages 1255
OSSEGE ET AL., J. NEUROLOG. SCI., vol. 144, 1996, pages 1
OSTMAN, A., C.H. HELDIN, ADV. CANCER RES., vol. 80, 2001, pages 1 - 38
OTTMANN ET AL., PROCEEDINGS OF THE AMERICAN SOCIETY FOR CLINICAL ONCOLOGY, 2004, pages 23
OVERALL ET AL., J. PERIODONTAL RES., vol. 22, 1987, pages 81
PACIFICI ET AL., J. BONE MINERAL RES., vol. 11, 1996, pages 1043
PACIFICI ET AL., J. CLIN. ENDOCRINOL. METABOL., vol. 82, 1997, pages 29
PAN ET AL., PATHOL. INT., vol. 46, 1996, pages 91
PARGELLIS C. ET AL., CURR. OPIN. INVEST. DRUGS, vol. 4, 2003, pages 566 - 571
PARGELLIS ET AL., NATURE STRUCT. BIOL., vol. 9, no. 4, 2002, pages 268 - 272
PARK ET AL., ONCOGENE, vol. 10, 1995, pages 135
PARUMS ET AL., J. PATHOL., vol. 179, 1996, pages A46
PEACOCK ET AL., J. EXPER. MED., vol. 175, 1992, pages 1135
PEER ET AL., LAB. INVEST., vol. 72, 1995, pages 638
PERLMANN ET AL., INFECT. IMMUNIT., vol. 65, 1997, pages 116
PHILLIPS ET AL., INT. J. ONCOL., vol. 2, 1993, pages 913
PIEKARZ ET AL., PROCEEDINGS OF THE AMERICAN SOCIETY FOR CLINICAL ONCOLOGY, 2004, pages 23
PIETRAS, K. ET AL., CANCER RES., vol. 61, no. 7, 2001, pages 2929 - 34
PIETRAS, K. ET AL., CANCER RES., vol. 62, no. 19, 2002, pages 5476 - 84
PIGUET ET AL., IMMUNOL. SER., vol. 56, 1992, pages 409
PLATE ET AL., NATURE, vol. 359, 1992, pages 845
POLI, PROC. NAT'L. ACAD. SCI. USA, vol. 87, 1990, pages 782
POLLACK ET AL., J. PHARM. EXP. THER., vol. 291, no. 2, 1999, pages 739 - 748
POLVERINI, P. J. ET AL., METHODS ENZYMOL., vol. 198, 1991, pages 440 - 450
POWELL, M.F. ET AL.: "Compendium of Excipients for Parenteral Formulations", PDA JOURNAL OF PHARMACEUTICAL SCIENCE & TECHNOLOGY, vol. 52, no. 5, 1998, pages 238 - 311, XP009119027
PRASHAR, WEISSMAN, PROC. NATL. ACAD. SCI., vol. 93, pages 659 - 663
QUINN ET AL., J. CLIN. ONCOLOGY, vol. 21, no. 4, 2003, pages 646 - 651
RABINDRAN ET AL., CANCER RES., vol. 64, 2004, pages 3958 - 3965
RAK ET AL., CANCER RES., vol. 55, 1995, pages 4575
RANGES ET AL.: "Book of Abstracts, 220th ACS National Meeting"
RAYMOND ET AL., DRUGS, vol. 60, no. 1, 2000, pages 15 - 23
RAZA ET AL., INT. J. HEMATOL., vol. 63, 1996, pages 265
REDLICH ET AL., J. IMMUNOL., vol. 157, 1996, pages 1705
REDMAN ET AL., BIOORG. MED. CHEM. LETT., vol. 11, 2001, pages 9 - 12
REGAN ET AL., J. MED. CHEM., vol. 45, 2002, pages 2994 - 3008
REICH ET AL., CANCER RES., vol. 48, 1988, pages 3307
RIEDL ET AL.: "Book of Abstracts, 92nd AACR Meeting"
ROBERTS ET AL., PROCEEDINGS OF THE AMERICAN ASSOCIATION OF CANCER RESEARCH, 2004, pages 45
ROCKWELL ET AL., MOL. CELL. DIFFER., vol. 3, 1995, pages 315
ROOK ET AL., MED. MALAD. INFECT., vol. 26, 1996, pages 904
ROTEN ET AL., AM. REV. RESPIR. DIS., vol. 143, 1991, pages 590
RUDIN ET AL., AM. J. PATHOL., vol. 150, 1997, pages 257
RYAN ET AL., PROCEEDINGS OF THE AMERICAN ASSOCIATION OF CANCER RESEARCH, 2004, pages 45
S. M. BERGE ET AL.: "Pharmaceutical Salts", J. PHARM. SCI., vol. 66, 1977, pages 1 - 19, XP002675560, DOI: doi:10.1002/jps.2600660104
SAHA ET AL., J. IMMUNOL., vol. 157, 1996, pages 3869
SAIKI ET AL., SCIENCE, vol. 241, 1988, pages 53
SATO ET AL., NATURE, vol. 376, 1995, pages 70 - 74
SCHAEFER, GENE CLONING AND ANALYSIS: CURRENT INNOVATIONS, 1997, pages 99 - 115
SEBOLT-LEOPOLD ET AL., PROCEEDINGS OF THE AMERICAN ASSOCIATION OF CANCER RESEARCH, 2004, pages 45
SHAHEEN, R.M. ET AL., CANCER RES., vol. 59, no. 21, 1999, pages 5412 - 6
SHAHEEN, R.M. ET AL., CANCER RES., vol. 61, no. 4, 2001, pages 1464 - 8
SHEMIRANI, B. ET AL., ORAL ONCOLOGY, vol. 38, 2002, pages 251 - 257
SHEN ET AL., BLOOD, vol. 102, 2003, pages 11
SHWEIKI ET AL., NATURE, vol. 359, 1992, pages 843
SHWEIKI ET AL., PROC. NAT'L. ACAD. SCI., vol. 92, 1995, pages 768
SIERRA ET AL., IMMUNOLOGY, vol. 78, 1993, pages 399
SIMON, C. ET AL., EXP. CELL RES., vol. 271, 2001, pages 344 - 355
SKOBE, M. ET AL., NATURE MED., vol. 7, no. 2, 2001, pages 192 - 198
SKOBE, M., N.E. FUSENIG, PROC. NATL. ACAD. SCI. U S A., vol. 95, no. 3, 1998, pages 1050 - 5
SMITH ET AL., BIOORG. MED. CHEM. LETT., vol. 11, 2001, pages 2775 - 2778
SOSKIC, V. ET AL., BIOCHEMISTRY, vol. 38, no. 6, 1999, pages 1757 - 64
STACKER, S.A. ET AL., NATURE MED., vol. 7, no. 2, 2001, pages 186 - 191
STEPHENS ET AL., J. BIOL. CHEM., vol. 272, 1997, pages 971
STEVENS ET AL., TRANSPLANT. PROC., vol. 22, 1990, pages 1924
STOKKERS ET AL., J. INFLAMM., June 1995 (1995-06-01), pages 47,97
STRICKLEY, R.G: "Parenteral Formulations of Small Molecule Therapeutics Marketed in the United States (1999)-Part-1", PDA JOURNAL OF PHARMACEUTICAL SCIENCE & TECHNOLOGY, vol. 53, no. 6, 1999, pages 324 - 349
STRUMBERG D ET AL., J CLIN ONCOL, vol. 27, 2009, pages 161S
STRUMBERG ET AL.: "Book of Abstracts, 38th ASCO Meeting"
SUN ET AL., J. CLIN. INVEST., vol. 81, 1988, pages 1328
SUNDBERG, C. ET AL., AM. J. PATHOL., vol. 151, no. 2, 1997, pages 479 - 92
SUTER ET AL., AM. REV. RESPIR. DIS., vol. 145, 1992, pages 1016
SUZUKI ET AL., CANCER RES., vol. 56, 1996, pages 3004
SZUBA ET AL., FASEB J., vol. 16, no. 14, 2002, pages 1985 - 7
TAGUCHI ET AL., 95TH AACR MEETING, 2004
TAVERNE ET AL., PARASITOL. TODAY, vol. 12, 1996, pages 290
TAYLOR, FOLKMAN, NATURE, vol. 297, 1982, pages 307 - 312
TRACEY ET AL., NATURE, vol. 330, 1987, pages 662
TYAGI, KRAMER, NATURE BIOTECH., vol. 14, 1996, pages 303 - 309
VAN DEVENTER ET AL., ALIMENT. PHARMACOL. THERAPEU., vol. 10, no. 2, 1996, pages 107
VAN DULLEMEN ET AL., GASTROENTEROLOGY, vol. 109, 1995, pages 129
VANHEE ET AL., EUR. RESPIR. J., vol. 8, 1995, pages 834
VIDAL ET AL., PROCEEDINGS OF THE AMERICAN SOCIETY FOR CLINICAL ONCOLOGY, 2004, pages 23
VIGLIETTO ET AL., ONCOGENE, vol. 11, 1995, pages 1569
VIGNAUD, J.M. ET AL., CANCER RES., vol. 54, no. 20, 1994, pages 5455 - 63
VINCENT ET AL.: "Book of Abstracts, 38th ASCO Meeting"
VINE ET AL., CLIN. SCI., vol. 81, 1991, pages 233
VYAKARAM ET AL., AIDS, vol. 4, 1990, pages 21
WAAGE ET AL., LANCET, 1987, pages 355
WALLACE ET AL., PROCEEDINGS OF THE AMERICAN ASSOCIATION OF CANCER RESEARCH, 2004, pages 45
WALTENBERGER ET AL., J. BIOL. CHEM., vol. 269, 1994, pages 26988
WANG, J., M.D. COLTRERA, A.M. GOWN, CANCER RES., vol. 54, no. 2, 1994, pages 560 - 4
WELSH ET AL., NUCLEIC ACID RES., vol. 20, 1992, pages 4965 - 4970
WESTERMARCK, J. ET AL., CANCER RES., vol. 60, 2000, pages 7156 - 7162
WILHELM S ET AL., MOL CANCER THER, vol. 8, 2009
WILHELM S ET AL.: "Regorafenib (BAY 73-4506): identification of clinically relevant metabolites and their preclinical pharmacology", AMERICAN SOCIETY OF CLINICAL ONCOLOGY (ASCO) ANNUAL MEETING, 2010
WILLIAMS ET AL., ARTHRITIS RHEUM., vol. 33, 1990, pages 533
WOESSNER ET AL., J. BIOL. CHEM., vol. 259, 1984, pages 3633
WOESSNER ET AL., STEROIDS T989, vol. 54, pages 491
WOOD ET AL., CANCER RES., vol. 60, no. 8, 2000, pages 2178 - 2189
WOOD ET AL., CURR. OPIN. PHARMACOL., vol. 1, 2001, pages 370 - 377
WOOLLEY ET AL., ARTHRITIS RHEUM., vol. 20, 1977, pages 1231
WU ET AL., PROCEEDINGS OF THE AMERICAN ASSOCIATION OF CANCER RESEARCH, 2004, pages 45
XIANG ET AL., DEV. BIO., vol. 206, 1999, pages 123 - 141
YEGIN ET AL., LANCET, vol. 349, 1997, pages 170
YU, J., A. MOON, H.R. KIM, BIOCHEM. BIOPHYS. RES. COMMUN., vol. 282, no. 3, 2001, pages 697 - 700
ZOPF DIETER ET AL: "Regorafenib (BAY 73-4506): preclinical pharmacology and clinical identification and quantification of its major metabolites", PROCEEDINGS OF THE ANNUAL MEETING OF THE AMERICAN ASSOCIATION FOR CANCER RESEARCH; 101ST ANNUAL MEETING OF THE AMERICAN-ASSOCIATION-FOR-CANCER-RESEARCH, AMERICAN ASSOCIATION FOR CANCER RESEARCH, US; WASHINGTON, DC, USA, vol. 51, 17 April 2010 (2010-04-17), pages 401 - 402, XP008138087, ISSN: 0197-016X *
ZVELEBIL, CURR. OPIN. ONCOL., ENDOCR. METAB. INVEST. DRUGS, vol. 2, no. 1, 2000, pages 74 - 82

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102643229A (en) * 2012-01-17 2012-08-22 湖南有色凯铂生物药业有限公司 N-((4-chloro-3-trifluoromethyl) phenyl)-N'-((2-fluoro-4-(2-formamyl)-4-pyridyloxy) phenyl) urea and application thereof serving as anticancer medicament
CN102885814A (en) * 2012-01-17 2013-01-23 湖南有色凯铂生物药业有限公司 Compound and use of compound as anti-cancer medicine
WO2013107225A1 (en) * 2012-01-17 2013-07-25 湖南有色凯铂生物药业有限公司 N-((4-chloro-3-trifluoromethyl)phenyl)-n'-(2-fluoro-4-((2-hydroxymethylaminoformyl)-4-pyridyloxo)phenyl)urea and use thereof as anti-cancer drug
CN103508944A (en) * 2012-06-25 2014-01-15 湖南有色凯铂生物药业有限公司 Crystal form A of N-((4-chloro-3-trifluoromethyl)phenyl)-N'-((2-fluoro-4-(2-formamyl)-4-pyridyloxy)phenyl)urea and preparation method thereof
CN103058922A (en) * 2012-09-06 2013-04-24 湖南有色凯铂生物药业有限公司 Crystal form of aromatic urea for antitumor drug and preparation method of crystal form
WO2014036953A1 (en) * 2012-09-06 2014-03-13 湖南有色凯铂生物药业有限公司 Polymorphs of aromatic urea for use in antineoplastic medicament and methods for preparing the polymorphs
CN103058922B (en) * 2012-09-06 2014-07-16 湖南有色凯铂生物药业有限公司 Crystal form of aromatic urea for antitumor drug and preparation method of crystal form

Also Published As

Publication number Publication date
CA2796744A1 (en) 2011-10-20
CN103079567A (en) 2013-05-01

Similar Documents

Publication Publication Date Title
EP1663978B1 (en) Fluoro substituted omega-carboxyaryl diphenyl urea for the treatment and prevention of diseases and conditions
WO2011130728A1 (en) Synthetic metabolites of fluoro substituted omega-carboxyaryl diphenyl urea for the treatment and prevention diseases and conditions
CA2526617C (en) Diaryl ureas with kinase inhibiting activity
EP2111401B1 (en) 4-{4-[({3-tert-butyl-1-[3-(hydroxymethyl) phenyl]-1h-pyrazol-5-yl } carbamoyl)-amino]-3-fluorophenoxy} -n-methylpyridine-2-carboxamide as well as prodrugs and salts thereof for the treatment of cancer
EP1599466A2 (en) 2-oxo-1,3,5-perhydrotriazapine derivatives useful in the treatment of hyper-proliferative, angiogenesis, and inflammatrory disorders
EP2134677B1 (en) 4-{4-[({3-tert-butyl-1-[3-(hydroxymethyl)phenyl]-1h-pyrazol-5-yl}carbamoyl)-amino]-3-chlorophenoxy}-n-methylpyridine-2-carboxamide as an inhibitor of the vegfr kinase for the treatment of cancer
AU2013200394B2 (en) Fluoro substituted omega-carboxyaryl diphenyl urea for the treatment and prevention of diseases and conditions
ZA200600609B (en) Fluoro substituted omega-carboxyaryl diphenyl urea for the treatment and prevention of diseases and conditions
MXPA06000860A (en) Fluoro substituted omega-carboxyaryl diphenyl urea for the treatment and prevention of diseases and conditions
MXPA05012491A (en) Diaryl ureas with kinase inhibiting activity

Legal Events

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

Ref document number: 201180029830.1

Country of ref document: CN

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

Ref document number: 11717412

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2796744

Country of ref document: CA

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 11717412

Country of ref document: EP

Kind code of ref document: A1