US20110086868A1 - Specific inhibitors for vascular endothelial growth factor receptors - Google Patents

Specific inhibitors for vascular endothelial growth factor receptors Download PDF

Info

Publication number
US20110086868A1
US20110086868A1 US12/969,848 US96984810A US2011086868A1 US 20110086868 A1 US20110086868 A1 US 20110086868A1 US 96984810 A US96984810 A US 96984810A US 2011086868 A1 US2011086868 A1 US 2011086868A1
Authority
US
United States
Prior art keywords
alkyl
ring
groups
substituted
phenyl
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US12/969,848
Inventor
Appu Rathinavelu
Sivanesan Dakshanamurthy
Nagarajan Pattabiraman
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Nova Southeastern University
Original Assignee
Nova Southeastern University
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 Nova Southeastern University filed Critical Nova Southeastern University
Priority to US12/969,848 priority Critical patent/US20110086868A1/en
Publication of US20110086868A1 publication Critical patent/US20110086868A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D473/00Heterocyclic compounds containing purine ring systems
    • C07D473/02Heterocyclic compounds containing purine ring systems with oxygen, sulphur, or nitrogen atoms directly attached in positions 2 and 6
    • C07D473/04Heterocyclic compounds containing purine ring systems with oxygen, sulphur, or nitrogen atoms directly attached in positions 2 and 6 two oxygen atoms
    • C07D473/06Heterocyclic compounds containing purine ring systems with oxygen, sulphur, or nitrogen atoms directly attached in positions 2 and 6 two oxygen atoms with radicals containing only hydrogen and carbon atoms, attached in position 1 or 3
    • C07D473/08Heterocyclic compounds containing purine ring systems with oxygen, sulphur, or nitrogen atoms directly attached in positions 2 and 6 two oxygen atoms with radicals containing only hydrogen and carbon atoms, attached in position 1 or 3 with methyl radicals in positions 1 and 3, e.g. theophylline
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/16Amides, e.g. hydroxamic acids
    • A61K31/164Amides, e.g. hydroxamic acids of a carboxylic acid with an aminoalcohol, e.g. ceramides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • 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
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • 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
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • A61K31/52Purines, e.g. adenine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C235/00Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms
    • C07C235/70Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms having carbon atoms of carboxamide groups and doubly-bound oxygen atoms bound to the same carbon skeleton
    • C07C235/72Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms having carbon atoms of carboxamide groups and doubly-bound oxygen atoms bound to the same carbon skeleton with the carbon atoms of the carboxamide groups bound to acyclic carbon atoms
    • C07C235/76Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms having carbon atoms of carboxamide groups and doubly-bound oxygen atoms bound to the same carbon skeleton with the carbon atoms of the carboxamide groups bound to acyclic carbon atoms of an unsaturated carbon skeleton
    • C07C235/78Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms having carbon atoms of carboxamide groups and doubly-bound oxygen atoms bound to the same carbon skeleton with the carbon atoms of the carboxamide groups bound to acyclic carbon atoms of an unsaturated carbon skeleton the carbon skeleton containing rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C251/00Compounds containing nitrogen atoms doubly-bound to a carbon skeleton
    • C07C251/72Hydrazones
    • C07C251/86Hydrazones having doubly-bound carbon atoms of hydrazone groups bound to carbon atoms of six-membered aromatic rings

Definitions

  • This invention relates generally to substituted isoindoles, which are vascular endothelial growth factor receptor (VEGFR) inhibitors, pharmaceutical compositions containing the same, and methods of using the same as anti-tumor agents for treatment of cancer (e.g., breast, colorectal, lung, prostate, and ovarian).
  • cancer e.g., breast, colorectal, lung, prostate, and ovarian.
  • Tumor angiogenesis is one of the essential steps that is required for the growth and metastasis of solid tumors in human.
  • Angiogenesis or neovascularization is the process of generating new capillary of blood vessels derived as extensions from an existing vasculature.
  • the cells that are primarily involved in the process of angiogenesis are endothelial cells that proliferate and organize to form new blood vessels. To achieve the new blood vessel formation, endothelial cells must first escape from their stable location by breaking through the basement membrane, and this degradation is associated with migration of endothelial cells out of the vascular channel toward the angiogenic stimulus.
  • the sub-endothelial basement membrane a dense meshwork of collagen, glycoproteins, and proteoglycans, is proteolytically disrupted to allow formation of new capillaries. Though it is an integral component of normal processes such as reproduction and wound healing, it is known to play an important role in other pathological processes ranging from tumor growth, metastasis to inflammation, and ocular diseases.
  • VEGF vascular endothelial growth factors
  • VEGF receptor-1 (known also as Flt-1, fms-like tyrosine kinase 1)
  • VEGF receptor-2 known as Flk-1/KDR (fetal liver kinase-1/kinase insert domain containing receptor)
  • Flt-4 VEGF receptor-3
  • VEGFR1 has an important role in vasculogenesis and angiogenesis by fixing the ligand-binding domain to the cell membrane and directly regulating the levels of ligands near the cell surface.
  • VEGFR activity Due to the paramount importance of angiogenesis in the control of tumor growth, it was envisioned that the development of anti-angiogenic drugs will potentially lead to novel therapies against all types of cancers.
  • two approaches are available to inhibit VEGFR activity for use in clinical practice: therapeutic monoclonal antibodies (mAbs) target the extracellular region to block dimerization and small-molecule agents block the kinase activity that is required for VEGFR-mediated signal transduction.
  • mAbs therapeutic monoclonal antibodies
  • small-molecule agents block the kinase activity that is required for VEGFR-mediated signal transduction.
  • Genentech has developed an anti-VEGF antibody that has antiangiogenic and antitumorigenic effects in animal models. So far some success has been achieved through this approach in clinical trials of patients with colorectal cancer.
  • small molecules targeted to the kinase domain have shown some success in the clinic.
  • Sugen's SU5416 and SU 6668, Astra-Zeneca's ZD4190, and Novartis' PTK787/ZK2284 are compounds that belong to this category and some are presently being tested in clinical trials. More recently, novel low molecular weight VEGFR antagonists, 5- ⁇ 3-[4-(octadecyloxy)phenyl]propionylamino ⁇ -2,4′-oxydibenzoic acid (VGA1102) and 5-[N-methyl-N-(4-octadecyloxyphenyl)acetyl]amino-2-ethylthiobenzoic acid (VGA1155) that prevent angiogenesis by binding to both VEGF receptor 1 (fms-like tyrosine kinase-1 expressing NIH3T3-cells) and VEGF receptor 2 (KDR/flk-1; VEGF receptor 2 transfected) cells at ⁇ M range have been reported.
  • VGA1102 and VGA1155 appear to be a very specific inhibitors for VEGFR-1 (flt-1) and VEGFR-2 (KDR/flk-1). These compounds do not inhibit the binding of other ligands to their receptors, such as EGF, PDGF, IL-8, PAF, IL-1b, IL-2, IL-4, IL-6, MIPs, TNF-a, and insulin.
  • VEGFR inhibitors are needed as potentially valuable therapeutic agents for the treatment of cancer. It is thus desirable to discover new VEGFR inhibitors.
  • one object of the present invention is to provide novel isoindoles that are useful as VEGFR inhibitors or pharmaceutically acceptable salts thereof.
  • It is another object of the present invention to provide pharmaceutical compositions comprising a pharmaceutically acceptable carrier and a therapeutically effective amount of at least one of the compounds of the present invention or a pharmaceutically acceptable salt thereof.
  • cancer e.g., breast, colorectal, lung, prostate, and ovarian
  • the present invention provides a novel method of treating cancer, comprising: administering a therapeutically effective amount of a compound of formula I or a stereoisomer or pharmaceutically acceptable salt thereof:
  • the present invention provides novel method of treating cancer, comprising: administering a therapeutically effective amount of a compound of formula I or a stereoisomer or pharmaceutically acceptable salt thereof:
  • ring A is selected from phenyl, pyridyl, and pyrimidyl;
  • ring A is substituted with:
  • R is selected from halogen, NO 2 , NR a R b , —CN, C 1-2 haloalkyl, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 1-6 alkoxy, CHO, C(O)C 1-6 alkyl, CO 2 -C 1-6 alkyl, C(O)NR a R b , S(O) 2 NR a R b , S(O) p —C 1-6 alkyl, phenyl, benzyl, and C 3-6 cycloalkyl;
  • L is a linear chain selected from n-propylene, n-butylene, and n-pentylene, wherein
  • L is substituted with 0-2 groups selected from C 1-4 alkyl, phenyl, benzyl, C 3-6 cycloalkyl, and NR a R b ;
  • three of the chain atoms optionally combine with a 2 atom bridge to form a 5 membered ring, the ring consisting of carbon atoms and 0-2 heteroatoms selected from O, N, and S(O) p , wherein the ring has 0-2 ring double bonds and from 0-1 atom of the 2 atom bridge is replaced by as carbonyl group;
  • a carbon atom in ring A that is adjacent to the carbon atom to which linker L is attached can be attached to linker L through a (CH 2 ) 1-2 bridge to form a 5-6 membered ring, wherein optionally 1 methylene of the bridge is replaced by a carbonyl group;
  • R a and R b are independently at each occurrence selected from H and C 1-6 alkyl;
  • NR a R b independently at each occurrence, forms a 5-6 membered cyclic amine consisting of the shown nitrogen atom and 4-5 methylenes;
  • ring B is selected from phenyl, pyridyl, pyrimidyl, and
  • R 1 is independently selected from CO 2 H, halogen, NO 2 , NR a R b , —CN, C 1-2 haloalkyl, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 1-6 alkoxy, CHO, C(O)C 1-6 alkyl, CO 2 —C 1-6 alkyl, C(O)NR a R b , S(O) 2 NR a R b , S(O) p -C 1-6 alkyl, C 3-6 cycloalkyl, phenyl substituted with 0-2 R 2 groups, benzyl substituted with 0-2 R 2 groups, S-phenyl substituted with 0-2 R 2 groups, O-phenyl substituted with 0-2 R 2 groups, and NR a -phenyl substituted with 0-2 R 2 groups;
  • ring B is substituted with 0-1 groups selected from methylene-dioxyl (—OCH 2 O—) and ethylene-dioxyl (—OCH 2 CH 2 O—);
  • R 2 is independently selected from CO 2 H, halogen, NH 2 , C 1-2 haloalkyl, C 1-6 alkyl, C 1-6 alkoxy, C(O)C 1-6 alkyl, and CO 2 C 1-6 alkyl; and,
  • p is independently selected from 0, 1, and 2.
  • the present invention provides novel method, wherein:
  • ring A is selected from phenyl and pyridyl
  • ring A is substituted with 0-3 R groups
  • ring A is substituted with 0-1 methylene-dioxyl (—OCH 2 O—);
  • R is selected from halogen, NO 2 , NR a R b , —CN, CF 3 , C 1-4 alkyl, C 2-4 alkenyl, C 2-4 alkynyl, C 1-4 alkoxy, CHO, C(O)C 1-4 alkyl, CO 2 —C 1-4 alkyl, C(O)NR a R b , S(O) 2 NR a R b , and S(O) p -C 1-6 alkyl;
  • L is a linear chain selected from n-propylene and n-butylene, wherein
  • L is substituted with 0-2 groups selected from C 1-4 alkyl, phenyl, and benzyl;
  • L is 4 atoms in length
  • three of the chain atoms optionally combine with a 2 atom bridge to form a 5 membered ring, the ring consisting of carbon atoms and 0-2 heteroatoms selected from O, N, and S(O) p , wherein the ring has 0-1 ring double bonds;
  • a carbon atom in ring A that is adjacent to the carbon atom to which linker L is attached can be attached to linker L through a (CH 2 ) 1-2 bridge to form a 5-6 membered ring, wherein optionally 1 methylene of the bridge is replaced by a carbonyl group;
  • R a and R b are independently at each occurrence selected from H and C 1-5 alkyl;
  • p is independently selected from 0, 1, and 2.
  • the present invention provides a novel method, wherein the compound is selected from:
  • the present invention provides novel method, wherein the cancer is selected from: breast, colorectal, lung, prostate, and ovarian.
  • the present invention provides novel method, wherein:
  • ring A is selected from phenyl and pyridyl
  • ring A is substituted with 1 group selected from O—C 7-20 alkyl, O—C 7-20 alkenyl, and O—C 7-20 alkynyl;
  • ring A is substituted with 0-1 R groups
  • R is selected from halogen, CF 3 , C 1-4 alkyl, C 2-4 alkenyl, C 2-4 alkynyl, C(O)C 1-4 alkyl, and CO 2 —C 1-4 alkyl;
  • L is a linear chain selected from n-propylene, n-butylene, and n-pentylene, wherein
  • L is substituted with 0-2 groups selected from C 1-4 alkyl, C 3-6 cycloalkyl, and NR a R b ;
  • R a and R b are independently at each occurrence selected from H and C 1-6 alkyl;
  • NR a R b forms a 5-6 membered cyclic amine consisting of the shown nitrogen atom and 4-5 methylenes;
  • ring B is selected from phenyl and pyridyl, wherein the phenyl and pyridyl rings are substituted with 1-2 R 1 groups;
  • R 1 is independently selected from CO 2 H, halogen, NR a R b , CF 3 , C 1-4 alkyl, C 1-6 alkoxy, C 3-6 cycloalkyl, S-phenyl substituted with 0-2 R 2 groups, O-phenyl substituted with 0-2 R 2 groups, and NR a -phenyl substituted with 0-2 R 2 groups;
  • R 2 is independently selected from CO 2 H, halogen, CF 3 , C 1-4 alkyl, and C 1-4 alkoxy;
  • p is independently selected from 0, 1, and 2.
  • the present invention provides novel method, wherein the compound is selected from:
  • the present invention provides novel method, wherein the cancer is selected from: breast, colorectal, lung, prostate, and ovarian.
  • the present invention provides novel compound of formula I or a stereoisomer or pharmaceutically acceptable salt thereof:
  • ring A is selected from phenyl, pyridyl, and pyrimidyl;
  • ring A is substituted with 0-1 groups selected from O—C 7-20 alkyl, O—C 7-20 alkenyl, and O—C 7-20 alkynyl;
  • ring A is substituted with 0-3 groups selected from halogen, NO 2 , NR a R b , —CN, C 1-2 haloalkyl, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 1-6 alkoxy, CHO, C(O)C 1-6 alkyl, CO 2 —C 1-6 alkyl, C(O)NR a R b , S(O) 2 NR a R b , S(O) p —C 1-6 alkyl, phenyl, benzyl, and C 3-6 cycloalkyl;
  • ring A is substituted with 0-1 groups selected from methylene-dioxyl (—OCH 2 O—) and ethylene-dioxyl (—OCH 2 CH 2 O—);
  • L is a linear chain selected from n-propylene, n-butylene, and n-pentylene, wherein
  • L is substituted with 0-2 groups selected from C 1-4 alkyl, phenyl, benzyl, C 3-6 cycloalkyl, and NR a R b ;
  • three of the chain atoms optionally combine with a 2 atom bridge to form a 5 membered ring, the ring consisting of carbon atoms and 0-2 heteroatoms selected from O, N, and S(O) p , wherein the ring has 0-2 ring double bonds and from 0-1 atom of the 2 atom bridge is replaced by as carbonyl group;
  • a carbon atom in ring A that is adjacent to the carbon atom to which linker L is attached can be attached to linker L through a (CH 2 ) 1-2 bridge to form a 5-6 membered ring, wherein optionally 1 methylene of the bridge is replaced by a carbonyl group;
  • R a and R b are independently at each occurrence selected from H and C 1-6 alkyl;
  • NR a R b forms a 5-6 membered cyclic amine consisting of the shown nitrogen atom and 4-5 methylenes;
  • ring B is selected from phenyl, pyridyl, pyrimidyl, and
  • R is independently selected from CO 2 H, halogen, NO 2 , NR a R b , —CN, C 1-2 haloalkyl, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 1-6 alkoxy, CHO, C(O)C 1-6 alkyl, CO 2 —C 1-6 alkyl, C(O)NR a R b , S(O) 2 NR a R b , S(O) p —C 1-6 alkyl, C 3-6 cycloalkyl, phenyl substituted with 0-2 R 1 groups, benzyl substituted with 0-2 R 1 groups, S-phenyl substituted with 0-2 R 1 groups, O-phenyl substituted with 0-2 R 1 groups, and NR a -phenyl substituted with 0-2 R 1 groups;
  • ring B is substituted with 0-1 groups selected from methylene-dioxyl (—OCH 2 O—) and ethylene-dioxyl (—OCH 2 CH 2 O—);
  • R 1 is independently selected from CO 2 H, halogen, NH 2 , C 1-2 haloalkyl, C 1-6 alkyl, C 1-6 alkoxy, C(O)C 1-6 alkyl, and CO 2 C 1-6 alkyl; and,
  • p is independently selected from 0, 1, and 2;
  • the present invention provides novel compound, wherein the compound is selected from Table 2 or a stereoisomer or pharmaceutically acceptable salt thereof.
  • the present invention provides a novel method of treating cancer, wherein the cancer is selected from: breast, colorectal, lung, prostate and ovarian.
  • the present invention provides novel pharmaceutical compositions, comprising: a pharmaceutically acceptable carrier and a therapeutically effective amount of a compound of the present invention or a pharmaceutically acceptable salt form thereof.
  • the present invention provides a novel method for treating cancer (e.g., breast, colorectal, lung, prostate and ovarian), comprising: administering to a patient in need thereof a therapeutically effective amount of a compound of the present invention or a pharmaceutically acceptable salt form thereof.
  • cancer e.g., breast, colorectal, lung, prostate and ovarian
  • the present invention provides a compound of the present invention for use in therapy.
  • the present invention provides the use of a compound of the present invention for the manufacture of a medicament for the treatment of cancer.
  • the compounds herein described may have asymmetric centers.
  • Compounds of the present invention containing an asymmetrically substituted atom may be isolated in optically active or racemic forms. It is well known in the art how to prepare optically active forms, such as by resolution of racemic forms or by synthesis from optically active starting materials. Many geometric isomers of olefins, C ⁇ N double bonds, and the like can also be present in the compounds described herein, and all such stable isomers are contemplated in the present invention. C is and trans geometric isomers of the compounds of the present invention are described and may be isolated as a mixture of isomers or as separated isomeric forms.
  • Examples of the molecular weight of compounds of the present invention include those that are (a) less than 500, 550, 600, 650, 700, 750, 800, 850, 900, 950, or 1000 grams per mole, (b) less than 950 grams per mole, (c) less than 850 grams per mole, and, (d) less than 750 grams per mole.
  • “Substituted” means that any one or more hydrogens on the designated atom is replaced with a selection from the indicated group, provided that the designated atom's normal valency is not exceeded, and that the substitution results in a stable compound.
  • a substituent is keto (i.e., ⁇ O)
  • 2 hydrogens on the atom are replaced.
  • Keto substituents are not present on aromatic moieties.
  • the present invention includes all isotopes of atoms occurring in the present compounds.
  • Isotopes include those atoms having the same atomic number but different mass numbers.
  • isotopes of hydrogen include tritium and deuterium.
  • isotopes of carbon include C-13 and C-14.
  • any variable e.g., R 4
  • its definition at each occurrence is independent of its definition at every other occurrence.
  • R 4 at each occurrence is selected independently from the definition of R 4 .
  • combinations of substituents and/or variables are permissible only if such combinations result in stable compounds.
  • Alkyl includes both branched and straight-chain saturated aliphatic hydrocarbon groups having the specified number of carbon atoms.
  • C 1-6 alkyl includes C 1 , C 2 , C 3 , C 4 , C 5 , and C 6 alkyl groups.
  • Examples of alkyl include methyl, ethyl, n-propyl, i-propyl, n-butyl, s-butyl, t-butyl, n-pentyl, and s-pentyl.
  • haloalkyl include trifluoromethyl, trichloromethyl, pentafluoroethyl, and pentachloroethyl.
  • Alkoxy represents an alkyl group as defined above with the indicated number of carbon atoms attached through an oxygen bridge.
  • C 1-6 alkoxy includes C 1 , C 2 , C 3 , C 4 , C 5 , and C 6 alkoxy groups.
  • alkoxy examples include methoxy, ethoxy, n-propoxy, i-propoxy, n-butoxy, s-butoxy, t-butoxy, n-pentoxy, and s-pentoxy.
  • Cycloalkyl includes saturated ring groups, such as cyclopropyl, cyclobutyl, or cyclopentyl.
  • C 3-7 cycloalkyl includes C 3 , C 4 , C 5 , C 6 , and C 7 cycloalkyl groups.
  • Alkenyl includes hydrocarbon chains of either straight or branched configuration and one or more unsaturated carbon-carbon bonds that may occur in any stable point along the chain, such as ethenyl and propenyl.
  • C 2-6 alkenyl includes C 2 , C 3 , C 4 , C 5 , and C 6 alkenyl groups.
  • “Alkynyl” includes hydrocarbon chains of either straight or branched configuration and one or more triple carbon-carbon bonds that may occur in any stable point along the chain, such as ethynyl and propynyl.
  • C 2-6 Alkynyl includes C 2 , C 3 , C 4 , C 5 , and C 6 alkynyl groups.
  • Halo or “halogen” refers to fluoro, chloro, bromo, and iodo; and “counterion” is used to represent a small, negatively charged species such as chloride, bromide, hydroxide, acetate, and sulfate.
  • Carbocycle means any stable 3, 4, 5, 6, or 7-membered monocyclic or bicyclic or 7, 8, 9, 10, 11, 12, or 13-membered bicyclic or tricyclic, any of which may be saturated, partially unsaturated, or aromatic.
  • Examples of such carbocycles include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, adamantyl, cyclooctyl, [3.3.0]bicyclooctane, [4.3.0]bicyclononane, [4.4.0]bicyclodecane, [2.2.2]bicyclooctane, fluorenyl, phenyl, naphthyl, indanyl, adamantyl, and tetrahydronaphthyl.
  • Heterocycle means a stable 5, 6, or 7-membered monocyclic or bicyclic or 7, 8, 9, or 10-membered bicyclic heterocyclic ring which is saturated, partially unsaturated or unsaturated (aromatic), and which consists of carbon atoms and 1, 2, 3, or 4 heteroatoms independently selected from the group consisting of N, O and S and including any bicyclic group in which any of the above-defined heterocyclic rings is fused to a benzene ring.
  • the nitrogen atom can be N, NH, or N-substituent depending on the ring and whether and the location of any substituent.
  • the nitrogen and sulfur heteroatoms may optionally be, oxidized.
  • the heterocyclic ring may be attached to its pendant group at any heteroatom or carbon atom that results in a stable structure.
  • the heterocyclic rings described herein may be substituted on carbon or on a nitrogen atom if the resulting compound is stable.
  • a nitrogen in the heterocycle may optionally be quaternized. When the total number of S and O atoms in the heterocycle exceeds 1, then these heteroatoms are not adjacent to one another. In a typical heterocycle, the total number of S and O atoms in the heterocycle is not more than 1.
  • Heteroaryl means a stable 5, 6, or 7-membered monocyclic or bicyclic or 7, 8, 9, or 10-membered bicyclic heterocyclic aromatic ring which consists of carbon atoms and 1, 2, 3, or 4 heteroatoms independently selected from the group consisting of N, O and S.
  • the nitrogen atom can be N, NH, or N-substituent depending on the ring and whether and the location of any substituent.
  • the total number of S and O atoms in the aromatic heterocycle is not more than 1. If the heteroaryl contains more than one ring, only one of the rings need be aromatic.
  • heterocycles and heteroaryls include acridinyl, azocinyl, benzimidazolyl, benzofuranyl, benzothiofuranyl, benzothiophenyl, benzoxazolyl, benzoxazolinyl, benzthiazolyl, benztriazolyl, benztetrazolyl, benzisoxazolyl, benzisothiazolyl, benzimidazolinyl, carbazolyl, 4aH-carbazolyl, carbolinyl, chromanyl, chromenyl, cinnolinyl, decahydroquinolinyl, 2H,6H-1,5,2-dithiazinyl, dihydrofuro[2,3-b]tetrahydrofuran, furanyl, furazanyl, imidazolidinyl, imidazolinyl, imidazolyl, 1H-indazolyl, indolenyl
  • phrases “pharmaceutically acceptable” refers to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
  • “Pharmaceutically acceptable salt” refers to derivatives of the disclosed compounds wherein the parent compound is modified by making acid or base salts thereof.
  • Examples of pharmaceutically acceptable salts include mineral or organic acid salts of basic residues such as amines; alkali or organic salts of acidic residues such as carboxylic acids; and the like.
  • the pharmaceutically acceptable salts include the conventional non-toxic salts or the quaternary ammonium salts of the parent compound formed, for example, from non-toxic inorganic or organic acids.
  • such conventional non-toxic salts include those derived from inorganic acids such as hydrochloric, hydrobromic, sulfuric, sulfamic, phosphoric, nitric and the like; and the salts prepared from organic acids such as acetic, propionic, succinic, glycolic, stearic, lactic, malic, tartaric, citric, ascorbic, pamoic, maleic, hydroxymaleic, phenylacetic, glutamic, benzoic, salicylic, sulfanilic, 2-acetoxybenzoic, fumaric, toluenesulfonic, methanesulfonic, ethane disulfonic, oxalic, isethionic, and the like.
  • inorganic acids such as hydrochloric, hydrobromic, sulfuric, sulfamic, phosphoric, nitric and the like
  • organic acids such as acetic, propionic, succinic, glycolic, stearic, lactic,
  • the pharmaceutically acceptable salts of the present invention can be synthesized from the parent compound that contains a basic or acidic moiety by conventional chemical methods.
  • such salts can be prepared by reacting the free acid or base forms of these compounds with a stoichiometric amount of the appropriate base or acid in water or in an organic solvent, or in a mixture of the two; generally, non-aqueous media like ether, ethyl acetate, ethanol, isopropanol, or acetonitrile are preferred.
  • Lists of suitable salts are found in Remington's Pharmaceutical Sciences, 17th ed., Mack Publishing Company, Easton, Pa., 1985, p. 1418, the disclosure of which is hereby incorporated by reference.
  • prodrugs are known to enhance numerous desirable qualities of pharmaceuticals (e.g., solubility, bioavailability, manufacturing, etc.) the compounds of the present invention may be delivered in prodrug form.
  • the present invention is intended to cover prodrugs of the presently claimed compounds, methods of delivering the same and compositions containing the same.
  • “Prodrugs” are intended to include any covalently bonded carriers that release an active parent drug of the present invention in vivo when such prodrug is administered to a mammalian subject.
  • Prodrugs of the present invention are prepared by modifying functional groups present in the compound in such a way that the modifications are cleaved, either in routine manipulation or in vivo, to the parent compound.
  • Prodrugs include compounds of the present invention wherein a hydroxy, amino, or sulfhydryl group is bonded to any group that, when the prodrug of the present invention is administered to a mammalian subject, it cleaves to form a free hydroxyl, free amino, or free sulfhydryl group, respectively.
  • prodrugs include acetate, formate and benzoate derivatives of alcohol and amine functional groups in the compounds of the present invention.
  • “Stable compound” and “stable structure” mean a compound that is sufficiently robust to survive isolation to a useful degree of purity from a reaction mixture, and formulation into an efficacious therapeutic agent.
  • “Substituted” means that one or more hydrogens on the atom indicated in the expression using “substituted” is replaced with a selection from the indicated group(s), provided that the indicated atom's normal valency is not exceeded, and that the substitution results in a stable compound.
  • a substituent is keto (i.e., ⁇ O) group, then 2 hydrogens on the atom are replaced.
  • Treating” or “treatment” cover the treatment of a disease-state in a mammal, particularly in a human, and include: (a) preventing the disease-state from occurring in a mammal, in particular, when such mammal is predisposed to the disease-state but has not yet been diagnosed as having it; (b) inhibiting the disease-state, i.e., arresting it development; and/or (c) relieving the disease-state, i.e., causing regression of the disease state.
  • “Therapeutically effective amount” includes an amount of a compound of the present invention that is effective when administered alone or in combination to inhibit a VEGFR. “Therapeutically effective amount” includes an amount of the combination of compounds claimed that is effective to inhibit a VEGFR.
  • the combination of compounds is preferably a synergistic combination. Synergy, as described, for example, by Chou and Talalay, Adv. Enzyme Regul. 1984, 22:27-55, occurs when the effect of the compounds when administered in combination is greater than the additive effect of the compounds when administered alone as a single agent. In general, a synergistic effect is most clearly demonstrated at sub-optimal concentrations of the compounds. Synergy can be in terms of lower cytotoxicity, increased antiviral effect, or some other beneficial effect of the combination compared with the individual components.
  • the compounds of the present invention can be prepared in a number of ways known to one skilled in the art of organic synthesis.
  • the compounds of the present invention can be synthesized using the methods described below, together with synthetic methods known in the art of synthetic organic chemistry, or by variations thereon as appreciated by those skilled in the art.
  • the reactions are performed in a solvent appropriate to the reagents and materials employed and suitable for the transformations being effected. It will be understood by those skilled in the art of organic synthesis that the functionality present on the molecule should be consistent with the transformations proposed. This will sometimes require a judgment to modify the order of the synthetic steps or to select one particular process scheme over another in order to obtain a desired compound of the invention.
  • the compounds of this invention are expected to be useful as antitumor agents anticoagulants for the treatment of cancer (e.g., breast, colorectal, lung, prostate and ovarian).
  • cancer e.g., breast, colorectal, lung, prostate and ovarian.
  • the compounds of the present invention can be administered alone or in combination with one or more additional therapeutic agents. These include additional compounds of the present invention, other antitumor agents, or other agents that may be beneficially administered with compounds of the present invention.
  • administering means that a compound of the present invention and one or more additional therapeutic agents are administered concurrently to the mammal being treated.
  • each component may be administered at the same time or sequentially in any order at different points in time.
  • each component may be administered separately but sufficiently closely in time so as to provide the desired therapeutic effect.
  • Administration of the compounds of the present invention in combination with such additional therapeutic agent may afford an efficacy advantage over the compounds and agents alone, and may do so while permitting the use of lower doses of each.
  • a lower dosage minimizes the potential of side effects, thereby providing an increased margin of safety.
  • the compounds of this invention can be administered in such oral dosage forms as tablets, capsules (each of which includes sustained release or timed release formulations), pills, powders, granules, elixirs, tinctures, suspensions, syrups, and emulsions. They may also be administered in intravenous (bolus or infusion), intraperitoneal, subcutaneous, or intramuscular form, all using dosage forms well known to those of ordinary skill in the pharmaceutical arts. They can be administered alone, but generally will be administered with a pharmaceutical carrier selected on the basis of the chosen route of administration and standard pharmaceutical practice.
  • the dosage regimen for the compounds of the present invention will, of course, vary depending upon known factors, such as the pharmacodynamic characteristics of the particular agent and its mode and route of administration; the species, age, sex, health, medical condition, and weight of the recipient; the nature and extent of the symptoms; the kind of concurrent treatment; the frequency of treatment; the route of administration, the renal and hepatic function of the patient, and the effect desired.
  • a physician or veterinarian can determine and prescribe the effective amount of the drug required to prevent, counter, or arrest the progress of the cancer.
  • Examples of the daily oral dosage of each active ingredient when used for the indicated effects, can range between (a) 0.001 to 1000 mg/kg of body weight, (b) 0.01 to 100 mg/kg of body weight per day, and (c) 1.0 to 20 mg/kg/day.
  • Intravenously doses can range from 1 to about 10 mg/kg/minute during a constant rate infusion (the dosage can also be calculated using the body's surface area).
  • Compounds of this invention may be administered in a single daily dose, or the total daily dosage may be administered in divided doses of two, three, or four times daily.
  • Compounds of this invention can be administered in intranasal form via topical use of suitable intranasal vehicles, or via transdermal routes, using transdermal skin patches.
  • suitable intranasal vehicles or via transdermal routes, using transdermal skin patches.
  • the dosage administration will, of course, be continuous rather than intermittent throughout the dosage regimen.
  • the compounds are typically administered in admixture with suitable pharmaceutical diluents, excipients, or carriers (collectively referred to herein as pharmaceutical carriers) suitably selected with respect to the intended form of administration, that is, oral tablets, capsules, elixirs, syrups and the like, and consistent with conventional pharmaceutical practices.
  • suitable pharmaceutical diluents, excipients, or carriers suitably selected with respect to the intended form of administration, that is, oral tablets, capsules, elixirs, syrups and the like, and consistent with conventional pharmaceutical practices.
  • the active drug component can be combined with an oral, non-toxic, pharmaceutically acceptable, inert carrier such as lactose, starch, sucrose, glucose, methyl callulose, magnesium stearate, dicalcium phosphate, calcium sulfate, mannitol, sorbitol and the like; for oral administration in liquid form, the oral drug components can be combined with any oral, non-toxic, pharmaceutically acceptable inert carrier such as ethanol, glycerol, water, and the like.
  • suitable binders, lubricants, disintegrating agents, and coloring agents can also be incorporated into the mixture.
  • Suitable binders include starch, gelatin, natural sugars such as glucose or beta-lactose, corn sweeteners, natural and synthetic gums such as acacia, tragacanth, or sodium alginate, carboxymethylcellulose, polyethylene glycol, waxes, and the like.
  • Lubricants used in these dosage forms include sodium oleate, sodium stearate, magnesium stearate, sodium benzoate, sodium acetate, sodium chloride, and the like.
  • Disintegrators include, without limitation, starch, methyl cellulose, agar, bentonite, xanthan gum, and the like.
  • the compounds of the present invention can also be administered in the form of liposome delivery systems, such as small unilamellar vesicles, large unilamellar vesicles, and multilamellar vesicles.
  • Liposomes can be formed from a variety of phospholipids, such as cholesterol, stearylamine, or phosphatidylcholines.
  • Compounds of the present invention may also be coupled with soluble polymers as targetable drug carriers.
  • soluble polymers can include polyvinylpyrrolidone, pyran copolymer, polyhydroxypropylmethacrylamide-phenol, polyhydroxyethylaspartamidephenol, or polyethyleneoxide-polylysine substituted with palmitoyl residues.
  • the compounds of the present invention may be coupled to a class of biodegradable polymers useful in achieving controlled release of a drug, for example, polylactic acid, polyglycolic acid, copolymers of polylactic and polyglycolic acid, polyepsilon caprolactone, polyhydroxy butyric acid, polyorthoesters, polyacetals, polydihydropyrans, polycyanoacylates, and crosslinked or amphipathic block copolymers of hydrogels.
  • a drug for example, polylactic acid, polyglycolic acid, copolymers of polylactic and polyglycolic acid, polyepsilon caprolactone, polyhydroxy butyric acid, polyorthoesters, polyacetals, polydihydropyrans, polycyanoacylates, and crosslinked or amphipathic block copolymers of hydrogels.
  • Dosage forms suitable for administration may contain from about 1 milligram to about 100 milligrams of active ingredient per dosage unit.
  • the active ingredient will ordinarily be present in an amount of about 0.5-95% by weight based on the total weight of the composition.
  • Gelatin capsules may contain the active ingredient and powdered carriers, such as lactose, starch, cellulose derivatives, magnesium stearate, stearic acid, and the like. Similar diluents can be used to make compressed tablets. Both tablets and capsules can be manufactured as sustained release products to provide for continuous release of medication over a period of hours. Compressed tablets can be sugar coated or film coated to mask any unpleasant taste and protect the tablet from the atmosphere, or enteric coated for selective disintegration in the gastrointestinal tract.
  • powdered carriers such as lactose, starch, cellulose derivatives, magnesium stearate, stearic acid, and the like. Similar diluents can be used to make compressed tablets. Both tablets and capsules can be manufactured as sustained release products to provide for continuous release of medication over a period of hours. Compressed tablets can be sugar coated or film coated to mask any unpleasant taste and protect the tablet from the atmosphere, or enteric coated for selective disintegration in the gastrointestinal tract.
  • Liquid dosage forms for oral administration can contain coloring and flavoring to increase patient acceptance.
  • parenteral solutions In general, water, a suitable oil, saline, aqueous dextrose (glucose), and related sugar solutions and glycols such as propylene glycol or polyethylene glycols are suitable carriers for parenteral solutions.
  • Solutions for parenteral administration preferably contain a water soluble salt of the active ingredient, suitable stabilizing agents, and if necessary, buffer substances.
  • Antioxidizing agents such as sodium bisulfate, sodium sulfite, or ascorbic acid, either alone or combined, are suitable stabilizing agents.
  • citric acid and its salts and sodium EDTA are also used.
  • parenteral solutions can contain preservatives, such as benzalkonium chloride, methyl-or propyl-paraben, and chlorobutanol.
  • Suitable pharmaceutical carriers are described in Remington's the Science and Practice of Pharmacy, Lippincott Williams & Wilkins, a standard reference text in this field.
  • Representative useful pharmaceutical dosage-forms for administration of the compounds of this invention can be illustrated as follows:
  • a large number of unit capsules can be prepared by filling standard two-piece hard gelatin capsules each with 100 milligrams of powdered active ingredient, 150 milligrams of lactose, 50 milligrams of cellulose, and 6 milligrams magnesium stearate.
  • a mixture of active ingredient in a digestible oil such as soybean oil, cottonseed oil or olive oil may be prepared and injected by means of a positive displacement pump into gelatin to form soft gelatin capsules containing 100 milligrams of the active ingredient.
  • the capsules should be washed and dried.
  • Tablets may be prepared by conventional procedures so that the dosage unit is 100 milligrams of active ingredient, 0.2 milligrams of colloidal silicon dioxide, 5 milligrams of magnesium stearate, 275 milligrams of microcrystalline cellulose, 11 milligrams of starch and 98.8 milligrams of lactose. Appropriate coatings may be applied to increase palatability or delay absorption.
  • a parenteral composition suitable for administration by injection may be prepared by stirring 1.5% by weight of active ingredient in 10% by volume propylene glycol and water. The solution should be made isotonic with sodium chloride and sterilized.
  • An aqueous suspension can be prepared for oral administration so that each 5 mL contain 100 mg of finely divided active ingredient, 200 mg of sodium carboxymethyl cellulose, 5 mg of sodium benzoate, 1.0 g of sorbitol solution, U.S.P, and 0.025 mL of vanillin.
  • a daily dosage may be about 0.1 to 100 milligrams of the compound of the present invention and about 1 to 7.5 milligrams of the second agent, per kilogram of patient body weight.
  • the compounds of this invention generally may be present in an amount of about 5 to 10 milligrams per dosage unit, and the second anti-coagulant in an amount of about 1 to 5 milligrams per dosage unit.
  • Human umbilical vein endothelial cell was purchased from Cambrex Co. (East Rutherford, N.J.) and maintained in EGM (Endothelial Growth Medium) supplemented with 2% FBS, 0.1% EGF, 0.1% Hydrocortisone, 0.1% GA-1000 and 0.4% BBE.
  • the morphogenesis assay on Matrigel was performed according to the manufacturer's instructions (Chemicon International).
  • the ECMatrixTM kit consists of laminin, collagen type IV, heparan sulfate, proteoglycans, entactin and nidogen. It also contains various growth factors (TGF- ⁇ , FGF) and proteolytic enzymes (plasminogen, tPA, MMPs) that are normally produced in EHS tumors.
  • the incubation condition was optimized for maximal tube-formation as follows: 50 ⁇ l of EC MatrixTM was suitably diluted in the ratio 9:1 with 10 ⁇ diluent buffer and used for coating the 96-well plate. The coated plates were incubated at 37° C.
  • the HUVECs that were cultured for 24 hours in EGM with 2% FBS was trypsinised and re-suspended in the growth media and the cells were counted.
  • the HUVECs were plated at 10 4 cells/well in the absence or in the presence of different VEGFR inhibitors (1 mM and 10 mM).
  • the cell three-dimensional organization was examined under an inverted photomicroscope. Each treatment was performed in triplicate.
  • Activated endothelial cells form cellular networks (mesh like structures) resembling capillary tubes sprouting into the stromal space.
  • the formation of these cellular networks is a dynamic process, which started migration and alignment of cells followed by the development of capillary tubes like structures, sprouting of new branches and finally the formation of the cellular networks.
  • this in vitro angiogenesis kit is designed only as a qualitative assay, we have made an attempt to quantitate the degree of angiogenesis using a scoring method. The scoring was based on the extent of the cellular networks that were observed as follows:
  • Table 1 shows the results for Examples 1-6, which are commercially available.
  • the EC 50 value for anti-antigenic effect of Example 1 was estimated to be 0.25 ⁇ M.
  • PTKs Protein Tyrosine Kinases
  • Phosphorylation of proteins by PTKs is essential for the regulation of these biological mechanisms and defects in these pathways may result in a number of human diseases, including cancer.
  • the important aspect of these enzymes in cellular regulation is accentuated by the fact that for a large number of kinases a corresponding viral oncogene product has been identified.
  • Protein tyrosine kinase activity is often associated with membrane receptor protein tyrosine kinases (e.g.
  • EGF-, PDGF-, CFS-, IGF-1 and insulin receptor EGF-, PDGF-, CFS-, IGF-1 and insulin receptor
  • soluble non-receptor tyrosine kinases e.g. p60c-Src, yes, lck, lyn, fyn.
  • Assaying of PTK activity allows for purification and characterization of protein tyrosine kinases, elucidation of their biological functions as well as aiding in development of specific PTK inhibitors.
  • CHEMICON's non-radioactive Tyrosine Kinase Activity Assay provides a simple, convenient and specific method for quantification and comparative determination of a wide range of PTKs.
  • the non-radioactive Tyrosine Kinase Activity Assay Kit consists of a synthetic Biotinylated Peptide Substrate, a Biotinylated Phosphopeptide, purified Phosphotyrosine specific monoclonal antibody conjugated to horseradish peroxidase (HRP) and other components required to perform 96 ELISA-based assays.
  • the synthetic Biotinylated Substrate, poly [Glu:Tyr], 4:1 contains multiple tyrosine residues and can be phosphorylated by a wide range of PTKs. After quenching the enzyme reaction with an inhibitor, both the phosphorylated and dephosphorylated substrates are immobilized by binding to the streptavidin-coated plate.
  • the fraction of phosphorylated substrate is visualized using a phosphotyrosine monoclonal antibody conjugated to HRP and an ensuing chromagenic substrate reaction.
  • the quantity of phosphate incorporated into the tyrosine kinase substrate is determined utilizing the phosphopeptide standard curve.
  • the assay mixture and start the reaction was be prepared by adding 10 ⁇ l of ATP/MgCl 2 Solution.
  • the reaction mixture was pre-incubate at 30° C.
  • the reaction time was dependent on the individual tyrosine kinase and was standardized.
  • the enzyme reaction was terminated by adding 10 ⁇ l of kinase inhibitor, such as 120 mM EDTA.
  • reaction mixture was transferred to Streptavidin-coated strip wells and incubated at 37° C. for 30 minutes.
  • the wells were washed four times using 1 ⁇ wash buffer and then 200 ⁇ l of blocking buffer solution will be added to each well and incubate at 37° C. for 30 minutes.
  • 100 ⁇ L of diluted mouse anti-Phosphotyrosine HRP conjugate was added to each well and incubated at room temperature for 60 minutes.
  • the wells were washed four times using 1 ⁇ Wash Buffer and then 100 ⁇ l of TMB substrate solution were added and the plates are incubate at room temperature for 5-15 minutes.
  • the assay reaction would be terminated by the addition of 100 ⁇ l of stop solution and finally, the absorbance was measured on a standard microplate reader at 450 nm.
  • Example 1 After testing the anti-antigenic effects of Example 1, the ability of this compound to inhibit the VEGFR linked tyrosine kinase activity was measured.
  • the VEGFR activity was measured by specifically immunoprecipitating the cell lysate using VEGFR specific antibody (Upstate, VA) and measuring its activity using the PTK assay.
  • Example 1 was able to inhibit the VEGFR associated tyrosine kinase activity, and the level of inhibition increased as the concentration of the compound increased.
  • the IC 50 for inhibiting VEGFR kinase was determined to be 0.4 ⁇ M.
  • Example 1 In addition to inhibiting the angiogenesis under in vitro conditions Example 1 also produced cytotoxicity towards GI-101A and MCF-7, breast carcinoma cell lines.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Epidemiology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The present application describes isoindoles and derivatives thereof, or pharmaceutically acceptable salt forms thereof, which are useful inhibitors of VEGFR.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • This application claims the priority benefit of U.S. Provisional Application No. 60/826,390 filed Sep. 21, 2006, now pending, which is incorporated herein by reference.
  • FIELD OF THE INVENTION
  • This invention relates generally to substituted isoindoles, which are vascular endothelial growth factor receptor (VEGFR) inhibitors, pharmaceutical compositions containing the same, and methods of using the same as anti-tumor agents for treatment of cancer (e.g., breast, colorectal, lung, prostate, and ovarian).
  • BACKGROUND OF THE INVENTION
  • Tumor angiogenesis is one of the essential steps that is required for the growth and metastasis of solid tumors in human. Angiogenesis or neovascularization is the process of generating new capillary of blood vessels derived as extensions from an existing vasculature. The cells that are primarily involved in the process of angiogenesis are endothelial cells that proliferate and organize to form new blood vessels. To achieve the new blood vessel formation, endothelial cells must first escape from their stable location by breaking through the basement membrane, and this degradation is associated with migration of endothelial cells out of the vascular channel toward the angiogenic stimulus. During this process, the sub-endothelial basement membrane, a dense meshwork of collagen, glycoproteins, and proteoglycans, is proteolytically disrupted to allow formation of new capillaries. Though it is an integral component of normal processes such as reproduction and wound healing, it is known to play an important role in other pathological processes ranging from tumor growth, metastasis to inflammation, and ocular diseases.
  • The angiogenesis process is strongly supported by one of an important series of endothelial cell mitogens called VEGF. The vascular endothelial growth factors (VEGF) play a crucial role in neovascularization of solid tumors. The expression of VEGF has been shown to correlate with the density of micro vessels in various tumors and exhibit higher metastatic ability. Several members of the VEGF family (i.e., A, B, C, and D) and several VEGF receptors: VEGF receptor-1 (known also as Flt-1, fms-like tyrosine kinase 1), VEGF receptor-2 known as Flk-1/KDR (fetal liver kinase-1/kinase insert domain containing receptor) and VEGF receptor-3 (known as Flt-4) have been identified. All of them have seven immunoglobulin homology domains in their extracellular part and an intracellular tyrosine kinase signaling domain split by a kinase insert. By binding to one or more of these receptors, VEGF induces angiogenesis as well as permeabilization of blood vessels and thereby plays a central role in the regulation of vasculogenesis. Recently, it has been demonstrated that the extra cellular domain of VEGFR1 has an important role in vasculogenesis and angiogenesis by fixing the ligand-binding domain to the cell membrane and directly regulating the levels of ligands near the cell surface.
  • Due to the paramount importance of angiogenesis in the control of tumor growth, it was envisioned that the development of anti-angiogenic drugs will potentially lead to novel therapies against all types of cancers. At present, two approaches are available to inhibit VEGFR activity for use in clinical practice: therapeutic monoclonal antibodies (mAbs) target the extracellular region to block dimerization and small-molecule agents block the kinase activity that is required for VEGFR-mediated signal transduction. For example, Genentech has developed an anti-VEGF antibody that has antiangiogenic and antitumorigenic effects in animal models. So far some success has been achieved through this approach in clinical trials of patients with colorectal cancer. Also, small molecules targeted to the kinase domain have shown some success in the clinic. Sugen's SU5416 and SU 6668, Astra-Zeneca's ZD4190, and Novartis' PTK787/ZK2284 are compounds that belong to this category and some are presently being tested in clinical trials. More recently, novel low molecular weight VEGFR antagonists, 5-{3-[4-(octadecyloxy)phenyl]propionylamino}-2,4′-oxydibenzoic acid (VGA1102) and 5-[N-methyl-N-(4-octadecyloxyphenyl)acetyl]amino-2-ethylthiobenzoic acid (VGA1155) that prevent angiogenesis by binding to both VEGF receptor 1 (fms-like tyrosine kinase-1 expressing NIH3T3-cells) and VEGF receptor 2 (KDR/flk-1; VEGF receptor 2 transfected) cells at μM range have been reported. VGA1102 and VGA1155 (VGA compounds) appear to be a very specific inhibitors for VEGFR-1 (flt-1) and VEGFR-2 (KDR/flk-1). These compounds do not inhibit the binding of other ligands to their receptors, such as EGF, PDGF, IL-8, PAF, IL-1b, IL-2, IL-4, IL-6, MIPs, TNF-a, and insulin.
  • Therefore, efficacious and specific inhibitors of VEGFR are needed as potentially valuable therapeutic agents for the treatment of cancer. It is thus desirable to discover new VEGFR inhibitors.
  • SUMMARY OF THE INVENTION
  • Accordingly, one object of the present invention is to provide novel isoindoles that are useful as VEGFR inhibitors or pharmaceutically acceptable salts thereof.
  • It is another object of the present invention to provide pharmaceutical compositions comprising a pharmaceutically acceptable carrier and a therapeutically effective amount of at least one of the compounds of the present invention or a pharmaceutically acceptable salt thereof.
  • It is another object of the present invention to provide a method for treating cancer (e.g., breast, colorectal, lung, prostate, and ovarian), comprising: administering to a host in need of such treatment a therapeutically effective amount of at least one of the compounds of the present invention or a pharmaceutically acceptable salt thereof.
  • It is another object of the present invention to provide novel isoindoles for use in therapy.
  • It is another object of the present invention to provide the use of novel isoindoles for the manufacture of a medicament for the treatment of cancer.
  • These and other objects, which will become apparent during the following detailed description, have been achieved by the inventor's discovery that the presently claimed isoindoles, or pharmaceutically acceptable salts thereof, are VEGFR inhibitors.
  • DETAILED DESCRIPTION OF PREFERRED EMBODIMENTS
  • Thus, in an embodiment, the present invention provides a novel method of treating cancer, comprising: administering a therapeutically effective amount of a compound of formula I or a stereoisomer or pharmaceutically acceptable salt thereof:
  • In another embodiment, the present invention provides novel method of treating cancer, comprising: administering a therapeutically effective amount of a compound of formula I or a stereoisomer or pharmaceutically acceptable salt thereof:
  • Figure US20110086868A1-20110414-C00001
  • wherein:
  • ring A is selected from phenyl, pyridyl, and pyrimidyl;
  • ring A is substituted with:
    • (a) 0-1 groups selected from O—C7-20 alkyl, O—C7-20 alkenyl, and O—C7-20 alkynyl;
    • (b) 0-3 R groups; and,
    • (c) 0-1 groups selected from methylene-dioxyl (—OCH2O—) and ethylene-dioxyl (—OCH2CH2O—);
  • R is selected from halogen, NO2, NRaRb, —CN, C1-2 haloalkyl, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 alkoxy, CHO, C(O)C1-6 alkyl, CO2-C1-6 alkyl, C(O)NRaRb, S(O)2NRaRb, S(O)p—C1-6 alkyl, phenyl, benzyl, and C3-6 cycloalkyl;
  • L is a linear chain selected from n-propylene, n-butylene, and n-pentylene, wherein
    • (a) from 0-2 of the methylene units are replaced by C═O;
    • (b) from 0-3 methylene units of L are replaced with a heteroatom selected from O, N, and S(O)p, provided that at least one methylene is present and other than an N—O or O—O bond is formed within L or at either attachment point of L; and,
    • (c) from 0-1 double bonds are present between the chain atoms of L;
  • L is substituted with 0-2 groups selected from C1-4 alkyl, phenyl, benzyl, C3-6 cycloalkyl, and NRaRb;
  • alternatively, when L is 4-5 atoms in length, then three of the chain atoms optionally combine with a 2 atom bridge to form a 5 membered ring, the ring consisting of carbon atoms and 0-2 heteroatoms selected from O, N, and S(O)p, wherein the ring has 0-2 ring double bonds and from 0-1 atom of the 2 atom bridge is replaced by as carbonyl group;
  • alternatively, a carbon atom in ring A that is adjacent to the carbon atom to which linker L is attached can be attached to linker L through a (CH2)1-2 bridge to form a 5-6 membered ring, wherein optionally 1 methylene of the bridge is replaced by a carbonyl group;
  • Ra and Rb are independently at each occurrence selected from H and C 1-6 alkyl;
  • alternatively, NRaRb, independently at each occurrence, forms a 5-6 membered cyclic amine consisting of the shown nitrogen atom and 4-5 methylenes;
  • ring B is selected from phenyl, pyridyl, pyrimidyl, and
  • Figure US20110086868A1-20110414-C00002
  • wherein the phenyl, pyridyl, and pyrimidyl rings are substituted with 1-3 R1 groups;
  • R1 is independently selected from CO2H, halogen, NO2, NRaRb, —CN, C1-2 haloalkyl, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 alkoxy, CHO, C(O)C1-6 alkyl, CO2—C1-6 alkyl, C(O)NRaRb, S(O)2NRaRb, S(O)p-C1-6 alkyl, C3-6 cycloalkyl, phenyl substituted with 0-2 R2 groups, benzyl substituted with 0-2 R2 groups, S-phenyl substituted with 0-2 R2 groups, O-phenyl substituted with 0-2 R2 groups, and NRa-phenyl substituted with 0-2 R2 groups;
  • alternatively, ring B is substituted with 0-1 groups selected from methylene-dioxyl (—OCH2O—) and ethylene-dioxyl (—OCH2CH2O—);
  • R2 is independently selected from CO2H, halogen, NH2, C1-2 haloalkyl, C1-6 alkyl, C1-6 alkoxy, C(O)C1-6 alkyl, and CO2C1-6 alkyl; and,
  • p is independently selected from 0, 1, and 2.
  • In another embodiment, the present invention provides novel method, wherein:
  • ring A is selected from phenyl and pyridyl;
  • ring A is substituted with 0-3 R groups;
  • ring A is substituted with 0-1 methylene-dioxyl (—OCH2O—);
  • R is selected from halogen, NO2, NRaRb, —CN, CF3, C1-4 alkyl, C2-4 alkenyl, C2-4 alkynyl, C1-4 alkoxy, CHO, C(O)C1-4 alkyl, CO2—C1-4 alkyl, C(O)NRaRb, S(O)2NRaRb, and S(O)p-C1-6 alkyl;
  • L is a linear chain selected from n-propylene and n-butylene, wherein
    • (a) from 0-1 of the methylene units are replaced by C═O; and,
    • (b) from 0-2 methylene units of L are replaced with a heteroatom selected from O and N, provided that other than an N—O or O—O bond is formed within L or at either attachment point of L;
  • L is substituted with 0-2 groups selected from C1-4 alkyl, phenyl, and benzyl;
  • alternatively, when L is 4 atoms in length, then three of the chain atoms optionally combine with a 2 atom bridge to form a 5 membered ring, the ring consisting of carbon atoms and 0-2 heteroatoms selected from O, N, and S(O)p, wherein the ring has 0-1 ring double bonds;
  • alternatively, a carbon atom in ring A that is adjacent to the carbon atom to which linker L is attached can be attached to linker L through a (CH2)1-2 bridge to form a 5-6 membered ring, wherein optionally 1 methylene of the bridge is replaced by a carbonyl group;
  • Ra and Rb are independently at each occurrence selected from H and C1-5 alkyl;
  • ring B is
  • Figure US20110086868A1-20110414-C00003
  • p is independently selected from 0, 1, and 2.
  • In another embodiment, the present invention provides a novel method, wherein the compound is selected from:
  • Figure US20110086868A1-20110414-C00004
  • or a pharmaceutically acceptable salt thereof.
  • In another embodiment, the present invention provides novel method, wherein the cancer is selected from: breast, colorectal, lung, prostate, and ovarian.
  • In another embodiment, the present invention provides novel method, wherein:
  • ring A is selected from phenyl and pyridyl;
  • ring A is substituted with 1 group selected from O—C7-20 alkyl, O—C7-20 alkenyl, and O—C7-20 alkynyl;
  • ring A is substituted with 0-1 R groups;
  • R is selected from halogen, CF3, C1-4 alkyl, C2-4 alkenyl, C2-4 alkynyl, C(O)C1-4 alkyl, and CO2—C1-4 alkyl;
  • L is a linear chain selected from n-propylene, n-butylene, and n-pentylene, wherein
    • (a) from 0-2 of the methylene units are replaced by C═O;
    • (b) from 0-3 methylene units of L are replaced with a heteroatom selected from O, N, and S(O)p, provided that at least one methylene is present; and,
    • (c) from 0-1 double bonds are present between the chain atoms of L;
  • L is substituted with 0-2 groups selected from C1-4 alkyl, C3-6 cycloalkyl, and NRaRb;
  • Ra and Rb are independently at each occurrence selected from H and C1-6 alkyl;
  • alternatively, independently at each occurrence, NRaRb forms a 5-6 membered cyclic amine consisting of the shown nitrogen atom and 4-5 methylenes;
  • ring B is selected from phenyl and pyridyl, wherein the phenyl and pyridyl rings are substituted with 1-2 R1 groups;
  • R1 is independently selected from CO2H, halogen, NRaRb, CF3, C1-4 alkyl, C1-6 alkoxy, C3-6 cycloalkyl, S-phenyl substituted with 0-2 R2 groups, O-phenyl substituted with 0-2 R2 groups, and NRa-phenyl substituted with 0-2 R2 groups;
  • R2 is independently selected from CO2H, halogen, CF3, C1-4 alkyl, and C1-4 alkoxy;
  • p is independently selected from 0, 1, and 2.
  • In another embodiment, the present invention provides novel method, wherein the compound is selected from:
  • Figure US20110086868A1-20110414-C00005
  • or a pharmaceutically acceptable salt thereof.
  • In another embodiment, the present invention provides novel method, wherein the cancer is selected from: breast, colorectal, lung, prostate, and ovarian.
  • In another embodiment, the present invention provides novel compound of formula I or a stereoisomer or pharmaceutically acceptable salt thereof:
  • Figure US20110086868A1-20110414-C00006
  • wherein:
  • ring A is selected from phenyl, pyridyl, and pyrimidyl;
  • ring A is substituted with 0-1 groups selected from O—C7-20 alkyl, O—C7-20 alkenyl, and O—C7-20 alkynyl;
  • ring A is substituted with 0-3 groups selected from halogen, NO2, NRaRb, —CN, C1-2 haloalkyl, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 alkoxy, CHO, C(O)C1-6 alkyl, CO2—C1-6 alkyl, C(O)NRaRb, S(O)2NRaRb, S(O)p—C1-6 alkyl, phenyl, benzyl, and C3-6 cycloalkyl;
  • ring A is substituted with 0-1 groups selected from methylene-dioxyl (—OCH2O—) and ethylene-dioxyl (—OCH2CH2O—);
  • L is a linear chain selected from n-propylene, n-butylene, and n-pentylene, wherein
    • (a) from 0-2 of the methylene units are replaced by C═O;
    • (b) from 0-3 methylene units of L are replaced with a heteroatom selected from O, N, and S(O)p, provided that at least one methylene is present and other than an N—O or O—O bond is formed within L or at either attachment point of L; and,
    • (c) from 0-1 double bonds are present between the chain atoms of L;
  • L is substituted with 0-2 groups selected from C1-4 alkyl, phenyl, benzyl, C3-6 cycloalkyl, and NRaRb;
  • alternatively, when L is 4-5 atoms in length, then three of the chain atoms optionally combine with a 2 atom bridge to form a 5 membered ring, the ring consisting of carbon atoms and 0-2 heteroatoms selected from O, N, and S(O)p, wherein the ring has 0-2 ring double bonds and from 0-1 atom of the 2 atom bridge is replaced by as carbonyl group;
  • alternatively, a carbon atom in ring A that is adjacent to the carbon atom to which linker L is attached can be attached to linker L through a (CH2)1-2 bridge to form a 5-6 membered ring, wherein optionally 1 methylene of the bridge is replaced by a carbonyl group;
  • Ra and Rb are independently at each occurrence selected from H and C1-6 alkyl;
  • alternatively, independently at each occurrence, NRaRb forms a 5-6 membered cyclic amine consisting of the shown nitrogen atom and 4-5 methylenes;
  • ring B is selected from phenyl, pyridyl, pyrimidyl, and
  • Figure US20110086868A1-20110414-C00007
  • wherein the phenyl, pyridyl, and pyrimidyl rings are substituted with 1-3 R groups;
  • R is independently selected from CO2H, halogen, NO2, NRaRb, —CN, C1-2 haloalkyl, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 alkoxy, CHO, C(O)C1-6 alkyl, CO2—C1-6 alkyl, C(O)NRaRb, S(O)2NRaRb, S(O)p—C1-6 alkyl, C3-6 cycloalkyl, phenyl substituted with 0-2 R1 groups, benzyl substituted with 0-2 R1 groups, S-phenyl substituted with 0-2 R1 groups, O-phenyl substituted with 0-2 R1 groups, and NRa-phenyl substituted with 0-2 R1 groups;
  • alternatively, ring B is substituted with 0-1 groups selected from methylene-dioxyl (—OCH2O—) and ethylene-dioxyl (—OCH2CH2O—);
  • R1 is independently selected from CO2H, halogen, NH2, C1-2 haloalkyl, C1-6 alkyl, C1-6 alkoxy, C(O)C1-6 alkyl, and CO2C1-6 alkyl; and,
  • p is independently selected from 0, 1, and 2;
  • provided that the compound of formula I is other than one of the following compounds:
  • Figure US20110086868A1-20110414-C00008
  • In another embodiment, the present invention provides novel compound, wherein the compound is selected from Table 2 or a stereoisomer or pharmaceutically acceptable salt thereof.
  • In another embodiment, the present invention provides a novel method of treating cancer, wherein the cancer is selected from: breast, colorectal, lung, prostate and ovarian.
  • In another embodiment, the present invention provides novel pharmaceutical compositions, comprising: a pharmaceutically acceptable carrier and a therapeutically effective amount of a compound of the present invention or a pharmaceutically acceptable salt form thereof.
  • In another embodiment, the present invention provides a novel method for treating cancer (e.g., breast, colorectal, lung, prostate and ovarian), comprising: administering to a patient in need thereof a therapeutically effective amount of a compound of the present invention or a pharmaceutically acceptable salt form thereof.
  • In another embodiment, the present invention provides a compound of the present invention for use in therapy.
  • In another embodiment, the present invention provides the use of a compound of the present invention for the manufacture of a medicament for the treatment of cancer.
  • The present invention may be embodied in other specific forms without departing from the spirit or essential attributes thereof. This invention encompasses all combinations of preferred aspects of the invention noted herein. It is understood that any and all embodiments of the present invention may be taken in conjunction with any other embodiment or embodiments to describe additional more preferred embodiments. It is also to be understood that each individual element of the preferred embodiments is intended to be taken individually as its own independent preferred embodiment. Furthermore, any element of an embodiment is meant to be combined with any and all other elements from any embodiment to describe an additional embodiment.
  • DEFINITIONS
  • All references cited herein are hereby incorporated in their entirety herein by reference.
  • All examples provided herein are not intended to be limiting, unless stated.
  • The compounds herein described may have asymmetric centers. Compounds of the present invention containing an asymmetrically substituted atom may be isolated in optically active or racemic forms. It is well known in the art how to prepare optically active forms, such as by resolution of racemic forms or by synthesis from optically active starting materials. Many geometric isomers of olefins, C═N double bonds, and the like can also be present in the compounds described herein, and all such stable isomers are contemplated in the present invention. C is and trans geometric isomers of the compounds of the present invention are described and may be isolated as a mixture of isomers or as separated isomeric forms. All chiral, diastereomeric, racemic forms and all geometric isomeric forms of a structure are intended, unless the specific stereochemistry or isomeric form is specifically indicated. All processes used to prepare compounds of the present invention and intermediates made therein are considered to be part of the present invention. All tautomers of shown or described compounds are also considered to be part of the present invention.
  • Examples of the molecular weight of compounds of the present invention include those that are (a) less than 500, 550, 600, 650, 700, 750, 800, 850, 900, 950, or 1000 grams per mole, (b) less than 950 grams per mole, (c) less than 850 grams per mole, and, (d) less than 750 grams per mole.
  • “Substituted” means that any one or more hydrogens on the designated atom is replaced with a selection from the indicated group, provided that the designated atom's normal valency is not exceeded, and that the substitution results in a stable compound. When a substituent is keto (i.e., ═O), then 2 hydrogens on the atom are replaced. Keto substituents are not present on aromatic moieties.
  • The present invention includes all isotopes of atoms occurring in the present compounds. Isotopes include those atoms having the same atomic number but different mass numbers. For example isotopes of hydrogen include tritium and deuterium. Isotopes of carbon include C-13 and C-14.
  • When any variable (e.g., R4) occurs more than one time in any constituent or formula for a compound, its definition at each occurrence is independent of its definition at every other occurrence. Thus, for example, if a group is shown to be substituted with 0-2 R4, then said group may optionally be substituted with up to two R4 groups and R4 at each occurrence is selected independently from the definition of R4. Also, combinations of substituents and/or variables are permissible only if such combinations result in stable compounds.
  • When a bond to a substituent is shown to cross a bond connecting two atoms in a ring, then such substituent may be bonded to any atom on the ring. When a substituent is listed without indicating the atom via which such substituent is bonded to the rest of the compound of a given formula, then such substituent may be bonded via any atom in such substituent. Combinations of substituents and/or variables are permissible only if such combinations result in stable compounds.
  • In cases wherein there are amines on the compounds of this invention, these can be converted to amine N-oxides by treatment with MCPBA and or hydrogen peroxides to afford other compounds of this invention. Thus, all shown amines cover both the shown amine and its N-oxide (N→O) derivative.
  • “Alkyl” includes both branched and straight-chain saturated aliphatic hydrocarbon groups having the specified number of carbon atoms. C1-6 alkyl, includes C1, C2, C3, C4, C5, and C6 alkyl groups. Examples of alkyl include methyl, ethyl, n-propyl, i-propyl, n-butyl, s-butyl, t-butyl, n-pentyl, and s-pentyl. “Haloalkyl” includes both branched and straight-chain saturated aliphatic hydrocarbon groups having the specified number of carbon atoms, substituted with 1 or more halogen (for example —CvFw where v=1 to 3 and w=1 to (2v+1)). Examples of haloalkyl include trifluoromethyl, trichloromethyl, pentafluoroethyl, and pentachloroethyl. “Alkoxy” represents an alkyl group as defined above with the indicated number of carbon atoms attached through an oxygen bridge. C1-6 alkoxy, includes C1, C2, C3, C4, C5, and C6 alkoxy groups. Examples of alkoxy include methoxy, ethoxy, n-propoxy, i-propoxy, n-butoxy, s-butoxy, t-butoxy, n-pentoxy, and s-pentoxy. “Cycloalkyl” includes saturated ring groups, such as cyclopropyl, cyclobutyl, or cyclopentyl. C3-7 cycloalkyl includes C3, C4, C5, C6, and C7 cycloalkyl groups. Alkenyl” includes hydrocarbon chains of either straight or branched configuration and one or more unsaturated carbon-carbon bonds that may occur in any stable point along the chain, such as ethenyl and propenyl. C2-6 alkenyl includes C2, C3, C4, C5, and C6 alkenyl groups. “Alkynyl” includes hydrocarbon chains of either straight or branched configuration and one or more triple carbon-carbon bonds that may occur in any stable point along the chain, such as ethynyl and propynyl. C2-6 Alkynyl includes C2, C3, C4, C5, and C6 alkynyl groups.
  • “Halo” or “halogen” refers to fluoro, chloro, bromo, and iodo; and “counterion” is used to represent a small, negatively charged species such as chloride, bromide, hydroxide, acetate, and sulfate.
  • “Carbocycle” means any stable 3, 4, 5, 6, or 7-membered monocyclic or bicyclic or 7, 8, 9, 10, 11, 12, or 13-membered bicyclic or tricyclic, any of which may be saturated, partially unsaturated, or aromatic. Examples of such carbocycles include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, adamantyl, cyclooctyl, [3.3.0]bicyclooctane, [4.3.0]bicyclononane, [4.4.0]bicyclodecane, [2.2.2]bicyclooctane, fluorenyl, phenyl, naphthyl, indanyl, adamantyl, and tetrahydronaphthyl.
  • “Heterocycle” means a stable 5, 6, or 7-membered monocyclic or bicyclic or 7, 8, 9, or 10-membered bicyclic heterocyclic ring which is saturated, partially unsaturated or unsaturated (aromatic), and which consists of carbon atoms and 1, 2, 3, or 4 heteroatoms independently selected from the group consisting of N, O and S and including any bicyclic group in which any of the above-defined heterocyclic rings is fused to a benzene ring. The nitrogen atom can be N, NH, or N-substituent depending on the ring and whether and the location of any substituent. The nitrogen and sulfur heteroatoms may optionally be, oxidized. The heterocyclic ring may be attached to its pendant group at any heteroatom or carbon atom that results in a stable structure. The heterocyclic rings described herein may be substituted on carbon or on a nitrogen atom if the resulting compound is stable. A nitrogen in the heterocycle may optionally be quaternized. When the total number of S and O atoms in the heterocycle exceeds 1, then these heteroatoms are not adjacent to one another. In a typical heterocycle, the total number of S and O atoms in the heterocycle is not more than 1.
  • “Heteroaryl” means a stable 5, 6, or 7-membered monocyclic or bicyclic or 7, 8, 9, or 10-membered bicyclic heterocyclic aromatic ring which consists of carbon atoms and 1, 2, 3, or 4 heteroatoms independently selected from the group consisting of N, O and S. The nitrogen atom can be N, NH, or N-substituent depending on the ring and whether and the location of any substituent. The total number of S and O atoms in the aromatic heterocycle is not more than 1. If the heteroaryl contains more than one ring, only one of the rings need be aromatic.
  • Examples of heterocycles and heteroaryls include acridinyl, azocinyl, benzimidazolyl, benzofuranyl, benzothiofuranyl, benzothiophenyl, benzoxazolyl, benzoxazolinyl, benzthiazolyl, benztriazolyl, benztetrazolyl, benzisoxazolyl, benzisothiazolyl, benzimidazolinyl, carbazolyl, 4aH-carbazolyl, carbolinyl, chromanyl, chromenyl, cinnolinyl, decahydroquinolinyl, 2H,6H-1,5,2-dithiazinyl, dihydrofuro[2,3-b]tetrahydrofuran, furanyl, furazanyl, imidazolidinyl, imidazolinyl, imidazolyl, 1H-indazolyl, indolenyl, indolinyl, indolizinyl, indolyl, 3H-indolyl, isatinoyl, isobenzofuranyl, isochromanyl, isoindazolyl, isoindolinyl, isoindolyl, isoquinolinyl, isothiazolyl, isoxazolyl, methylenedioxyphenyl, morpholinyl, naphthyridinyl, octahydroisoquinolinyl, oxadiazolyl, 1,2,3-oxadiazolyl, 1,2,4-oxadiazolyl, 1,2,5-oxadiazolyl, 1,3,4-oxadiazolyl, oxazolidinyl, oxazolyl, oxindolyl, pyrimidinyl, phenanthridinyl, phenanthrolinyl, phenazinyl, phenothiazinyl, phenoxathinyl, phenoxazinyl, phthalazinyl, piperazinyl, piperidinyl, piperidonyl, 4-piperidonyl, piperonyl, pteridinyl, purinyl, pyranyl, pyrazinyl, pyrazolidinyl, pyrazolinyl, pyrazolyl, pyridazinyl, pyridooxazole, pyridoimidazole, pyridothiazole, pyridinyl, pyridyl, pyrimidinyl, pyrrolidinyl, pyrrolinyl, 2H-pyrrolyl, pyrrolyl, quinazolinyl, quinolinyl, 4H-quinolizinyl, quinoxalinyl, quinuclidinyl, tetrahydrofuranyl, tetrahydroisoquinolinyl, tetrahydroquinolinyl, tetrazolyl, 6H-1,2,5-thiadiazinyl, 1,2,3-thiadiazolyl, 1,2,4-thiadiazolyl, 1,2,5-thiadiazolyl, 1,3,4-thiadiazolyl, thianthrenyl, thiazolyl, thienyl, thienothiazolyl, thienooxazolyl, thienoimidazolyl, thiophenyl, triazinyl, 1,2,3-triazolyl, 1,2,4-triazolyl, 1,2,5-triazolyl, 1,3,4-triazolyl, and xanthenyl. Also included are fused ring and Spiro compounds containing, for example, the above heterocycles.
  • The phrase “pharmaceutically acceptable” refers to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
  • “Pharmaceutically acceptable salt” refers to derivatives of the disclosed compounds wherein the parent compound is modified by making acid or base salts thereof. Examples of pharmaceutically acceptable salts include mineral or organic acid salts of basic residues such as amines; alkali or organic salts of acidic residues such as carboxylic acids; and the like. The pharmaceutically acceptable salts include the conventional non-toxic salts or the quaternary ammonium salts of the parent compound formed, for example, from non-toxic inorganic or organic acids. For example, such conventional non-toxic salts include those derived from inorganic acids such as hydrochloric, hydrobromic, sulfuric, sulfamic, phosphoric, nitric and the like; and the salts prepared from organic acids such as acetic, propionic, succinic, glycolic, stearic, lactic, malic, tartaric, citric, ascorbic, pamoic, maleic, hydroxymaleic, phenylacetic, glutamic, benzoic, salicylic, sulfanilic, 2-acetoxybenzoic, fumaric, toluenesulfonic, methanesulfonic, ethane disulfonic, oxalic, isethionic, and the like.
  • The pharmaceutically acceptable salts of the present invention can be synthesized from the parent compound that contains a basic or acidic moiety by conventional chemical methods. Generally, such salts can be prepared by reacting the free acid or base forms of these compounds with a stoichiometric amount of the appropriate base or acid in water or in an organic solvent, or in a mixture of the two; generally, non-aqueous media like ether, ethyl acetate, ethanol, isopropanol, or acetonitrile are preferred. Lists of suitable salts are found in Remington's Pharmaceutical Sciences, 17th ed., Mack Publishing Company, Easton, Pa., 1985, p. 1418, the disclosure of which is hereby incorporated by reference.
  • Since prodrugs are known to enhance numerous desirable qualities of pharmaceuticals (e.g., solubility, bioavailability, manufacturing, etc.) the compounds of the present invention may be delivered in prodrug form. Thus, the present invention is intended to cover prodrugs of the presently claimed compounds, methods of delivering the same and compositions containing the same. “Prodrugs” are intended to include any covalently bonded carriers that release an active parent drug of the present invention in vivo when such prodrug is administered to a mammalian subject. Prodrugs of the present invention are prepared by modifying functional groups present in the compound in such a way that the modifications are cleaved, either in routine manipulation or in vivo, to the parent compound. Prodrugs include compounds of the present invention wherein a hydroxy, amino, or sulfhydryl group is bonded to any group that, when the prodrug of the present invention is administered to a mammalian subject, it cleaves to form a free hydroxyl, free amino, or free sulfhydryl group, respectively.
  • Examples of prodrugs include acetate, formate and benzoate derivatives of alcohol and amine functional groups in the compounds of the present invention.
  • “Stable compound” and “stable structure” mean a compound that is sufficiently robust to survive isolation to a useful degree of purity from a reaction mixture, and formulation into an efficacious therapeutic agent.
  • “Substituted” means that one or more hydrogens on the atom indicated in the expression using “substituted” is replaced with a selection from the indicated group(s), provided that the indicated atom's normal valency is not exceeded, and that the substitution results in a stable compound. When a substituent is keto (i.e., ═O) group, then 2 hydrogens on the atom are replaced.
  • “Treating” or “treatment” cover the treatment of a disease-state in a mammal, particularly in a human, and include: (a) preventing the disease-state from occurring in a mammal, in particular, when such mammal is predisposed to the disease-state but has not yet been diagnosed as having it; (b) inhibiting the disease-state, i.e., arresting it development; and/or (c) relieving the disease-state, i.e., causing regression of the disease state.
  • “Therapeutically effective amount” includes an amount of a compound of the present invention that is effective when administered alone or in combination to inhibit a VEGFR. “Therapeutically effective amount” includes an amount of the combination of compounds claimed that is effective to inhibit a VEGFR. The combination of compounds is preferably a synergistic combination. Synergy, as described, for example, by Chou and Talalay, Adv. Enzyme Regul. 1984, 22:27-55, occurs when the effect of the compounds when administered in combination is greater than the additive effect of the compounds when administered alone as a single agent. In general, a synergistic effect is most clearly demonstrated at sub-optimal concentrations of the compounds. Synergy can be in terms of lower cytotoxicity, increased antiviral effect, or some other beneficial effect of the combination compared with the individual components.
  • SYNTHESIS
  • The compounds of the present invention can be prepared in a number of ways known to one skilled in the art of organic synthesis. The compounds of the present invention can be synthesized using the methods described below, together with synthetic methods known in the art of synthetic organic chemistry, or by variations thereon as appreciated by those skilled in the art. The reactions are performed in a solvent appropriate to the reagents and materials employed and suitable for the transformations being effected. It will be understood by those skilled in the art of organic synthesis that the functionality present on the molecule should be consistent with the transformations proposed. This will sometimes require a judgment to modify the order of the synthetic steps or to select one particular process scheme over another in order to obtain a desired compound of the invention. It will also be recognized that another major consideration in the planning of any synthetic route in this field is the judicious choice of the protecting group used for protection of the reactive functional groups present in the compounds described in this invention. An authoritative account describing the many alternatives to the trained practitioner is Greene and Wuts (Protective Groups In Organic Synthesis, Wiley and Sons, 1991).
  • Examples of typical synthetic routes to compounds of the present invention are shown in Schemes 1-2 below.
  • Figure US20110086868A1-20110414-C00009
  • Figure US20110086868A1-20110414-C00010
  • UTILITY
  • The compounds of this invention are expected to be useful as antitumor agents anticoagulants for the treatment of cancer (e.g., breast, colorectal, lung, prostate and ovarian).
  • The effectiveness of some of the compounds of the present invention as inhibitors of a VEGFR was determined in an in vitro angiogenesis assay as well as a VEGF receptor tyrosine kinase assay as described in the Examples.
  • The compounds of the present invention can be administered alone or in combination with one or more additional therapeutic agents. These include additional compounds of the present invention, other antitumor agents, or other agents that may be beneficially administered with compounds of the present invention.
  • “Administered in combination” or “combination therapy” means that a compound of the present invention and one or more additional therapeutic agents are administered concurrently to the mammal being treated. When administered in combination each component may be administered at the same time or sequentially in any order at different points in time. Thus, each component may be administered separately but sufficiently closely in time so as to provide the desired therapeutic effect.
  • Administration of the compounds of the present invention in combination with such additional therapeutic agent, may afford an efficacy advantage over the compounds and agents alone, and may do so while permitting the use of lower doses of each. A lower dosage minimizes the potential of side effects, thereby providing an increased margin of safety.
  • Dosage and Formulation
  • The compounds of this invention can be administered in such oral dosage forms as tablets, capsules (each of which includes sustained release or timed release formulations), pills, powders, granules, elixirs, tinctures, suspensions, syrups, and emulsions. They may also be administered in intravenous (bolus or infusion), intraperitoneal, subcutaneous, or intramuscular form, all using dosage forms well known to those of ordinary skill in the pharmaceutical arts. They can be administered alone, but generally will be administered with a pharmaceutical carrier selected on the basis of the chosen route of administration and standard pharmaceutical practice.
  • The dosage regimen for the compounds of the present invention will, of course, vary depending upon known factors, such as the pharmacodynamic characteristics of the particular agent and its mode and route of administration; the species, age, sex, health, medical condition, and weight of the recipient; the nature and extent of the symptoms; the kind of concurrent treatment; the frequency of treatment; the route of administration, the renal and hepatic function of the patient, and the effect desired. A physician or veterinarian can determine and prescribe the effective amount of the drug required to prevent, counter, or arrest the progress of the cancer.
  • Examples of the daily oral dosage of each active ingredient, when used for the indicated effects, can range between (a) 0.001 to 1000 mg/kg of body weight, (b) 0.01 to 100 mg/kg of body weight per day, and (c) 1.0 to 20 mg/kg/day. Intravenously doses can range from 1 to about 10 mg/kg/minute during a constant rate infusion (the dosage can also be calculated using the body's surface area). Compounds of this invention may be administered in a single daily dose, or the total daily dosage may be administered in divided doses of two, three, or four times daily.
  • Compounds of this invention can be administered in intranasal form via topical use of suitable intranasal vehicles, or via transdermal routes, using transdermal skin patches. When administered in the form of a transdermal delivery system, the dosage administration will, of course, be continuous rather than intermittent throughout the dosage regimen.
  • The compounds are typically administered in admixture with suitable pharmaceutical diluents, excipients, or carriers (collectively referred to herein as pharmaceutical carriers) suitably selected with respect to the intended form of administration, that is, oral tablets, capsules, elixirs, syrups and the like, and consistent with conventional pharmaceutical practices.
  • For instance, for oral administration in the form of a tablet or capsule, the active drug component can be combined with an oral, non-toxic, pharmaceutically acceptable, inert carrier such as lactose, starch, sucrose, glucose, methyl callulose, magnesium stearate, dicalcium phosphate, calcium sulfate, mannitol, sorbitol and the like; for oral administration in liquid form, the oral drug components can be combined with any oral, non-toxic, pharmaceutically acceptable inert carrier such as ethanol, glycerol, water, and the like. Moreover, when desired or necessary, suitable binders, lubricants, disintegrating agents, and coloring agents can also be incorporated into the mixture. Suitable binders include starch, gelatin, natural sugars such as glucose or beta-lactose, corn sweeteners, natural and synthetic gums such as acacia, tragacanth, or sodium alginate, carboxymethylcellulose, polyethylene glycol, waxes, and the like. Lubricants used in these dosage forms include sodium oleate, sodium stearate, magnesium stearate, sodium benzoate, sodium acetate, sodium chloride, and the like. Disintegrators include, without limitation, starch, methyl cellulose, agar, bentonite, xanthan gum, and the like.
  • The compounds of the present invention can also be administered in the form of liposome delivery systems, such as small unilamellar vesicles, large unilamellar vesicles, and multilamellar vesicles. Liposomes can be formed from a variety of phospholipids, such as cholesterol, stearylamine, or phosphatidylcholines.
  • Compounds of the present invention may also be coupled with soluble polymers as targetable drug carriers. Such polymers can include polyvinylpyrrolidone, pyran copolymer, polyhydroxypropylmethacrylamide-phenol, polyhydroxyethylaspartamidephenol, or polyethyleneoxide-polylysine substituted with palmitoyl residues. Furthermore, the compounds of the present invention may be coupled to a class of biodegradable polymers useful in achieving controlled release of a drug, for example, polylactic acid, polyglycolic acid, copolymers of polylactic and polyglycolic acid, polyepsilon caprolactone, polyhydroxy butyric acid, polyorthoesters, polyacetals, polydihydropyrans, polycyanoacylates, and crosslinked or amphipathic block copolymers of hydrogels.
  • Dosage forms (pharmaceutical compositions) suitable for administration may contain from about 1 milligram to about 100 milligrams of active ingredient per dosage unit. In these pharmaceutical compositions the active ingredient will ordinarily be present in an amount of about 0.5-95% by weight based on the total weight of the composition.
  • Gelatin capsules may contain the active ingredient and powdered carriers, such as lactose, starch, cellulose derivatives, magnesium stearate, stearic acid, and the like. Similar diluents can be used to make compressed tablets. Both tablets and capsules can be manufactured as sustained release products to provide for continuous release of medication over a period of hours. Compressed tablets can be sugar coated or film coated to mask any unpleasant taste and protect the tablet from the atmosphere, or enteric coated for selective disintegration in the gastrointestinal tract.
  • Liquid dosage forms for oral administration can contain coloring and flavoring to increase patient acceptance.
  • In general, water, a suitable oil, saline, aqueous dextrose (glucose), and related sugar solutions and glycols such as propylene glycol or polyethylene glycols are suitable carriers for parenteral solutions. Solutions for parenteral administration preferably contain a water soluble salt of the active ingredient, suitable stabilizing agents, and if necessary, buffer substances. Antioxidizing agents such as sodium bisulfate, sodium sulfite, or ascorbic acid, either alone or combined, are suitable stabilizing agents. Also used are citric acid and its salts and sodium EDTA. In addition, parenteral solutions can contain preservatives, such as benzalkonium chloride, methyl-or propyl-paraben, and chlorobutanol.
  • Suitable pharmaceutical carriers are described in Remington's the Science and Practice of Pharmacy, Lippincott Williams & Wilkins, a standard reference text in this field.
  • Representative useful pharmaceutical dosage-forms for administration of the compounds of this invention can be illustrated as follows:
  • Capsules
  • A large number of unit capsules can be prepared by filling standard two-piece hard gelatin capsules each with 100 milligrams of powdered active ingredient, 150 milligrams of lactose, 50 milligrams of cellulose, and 6 milligrams magnesium stearate.
  • Soft Gelatin Capsules
  • A mixture of active ingredient in a digestible oil such as soybean oil, cottonseed oil or olive oil may be prepared and injected by means of a positive displacement pump into gelatin to form soft gelatin capsules containing 100 milligrams of the active ingredient. The capsules should be washed and dried.
  • Tablets
  • Tablets may be prepared by conventional procedures so that the dosage unit is 100 milligrams of active ingredient, 0.2 milligrams of colloidal silicon dioxide, 5 milligrams of magnesium stearate, 275 milligrams of microcrystalline cellulose, 11 milligrams of starch and 98.8 milligrams of lactose. Appropriate coatings may be applied to increase palatability or delay absorption.
  • Injectable
  • A parenteral composition suitable for administration by injection may be prepared by stirring 1.5% by weight of active ingredient in 10% by volume propylene glycol and water. The solution should be made isotonic with sodium chloride and sterilized.
  • Suspension
  • An aqueous suspension can be prepared for oral administration so that each 5 mL contain 100 mg of finely divided active ingredient, 200 mg of sodium carboxymethyl cellulose, 5 mg of sodium benzoate, 1.0 g of sorbitol solution, U.S.P, and 0.025 mL of vanillin.
  • Where the compounds of this invention are combined with other antitumor agents, for example, a daily dosage may be about 0.1 to 100 milligrams of the compound of the present invention and about 1 to 7.5 milligrams of the second agent, per kilogram of patient body weight. For a tablet dosage form, the compounds of this invention generally may be present in an amount of about 5 to 10 milligrams per dosage unit, and the second anti-coagulant in an amount of about 1 to 5 milligrams per dosage unit.
  • Other features of the invention will become apparent in the course of the following descriptions of exemplary embodiments that are given for illustration of the invention and are not intended to be limiting thereof.
  • EXAMPLES
  • In Vitro Angiogenesis Assay
  • Human umbilical vein endothelial cell was purchased from Cambrex Co. (East Rutherford, N.J.) and maintained in EGM (Endothelial Growth Medium) supplemented with 2% FBS, 0.1% EGF, 0.1% Hydrocortisone, 0.1% GA-1000 and 0.4% BBE.
  • The morphogenesis assay on Matrigel was performed according to the manufacturer's instructions (Chemicon International). The ECMatrix™ kit consists of laminin, collagen type IV, heparan sulfate, proteoglycans, entactin and nidogen. It also contains various growth factors (TGF-β, FGF) and proteolytic enzymes (plasminogen, tPA, MMPs) that are normally produced in EHS tumors. The incubation condition was optimized for maximal tube-formation as follows: 50 μl of EC Matrix™ was suitably diluted in the ratio 9:1 with 10× diluent buffer and used for coating the 96-well plate. The coated plates were incubated at 37° C. for 1 hr. to allow the Matrix solution to solidify. In the meantime the HUVECs that were cultured for 24 hours in EGM with 2% FBS was trypsinised and re-suspended in the growth media and the cells were counted. After 1 hr. pre-incubation of the plate with Matrix solution, the HUVECs were plated at 104 cells/well in the absence or in the presence of different VEGFR inhibitors (1 mM and 10 mM). After 8 hours incubation at 37° C., the cell three-dimensional organization (cellular network structures) was examined under an inverted photomicroscope. Each treatment was performed in triplicate.
  • Activated endothelial cells form cellular networks (mesh like structures) resembling capillary tubes sprouting into the stromal space. The formation of these cellular networks is a dynamic process, which started migration and alignment of cells followed by the development of capillary tubes like structures, sprouting of new branches and finally the formation of the cellular networks. Although this in vitro angiogenesis kit is designed only as a qualitative assay, we have made an attempt to quantitate the degree of angiogenesis using a scoring method. The scoring was based on the extent of the cellular networks that were observed as follows:
  • Individual cells, well separated (+++++)
  • Cells begin to migrate and align themselves (++++)
  • Cells begin to line but not sprouting (+++)
  • Visible Sprouting (++)
  • Closed polygons begins to form (+).
  • According to our qualitative assessment, the higher the scoring the greater the efficiency of the compound for inhibiting endothelial cell mediated angiogenesis.
  • Table 1, below, shows the results for Examples 1-6, which are commercially available.
  • TABLE 1
    In Vitro Angiogenesis Assay
    Example # Structure Results
    1
    Figure US20110086868A1-20110414-C00011
    +++++
    2
    Figure US20110086868A1-20110414-C00012
    ++++
    3
    Figure US20110086868A1-20110414-C00013
    ++++
    4
    Figure US20110086868A1-20110414-C00014
    +++
    5
    Figure US20110086868A1-20110414-C00015
    +++
    6
    Figure US20110086868A1-20110414-C00016
    ++
    7
    Figure US20110086868A1-20110414-C00017
    ++
  • The EC50 value for anti-antigenic effect of Example 1 was estimated to be 0.25 μM.
  • Assay of VEGF Receptor Tyrosine Kinase Activity:
  • Protein Tyrosine Kinases (PTKs) perform a critical role in signal transduction pathways that control cell proliferation, differentiation, metabolism, and apoptosis. Phosphorylation of proteins by PTKs is essential for the regulation of these biological mechanisms and defects in these pathways may result in a number of human diseases, including cancer. The important aspect of these enzymes in cellular regulation is accentuated by the fact that for a large number of kinases a corresponding viral oncogene product has been identified. Protein tyrosine kinase activity is often associated with membrane receptor protein tyrosine kinases (e.g. EGF-, PDGF-, CFS-, IGF-1 and insulin receptor) and soluble non-receptor tyrosine kinases (e.g. p60c-Src, yes, lck, lyn, fyn). Assaying of PTK activity allows for purification and characterization of protein tyrosine kinases, elucidation of their biological functions as well as aiding in development of specific PTK inhibitors. CHEMICON's non-radioactive Tyrosine Kinase Activity Assay provides a simple, convenient and specific method for quantification and comparative determination of a wide range of PTKs. Testing of soluble and receptor tyrosine kinases, in vitro inhibitor screening and the study of PTK regulation can be performed with this assay. Our assay is immunoprecipitation compatible and will not cross-react with serine/threonine kinases.
  • The non-radioactive Tyrosine Kinase Activity Assay Kit consists of a synthetic Biotinylated Peptide Substrate, a Biotinylated Phosphopeptide, purified Phosphotyrosine specific monoclonal antibody conjugated to horseradish peroxidase (HRP) and other components required to perform 96 ELISA-based assays. The synthetic Biotinylated Substrate, poly [Glu:Tyr], 4:1 contains multiple tyrosine residues and can be phosphorylated by a wide range of PTKs. After quenching the enzyme reaction with an inhibitor, both the phosphorylated and dephosphorylated substrates are immobilized by binding to the streptavidin-coated plate. The fraction of phosphorylated substrate is visualized using a phosphotyrosine monoclonal antibody conjugated to HRP and an ensuing chromagenic substrate reaction. The quantity of phosphate incorporated into the tyrosine kinase substrate is determined utilizing the phosphopeptide standard curve. The assay mixture and start the reaction was be prepared by adding 10 μl of ATP/MgCl2 Solution. The reaction mixture was pre-incubate at 30° C. The reaction time was dependent on the individual tyrosine kinase and was standardized. The enzyme reaction was terminated by adding 10 μl of kinase inhibitor, such as 120 mM EDTA. After termination, 50 μL of reaction mixture was transferred to Streptavidin-coated strip wells and incubated at 37° C. for 30 minutes. The wells were washed four times using 1× wash buffer and then 200 μl of blocking buffer solution will be added to each well and incubate at 37° C. for 30 minutes. After removing the blocking buffer 100 μL of diluted mouse anti-Phosphotyrosine HRP conjugate was added to each well and incubated at room temperature for 60 minutes. The wells were washed four times using 1× Wash Buffer and then 100 μl of TMB substrate solution were added and the plates are incubate at room temperature for 5-15 minutes. The assay reaction would be terminated by the addition of 100 μl of stop solution and finally, the absorbance was measured on a standard microplate reader at 450 nm.
  • After testing the anti-antigenic effects of Example 1, the ability of this compound to inhibit the VEGFR linked tyrosine kinase activity was measured. The VEGFR activity was measured by specifically immunoprecipitating the cell lysate using VEGFR specific antibody (Upstate, VA) and measuring its activity using the PTK assay. Example 1 was able to inhibit the VEGFR associated tyrosine kinase activity, and the level of inhibition increased as the concentration of the compound increased. The IC50 for inhibiting VEGFR kinase was determined to be 0.4 μM.
  • In addition to inhibiting the angiogenesis under in vitro conditions Example 1 also produced cytotoxicity towards GI-101A and MCF-7, breast carcinoma cell lines.
  • Representative examples of the present invention include those in the following tables.
  • TABLE 2
    Figure US20110086868A1-20110414-C00018
    Ex. # L A
    1. CH2C(O)OCH2 Phenyl
    2. CH2C(O)NHCH2 4-Cl-phenyl
    3. CH2C(O)
    Figure US20110086868A1-20110414-C00019
    4. CH2C(O)NHCH2 4-NH2SO2-phenyl
    5. CH2C(O)NH 4-CH3O-phenyl
    6. CH2C(O)NH
    Figure US20110086868A1-20110414-C00020
    7. CH2C(O)NH 2-CH3O-phenyl
    8. CH2C(O)NH Phenyl
    9. CH2C(O)N(Et)CH2 Phenyl
    10. CH2CH(OH)CH2NH 3-CH3-phenyl
    11. CH2CH(OH)CH2NH 4-Cl-phenyl
    12. CH2CH(OH)CH2O Phenyl
    13. CH2C(O)N(CH3)CH2 Phenyl
    14.
    Figure US20110086868A1-20110414-C00021
    4-Br-phenyl
    15. CH2C(O)
    Figure US20110086868A1-20110414-C00022
    16. CH2C(O)NH 3,4-Di-CH3O-phenyl
    17. CH2C(O)NHCH(phenyl) Phenyl
    18. CH2C(O)NHNH 2,4-Di-NO2-phenyl
    19. CH2C(O)
    Figure US20110086868A1-20110414-C00023
  • Numerous modifications and variations of the present invention are possible in light of the above teachings. It is therefore to be understood that within the scope of the appended claims, the invention may be practiced otherwise that as specifically described herein.

Claims (11)

1. A method of treating cancer, comprising: administering a therapeutically effective amount of a compound of formula I or a stereoisomer or pharmaceutically acceptable salt thereof:
Figure US20110086868A1-20110414-C00024
wherein:
ring A is selected from phenyl, pyridyl, and pyrimidyl;
ring A is substituted with:
(a) 0-1 groups selected from O—C7-20 alkyl, O—C7-20 alkenyl, and O—C7-20 alkynyl;
(b) 0-3 R groups; and,
(c) 0-1 groups selected from methylene-dioxyl (—OCH2O—) and ethylene-dioxyl (—OCH2CH2O—);
R is selected from halogen, NO2, NRaRb, —CN, C1-2 haloalkyl, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 alkoxy, CHO, C(O)C1-6 alkyl, CO2—C1-6 alkyl, C(O)NRaRb, S(O)2NRaRb, S(O)p—C1-6 alkyl, phenyl, benzyl, and C3-6 cycloalkyl;
L is a linear chain selected from n-propylene, n-butylene, and n-pentylene, wherein
(a) from 0-2 of the methylene units are replaced by C═O;
(b) from 0-3 methylene units of L are replaced with a heteroatom selected from O, N, and S(O)p, provided that at least one methylene is present and other than an N—O or O—O bond is formed within L or at either attachment point of L; and,
(c) from 0-1 double bonds are present between the chain atoms of L;
L is substituted with 0-2 groups selected from C1-4 alkyl, phenyl, benzyl, C3-6 cycloalkyl, and NRaRb;
alternatively, when L is 4-5 atoms in length, then three of the chain atoms optionally combine with a 2 atom bridge to form a 5 membered ring, the ring consisting of carbon atoms and 0-2 heteroatoms selected from O, N, and S(O)p, wherein the ring has 0-2 ring double bonds and from 0-1 atom of the 2 atom bridge is replaced by as carbonyl group;
alternatively, a carbon atom in ring A that is adjacent to the carbon atom to which linker L is attached can be attached to linker L through a (CH2)1-2 bridge to form a 5-6 membered ring, wherein optionally 1 methylene of the bridge is replaced by a carbonyl group;
Ra and Rb are independently at each occurrence selected from H and C1-6 alkyl;
alternatively, NRaRb, independently at each occurrence, forms a 5-6 membered cyclic amine consisting of the shown nitrogen atom and 4-5 methylenes;
ring B is selected from phenyl, pyridyl, pyrimidyl, and
Figure US20110086868A1-20110414-C00025
wherein the phenyl, pyridyl, and pyrimidyl rings are substituted with 1-3 R1 groups;
R1 is independently selected from CO2H, halogen, NO2, NRaRb, —CN, C1-2 haloalkyl, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 alkoxy, CHO, C(O)C1-6 alkyl, CO2—C1-6 alkyl, C(O)NRaRb, S(O)2NRaRb, S(O)p—C1-6 alkyl, C3-6 cycloalkyl, phenyl substituted with 0-2 R2 groups, benzyl substituted with 0-2 R2 groups, S-phenyl substituted with 0-2 R2 groups, O-phenyl substituted with 0-2 R2 groups, and NRa-phenyl substituted with 0-2 R2 groups;
alternatively, ring B is substituted with 0-1 groups selected from methylene-dioxyl (—OCH2O—) and ethylene-dioxyl (—OCH2CH2O—);
R2 is independently selected from CO2H, halogen, NH2, C1-2 haloalkyl, C1-6 alkyl, C1-6 alkoxy, C(O)C1-6 alkyl, and CO2C1-6 alkyl; and,
p is independently selected from 0, 1, and 2.
2. A method of claim 1, wherein:
ring A is selected from phenyl and pyridyl;
ring A is substituted with 0-3 R groups;
ring A is substituted with 0-1 methylene-dioxyl (—OCH2O—);
R is selected from halogen, NO2, NRaRb, —CN, CF3, C1-4 alkyl, C2-4 alkenyl, C2-4 alkynyl, C1-4 alkoxy, CHO, C(O)C1-4 alkyl, CO2—C1-4 alkyl, C(O)NRaRb, S(O)2NRaRb, and S(O)p—C1-6 alkyl;
L is a linear chain selected from n-propylene and n-butylene, wherein
(a) from 0-1 of the methylene units are replaced by C═O; and,
(b) from 0-2 methylene units of L are replaced with a heteroatom selected from O and N, provided that other than an N—O or O—O bond is formed within L or at either attachment point of L;
L is substituted with 0-2 groups selected from C1-4 alkyl, phenyl, and benzyl;
alternatively, when L is 4 atoms in length, then three of the chain atoms optionally combine with a 2 atom bridge to form a 5 membered ring, the ring consisting of carbon atoms and 0-2 heteroatoms selected from O, N, and S(O)p, wherein the ring has 0-1 ring double bonds;
alternatively, a carbon atom in ring A that is adjacent to the carbon atom to which linker L is attached can be attached to linker L through a (CH2)1-2 bridge to form a 5-6 membered ring, wherein optionally 1 methylene of the bridge is replaced by a carbonyl group;
Ra and Rb are independently at each occurrence selected from H and C1-6 alkyl;
ring B is
Figure US20110086868A1-20110414-C00026
p is independently selected from 0, 1, and 2.
3. The method of claim 2, wherein the compound is selected from:
Figure US20110086868A1-20110414-C00027
or a pharmaceutically acceptable salt thereof.
4. The method of claim 3, wherein the cancer is selected from: breast, colorectal, lung, prostate, and ovarian.
5. A method of claim 1, wherein:
ring A is selected from phenyl and pyridyl;
ring A is substituted with 1 group selected from O—C7-20 alkyl, O—C7-20 alkenyl, and O—C7-20 alkynyl;
ring A is substituted with 0-1 R groups;
R is selected from halogen, CF3, C1-4 alkyl, C2-4 alkenyl, C2-4 alkynyl, C(O)C1-4 alkyl, and CO2—C1-4 alkyl;
L is a linear chain selected from n-propylene, n-butylene, and n-pentylene, wherein
(a) from 0-2 of the methylene units are replaced by C═O;
(b) from 0-3 methylene units of L are replaced with a heteroatom selected from O, N, and S(O)p, provided that at least one methylene is present; and,
(c) from 0-1 double bonds are present between the chain atoms of L;
L is substituted with 0-2 groups selected from C1-4 alkyl, C3-6 cycloalkyl, and NRaRb;
Ra and Rb are independently at each occurrence selected from H and C1-6 alkyl;
alternatively, independently at each occurrence, NRaRb forms a 5-6 membered cyclic amine consisting of the shown nitrogen atom and 4-5 methylenes;
ring B is selected from phenyl and pyridyl, wherein the phenyl and pyridyl rings are substituted with 1-2 R1 groups;
R1 is independently selected from CO2H, halogen, NRaRb, CF3, C1-4 alkyl, C1-6 alkoxy, C3-6 cycloalkyl, S-phenyl substituted with 0-2 R2 groups, 0-phenyl substituted with 0-2 R2 groups, and NRa-phenyl substituted with 0-2 R2 groups;
R2 is independently selected from CO2H, halogen, CF3, C1-4 alkyl, and C1-4 alkoxy;
p is independently selected from 0, 1, and 2.
6. The method of claim 5, wherein the compound is selected from:
Figure US20110086868A1-20110414-C00028
or a pharmaceutically acceptable salt thereof.
7. The method of claim 6, wherein the cancer is selected from: breast, colorectal, lung, prostate, and ovarian.
8. A compound of formula I or a stereoisomer or pharmaceutically acceptable salt thereof:
Figure US20110086868A1-20110414-C00029
wherein:
ring A is selected from phenyl, pyridyl, and pyrimidyl;
ring A is substituted with 0-1 groups selected from O—C7-20 alkyl, O—C7-20 alkenyl, and O—C7-20 alkynyl;
ring A is substituted with 0-3 groups selected from halogen, NO2, NRaRb, —CN, C1-2 haloalkyl, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 alkoxy, CHO, C(O)C1-6 alkyl, CO2—C1-6 alkyl, C(O)NRaRb, S(O)2NRaRb, S(O)p—C1-6 alkyl, phenyl, benzyl, and C3-6 cycloalkyl;
ring A is substituted with 0-1 groups selected from methylene-dioxyl (—OCH2O—) and ethylene-dioxyl (—OCH2CH2O—);
L is a linear chain selected from n-propylene, n-butylene, and n-pentylene, wherein
(a) from 0-2 of the methylene units are replaced by C═O;
(b) from 0-3 methylene units of L are replaced with a heteroatom selected from O, N, and S(O)p, provided that at least one methylene is present and other than an N—O or O—O bond is formed within L or at either attachment point of L; and,
(c) from 0-1 double bonds are present between the chain atoms of L;
L is substituted with 0-2 groups selected from C1-4 alkyl, phenyl, benzyl, C3-6 cycloalkyl, and NRaRb;
alternatively, when L is 4-5 atoms in length, then three of the chain atoms optionally combine with a 2 atom bridge to form a 5 membered ring, the ring consisting of carbon atoms and 0-2 heteroatoms selected from O, N, and S(O)p, wherein the ring has 0-2 ring double bonds and from 0-1 atom of the 2 atom bridge is replaced by as carbonyl group;
alternatively, a carbon atom in ring A that is adjacent to the carbon atom to which linker L is attached can be attached to linker L through a (CH2)1-2 bridge to form a 5-6 membered ring, wherein optionally 1 methylene of the bridge is replaced by a carbonyl group;
Ra and Rb are independently at each occurrence selected from H and C1-6 alkyl;
alternatively, independently at each occurrence, NRaRb forms a 5-6 membered cyclic amine consisting of the shown nitrogen atom and 4-5 methylenes;
ring B is selected from phenyl, pyridyl, pyrimidyl, and
Figure US20110086868A1-20110414-C00030
wherein the phenyl, pyridyl, and pyrimidyl rings are substituted with 1-3 R groups;
R is independently selected from CO2H, halogen, NO2, NRaRb, —CN, C1-2 haloalkyl, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 alkoxy, CHO, C(O)C1-6 alkyl, CO2—C1-6 alkyl, C(O)NRaRb, S(O)2NRaRb, S(O)p—C1-6 alkyl, C3-6 cycloalkyl, phenyl substituted with 0-2 R1 groups, benzyl substituted with 0-2 R1 groups, S-phenyl substituted with 0-2 R1 groups, O-phenyl substituted with 0-2 R1 groups, and NRa-phenyl substituted with 0-2 R1 groups;
alternatively, ring B is substituted with 0-1 groups selected from methylene-dioxyl (—OCH2O—) and ethylene-dioxyl (—OCH2CH2O—);
R1 is independently selected from CO2H, halogen, NH2, C1-2 haloalkyl, C1-6 alkyl, C1-6 alkoxy, C(O)C1-6 alkyl, and CO2C1-6 alkyl; and,
p is independently selected from 0, 1, and 2;
provided that the compound of formula I is other than one of the following compounds:
Figure US20110086868A1-20110414-C00031
9. A compound of claim 8, wherein the compound is selected from Table 2 or a stereoisomer or pharmaceutically acceptable salt thereof.
10. A pharmaceutical composition, comprising: a pharmaceutically acceptable carrier and a therapeutically effective amount of a compound of claim 8 or a pharmaceutically acceptable salt form thereof.
11. A pharmaceutical composition, comprising: a pharmaceutically acceptable carrier and a therapeutically effective amount of a compound of claim 9 or a pharmaceutically acceptable salt form thereof.
US12/969,848 2006-09-21 2010-12-16 Specific inhibitors for vascular endothelial growth factor receptors Abandoned US20110086868A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US12/969,848 US20110086868A1 (en) 2006-09-21 2010-12-16 Specific inhibitors for vascular endothelial growth factor receptors

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US82639006P 2006-09-21 2006-09-21
US11/859,235 US7875603B2 (en) 2006-09-21 2007-09-21 Specific inhibitors for vascular endothelial growth factor receptors
US12/969,848 US20110086868A1 (en) 2006-09-21 2010-12-16 Specific inhibitors for vascular endothelial growth factor receptors

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US11/859,235 Continuation US7875603B2 (en) 2006-09-21 2007-09-21 Specific inhibitors for vascular endothelial growth factor receptors

Publications (1)

Publication Number Publication Date
US20110086868A1 true US20110086868A1 (en) 2011-04-14

Family

ID=39498913

Family Applications (2)

Application Number Title Priority Date Filing Date
US11/859,235 Active US7875603B2 (en) 2006-09-21 2007-09-21 Specific inhibitors for vascular endothelial growth factor receptors
US12/969,848 Abandoned US20110086868A1 (en) 2006-09-21 2010-12-16 Specific inhibitors for vascular endothelial growth factor receptors

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US11/859,235 Active US7875603B2 (en) 2006-09-21 2007-09-21 Specific inhibitors for vascular endothelial growth factor receptors

Country Status (1)

Country Link
US (2) US7875603B2 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023214706A1 (en) * 2022-05-04 2023-11-09 엘림랜드 주식회사 Novel diaryl-propanedione derivative compound, preparation method therefor, and pharmaceutical composition comprising same for prevention or treatment of inflammatory or allergic diseases

Families Citing this family (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
MX2012007759A (en) 2009-12-29 2012-08-01 Mapi Pharma Ltd Intermediate compounds and processes for the preparation of tapentadol and related compounds.
WO2011156632A2 (en) * 2010-06-09 2011-12-15 Georgetown University Compositions and methods of treatment for tumors in the nervous system
LT2605652T (en) 2010-08-18 2018-03-12 Samumed, Llc Diketones and hydroxyketones as catenin signaling pathway activators
HUE054031T2 (en) 2012-04-24 2021-08-30 Vertex Pharma Dna-pk inhibitors
PE20151721A1 (en) 2013-02-22 2015-12-07 Samumed Llc Y-DICETONES AS ACTIVATORS OF THE WNT / B-CATENIN SIGNALING ROUTE
HUE041544T2 (en) 2013-03-12 2019-05-28 Vertex Pharma Dna-pk inhibitors
SI3057953T1 (en) 2013-10-17 2018-12-31 Vertex Pharmaceuticals Incorporated Co-crystals of (s)-n-methyl-8-(1-((2'-methyl-(4,5'-bipyrimidin)-6-yl)amino)propan-2-yl)quinoline-4-carboxamide and deuterated derivatives thereof as dna-pk inhibitors
CZ2014307A3 (en) * 2014-05-06 2016-01-13 Vysoká škola chemicko- technologická v Praze Coffeine-8-hydrazones as novel cytostatics for the treatment of oncologic diseases
RS59797B1 (en) 2014-08-20 2020-02-28 Samumed Llc Gamma-diketones for treatment and prevention of aging skin and wrinkles
CA3038657A1 (en) 2016-09-27 2018-04-05 Vertex Pharmaceuticals Incorporated Method for treating cancer using a combination of dna-damaging agents and dna-pk inhibitors
MD3448859T2 (en) 2017-03-20 2020-03-31 Forma Therapeutics Inc Pyrrolopyrrole compositions as pyruvate kinase (PKR) activators
GB201712110D0 (en) * 2017-07-27 2017-09-13 Thomas Helledays Stiftelse För Medicinsk Forskning New compounds and uses
BR112021005188A2 (en) 2018-09-19 2021-06-08 Forma Therapeutics, Inc. treating sickle cell anemia with a pyruvate kinase r activating compound
JP7450610B2 (en) 2018-09-19 2024-03-15 ノヴォ・ノルディスク・ヘルス・ケア・アーゲー Activation of pyruvate kinase R

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6878715B1 (en) * 1994-02-18 2005-04-12 Cell Therapeutics, Inc. Therapeutic compounds for inhibiting interleukin-12 signals and method for using same
WO2005074375A2 (en) * 2004-02-06 2005-08-18 Insight Biopharmaceuticals Ltd Heparanase inhibitors and uses thereof
US20070015752A1 (en) * 2004-12-28 2007-01-18 Hangauer David G Jr Biaryl compositions and methods for modulating a kinase cascade

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6878715B1 (en) * 1994-02-18 2005-04-12 Cell Therapeutics, Inc. Therapeutic compounds for inhibiting interleukin-12 signals and method for using same
WO2005074375A2 (en) * 2004-02-06 2005-08-18 Insight Biopharmaceuticals Ltd Heparanase inhibitors and uses thereof
US20070015752A1 (en) * 2004-12-28 2007-01-18 Hangauer David G Jr Biaryl compositions and methods for modulating a kinase cascade

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023214706A1 (en) * 2022-05-04 2023-11-09 엘림랜드 주식회사 Novel diaryl-propanedione derivative compound, preparation method therefor, and pharmaceutical composition comprising same for prevention or treatment of inflammatory or allergic diseases

Also Published As

Publication number Publication date
US7875603B2 (en) 2011-01-25
US20080139585A1 (en) 2008-06-12

Similar Documents

Publication Publication Date Title
EP2271341B1 (en) Specific inhibitors for vascular endothelial growth factor receptors
US7875603B2 (en) Specific inhibitors for vascular endothelial growth factor receptors
KR101991327B1 (en) Opioid Receptor Ligands and Methods of Using and Making Same
TWI525092B (en) Tetrahydrocarboline derivatives
BR112018004175B1 (en) PYRAZOLO[3,4-D]PYRIMIDINE COMPOUND, PHARMACEUTICAL COMPOSITION, HER2 INHIBITOR AND ANTITUMOR AGENT CONTAINING SAID COMPOUND AND THERAPEUTIC USES OF SAID COMPOUND
CN115057855B (en) Substituted five-membered and six-membered heterocyclic compound, preparation method, pharmaceutical composition and application thereof
CN101222850B (en) Methods for treating drug resistant cancer
CN107428758A (en) Acrylate derivative, its preparation method and its purposes in medicine
EP1914234A1 (en) Pyrido[2,3-d]pyrimidines and their use as kinase inhibitors
EA028175B1 (en) Pyrazolo-pyrrolidin-4-one derivatives as bet inhibitors and their use in the treatment of disease
ES2930106T3 (en) Pyrrole-pyridine derivative compound, process for preparing the same, and pharmaceutical composition containing the same as an active ingredient for the prevention or treatment of protein kinase-related diseases
EA028035B1 (en) Pyrazolo-pyrrolidin-4-one derivatives and their use in the treatment of disease
BRPI0613644A2 (en) thiene pyrimidine and thiene pyrimidine kinase modulators
JP2019512459A (en) Seven-membered ring compounds, process for their preparation, their pharmaceutical compositions and their use
CN103930408A (en) Morpholinyl benzotriazine for use in cancer therapy
TW201309698A (en) Tetrahydrocarboline derivative
TW201915004A (en) Thienopyridine compound, and preparing method, pharmaceutical composition and application thereof for preparing a medicament capable of preventing or treating tumor growth and metastasis
US7939557B2 (en) Vascular endothelial receptor specific inhibitors
JPWO2006132192A1 (en) New 2-quinolone derivatives
BR112017016278B1 (en) Use of a compound having btk inhibitory activity or a salt thereof to produce a preventive and/or therapeutic agent for the prevention or treatment of an immune disease
EP4363414A1 (en) Compounds that inhibit pi3k isoform alpha and methods for treating cancer
EA017817B1 (en) 3-ARYLSULFONYLPYRAZOLO[1,5-a]PYRIMIDINES - ANTAGONISTS OF SEROTONIN 5-HTRECEPTORS, METHODS FOR THE PRODUCTION AND THE USE THEREOF
EP2930167A1 (en) Indole full ketone derivative used as tyrosine-kinase inhibitor
KR20230031322A (en) Compositions of Compounds that Modulate Cellular Metabolism and Methods of Use
CN101423513B (en) Amine pyrimidine derivates, and production method thereof, and medicament composition and use

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

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