WO2006024034A1 - Heterocyclic compounds and methods of use - Google Patents

Heterocyclic compounds and methods of use Download PDF

Info

Publication number
WO2006024034A1
WO2006024034A1 PCT/US2005/030614 US2005030614W WO2006024034A1 WO 2006024034 A1 WO2006024034 A1 WO 2006024034A1 US 2005030614 W US2005030614 W US 2005030614W WO 2006024034 A1 WO2006024034 A1 WO 2006024034A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
substituted
group
cancer
alkyl
Prior art date
Application number
PCT/US2005/030614
Other languages
French (fr)
Inventor
Wolfgang Wrasidlo
Elena Dneprovskaia
Original Assignee
Targegen, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Targegen, Inc. filed Critical Targegen, Inc.
Priority to MX2007002208A priority Critical patent/MX2007002208A/en
Priority to CA002578283A priority patent/CA2578283A1/en
Priority to EP05792421A priority patent/EP1799656A4/en
Priority to JP2007530214A priority patent/JP5275628B2/en
Priority to AU2005276974A priority patent/AU2005276974B2/en
Priority to RU2007110934/04A priority patent/RU2468021C2/en
Priority to NZ553492A priority patent/NZ553492A/en
Publication of WO2006024034A1 publication Critical patent/WO2006024034A1/en
Priority to IL181524A priority patent/IL181524A/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/60Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D213/62Oxygen or sulfur atoms
    • C07D213/63One oxygen atom
    • C07D213/65One oxygen atom attached in position 3 or 5
    • 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
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/4439Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. omeprazole
    • 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
    • A61K31/50Pyridazines; Hydrogenated pyridazines
    • A61K31/501Pyridazines; Hydrogenated pyridazines not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic 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
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/08Drugs for disorders of the urinary system of the prostate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/10Drugs for disorders of the urinary system of the bladder
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/02Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings
    • C07D239/24Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members
    • C07D239/28Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, directly attached to ring carbon atoms
    • C07D239/46Two or more oxygen, sulphur or nitrogen atoms
    • C07D239/48Two nitrogen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D253/00Heterocyclic compounds containing six-membered rings having three nitrogen atoms as the only ring hetero atoms, not provided for by group C07D251/00
    • C07D253/02Heterocyclic compounds containing six-membered rings having three nitrogen atoms as the only ring hetero atoms, not provided for by group C07D251/00 not condensed with other rings
    • C07D253/061,2,4-Triazines
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D271/00Heterocyclic compounds containing five-membered rings having two nitrogen atoms and one oxygen atom as the only ring hetero atoms
    • C07D271/02Heterocyclic compounds containing five-membered rings having two nitrogen atoms and one oxygen atom as the only ring hetero atoms not condensed with other rings
    • C07D271/101,3,4-Oxadiazoles; Hydrogenated 1,3,4-oxadiazoles
    • C07D271/1131,3,4-Oxadiazoles; Hydrogenated 1,3,4-oxadiazoles with oxygen, sulfur or nitrogen atoms, directly attached to ring carbon atoms, the nitrogen atoms not forming part of a nitro radical
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D409/00Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms
    • C07D409/14Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/12Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing three or more hetero rings

Definitions

  • the present invention relates generally to the use of compounds to treat a variety of disorders, diseases and pathologic conditions and more specifically to the use of various heterocyclic compounds for therapeutic purposes.
  • kinases are a large family of cellular proteins involved in signal transduction of cascades which control cell growth and death, survival, migration, differentiation, gene expression, metabolism, protein synthesis and cell cycle regulation. A common mechanism by which these signals are transmitted is reversible phosphorylation, which induces conformational changes is these enzymes and alters their structure and function. The entire kinase genome discovered so far incorporates over 500 individual proteins and their isoforms. Different branches of this genomic tree have been characterized into groups specific for phosphorylating either serine/threonine residues or tyrosines. Some kinases exhibit dual specificity, capable of substrate phosphorylation of tyrosine as well as serine/threonines.
  • Transmembrane receptor protein kinases exhibit an extracellular domain, capable of ligand binding. These ligand binding mechanisms trigger activation of the kinase catalytic domain which initiates a cascade of signals that controls intracellular functions. Examples of a receptor protein kinase are growth factors such as EGF, FGF, PDGF and IGF. Nonreceptor protein kinases can be found in many compartments of a cell from inner-cell surface membranes to the cell nucleus.
  • a nonreceptor protein kinase is the mitogen activated protein kinase (MAPK) which regulates a pathway, which is important in cell signaling initiated on the exterior cell surfaces via growth factors, for example, VEGF, or hormones, and extending to the cell nucleus by activating transcription factors. These nuclear factors in turn control gene expression in the regulation of cell cycle progression and ultimately cell proliferation, and differentiation.
  • MAPK mitogen activated protein kinase
  • the MAPK cell signaling pathway is important for drug targeting as this path impinges on nearly all functional hallmarks of cancer cells such as immortalization, growth factor independent proliferation, insensitivity to growth inhibitory signals, metastasis, blood vessel attraction, evasion of apoptosis, and other functional hallmarks.
  • the present invention provides compounds which affect the MAPK pathway.
  • the compounds of the invention are useful as pharmaceutical compositions, for example where modulation of the MAPK pathway is indicated for the treatment of various human diseases, such as cancer.
  • compounds having the structure (A) are provided, or an N-oxide, N,N'-dioxide, N,N',N"-trioxide, or a pharmaceutically acceptable salt thereof:
  • compounds having the structure (B) are provided, or an N-oxide, N,N'-dioxide, N,N',N"-trioxide, or a pharmaceutically acceptable salt thereof:
  • X can be absent or can be NH
  • Y can be absent or can be one of the following moieties:
  • the substituted R 1 can be an aryl, a substituted aryl, a heterocycle, a heteroaryl, a substituted heterocycle, and a substituted heteroaryl.
  • R 1 can be one of C 6 -Cj 2 aryl; C 3 -Ci 2 heteroaryl having 1-3 heteroatoms such as N, S and O; substituted C 3 -Ci O cycloalkyl having 0-3 heteroatoms such as N, S, and O; substituted C 6 -Ci 2 aryl; substituted C 3 -Ci 2 heteroaryl having 1-3 heteroatoms such as N, S and O; C 7 -C 24 aralkyl; C 7 -C 24 alkylaryl; substituted C 7 -C 24 aralkyl; and substituted C 7 -C 24 alkaryl.
  • the substitutent Ri can be, independently of the substitutent Ri present in the structure (A), an unsubstituted or a substituted C 3 -C 12 heteroaryl having 1-3 heteroatoms such as N, S or O.
  • the substituent Ri that can be present in compounds having structure (B), can include a substituted pyridyl group.
  • the substituents in the substituted pyridyl group can include an amido moiety, an aminoalkyl group (e.g., aminomethyl), or a carboxyl group, or a carboxylate group.
  • the amido moiety attached to the pyridyl group can be in turn also substituted by attaching to the nitrogen in the amido moiety a substitutent selected from an alkyl (e.g., methyl), an alkylaminoalkyl (e.g., diethylamino alkyl), a pyridyl, an alkyl pyrrolidine, an alkyl morpholine, and an alkyl piperazine groups.
  • substitutent R 1 that can be present in compounds having either structure (A) or structure (B), can be selected from one of the following moieties:
  • n can be an integer selected from a group consisting of 0, 1, 2, and 3.
  • R 2 can be, independently, any one of hydrogen, halogen, C 1 -C 18 alkyl (e.g., methyl), -OH, -NO 2 , -CN, Ci-C 18 alkoxy (e.g., methoxy), -NHSO 2 R 5 , -SO 2 NHR 5 , -NHCOR 5 , -NH 2 , -NR 5 R 6 , -S(O)R 5 , -S(O) 2 R 5 , -CO 2 R 5 , - CONR 5 R 6 , and where R 5 and R 6 are independently selected from hydrogen, a Ci-Ci 2 alkyl and a substituted Ci-Ci 2 alkyl.
  • the substituent R 3 can be an aryl, a substituted aryl, a heterocycle, a heteroaryl, a substituted heterocycle, and a substituted heteroaryl.
  • R 3 can be one of Ce-Ci 2 aryl; C 3 -Ci 2 heteroaryl having 1-3 heteroatoms such as N, S and O; substituted C 3 -Ci 0 cycloalkyl having 0-3 heteroatoms such as N, S, and O; substituted C 6 -C 12 aryl; substituted C 3 -Ci 2 heteroaryl having 1-3 heteroatoms such as N, S and O; C 7 -C 24 aralkyl; C 7 -C 24 alkylaryl; substituted C 7 -C 24 aralkyl; and substituted C 7 -C 24 alkaryl.
  • the substitutent R 3 can be, independently of the substitutent R 3 present in the structure (A), hydrogen, a Ci-Ci 8 alkyl, a substituted Ci-C 18 alkyl, a Ci-Ci 2 cycloalkyl, a substituted Ci-Ci 2 cycloalkyl, a substituted C 3 -Ci O cycloalkyl having 0-3 heteroatoms such as N, S, or O, an aryl such as a C 6 -Ci 2 aryl, a substituted aryl such as a substituted C 6 -Ci 2 aryl, a heterocycle, a substituted heterocycle, a heteroaryl such as a C 3 -Ci 2 heteroaryl having 1-3 heteroatoms such as N, S or O, a substituted heteroaryl such as substituted C 3 -Ci 2 heteroaryl having 1-3 heteroatoms such as N, S or O, a substituted heteroaryl such as substituted C 3 -Ci 2 hetero
  • substituent R 3 examples include tert-butyl phenyl, trifluoromethoxyphenyl, methoxyphenyl, dimethylarninophenyl, aminophenyl, trifluoroethoxyphenyl, trifluoromethoxychlorophenyl, trifluoromethoxybromophenyl, trifluoroethoxychlorophenyl, chlorophenyl, dichlorophenyl, trifluoromethyl phenyl, trifluoromethylchlorophenyl, chlorotoluyl, N-phenylacetaniide, N,N-alkyl-benzamide, isopropoxyphenyl, alkoxyphenyl, dialkoxyphenyl, or acetylphenyl.
  • substitutent R 3 that can be present in compounds having either structure (A) or structure (B), can be selected from one of the following moieties:
  • compounds that are derivatives of benzotriazine including a benzotriazine moiety having at least a first substituent attached to the benzene ring of benzotriazine and a second substituent attached to the triazine ring of the benzotriazine, where the first substituent includes a substituted pyridyl group, and the second substituent includes a secondary amino group, a substituted amide group, or a substituted sulfonylamino group.
  • compounds including a benzene-derived moiety bridged to a heterocyclic moiety are provided, where the benzene- derived moiety includes a molecule of benzene substituted with either a sulfonyl group or a pyridyl group connected to the benzene molecule via an oxygen link, and the heterocyclic moiety includes triazole, oxadiazole, oxazole, pyrazol, imidazole, thiadiazole and triazine.
  • articles of manufacture including packaging material and a pharmaceutical composition contained within the packaging material, wherein the packaging material includes a label which indicates that the pharmaceutical composition can be used for treatment of disorders associated with cancer.
  • the pharmaceutical composition can include at least one compound set forth in Structures (A) and (B) or any combination thereof.
  • a method for treating a disorder including administering to a subject in need thereof an effective amount of a compound, wherein the compound is set forth in Structures (A) and (B) or any combination thereof.
  • the disorder is cancer, eye disease, inflammation, psoriasis, or a viral infection, for example.
  • the cancer is an alimentary/gastrointestinal tract cancer, colon cancer, liver cancer, skin cancer, breast cancer, ovarian cancer, prostate cancer, lymphoma, leukemia, kidney cancer, lung cancer, muscle cancer, bone cancer, bladder cancer or brain cancer.
  • a pharmaceutical composition is provided, at least one compound set forth in structures (A) and (B) or any combination thereof, in a pharmaceutically acceptable carrier.
  • FIG. 1 shows the results of the Rafl direct assay of invention compounds
  • FIG. 2 shows the results of XTT cell viability assay of invention compounds
  • the present invention is directed to heterocyclic compounds, such as heterocyclic compounds derived from benzotriazine, triazines, triazoles, oxadiazoles, imidazoles and tliiadiazole and to use of the heterocyclic compounds for therapeutic purposes.
  • heterocyclic compounds such as heterocyclic compounds derived from benzotriazine, triazines, triazoles, oxadiazoles, imidazoles and tliiadiazole and to use of the heterocyclic compounds for therapeutic purposes.
  • IUPAC International Union of Pure and Applied Chemistry
  • heterocyclic when used to describe an aromatic ring, means that the aromatic ring contains at least one heteroatom.
  • the abbreviation “Het” is sometimes used to signify a heterocyclic structure.
  • heteroatom is defined to include any atom other than carbon, for example, N, O, or S.
  • aromatic or aryl is defined to include a cyclically conjugated molecular entity with a stability, due to derealization, significantly greater than that of a hypothetical localized structure, such as the Kekule structure.
  • heterocyclic when not used to describe an aromatic ring, is defined to include cyclic (i.e., ring-containing) groups other than aromatic groups, the cyclic group being formed by between 3 and about 14 carbon atoms and at least one heteroatom described above.
  • substituted heterocyclic is defined to include both aromatic and non- aromatic structures to heterocyclic groups further bearing one or more substituents described above.
  • alkyl is defined to include a monovalent straight or branched chain hydrocarbon group having from one to about 12 carbon atoms, for example, methyl, ethyl, n- propyl, wo-propyl, w-butyl, ⁇ o-butyl, tert-butyl, 7i-pentyl (also known as rc-amyl), n-hexyl, and the like.
  • the abbreviations "Me” and "Et” stand for the methyl and ethyl groups, respectively.
  • aralkyl refers to an aryl or heteroaryl group, as defined herein, attached through a C1-C6 alkyl linker.
  • aralkyl include, but are not limited to, benzyl, phenylpropyl, 2-pyridylmethyl, 3-isoxazolylmethyl, 2-imidazolylethyl.
  • halogen is defined to include an atom of fluorine, chlorine, bromine or iodine.
  • carboxyl or “carboxyl group” is defined as an acid moiety having the structure -COOH.
  • amino or “amino group” is defined to include moieties -NRR', where each of R and R' is hydrogen ("primary amino"), or one of them is an organic radical ("secondary amino”), or each is an organic radical ("tertiary amino").
  • aminoalkyl or “aminoalkyl group” is defined to include moieties -R-N(R 1 R"), wherein R is an organic radical and each of R' and R" is hydrogen or an organic radical. If at least one of R' and R" is an organic radical, the moiety is defined as “alkylaminoalkyl” or “alkylaminoalkyl group.”
  • sulfonyl or "sulfonyl group” is defined to include moieties that comprise structure (S), in which R is an organic radical:
  • sulfonylamino or "sulfonylamino group” is defined to include moieties that comprise structure (SA), in which R is an organic radical:
  • substituted amide is defined to include moieties containing a structure RNH-CO-, in which R is an organic radical.
  • phenyl is defined to include moieties having structure (Ph):
  • toluyl is defined to include moieties having structure (Tl):
  • heteroaryl is defined to include aromatic rings, where the ring structure is formed by between 3 and about 14 carbon atoms and by at least one heteroatom described above, and the term “substituted heteroaryl” refers to heteroaryl groups further bearing one or more substituents described above.
  • triazine is defined to include moieties containing the aromatic 6- member heterocycle having three atoms of nitrogen in the ring. Two examples of such heterocycle are shown as the following structures (Tr):
  • benzotriazine is defined to include moieties containing a heterocyclic structure in which the triazine ring is fused with the benzene ring, as shown by structure (BTr): [0046]
  • N-oxide nitrogen-containing heterocyclic moieties in which at least one nitrogen atom is associated with oxygen to form the structures N ⁇ O.
  • the heterocyclic moiety can be any nitrogen-containing heterocycle, for example, benzotriazine, triazine, pyridine, pyrimidine, etc. Where the heterocyclic structure is benzotriazine, for example, some N-oxides or dioxides can be described as the following structures:
  • heterocyclic structure having two fused 6-member rings, each ring containing two atoms of nitrogen, as shown by structure (PTr):
  • pyridazine is defined to include moieties containing the aromatic 6- member heterocycle having two atoms of nitrogen in the ring in ortho position, as shown by the structure (PAz):
  • pyrimidine is defined to include moieties containing the aromatic 6- member heterocycle having two atoms of nitrogen in the ring in meta position, as shown by the structure (PRm):
  • thiadiazole is defined to include moieties containing the aromatic 5- member thiophene-based heterocycle, having two atoms of nitrogen and one atom of sulfur, as shown by the structure (TDa):
  • pyridyl is defined to include moieties containing a radical derived from pyridine.
  • One structure of pyridyl is shown as the structure (Py):
  • alkyl pyrrolidine is defined to include moieties containing a radical derived from pyrrolidine (a 5 -member saturated heterocycle having one nitrogen atom), where an alkylene group R is attached to the nitrogen atom of the pyrrolidine ring.
  • alkyl pyrrolidine is shown as the structure (APy):
  • alkyl morpholine is defined to include moieties containing a radical derived from morholine, (a 6-member saturated heterocycle having one nitrogen atom and one oxygen atom), where an alkylene group R is attached to the nitrogen atom of the morpholine ring.
  • Ar alkyl morpholine
  • alkyl piperazine is defined to include moieties containing a radical derived from piperazine (a 6-member saturated heterocycle having two nitrogen atoms), where an alkylene group R is attached to one nitrogen atom of the piperazine ring.
  • APi One structure of alkyl piperazine is shown as the structure (APi):
  • isoxazole is defined to include moieties containing the aromatic 5- meniber heterocycle, having one atoms of nitrogen and one atom of oxygen, as shown by the structure (ISo):
  • hydrophobic is defined as a group or structure free of strongly polar groups such as -OH, -COOH, -NH 2 , -NH-CO-, halogens, or the like.
  • kinase is defined to include any enzyme that catalyzes the addition of phosphate groups to a protein residue; for example, serine and threonine kinases catalyze the addition of phosphate groups to serine and threonine residues.
  • terapéuticaally effective amount is defined as the amount of the compound or pharmaceutical composition that will elicit the biological or medical response of a tissue, system, animal or human that is being sought by the researcher, veterinarian, medical doctor or other clinician, e.g., restoration or maintenance of vasculostasis or prevention of the compromise or loss or vasculostasis; reduction of tumor burden; reduction of morbidity and/or mortality.
  • pharmaceutically acceptable is defined as a carrier, whether diluent or excipient, that is compatible with the other ingredients of the formulation and not deleterious to the recipient thereof.
  • administering a compound is defined to include an act of providing a compound of the invention or pharmaceutical composition to the subject in need of treatment.
  • heterocyclic compounds are provided for treatment of various diseases, disorders, and pathologies, including cancer.
  • the heterocyclic compounds of the invention can inhibit the activity of a kinase, such as any kinase in the MAPK signaling pathway.
  • a first type of compounds for treatment of various diseases, disorders, and pathologies, including cancer.
  • the first type of compounds can include derivatives of benzotriazine.
  • the derivatives of benzotriazine that can be used can comprise the compounds that include a benzotriazine moiety having at least one substituent attached to the benzene ring of benzotriazine and a at least one substituent attached to the triazine ring of the benzotriazine, and an N-oxide, N,N'- dioxide, N,N',N"-trioxide, or pharmaceutically acceptable salts thereof.
  • a substituent attached to the benzene ring of benzotriazine can include a substituted pyridyl group.
  • the substituents in the substituted pyridyl group can include an amido moiety, an aminoalkyl group (e.g., aminomethyl), or a carboxyl group, or a carboxylate group.
  • the amido moiety attached to the pyridyl group can be in turn also substituted by attaching to the nitrogen in the amido moiety a substituent selected from an alkyl (e.g., methyl), an alkylaminoalkyl (e.g., diethylamino alkyl), a pyridyl, an alkyl pyrrolidine, an alkyl morpholine, and an alkyl piperazine groups.
  • the benzene ring of the compounds of the first type can contain a second substituent located in any available position of the ring, for example, methyl, halogen or methoxy.
  • a second substituent located in any available position of the ring, for example, methyl, halogen or methoxy.
  • benzene-ring containing moieties that can be used include tert-h ⁇ tyl phenyl, trifluoromethoxyphenyl, methoxyphenyl, dimethylamino, dimethylaminophenyl, aminophenyl, trifluoroethoxyphenyl, trifluoromethoxychlorophenyl, trifluoromethoxybromophenyl, trifluoroethoxychlorophenyl, chlorophenyl, dichlorophenyl, trifluoromethyl phenyl, trifluoromethylchloro phenyl, chlorotoluyl, N-phenylacetamide, N 5 N- alky
  • a substituent attached to the triazine ring of benzotriazine in the compounds of the first type can include a secondary amino group, a substituted amide group, or a substituted sulfonylamino group; each of these groups can further contain a moiety derived from benzene, thiophene, or isoxazole. If the substituent attached to the triazine ring of benzotriazine in the compounds of the first type is a secondary amino group, the moiety derived from benzene, thiophene, or isoxazole can be attached to the nitrogen of the secondary amino group.
  • the substituent in the triazine ring is a substituted amide group or the substituted sulfonylamino group
  • the moiety derived from benzene, thiophene, or isoxazole can be attached via the acyl group or the sulfonyl group, respectively.
  • Moieties derived from benzene, thiophene, or isoxazole can further include alkyls, e.g., £-butyl phenyl, chlorophenyl, dichlorophenyl, trifluoromethyl phenyl, trifluoromethylchloro phenyl, and chlorotoluyl.
  • Compounds of the first type can be described as compounds having the general structure (A), and an N-oxide, N,N'-dioxide, N,N',N"-trioxide, or a pharmaceutically acceptable salt thereof.
  • the general structure (A) is as follows:
  • Y can be absent or can be one of the following moieties:
  • R 1 can be an aryl, a substituted aryl, a heterocycle, a heteroaryl, a substituted heterocycle, and a substituted heteroaryl, such as C 6 -Ci 2 aryl; C 3 -Ci 2 heteroaryl having 1-3 heteroatoms such as N, S and O; substituted C 3 -Ci 0 cycloalkyl having 0-3 heteroatoms such as N, S, and O; substituted C 6 -Ci 2 aryl; substituted C 3 -Ci 2 heteroaryl having 1-3 heteroatoms such as N, S and O; C 7 -C 24 aralkyl; C 7 -C 24 alkylaryl; substituted C 7 - C 24 aralkyl; and substituted C 7 -C 24 alkaryl.
  • Ri can be any one of the following moieties:
  • n can be an integer selected from a group consisting of 0, 1, 2, and 3.
  • the substituent R 2 can be any one of hydrogen, halogen, C 1 -Ci 8 alkyl (e.g., methyl), -OH, -NO 2 , -CN, Ci-Ci 8 alkoxy (e.g., methoxy), -NHSO 2 R 5 , -SO 2 NHR 5 , -NHCOR 5 , -NH 2 , -NR 5 R 6 , -S(O)R 5 , -S(O) 2 R 5 , -CO 2 R 5 , -CONR 5 R 6 , and where R 5 and R 6 are independently selected from hydrogen, a Ci-Ci 2 alkyl and a substituted Ci-Ci 2 alkyl.
  • group R 3 can be an aryl, a substituted aryl, a heterocycle, a heteroaryl, a substituted heterocycle, and a substituted heteroaryl.
  • R 3 can be one of C 6 -Ci 2 aryl; C 3 -Ci 2 heteroaryl having 1-3 heteroatoms such as N, S and O; substituted C 3 -Ci O cycloalkyl having 0-3 heteroatoms such as N, S, and O; substituted C 6 -Ci 2 aryl; substituted C 3 -Ci 2 heteroaryl having 1-3 heteroatoms such as N, S and O; C 7 -C 24 aralkyl; C 7 - C 24 alkylaryl; substituted C 7 -C 24 aralkyl; and substituted C 7 -C 24 alkaryl.
  • the substitutent R 3 can be one of the following:
  • Some general examples of compounds described by the general structure (A), include a compound having the general structure (I), a compound having the general structure (II), a compound having the general structure (III) or a compound having the general structure (IV):
  • a second type of compounds for treatment of various diseases, disorders, and pathologies, including cancer.
  • the second type of compounds can include a benzene derived moiety bridged to a heterocyclic moiety, or pharmaceutically acceptable salts thereof.
  • the bridge between the benzene- derived moiety and the heterocyclic moiety can include a single bond or a nitrogen atom. If the heterocyclic moiety contains at least one nitrogen, the second type of compounds can be an N-oxide, or N,N'-dioxide, or N,N',N"-trioxide.
  • the compound can be an N-oxide, or N,N'-dioxide, or N,N',N"-trioxide, depends on the number of nitrogen atoms contained in the heterocyclic moiety. For example, if the heterocyclic moiety has only one nitrogen, the second type of compounds can be an N- oxide. If the heterocyclic moiety has two atoms of nitrogen, the second type of compounds can be an N-oxide or N,N'-dioxide. If the heterocyclic moiety has three atoms of nitrogen, the second type of compounds can be any of an N-oxide, an N,N'-dioxide, an N,N',N"- trioxide.
  • the benzene-derived moiety can include a substituent sucfr as a pyridyl group connected to the benzene molecule via an oxygen link, or a sulfonyl
  • the pyridyl group connected to the benzene molecule can be further substituted.
  • the siibstituents in the pyridyl group can include the same moieties as described above for the first type of compounds of the present invention.
  • the sulfonyl group connected to the benzene molecule can be also further substituted.
  • the substituents in the sulfonyl group can include the substituted pyridyl group described above for the first type of compounds of the present invention.
  • the benzene-derived moiety of the compounds of the second type can contain a second substituent, e.g., methyl, halogen or methoxy, which can be located in any position of the benzene ring.
  • a second substituent e.g., methyl, halogen or methoxy
  • Some exemplary benzene-derived moieties that can be included in the second type of compounds can include tert-butyl phenyl, trifluoromethoxyphenyl, methoxyphenyl, dimethylamino, dimethylaminophenyl, aminophenyl, trifiuoroethoxyphenyl, trifluoromethoxychlorophenyl, trifluoromethoxybromophenyl, trifluoroethoxychlorophenyl, chlorophenyl, dichlorophenyl, trifluoromethyl phenyl, trifiuoromethylchloro phenyl, chlorotoluyl, N-phenylacetamide, N,N-alkyl-benzamide, isopropoxyphenyl, alkoxyphenyl, dialkoxyphenyl, acetylphenyl.
  • the compounds of the second type include heterocyclic compounds having the general structure (B), or an N-oxide, or N,N'-dioxide, N,N',N"-trioxide, or a pharmaceutically acceptable salt thereof, and can inhibit the activity of a kinase, such as any kinase in the MAPK signaling pathway.
  • the general structure (B) can be represented as follows:
  • X can be absent or be NH;
  • Y can be absent or can be one of the following moieties:
  • substitutents R 1 , R 2 , and R 3 can be as follows:
  • R 1 can be an unsubstituted or a substituted C 3 -C 12 heteroaryl having 1 -3 heteroatoms such as N, S or O;
  • R 2 can be any one of hydrogen, halogen, C 1 -Ci 8 alkyl (e.g., methyl), -OH, -NO 2 , - CN, C 1 -Ci 8 alkoxy (e.g., methoxy), -NHSO 2 R 5 , -SO 2 NHR 5 , -NHCOR 5 , -NH 2 , -NR 5 R 6 , - S(O)R 5 , -S(O) 2 R 5 , -CO 2 R 5 , -CONR 5 R 6 , and where R 5 and R 6 are independently selected from hydrogen, a Ci-Cis alkyl and a substituted Ci-Ci 2 alkyl; and
  • R 3 can be hydrogen, a Ci-Cis alkyl, a substituted Ci-Ci 2 alkyl, a C 1 -C 12 cycloalkyl, a substituted C 1 -Ci 2 cycloalkyl, a substituted C 3 -Ci 0 cycloalkyl having 0-3 heteroatoms such as N, S, or O, an aryl such as a C ⁇ -Ci 2 aryl, a substituted aryl such as a substituted C 6 -Ci 2 aryl, a heterocycle, a substituted heterocycle, a heteroaryl such as a C 3 -Ci 2 heteroaryl having 1-3 heteroatoms such as N, S or O, a substituted heteroaryl such as substituted C 3 -Ci 2 heteroaryl having 1-3 heteroatoms such as N, S or O, a substituted heteroaryl such as substituted C 3 -Ci 2 heteroaryl having 1-3 heteroatoms such as N, S or O
  • the substituent Ri can include a substituted pyridyl or a substituted pyrimidyl group.
  • the substituents in the substituted pyridyl or substituted pyrimidyl group can include an amido moiety, an aminoalkyl group (e.g., aminomethyl), or a carboxyl group, or a carboxylate group.
  • the amido moiety attached to the pyridyl/pyrimidyl group can be in turn also substituted by attaching to the nitrogen in the amido moiety a substitutent selected from an alkyl (e.g., methyl), an alkylaminoalkyl (e.g., diethylamino alkyl), a pyridyl, an alkyl pyrrolidine, an alkyl morpholine, and an alkyl piperazine groups.
  • Ri that can be used in compounds having the structure (B) include any of the following moieties:
  • n can be an integer selected from a group consisting of 0, 1, 2, and 3.
  • substituent R 3 examples include tert-butyl phenyl, trifluoromethoxyphenyl, methoxyphenyl, dimethylaminophenyl, aminophenyl, trifluoroethoxyphenyl, trifluoromethoxychlorophenyl, trifluoromethoxybromophenyl, trifluoroethoxychlorophenyl, chlorophenyl, dichlorophenyl, trifluoromethyl phenyl, trifluoromethylchlorophenyl, chlorotoluyl, N-phenylacetamide, N,N-alkyl-benzamide, isopropoxyphenyl, alkoxyphenyl, dialkoxyphenyl, and acetylphenyl.
  • n can be an integer selected from a group consisting of 0, 1, 2, and 3, and R' is hydrogen, a C 1 -Ci 8 alkyl, or a substituted Ci-Ci 8 alkyl.
  • 1,2,4-triazoles of the type (VI) described above can be made as shown in Scheme VIII or by an alternative route as outlined in Scheme IX.
  • Scheme XII [0096] Appropriately substituted 2-amino- 1 ,3 ,4-oxadiazoles of type (IX), can be synthesized using one of several reaction schemes, for example, as shown by Schemes XIII and XIV.
  • the compounds and methods of the present invention are useful in treating a variety of disorders, including, but not limited to, for example, cancer, eye disease, inflammation, psoriasis, and a viral infection.
  • agents e.g., chemotherapeutic agents or protein therapeutic agents described below
  • the kinds of cancer that can be treated include, but are not limited to, an alimentary/gastrointestinal tract cancer, colon cancer, liver cancer, skin cancer, breast cancer, ovarian cancer, prostate cancer, lymphoma, leukemia, kidney cancer, lung cancer, muscle cancer, bone cancer, bladder cancer or brain cancer.
  • Embodiments of the present invention also provide articles of manufacture that can include a packaging material and a pharmaceutical composition contained within the packaging material.
  • the packaging material can comprise a label which indicates that the pharmaceutical composition can be used for treatment of one or more disorders identified above.
  • the pharmaceutical composition can include a compound according to the present invention.
  • the pharmaceutical may also contain other therapeutic agents, and may be formulated, for example, by employing conventional solid or liquid vehicles or diluents, as well as pharmaceutical additives of a type appropriate to the mode of desired administration (for example, excipients, binders, preservatives, stabilizers, flavors, etc.) according to techniques known in the art of pharmaceutical formulation.
  • the invention provides a pharmaceutical composition including a therapeutic agent and a compound of the invention. The compound is present in a concentration effective to treat cancer.
  • the compounds of the invention may be formulated into therapeutic compositions as natural or salt forms.
  • Pharmaceutically acceptable non-toxic salts include the base addition salts (formed with free carboxyl or other anionic groups) which may be derived from inorganic bases such as, for example, sodium, potassium, ammonium, calcium, or ferric hydroxides, and such organic bases as isopropylamine, trimethylamine, 2-ethylamino- ethanol, histidine, procaine, and the like.
  • Such salts may also be formed as acid addition salts with any free cationic groups and will generally be formed with inorganic acids such as, for example, hydrochloric, sulfuric, or phosphoric acids, or organic acids such as acetic, citric, p- toluenesulfonic, methanesulfonic acid, oxalic, tartaric, mandelic, and the like.
  • inorganic acids such as, for example, hydrochloric, sulfuric, or phosphoric acids
  • organic acids such as acetic, citric, p- toluenesulfonic, methanesulfonic acid, oxalic, tartaric, mandelic, and the like.
  • Salts of the invention can include amine salts formed by the protonation of an amino group with inorganic acids such as hydrochloric acid, hydrobromic acid, hydroiodic acid, sulfuric acid, phosphoric acid, and the like. Salts of the invention can also include amine salts formed by the protonation of an amino group with suitable organic acids, such as p-toluenesulfonic acid, acetic acid, methanesulfonic acid and the like. Additional excipients which are contemplated for use in the practice of the present invention are those available to those of ordinary skill in the art, for example, those found in the United States Pharmacopeia Vol. XXII and National Formulary Vol. XVII, U.S. Pharmacopeia Convention, Inc., Rockville, MD (1989), the relevant contents of which is incorporated herein by reference. In addition, polymorphs of the invention compounds are included in the present invention.
  • compositions of the invention may be administered by any suitable means, for example, orally, such as in the form of tablets, capsules, granules or powders; sublingually; buccally; parenterally, such as by subcutaneous, intravenous, intramuscular, intrathecal, or intracisternal injection or infusion techniques (e.g., as sterile injectable aqueous or non-aqueous solutions or suspensions); nasally such as by inhalation spray; topically, such as in the form of a cream or ointment; or rectally such as in the form of suppositories; in dosage unit formulations containing non-toxic, pharmaceutically acceptable vehicles or diluents.
  • suitable means for example, orally, such as in the form of tablets, capsules, granules or powders; sublingually; buccally; parenterally, such as by subcutaneous, intravenous, intramuscular, intrathecal, or intracisternal injection or infusion techniques (e.g., as
  • the present compounds may, for example, be administered in a form suitable for immediate release or extended release. Immediate release or extended release may be achieved by the use of suitable pharmaceutical compositions comprising the present compounds, or, particularly in the case of extended release, by the use of devices such as subcutaneous implants or osmotic pumps.
  • the present compounds may also be administered liposomally.
  • mammals including, but not limited to, cows, sheep, goats, horses, dogs, cats, guinea pigs, rats or other bovine, ovine, equine, canine, feline, rodent or murine species can be treated.
  • the method can also be practiced in other species, such as avian species (e.g., chickens).
  • compositions for the administration of the compounds of this embodiment may conveniently be presented in dosage unit form and may be prepared by any of the methods well known in the art of pharmacy. All methods include bringing the active ingredient into association with the carrier which constitutes one or more accessory ingredients. In general, the pharmaceutical compositions are prepared by uniformly and intimately bringing the active ingredient into association with a liquid carrier or a finely divided solid carrier or both, and then, if necessary, shaping the product into the desired formulation. In the pharmaceutical composition the active object compound is included in an amount sufficient to produce the desired effect upon the process or condition of diseases.
  • compositions containing the active ingredient may be in a form suitable for oral use, for example, as tablets, troches, lozenges, aqueous or oily suspensions, dispersible powders or granules, emulsions, hard or soft capsules, or syrups or elixirs.
  • compositions intended for oral use may be prepared according to any method known to the art for the manufacture of pharmaceutical compositions and such compositions may contain one or more agents selected from the group consisting of sweetening agents, flavoring agents, coloring agents and preserving agents in order to provide pharmaceutically elegant and palatable preparations.
  • Tablets contain the active ingredient in admixture with non-toxic pharmaceutically acceptable excipients which are suitable for the manufacture of tablets.
  • excipients may be for example, inert diluents, such as calcium carbonate, sodium carbonate, lactose, calcium phosphate or sodium phosphate; granulating and disintegrating agents, for example, corn starch, or alginic acid; binding agents, for example starch, gelatin or acacia, and lubricating agents, for example magnesium stearate, stearic acid or talc.
  • the tablets may be uncoated or they may be coated by known techniques to delay disintegration and absorption in the gastrointestinal tract and thereby provide a sustained action over a longer period.
  • a time delay material such as glyceryl monostearate Ji-" IL.
  • i'i / U '.::::» n.j ;;;; ⁇ > >• ⁇ .».* ««.• ⁇ > «. «.. » or glyceryl distearate may be employed. They may also be coated to form osmotic therapeutic tablets for control release.
  • Formulations for oral use may also be presented as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent, for example, calcium carbonate, calcium phosphate or kaolin, or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium, for example peanut oil, liquid paraffin, or olive oil.
  • an inert solid diluent for example, calcium carbonate, calcium phosphate or kaolin
  • water or an oil medium for example peanut oil, liquid paraffin, or olive oil.
  • Aqueous suspensions contain the active materials in admixture with excipients suitable for the manufacture of aqueous suspensions.
  • excipients are suspending agents, for example sodium carboxymethylcellulose, methylcellulose, hydroxy- propylmethylcellulose, sodium alginate, polyvinyl-pyrrolidone, gum tragacanth and gum acacia; dispersing or wetting agents may be a naturally-occurring phosphatide, for example lecithin, or condensation products of an alkylene oxide with fatty acids, for example polyoxyethylene stearate, or condensation products of ethylene oxide with long chain aliphatic alcohols, for example heptadecaethyleneoxycetanol, or condensation products of ethylene oxide with partial esters derived from fatty acids and a hexitol such as polyoxyethylene sorbitol monooleate, or condensation products of ethylene oxide with partial esters derived from fatty acids and hexitol anhydrides, for example polyethylene sorbitan
  • the aqueous suspensions may also contain one or more preservatives, for example ethyl, or / ⁇ -propyl, p- hydroxybenzoate, one or more coloring agents, one or more flavoring agents, and one or more sweetening agents, such as sucrose or saccharin.
  • Oily suspensions may be formulated by suspending the active ingredient in a vegetable oil, for example arachis oil, olive oil, sesame oil or coconut oil, or in a mineral oil such as liquid paraffin.
  • the oily suspensions may contain a thickening agent, for example beeswax, hard paraffin or cetyl alcohol. Sweetening agents such as those set forth above, and flavoring agents may be added to provide a palatable oral preparation. These compositions may be preserved by the addition of an anti-oxidant such as ascorbic acid.
  • Dispersible powders and granules suitable for preparation of an aqueous suspension by the addition of water provide the active ingredient in admixture with a dispersing or wetting agent, suspending agent and one or more preservatives.
  • a dispersing or wetting agent e.g., sodium EDTA
  • suspending agent e.g., sodium EDTA
  • preservatives e.g., sodium EDTA, sodium bicarbonate, sodium bicarbonate
  • Syrups and elixirs may be formulated with sweetening agents, for example glycerol, propylene glycol, sorbitol or sucrose. Such formulations may also contain a demulcent, a preservative and flavoring and coloring agents.
  • sweetening agents for example glycerol, propylene glycol, sorbitol or sucrose.
  • Such formulations may also contain a demulcent, a preservative and flavoring and coloring agents.
  • compositions may be in the form of a sterile injectable aqueous or oleagenous suspension.
  • This suspension may be formulated according to the known art using those suitable dispersing or wetting agents and suspending agents which have been mentioned above.
  • the sterile injectable preparation may also be a sterile injectable solution or suspension in a parenterally-acceptable diluent or solvent or cosolvent or complexing agent or dispersing agent or excipient or combination thereof, for example 1,3-butanediol, polyethylene glycols, polypropylene glycols, ethanol or other alcohols, povidones, various brands of TWEEN surfactant, sodium dodecyl sulfate, sodium deoxycholate, dimethylacetamide, polysorbates, poloxamers, cyclodextrins, lipids, and excipients such as inorganic salts (e.g., sodium chloride), buffering agents (e.g., sodium citrate, sodium phosphate), and sugars (e.g., saccharose and dextrose).
  • a parenterally-acceptable diluent or solvent or cosolvent or complexing agent or dispersing agent or excipient or combination thereof for example 1,3-butan
  • Suitable vehicles and solvents that may be employed are water, dextrose solutions, Ringer's solutions and isotonic sodium chloride solution.
  • sterile, fixed oils are conventionally employed as a solvent or suspending medium.
  • any bland fixed oil may be employed including synthetic mono- or diglycerides.
  • fatty acids such as oleic acid find use in the preparation of injectables.
  • these pharmaceutical compositions may be formulated and administered systemically or locally. Techniques for formulation and administration may be found in the latest edition of "Remington's Pharmaceutical Sciences” (Mack Publishing Co, Easton Pa.). Suitable routes may, for example, include oral or transniucosal administration; as well as parenteral delivery, including intramuscular, subcutaneous, intramedullary, intrathecal, intraventricular, intravenous, intraperitoneal, or intranasal administration.
  • the pharmaceutical compositions of the invention may be formulated in aqueous solutions, preferably in physiologically compatible buffers such as Hanks' solution, Ringer's solution, or physiologically buffered saline.
  • penetrants appropriate to the particular barrier to be permeated are used in the formulation. Such penetrants are generally known in the art.
  • Pharmaceutical formulations for parenteral administration include aqueous solutions of the active compounds If" «... ii in water-soluble form.
  • suspensions of the active compounds may be prepared as appropriate oily injection suspensions.
  • Suitable lipophilic solvents or vehicles include fatty oils such as sesame oil, or synthetic fatty acid esters, such as ethyl oleate or triglycerides, or liposomes.
  • Aqueous injection suspensions may contain substances that increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, or dextran.
  • the suspension may also contain suitable stabilizers or agents that increase the solubility of the compounds to allow for the preparation of highly concentrated solutions.
  • compositions of the present invention may also be administered in the form of suppositories for rectal administration of the drug.
  • suppositories for rectal administration of the drug.
  • These compositions can be prepared by mixing the drug with a suitable non-irritating excipient which is solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum to release the drug.
  • suitable non-irritating excipient which is solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum to release the drug.
  • Such materials are cocoa butter and polyethylene glycols.
  • creams, ointments, jellies, solutions or suspensions, etc., containing the compounds of the present invention are employed.
  • topical application shall include mouthwashes and gargles).
  • the invention compounds are administered in combination with an anti-inflammatory agent, antihistamines, chemotherapeutic agent, immunornodulator, therapeutic antibody or a protein kinase inhibitor, e.g., a tyrosine kinase inhibitor, to a subject in need of such treatment.
  • chemotherapeutic agents include antimetabolites, such as methotrexate, DNA cross-linking agents, such as cisplatin/carboplatin; alkylating agents, such as canbusil; topoisomerase I inhibitors such as dactinomicin; microtubule inhibitors such as taxol (paclitaxol), and the like.
  • chemotherapeutic agents include, for example, a vinca alkaloid, mitomycin-type antibiotic, bleomycin-type antibiotic, antifolate, colchicine, demecoline, etoposide, taxane, anthracycline antibiotic, doxorubicin, daunorubicin, carminomycin, epirubicin, idarubicin, mithoxanthrone, 4-dimethoxy-daunomycin, 11-deoxydaunorubicin, 13-deoxydaunorubicin, adriamycin- 14-benzoate, adriamycin- 14-octanoate, adriamycin- 14-naphthaleneacetate, amsacrine, carmustine, cyclophosphamide, cytarabine, etoposide, lovastatin, melphalan, topetecan, oxalaplatin, chlorambucil, meth
  • therapeutic antibodies include antibodies directed against the HER2 protein, such as trastuzumab; antibodies directed against growth factors or growth factor receptors, such as bevacizumab, which targets vascular endothelial growth factor, and OSI-774, which targets epidermal growth factor; antibodies targeting integrin receptors, such as Vitaxin (also known as MEDI-522), and the like.
  • Classes of anticancer agents suitable for use in compositions and methods of the present invention include, but are not limited to: 1) alkaloids, including, microtubule inhibitors (e.g., Vincristine, Vinblastine, and Vindesine, etc.), microtubule stabilizers (e.g., Paclitaxel [Taxol], and Docetaxel, Taxotere, etc.), and chromatin function inhibitors, including, topoisomerase inhibitors, such as, epipodophyllotoxins (e.g., Etoposide [VP-16], and Teniposide [VM-26], etc.), and agents that target topoisomerase I (e.g., Camptothecin and Isirinotecan [CPT-Il], etc.); 2) covalent DNA-binding agents [alkylating agents], including, nitrogen mustards (e.g., Mechlorethamine, Chlorambucil, Cyclophosphamide, Ifosphamide, and Busulfan [
  • composition and method of the present invention may further comprise other therapeutically active compounds as noted herein which are usually applied in 005/030614
  • cyclosporins e.g., cyclosporin A
  • CTLA4-Ig antibodies such as ICAM-3, anti-IL-2 receptor (Anti-Tac), anti-CD45RB, anti-CD2, anti-CD3 (OKT-3), anti- CD4, anti-CD80, anti-CD86, agents blocking the interaction between CD40 and gp39, such as antibodies specific for CD40 and/or gp39 (i.e., CD154), fusion proteins constructed from CD40 and gp39 (CD40Ig and CD8gp39), inhibitors, such as nuclear translocation inhibitors, of NF-kappa B function, such as deoxyspergualin (DSG), cholesterol biosynthesis inhibitors such as HMG CoA reductase inhibitors (lovastatin and simvastat
  • cytokine encompasses chemokines, interleukins, lymphokines, monokines, colony stimulating factors, and receptor associated proteins, and functional fragments thereof.
  • functional fragment refers to a polypeptide or peptide which possesses biological function or activity that is identified through a defined functional assay.
  • the cytokines include endothelial monocyte activating polypeptide II (EMAP-II), granulocyte-macrophage-CSF (GM-CSF), granulocyte-CSF (G-CSF), macrophage-CSF (M- CSF), IL-I, IL-2, IL-3, IL-4, IL-5, IL-6, IL-12, and IL-13, interferons, and the like and which is associated with a particular biologic, morphologic, or phenotypic alteration in a cell or cell mechanism.
  • EMP-II endothelial monocyte activating polypeptide II
  • GM-CSF granulocyte-macrophage-CSF
  • G-CSF granulocyte-CSF
  • M- CSF macrophage-CSF
  • IL-I IL-2, IL-3, IL-4, IL-5, IL-6, IL-12, and IL-13
  • interferons interferons
  • an appropriate dosage level can generally be between about 0.01 and about 1000 mg per 1 kg of patient body weight per day which can be administered in single or multiple doses.
  • the dosage level can be between about 0.01 and about 250 mg/kg per day; more narrowly, between about 0.5 and about 100 mg/kg per day.
  • a suitable dosage level can be between about 0.01 and about 250 mg/kg per day, between about 0.05 and about 100 mg/kg per day, or between about 0.1 and about 50 mg/kg per day, or about 1.0 mg/kg per day.
  • the dosage can be between about 0.05 and about 0.5 mg/kg per day, or between about 0.5 and about 5 mg/kg per day, or between about 5 and about 50 mg/kg per day.
  • the compositions can be provided in the form of tablets containing between about 1.0 and about 1,000 mg of the active ingredient, for example, about 1.0, about 5.0, about 10.0, about 15.0, about 20.0, about 25.0, about 50.0, about 75.0, about 100.0, about 150.0, about 200.0, about 250.0, about 300.0, about 400.0, about 500.0, about 600.0, about 750.0, about 800.0, about 900.0, and about 1,000.0 mg of the active ingredient for the symptomatic adjustment of the dosage to the patient to be treated.
  • the compounds can be administered on a regimen of 1 to 4 times per day, such as once or twice per day. There may be a period of no administration followed by another regimen of administration.
  • Compounds of the present invention can be used, alone or in combination with an effective amount of a therapeutic antibody or chemically attached to a tumor tissue targeting antibody (or therapeutic fragment thereof), a chemotherapeutic or an immunotoxic agent, for treatment of tumors.
  • chemotherapeutic agents that can be used for this purpose include doxorubicin, docetaxel, or taxol.
  • the invention includes combination therapy including a compound of the invention, including but not limited to vasculostatic agents, such as tyrosine, serine or threonine kinase inhibitors, for example, Src-family inhibitors, and any chemotherapeutic agent or therapeutic antibody.
  • the present invention also provides screening assays using appropriate cells which express any kinases within the MAPK pathway.
  • Such cells include cells from mammals, yeast, Drosophila or E. coli.
  • the cells which are contacted with the candidate compound are compared with the same cells which are not contacted for Raf polypeptide or MAPK pathway polypeptide activity
  • This invention contemplates the treatment and/or amelioration of such diseases by administering a MAPK pathway polypeptides inhibiting amount of a compound.
  • a MAPK pathway polypeptides inhibiting amount of a compound is believed that among the useful inhibitors of MAPK pathway polypeptides function are those compounds which inhibit the kinase activity of the MAPK pathway polypeptides. Other sites of inhibition are, of course, possible owing to its position in a signal transduction cascade. Inhibitors of protein-protein interactions between, for example Raf polypeptide or MAPK pathway polypeptides and other factors could lead to the development of pharmaceutical agents for the modulation of Raf polypeptide or MAPK pathway polypeptides activity.
  • the assays of the invention may test binding of a candidate compound wherein adherence to the cells bearing the Raf polypeptide or MAPK pathway polypeptides is detected by means of a label directly or indirectly associated with the candidate compound or in an assay involving competition with a labeled competitor. Further, these assays may test whether the candidate compound results in a signal generated by activation of MAPK pathway polypeptides, using detection systems appropriate to the cells bearing the Raf polypeptide or MAPK pathway polypeptides.
  • Inhibitors of activation are generally assayed in the presence of a known agonist and the effect on activation by the agonist by the presence of the candidate compound is observed. Standard methods for conducting such screening assays are well understood in the art and are also illustrated in the examples below (e.g., direct and raf 1 -MEKl assays or MAPK pathway cellular assays).
  • Example 1 describes general synthetic procedures that were used to make the compounds described in the subsequent examples. AU solvents were used without further purification. Reactions can be usually conducted without an inert gas atmosphere unless specified otherwise. The reported yields are based on unoptimized conditions and single test runs. The yields can be optimized by changing the reaction conditions, such as solvent, use of base or acid, temperature, use of catalyst and the time of the reaction. Microwave reactions were run in EmrysTM Process vials (2-5 mL) using Initiator module (Biotage/Personal chemistry). All 1 H NMR were run on a 500 MHz Bruker NMR or Bruker Avance 400 MHz NMR.
  • a Waters LC/MS system is used in identity and purity analysis. This system includes a 2795 separation module, a 996 photodiode array detector and a ZQ2000 mass spectrometer. A Zorbax SB column (150 x 4.6 mm 3.5 ⁇ , Agilent Technologies) was used for the LC. Column temperature was 4O 0 C. Compounds were separated using gradient elution with mobile phases of water (0.05% TFA (A)) and acetonitrile (0.05% TFA (B)). Flow rate was lmL/min. The gradient program used in separation was 0-15 min: 5-60 % B; 15-15.5 min: 60-100 % B; 15.5-17 min: 100 % B.
  • reaction mixture changed colors from green to orange, then to purple over 2 hours, then resulted in an orange solution with a yellow precipitate. It was cooled down to ambient temperature and 150 mL of anhydrous toluene was added. The suspension was concentrated to about 50 mL total on rotovap. This process was repeated three times.
  • the formed precipitate was collected by filtration, washed with 50 mL of water, 50 mL of methanol, 50 mL of diethyl ether and dried in vacuum to produce 3.22 g of the product as a dirty-yellow solid.
  • the filtrate was extracted four times with 100 mL of ethyl acetate.
  • the combined extracts were washed 3 times with 100 mL of water, then with brine and dried over anhydrous sodium sulfate.
  • Solvent was removed in vacuum to yield additional 1.2 g of the product in a form of a yellow solid. Yield: 75% combined.
  • reaction P 1 C 1 / IJ hi U TM ii / -3 U O , .JL -ir mixture was filtered through 0.22 ⁇ syringe filter and purified by reverse-phase preperative HPLC using acetonitrile/water mixture with 0.1 % of TFA.
  • reaction mixture was filtered through 0.22 ⁇ syringe filter and purified by reverse-phase prep-HPLC using acetonitrile/water mixture with 0.1 % of TFA as a solvent system.
  • the Rafl -MEKl assay utilized 2.9U/well rafl and 0.25 ug/well inactive MEKl (MEKl inactive, Upstate, Lake Placid, NY) and 3uM ATP. After reacting for 60 minutes at 30 0 C, residual ATP was measured using a luciferase-based assay (KinaseGlo, Promega Corp.) as a measure of kinase activity. Data from four wells were then averaged and used to determine IC 5 O values for the test compounds (Prism software package, GraphPad Software, San Diego CA).
  • test results were as follows: a known Raf inhibitor, compound A, displayed an IC50 of 16 nM; a known Raf inhibitor, compound B, displayed an IC 50 of 43 nM; and an invention compound C, showed an IC50 of 76 nM.
  • HUVECs were treated with 50 ng/ml VEGF or FGF (Peprotech, Rocky Hill, NJ) for 6 min and the cells were immediately washed with ice-cold PBS.
  • Cells were lysed with ice-cold RIPA buffer containing 10OmM Tris pH 7.5, 150 mM NaCl, 1 niM EDTA, 1% deoxycholate, 1% Triton X-100, 0.1% SDS, 2 mM PMSF, one Complete-Mini protease inhibitor tablet (Roche, Indianapolis, IN; 1 tablet/ 7 ml of lysis buffer) and the phophatase inhibitors NaF (500 mM) and orthovanadate (1 mM).
  • the cells were scraped and lysates transferred and centrifuged at 15,000 g for 10 min. Supernatants were transferred to new tubes and protein concentration was quantitated using the BCA protein reagent (Pierce, Rockford, IL). Cell lysates containing 20 ⁇ g of total protein were separated by 10% SDS-PAGE, transferred to nitrocellulose, and blocked in 5% milk in TBST.
  • Anti phospho-ERK Thr 202/Tyr 204 (Cell Signaling, Beverly, MA), anti-phospho-MEK Ser217/221 (Cell Signaling), and c-Raf (BD Biosciences Pharmingen, San Diego, CA) used as primary antibodies were detected with horseradish peroxidase-conjugated goat anti-mouse or rabbit secondary antibodies and bands were visualized using the SuperSignal West Pico chemiluminescence reagent system (Pierce) and Kodak X-ray film (Rochester, NY).
  • Bay 43-9006 (Raf/FGF inhibitor) showed reduction of expression of p-MEK and p-ERK with IC50 between 200 and 300 nM when tested in this assay.
  • UO 126 (MEK inhibitor) showed reduction in p-Erk levels with IC 50 between 200 and 300 nM, while p- MEK levels were unaffected.
  • Table 1 As can be seen, compounds of the invention showed reduction in p-MEK and p-ERK levels with IC50 between 400 nM and 20 ⁇ M.
  • XTT assay HUVECs were seeded at 10,000 cells/well of a tissue culture treated 96-well plate treated with collagen type I and grown overnight in the complete EGM-2 media as described above. The following morning, the inhibitors were serial diluted with DMSO and added to the cells with a final DMSO concentration of 1%. After 24-48 hours cell viability was measured with an XTT assay (Sigma, St. Louis, MO). The cells were also photographed to compare morphological differences to the XTT trends observed. Determination of the IC 50 values was performed with quantitative software (Prism software package, GraphPad Software, San Diego CA). Several inhibitors blocked cell proliferation and induced apoptosis at concentrations below 1 ⁇ M and experiments were repeated three times to confirm the observations. The compounds of the invention displayed IC50 between 100 nM and 40 uM in this assay (Table 1).
  • reaction mixture was diluted with ca. 10 mL of MeOH, transferred into a round- bottom flask and concentrated in vacuo to ca. 20 mL.
  • the resulting reddish solution was loaded on a short pad of silica gel and eluted first with 100 % ethyl acetate to remove the unreacted starting material and then with 20 % MeOH in EtOAc to elute the product.
  • Tl ⁇ e product was further purified by ISCO system (80 g pre-packed column, 40 min method, 0% to 10 % MeOH gradient in ethyl acetate). Solvent was removed in vacuo to give the title product as an off-white solid (0.785 g). Yield 46.3 %.
  • reaction mixture was diluted with ca. ImL of MeOH, filtered through 0.22 um syringe filter and purified by reverse-phase preparative HPLC in acetonitrile/water system with 0.01% TFA.
  • the product was isolated as a TFA salt (46.6 mg).
  • the impregnated silica gel was loaded into 25 g ISCO sample cartridge and the product was purified using ISCO system (solid method, 80 g column, 45 min, 0% to 50% EtOAc gradient in hexane). Solvent was removed in vacuo to give the title product as a white solid (3.83 g). Yield 47.6%.
  • reaction mixture was diluted with ca. ImL of MeOH, filtered through 0.22 um syringe filter and purified by reverse-phase preparative HPLC in acetonitrile/water system with 0.01% TFA.
  • the product was isolated as a TFA salt (31.7 mg of white solid).
  • reaction mixture was diluted with ca. ImL of MeOH, filtered through 0.22 um syringe filter and purified by reverse-phase preparative ⁇ PLC in acetonitrile/water system with 0.01% TFA.
  • the product was isolated as a TFA salt (34.6 mg of beige solid).
  • the vial was capped and microwaved at 250 0 C for 20 min. Then the reaction mixture was diluted with ca. ImL of MeOH, filtered through 0.22 um syringe filter and purified by reverse-phase preparative ⁇ PLC in acetonitrile/water system with 0.01% TFA. The product was isolated as a TFA salt (35.5 mg of light-brown solid).
  • the filtrate was mixed with 10 g of dry silica gel and solvent was removed in vacuo.
  • the impregnated silica gel was loaded on silica gel column and the product was separated using a gradient of hexane: ethyl acetate mixture starting from 50:50 ratio and finishing at 0: 100. All fractions containing the product were combined; solvent was removed in vacuo to give a grey solid.
  • the solid was heated in 50 mL of 4:1 mixture of EtOAc/MeOH. The formed suspension was cooled down to ambient temperature and filtered to give the title product as a white crystalline solid (4.54 g, 60.7 % yield).
  • reaction mixture was microwaved at 200 0 C for 25 min (Initiator, Biotage). Then it was diluted with 1 mL of MeOH, filtered through 0.22 urn syringe filter and purified by preparative reverse- phase chromatography using acetonitrile/water gradient with 0.1 % TFA. The major peak having the mass of the product was collected; solvent was removed in vacuo to give the title product as a white fluffy solid (85.6 mg).
  • reaction mixture was diluted with ca. 1 mL of MeOH, filtered through 0.22 um syringe filter and purified by reverse-phase preparative HPLC in acetonitrile/water system with 0.01% TFA.
  • Fractions, containing the product were partitioned between ca. 40 mL of EtOAc and ca. 40 mL of saturated NaHCO 3 .
  • EtOAc layer was washed with brine, dried over anhydrous Na 2 SO 4 and filtered. Solvent was removed in vacuo to give the title product as a beige solid (61.6 mg).
  • reaction mixture was diluted with ca. 1 rnL of MeOH, filtered through 0.22 um syringe filter and purified by reverse-phase preparative HPLC in acetonitrile/water system with 0.01% TFA.
  • Fractions, containing the product were partitioned between ca. 50 niL of EtOAc and ca. 50 mL of saturated aqueous NaHCO 3 .
  • EtOAc layer was washed with brine, dried over anhydrous Na 2 SO 4 and filtered. Solvent was removed in vacuo to give the title product as a light-yellow solid (74.7 mg). Yield 40.8%.
  • N-amino-N'- [4-chloro-3 -(trifluoromethyl)phenyl] -guanidine hydroiodide can be reacted with about 1.0-1.5 equivalents of 4-hydroxy-phenylglyoxale in 1:1 mixture of methanol/water to yield the title product.
  • the product can be isolated by precipitation or by extraction with a number of solvents, such as ethyl acetate, methylene chloride or diethyl ether or by silica gel column chromatography.
  • the aqueous layer can be extracted with a number of solvents, such as ethyl acetate, methylene chloride or ether.
  • solvents such as ethyl acetate, methylene chloride or ether.
  • the combined extracts can be washed 3 times with 100 mL of water, then with brine and dried over anhydrous sodium sulfate.
  • Solvent can be removed in vacuum to yield crude 4-[4-(3-phenylamino- [l,2,4]triazin-6-yl)-phenoxy]-pyridine-2-carboxylic acid methylamide.
  • the product can be than purified using silica gel column chromatography. Those having ordinary skill in the art can determine which solvent system can be used as an eluents in the chromatographic purification.
  • 1.0-1.5 equivalent of 4-hydroxyphenylglyoxale can be reacted with 1-amino-S- ethylisothiourea hydrobromide in 1:1 mixture of methanol/water with 1.0-2.0 equivalent of K 2 CO 3 to yield 4-(3-ethylsulfanyl-[l,2,4]triazin-5-yl)-phenol.
  • the product can be isolated by a number of methods known to one skilled in the art, such as precipitation or by extraction with a number of solvents, such as ethyl acetate, methylene chloride or diethyl ether or by silica gel column chromatography.
  • the aqueous layer can be extracted with a number of solvents, such as ethyl acetate, methylene chloride or ether.
  • solvents such as ethyl acetate, methylene chloride or ether.
  • the combined extracts can be washed 3 times with 100 mL of water, then with brine and dried over anhydrous sodium sulfate.
  • Solvent can be removed in vacuum to give crude 4-[4-(3-amino-[l,2,4]triazin-5-yl)-phenoxy]-N-methylpyridine-2- carboxamide.
  • the product can be than purified using silica gel column chromatography. Those having ordinary skill in the art can determine which solvent system can be used as an eluents in the chromatographic purification.
  • the product can be isolated by a number of methods known to those skilled in the art, such as precipitation or by extraction with a number of solvents, such as ethyl acetate, methylene chloride or diethyl ether or by silica gel column chromatography, or by reverse-phase prep-HPLC chromatography.
  • Example 104 Synthesis of 4- ⁇ 4-r3-( " ⁇ r4-chloro-3-(trifluoromethyl)phenyllsulfonyl>ammoV r 1 ,2,41triazin-5-vlVphenoxv> -N-methylpyridine-2-carboxamide
  • the product can be isolated by a number of methods known to one skilled in the art, such as precipitation or by extraction with a number of solvents, such as ethyl acetate, methylene chloride or diethyl ether or by silica gel column chromatography, or by reverse-phase preparative HPLC.
  • solvents such as ethyl acetate, methylene chloride or diethyl ether or by silica gel column chromatography, or by reverse-phase preparative HPLC.
  • HUVECs Early passage primary human umbilical vein endothelial cells (HUVECs) were maintained in EGM-2 containing SingleQuots (Cambrex, East Rutherford, NJ), 10% FBS, 1OmM HEPES, and 50 ⁇ g/ml gentamicin. Prior to treatment of the cells with inhibitor, the HUVECs were starved for 18h by replacing serum-containing complete media with serum-free and SingleQuot-free media. The starved cells were pre-treated with inhibitors for 60 min at various concentrations (0-20 ⁇ M).
  • HUVECs were treated with 50 ng/ml VEGF or FGF (Peprotech, Rocky Hill, NJ) for 6 min and the cells were immediately washed with ice-cold PBS.
  • Cells were lysed with ice- cold RIPA buffer containing 10OmM Tris pH 7.5, 150 mM NaCl, 1 mM EDTA, 1% deoxycholate, 1% Triton X-100, 0.1% SDS, 2 mM PMSF, one Complete-Mini protease inhibitor tablet (Roche, Indianapolis, IN; 1 tablet/ 7 ml of lysis buffer) and the phophatase inhibitors NaF (500 mM) and orthovanadate (1 mM).
  • the cells were scraped and lysates transferred and centrifuged at 15,000 g for 10 min. Supernatants were transferred to new tubes and protein concentration was quantitated using the BCA protein reagent (Pierce, Rockford, IL). Cell lysates containing 20 ⁇ g of total protein were separated by 10% SDS- PAGE, transferred to nitrocellulose, and blocked in 5% milk in TBST.
  • Anti phospho-ERK Thr 202/Tyr 204 (Cell Signaling, Beverly, MA), anti-phospho-MEK Ser217/221 (Cell Signaling), and c-Raf (BD Biosciences Pharmingen, San Diego, CA) used as primary antibodies were detected with horseradish peroxidase-conjugated goat anti-mouse or rabbit secondary antibodies and bands were visualized using the SuperSignal West Pico chemiluminescence reagent system (Pierce) and Kodak X-ray film (Rochester, NY).
  • Bay 43-9006 (Raf/FGF inhibitor) showed reduction of expression of p-MEK and p-ERK with IC50 between 200 and 300 nM when tested in this assay.
  • U0126 (MEK inhibitor) showed reduction in p-Erk levels with IC 50 between 200 and 300 nM, while p- MEK levels were unaffected.
  • Table 1 As can be seen, compounds of the invention showed reduction in p-MEK and p-ERK levels with IC 50 between 400 nM and 20 ⁇ M.
  • XTT assay HUVECs were seeded at 10,000 cells/well of a tissue culture treated 96-well plate treated with collagen type I and grown overnight in the complete EGM-2 media as described above. The following morning, the inhibitors were serial diluted with DMSO and added to the cells with a final DMSO concentration of 1%. After 72 hours cell viability was measured with an XTT assay (Sigma, St. Louis, MO). The cells were also photographed to compare morphological differences to the XTT trends observed. Determination of the IC50 values was performed with quantitative software (Prism software package, GraphPad Software, San Diego CA).

Abstract

Heterocyclic compounds derived from benzotriazine, triazines, triazoles and oxadiazoles are disclosed. The methods of synthesis and of use of such heterocyclic compounds are also provided.

Description

HETEROCYCLIC COMPOUNDS AND METHODS OF USE
CROSS REFERENCE TO RELATED APPLICATIONS
[0001] This application claims the benefit of priority under 35 U.S.C. § 119(e) of patent applications U.S. Serial Number 60/604,298 filed August 25, 2004, and 60/696,168 filed July 1, 2005, the entire content of which is incorporated herein by reference.
FBELD OF THE INVENTION
[0002] The present invention relates generally to the use of compounds to treat a variety of disorders, diseases and pathologic conditions and more specifically to the use of various heterocyclic compounds for therapeutic purposes.
BACKGROUND
[0003] Kinases are a large family of cellular proteins involved in signal transduction of cascades which control cell growth and death, survival, migration, differentiation, gene expression, metabolism, protein synthesis and cell cycle regulation. A common mechanism by which these signals are transmitted is reversible phosphorylation, which induces conformational changes is these enzymes and alters their structure and function. The entire kinase genome discovered so far incorporates over 500 individual proteins and their isoforms. Different branches of this genomic tree have been characterized into groups specific for phosphorylating either serine/threonine residues or tyrosines. Some kinases exhibit dual specificity, capable of substrate phosphorylation of tyrosine as well as serine/threonines. Further differentiation can be made in terms of their location in cells. Transmembrane receptor protein kinases exhibit an extracellular domain, capable of ligand binding. These ligand binding mechanisms trigger activation of the kinase catalytic domain which initiates a cascade of signals that controls intracellular functions. Examples of a receptor protein kinase are growth factors such as EGF, FGF, PDGF and IGF. Nonreceptor protein kinases can be found in many compartments of a cell from inner-cell surface membranes to the cell nucleus. One example of a nonreceptor protein kinase is the mitogen activated protein kinase (MAPK) which regulates a pathway, which is important in cell signaling initiated on the exterior cell surfaces via growth factors, for example, VEGF, or hormones, and extending to the cell nucleus by activating transcription factors. These nuclear factors in turn control gene expression in the regulation of cell cycle progression and ultimately cell proliferation, and differentiation. [0004] The MAPK cell signaling pathway is important for drug targeting as this path impinges on nearly all functional hallmarks of cancer cells such as immortalization, growth factor independent proliferation, insensitivity to growth inhibitory signals, metastasis, blood vessel attraction, evasion of apoptosis, and other functional hallmarks. Inappropriate activation though mutation of this molecule is associated with nearly 30% of all human cancers. In general, the inhibition of disregulated kinases such as Ras, PBK and Raf is an important approach to discover novel treatments for cancer and other diseases. One approach is the discovery of small molecules capable of binding either to the kinase catalytic domain or a regulatory domain in order to modulate the function of protein kinases. Important in this respect is to discover molecules which inhibit a specific signaling path with a high degree of selectivity and a potency within a practical therapeutic window. While significant progress has been made in developing various compounds for the treatment of cancer and inflammatory diseases, there remains a need for specific chemical structures capable of modulating protein kinases, whose disregulated function has been implicated in these diseases.
SUMMARY OF THE INVENTION
[0005] The present invention provides compounds which affect the MAPK pathway. The compounds of the invention are useful as pharmaceutical compositions, for example where modulation of the MAPK pathway is indicated for the treatment of various human diseases, such as cancer.
[0006] According to one embodiment of the invention, compounds having the structure (A) are provided, or an N-oxide, N,N'-dioxide, N,N',N"-trioxide, or a pharmaceutically acceptable salt thereof:
Figure imgf000003_0001
(A)
wherein Y can be absent or c*an be one of the following moieties:
Figure imgf000004_0001
[0007] According to another embodiment of the invention, compounds having the structure (B) are provided, or an N-oxide, N,N'-dioxide, N,N',N"-trioxide, or a pharmaceutically acceptable salt thereof:
Figure imgf000004_0002
(B)
wherein X can be absent or can be NH, and Y can be absent or can be one of the following moieties:
Figure imgf000004_0003
[0008] In compounds having structure (B), each Of Z1, Z2 and Z3 can be, independently, N, N=CH, CH, O, S or N-R4, wherein R4 is hydrogen or lower alkyl, with the further proviso that at least one Of Z1, Z2 and Z3 is not CH.
[0009] In compounds having structure (A), the substituted R1 can be an aryl, a substituted aryl, a heterocycle, a heteroaryl, a substituted heterocycle, and a substituted heteroaryl. For example, R1 can be one of C6-Cj2 aryl; C3-Ci2 heteroaryl having 1-3 heteroatoms such as N, S and O; substituted C3-CiO cycloalkyl having 0-3 heteroatoms such as N, S, and O; substituted C6-Ci2 aryl; substituted C3-Ci2 heteroaryl having 1-3 heteroatoms such as N, S and O; C7-C24 aralkyl; C7-C24 alkylaryl; substituted C7-C24 aralkyl; and substituted C7-C24 alkaryl.
[0010] In compounds having structure (B), the substitutent Ri can be, independently of the substitutent Ri present in the structure (A), an unsubstituted or a substituted C3-C12 heteroaryl having 1-3 heteroatoms such as N, S or O. The substituent Ri that can be present in compounds having structure (B), can include a substituted pyridyl group. The substituents in the substituted pyridyl group can include an amido moiety, an aminoalkyl group (e.g., aminomethyl), or a carboxyl group, or a carboxylate group. The amido moiety attached to the pyridyl group can be in turn also substituted by attaching to the nitrogen in the amido moiety a substitutent selected from an alkyl (e.g., methyl), an alkylaminoalkyl (e.g., diethylamino alkyl), a pyridyl, an alkyl pyrrolidine, an alkyl morpholine, and an alkyl piperazine groups.
[0011] Some examples of the substitutent R1 that can be present in compounds having either structure (A) or structure (B), can be selected from one of the following moieties:
Figure imgf000005_0001
(IH7 W7 IJH2
Figure imgf000006_0001
Figure imgf000007_0001
where n can be an integer selected from a group consisting of 0, 1, 2, and 3.
[0012] In compounds having structure (A) or (B), R2 can be, independently, any one of hydrogen, halogen, C1-C18 alkyl (e.g., methyl), -OH, -NO2, -CN, Ci-C18 alkoxy (e.g., methoxy), -NHSO2R5, -SO2NHR5, -NHCOR5, -NH2, -NR5R6 , -S(O)R5, -S(O)2R5, -CO2R5, - CONR5R6, and where R5 and R6 are independently selected from hydrogen, a Ci-Ci2 alkyl and a substituted Ci-Ci2 alkyl.
[0013] In compounds having structure (A), the substituent R3 can be an aryl, a substituted aryl, a heterocycle, a heteroaryl, a substituted heterocycle, and a substituted heteroaryl. For example, R3 can be one of Ce-Ci2 aryl; C3-Ci2 heteroaryl having 1-3 heteroatoms such as N, S and O; substituted C3-Ci0 cycloalkyl having 0-3 heteroatoms such as N, S, and O; substituted C6-C12 aryl; substituted C3-Ci2 heteroaryl having 1-3 heteroatoms such as N, S and O; C7-C24 aralkyl; C7-C24 alkylaryl; substituted C7-C24 aralkyl; and substituted C7-C24 alkaryl.
[0014] In compounds having structure (B), the substitutent R3 can be, independently of the substitutent R3 present in the structure (A), hydrogen, a Ci-Ci8 alkyl, a substituted Ci-C18 alkyl, a Ci-Ci2 cycloalkyl, a substituted Ci-Ci2 cycloalkyl, a substituted C3-CiO cycloalkyl having 0-3 heteroatoms such as N, S, or O, an aryl such as a C6-Ci2 aryl, a substituted aryl such as a substituted C6-Ci2 aryl, a heterocycle, a substituted heterocycle, a heteroaryl such as a C3-Ci2 heteroaryl having 1-3 heteroatoms such as N, S or O, a substituted heteroaryl such as substituted C3-Ci2 heteroaryl having 1-3 heteroatoms such as N, S or O, a C7-C24 aralkyl, a substituted C7-C24 aralkyl, a C7-C24 alkylaryl, and a substituted C7-C24 alkaryl. Some particular examples of the substituent R3 than can be used include tert-butyl phenyl, trifluoromethoxyphenyl, methoxyphenyl, dimethylarninophenyl, aminophenyl, trifluoroethoxyphenyl, trifluoromethoxychlorophenyl, trifluoromethoxybromophenyl, trifluoroethoxychlorophenyl, chlorophenyl, dichlorophenyl, trifluoromethyl phenyl, trifluoromethylchlorophenyl, chlorotoluyl, N-phenylacetaniide, N,N-alkyl-benzamide, isopropoxyphenyl, alkoxyphenyl, dialkoxyphenyl, or acetylphenyl.
[0015] To summarize, some examples of the substitutent R3 that can be present in compounds having either structure (A) or structure (B), can be selected from one of the following moieties:
Figure imgf000008_0001
Figure imgf000008_0003
Figure imgf000008_0002
NIUIe2
Figure imgf000008_0004
Figure imgf000008_0005
Figure imgf000009_0001
Figure imgf000010_0001
Figure imgf000011_0001
[0016] According to another embodiment of the invention, compounds that are derivatives of benzotriazine are provided, the compounds including a benzotriazine moiety having at least a first substituent attached to the benzene ring of benzotriazine and a second substituent attached to the triazine ring of the benzotriazine, where the first substituent includes a substituted pyridyl group, and the second substituent includes a secondary amino group, a substituted amide group, or a substituted sulfonylamino group.
[0017] According to yet another embodiment of the invention, compounds including a benzene-derived moiety bridged to a heterocyclic moiety are provided, where the benzene- derived moiety includes a molecule of benzene substituted with either a sulfonyl group or a pyridyl group connected to the benzene molecule via an oxygen link, and the heterocyclic moiety includes triazole, oxadiazole, oxazole, pyrazol, imidazole, thiadiazole and triazine.
[0018] According to yet another embodiment, articles of manufacture are provided, the articles including packaging material and a pharmaceutical composition contained within the packaging material, wherein the packaging material includes a label which indicates that the pharmaceutical composition can be used for treatment of disorders associated with cancer. The pharmaceutical composition can include at least one compound set forth in Structures (A) and (B) or any combination thereof.
[0019] According to another embodiment, a method for treating a disorder including administering to a subject in need thereof an effective amount of a compound, wherein the compound is set forth in Structures (A) and (B) or any combination thereof.
[0020] In one aspect, the disorder is cancer, eye disease, inflammation, psoriasis, or a viral infection, for example. More particularly, the cancer is an alimentary/gastrointestinal tract cancer, colon cancer, liver cancer, skin cancer, breast cancer, ovarian cancer, prostate cancer, lymphoma, leukemia, kidney cancer, lung cancer, muscle cancer, bone cancer, bladder cancer or brain cancer.
[0021] According to yet another embodiment, a pharmaceutical composition is provided, at least one compound set forth in structures (A) and (B) or any combination thereof, in a pharmaceutically acceptable carrier. BRIEF DESCRIPTION OF THE DRAWINGS
[0022] FIG. 1 shows the results of the Rafl direct assay of invention compounds
[0023] FIG. 2 shows the results of XTT cell viability assay of invention compounds
DETAILED DESCRIPTION OF THE INVENTION
[0024] The present invention is directed to heterocyclic compounds, such as heterocyclic compounds derived from benzotriazine, triazines, triazoles, oxadiazoles, imidazoles and tliiadiazole and to use of the heterocyclic compounds for therapeutic purposes. [0025] The following terminology and definitions apply as used in the present application, generally in conformity with the terminology recommended by the International Union of Pure and Applied Chemistry (IUPAC).
[0026] The term "heterocyclic," when used to describe an aromatic ring, means that the aromatic ring contains at least one heteroatom. The abbreviation "Het" is sometimes used to signify a heterocyclic structure.
[0027] The term "heteroatom" is defined to include any atom other than carbon, for example, N, O, or S.
[0028] The term "aromatic" or "aryl" is defined to include a cyclically conjugated molecular entity with a stability, due to derealization, significantly greater than that of a hypothetical localized structure, such as the Kekule structure.
[0029] The term "heterocyclic," when not used to describe an aromatic ring, is defined to include cyclic (i.e., ring-containing) groups other than aromatic groups, the cyclic group being formed by between 3 and about 14 carbon atoms and at least one heteroatom described above.
[0030] The term "substituted heterocyclic" is defined to include both aromatic and non- aromatic structures to heterocyclic groups further bearing one or more substituents described above.
[0031] The term "alkyl" is defined to include a monovalent straight or branched chain hydrocarbon group having from one to about 12 carbon atoms, for example, methyl, ethyl, n- propyl, wo-propyl, w-butyl, ώo-butyl, tert-butyl, 7i-pentyl (also known as rc-amyl), n-hexyl, and the like. The abbreviations "Me" and "Et" stand for the methyl and ethyl groups, respectively.
[0032] The term "aralkyl" refers to an aryl or heteroaryl group, as defined herein, attached through a C1-C6 alkyl linker. Examples of "aralkyl" include, but are not limited to, benzyl, phenylpropyl, 2-pyridylmethyl, 3-isoxazolylmethyl, 2-imidazolylethyl.
[0033] The term "methoxy" is defined as the group -OCH3.
[0034] The term "halogen" is defined to include an atom of fluorine, chlorine, bromine or iodine.
[0035] The term "carboxyl" or "carboxyl group" is defined as an acid moiety having the structure -COOH.
[0036] The term "amino" or "amino group" is defined to include moieties -NRR', where each of R and R' is hydrogen ("primary amino"), or one of them is an organic radical ("secondary amino"), or each is an organic radical ("tertiary amino").
[0037] The term "aminoalkyl" or "aminoalkyl group" is defined to include moieties -R-N(R1R"), wherein R is an organic radical and each of R' and R" is hydrogen or an organic radical. If at least one of R' and R" is an organic radical, the moiety is defined as "alkylaminoalkyl" or "alkylaminoalkyl group."
[0038] The term "sulfonyl" or "sulfonyl group" is defined to include moieties that comprise structure (S), in which R is an organic radical:
Figure imgf000014_0001
(S)
[0039] The term "sulfonylamino" or "sulfonylamino group" is defined to include moieties that comprise structure (SA), in which R is an organic radical:
Figure imgf000014_0002
(Sa) [0040] The term "amide," or "amido," or "amide group," or "amido group" is defined to include moieties containing at least one acyl group >C=O attached to nitrogen. The term "substituted amide" is defined to include moieties containing a structure RNH-CO-, in which R is an organic radical.
[0041] The term "phenyl" is defined to include moieties having structure (Ph):
Figure imgf000015_0001
(Ph)
[0042] The term "toluyl" is defined to include moieties having structure (Tl):
Figure imgf000015_0002
(Tl)
[0043] The term "heteroaryl" is defined to include aromatic rings, where the ring structure is formed by between 3 and about 14 carbon atoms and by at least one heteroatom described above, and the term "substituted heteroaryl" refers to heteroaryl groups further bearing one or more substituents described above.
[0044] The term "triazine" is defined to include moieties containing the aromatic 6- member heterocycle having three atoms of nitrogen in the ring. Two examples of such heterocycle are shown as the following structures (Tr):
Figure imgf000015_0003
(Tr)
[0045] The term "benzotriazine" is defined to include moieties containing a heterocyclic structure in which the triazine ring is fused with the benzene ring, as shown by structure (BTr): [0046]
Figure imgf000016_0001
(BTr)
[0047] The terms "N-oxide," "N,N'-dioxide," and "N,N',N"-trioxide" are defined to include nitrogen-containing heterocyclic moieties in which at least one nitrogen atom is associated with oxygen to form the structures N→O. The heterocyclic moiety can be any nitrogen-containing heterocycle, for example, benzotriazine, triazine, pyridine, pyrimidine, etc. Where the heterocyclic structure is benzotriazine, for example, some N-oxides or dioxides can be described as the following structures:
Figure imgf000016_0002
[0048] The term "pteridine" is defined to include moieties containing a heterocyclic structure having two fused 6-member rings, each ring containing two atoms of nitrogen, as shown by structure (PTr):
Figure imgf000016_0003
(PTr)
[0049] The term "pyridazine" is defined to include moieties containing the aromatic 6- member heterocycle having two atoms of nitrogen in the ring in ortho position, as shown by the structure (PAz):
Figure imgf000016_0004
(PAz)
[0050] The term "pyrimidine" is defined to include moieties containing the aromatic 6- member heterocycle having two atoms of nitrogen in the ring in meta position, as shown by the structure (PRm):
Figure imgf000017_0001
(PRm)
[0051] The term "thiadiazole" is defined to include moieties containing the aromatic 5- member thiophene-based heterocycle, having two atoms of nitrogen and one atom of sulfur, as shown by the structure (TDa):
Figure imgf000017_0002
(TDa)
[0052] The term "pyridyl" is defined to include moieties containing a radical derived from pyridine. One structure of pyridyl is shown as the structure (Py):
Figure imgf000017_0003
(Py)
[0053] The term "alkyl pyrrolidine" is defined to include moieties containing a radical derived from pyrrolidine (a 5 -member saturated heterocycle having one nitrogen atom), where an alkylene group R is attached to the nitrogen atom of the pyrrolidine ring. One structure of alkyl pyrrolidine is shown as the structure (APy):
Figure imgf000017_0004
(APy)
[0054] The term "alkyl morpholine" is defined to include moieties containing a radical derived from morholine, (a 6-member saturated heterocycle having one nitrogen atom and one oxygen atom), where an alkylene group R is attached to the nitrogen atom of the morpholine ring. One structure of alkyl morpholine is shown as the structure (AMr):
Figure imgf000018_0001
(AMr)
[0055] The term "alkyl piperazine" is defined to include moieties containing a radical derived from piperazine (a 6-member saturated heterocycle having two nitrogen atoms), where an alkylene group R is attached to one nitrogen atom of the piperazine ring. One structure of alkyl piperazine is shown as the structure (APi):
Figure imgf000018_0002
(APi)
[0056] The term "isoxazole" is defined to include moieties containing the aromatic 5- meniber heterocycle, having one atoms of nitrogen and one atom of oxygen, as shown by the structure (ISo):
Figure imgf000018_0003
(Iso)
[0057] The term "hydrophobic" is defined as a group or structure free of strongly polar groups such as -OH, -COOH, -NH2, -NH-CO-, halogens, or the like.
[0058] The term "kinase" is defined to include any enzyme that catalyzes the addition of phosphate groups to a protein residue; for example, serine and threonine kinases catalyze the addition of phosphate groups to serine and threonine residues.
[0059] The term "therapeutically effective amount" is defined as the amount of the compound or pharmaceutical composition that will elicit the biological or medical response of a tissue, system, animal or human that is being sought by the researcher, veterinarian, medical doctor or other clinician, e.g., restoration or maintenance of vasculostasis or prevention of the compromise or loss or vasculostasis; reduction of tumor burden; reduction of morbidity and/or mortality.
[0060] The term "pharmaceutically acceptable" is defined as a carrier, whether diluent or excipient, that is compatible with the other ingredients of the formulation and not deleterious to the recipient thereof.
[0061] The terms "administration of a compound" or "administering a compound" is defined to include an act of providing a compound of the invention or pharmaceutical composition to the subject in need of treatment.
[0062] According to embodiments of the present invention, two types of heterocyclic compounds are provided for treatment of various diseases, disorders, and pathologies, including cancer. The heterocyclic compounds of the invention can inhibit the activity of a kinase, such as any kinase in the MAPK signaling pathway.
[0063] According to an embodiment of the invention, a first type of compounds is provided for treatment of various diseases, disorders, and pathologies, including cancer. The first type of compounds can include derivatives of benzotriazine. The derivatives of benzotriazine that can be used can comprise the compounds that include a benzotriazine moiety having at least one substituent attached to the benzene ring of benzotriazine and a at least one substituent attached to the triazine ring of the benzotriazine, and an N-oxide, N,N'- dioxide, N,N',N"-trioxide, or pharmaceutically acceptable salts thereof.
[0064] A substituent attached to the benzene ring of benzotriazine can include a substituted pyridyl group. The substituents in the substituted pyridyl group can include an amido moiety, an aminoalkyl group (e.g., aminomethyl), or a carboxyl group, or a carboxylate group. The amido moiety attached to the pyridyl group can be in turn also substituted by attaching to the nitrogen in the amido moiety a substituent selected from an alkyl (e.g., methyl), an alkylaminoalkyl (e.g., diethylamino alkyl), a pyridyl, an alkyl pyrrolidine, an alkyl morpholine, and an alkyl piperazine groups.
[0065] Optionally, the benzene ring of the compounds of the first type can contain a second substituent located in any available position of the ring, for example, methyl, halogen or methoxy. Some particular examples of benzene-ring containing moieties that can be used include tert-hυtyl phenyl, trifluoromethoxyphenyl, methoxyphenyl, dimethylamino, dimethylaminophenyl, aminophenyl, trifluoroethoxyphenyl, trifluoromethoxychlorophenyl, trifluoromethoxybromophenyl, trifluoroethoxychlorophenyl, chlorophenyl, dichlorophenyl, trifluoromethyl phenyl, trifluoromethylchloro phenyl, chlorotoluyl, N-phenylacetamide, N5N- alkyl-benzamide, isopropoxyphenyl, alkoxyphenyl, dialkoxyphenyl, acetylphenyl.
[0066] A substituent attached to the triazine ring of benzotriazine in the compounds of the first type can include a secondary amino group, a substituted amide group, or a substituted sulfonylamino group; each of these groups can further contain a moiety derived from benzene, thiophene, or isoxazole. If the substituent attached to the triazine ring of benzotriazine in the compounds of the first type is a secondary amino group, the moiety derived from benzene, thiophene, or isoxazole can be attached to the nitrogen of the secondary amino group. If the substituent in the triazine ring is a substituted amide group or the substituted sulfonylamino group, the moiety derived from benzene, thiophene, or isoxazole can be attached via the acyl group or the sulfonyl group, respectively. Moieties derived from benzene, thiophene, or isoxazole can further include alkyls, e.g., £-butyl phenyl, chlorophenyl, dichlorophenyl, trifluoromethyl phenyl, trifluoromethylchloro phenyl, and chlorotoluyl.
[0067] Compounds of the first type can be described as compounds having the general structure (A), and an N-oxide, N,N'-dioxide, N,N',N"-trioxide, or a pharmaceutically acceptable salt thereof. The general structure (A) is as follows:
Figure imgf000020_0001
(A)
wherein Y can be absent or can be one of the following moieties:
Figure imgf000020_0002
[0068] In structure (A), R1 can be an aryl, a substituted aryl, a heterocycle, a heteroaryl, a substituted heterocycle, and a substituted heteroaryl, such as C6-Ci2 aryl; C3-Ci2 heteroaryl having 1-3 heteroatoms such as N, S and O; substituted C3-Ci0 cycloalkyl having 0-3 heteroatoms such as N, S, and O; substituted C6-Ci2 aryl; substituted C3-Ci2 heteroaryl having 1-3 heteroatoms such as N, S and O; C7-C24 aralkyl; C7-C24 alkylaryl; substituted C7- C24 aralkyl; and substituted C7-C24 alkaryl. In particular, Ri can be any one of the following moieties:
Figure imgf000021_0001
Figure imgf000021_0002
1IH2 UH9, NH7
Figure imgf000022_0001
Figure imgf000023_0001
where n can be an integer selected from a group consisting of 0, 1, 2, and 3.
[0069] In structure (A), the substituent R2 can be any one of hydrogen, halogen, C1-Ci8 alkyl (e.g., methyl), -OH, -NO2, -CN, Ci-Ci8 alkoxy (e.g., methoxy), -NHSO2R5, -SO2NHR5, -NHCOR5, -NH2, -NR5R6 , -S(O)R5, -S(O)2R5, -CO2R5, -CONR5R6, and where R5 and R6 are independently selected from hydrogen, a Ci-Ci2 alkyl and a substituted Ci-Ci2 alkyl.
[0070] In structure (A), group R3 can be an aryl, a substituted aryl, a heterocycle, a heteroaryl, a substituted heterocycle, and a substituted heteroaryl. For example, R3 can be one of C6-Ci2 aryl; C3-Ci2 heteroaryl having 1-3 heteroatoms such as N, S and O; substituted C3-CiO cycloalkyl having 0-3 heteroatoms such as N, S, and O; substituted C6-Ci2 aryl; substituted C3-Ci2 heteroaryl having 1-3 heteroatoms such as N, S and O; C7-C24 aralkyl; C7- C24 alkylaryl; substituted C7-C24 aralkyl; and substituted C7-C24 alkaryl. In particular, the substitutent R3 can be one of the following:
Figure imgf000023_0002
Figure imgf000024_0001

Figure imgf000025_0001
Figure imgf000025_0002

Figure imgf000026_0001
Figure imgf000027_0001
[0071] Some general examples of compounds described by the general structure (A), include a compound having the general structure (I), a compound having the general structure (II), a compound having the general structure (III) or a compound having the general structure (IV):
Figure imgf000027_0002
(I) (H)
Figure imgf000028_0001
(HI) (IV)
[0072] Some non-limiting examples of particular compounds described by the general structure (A) that can be used include compounds having formulae (l)-(33):
Figure imgf000028_0002
(1) (2)
Figure imgf000028_0003
(3) (4)
Figure imgf000028_0004
(5) (6)
Figure imgf000028_0005
(9) (10)
Figure imgf000029_0001
(13) (14)
Figure imgf000029_0002
(17) (18)
Figure imgf000029_0003
(23) (24)
Figure imgf000030_0001
(25) (26)
Figure imgf000030_0002
(31) (32)
Figure imgf000030_0003
(33)
[0073] The benzotriazine derivatives described above and illustrated by the general structure (A) can be prepared as shown by Scheme I:
Figure imgf000030_0004
Figure imgf000030_0005
Intermediate A
Figure imgf000031_0001
in
Scheme I
[0074] To prepare the intermediate A, where Ri is, for example, 2-pyridine carboxamide, the synthetic route shown by Scheme II can be used:
Figure imgf000031_0002
Figure imgf000031_0003
Scheme II
[0075] As seen from Scheme II, the synthesis of the intermediate A requires using 4- chloro-2-pyridinecarboxamide 4, which can be separately preliminarily synthesized as shown by Scheme III:
Figure imgf000032_0001
8
Scheme III
The benzotriazine derivatives described in the formula (IV) and illustrated by the general structure (A) can be prepared as shown by Scheme IV:
NaNO2, CuCl, HCl Pd(PPh3)CI2, CO
Figure imgf000032_0003
Figure imgf000032_0002
Intermediate A
Figure imgf000032_0004
IV
Scheme IV.
[0076] According to an embodiment of the invention, a second type of compounds is provided for treatment of various diseases, disorders, and pathologies, including cancer. The second type of compounds can include a benzene derived moiety bridged to a heterocyclic moiety, or pharmaceutically acceptable salts thereof. The bridge between the benzene- derived moiety and the heterocyclic moiety can include a single bond or a nitrogen atom. If the heterocyclic moiety contains at least one nitrogen, the second type of compounds can be an N-oxide, or N,N'-dioxide, or N,N',N"-trioxide.
[0077] Whether the compound can be an N-oxide, or N,N'-dioxide, or N,N',N"-trioxide, depends on the number of nitrogen atoms contained in the heterocyclic moiety. For example, if the heterocyclic moiety has only one nitrogen, the second type of compounds can be an N- oxide. If the heterocyclic moiety has two atoms of nitrogen, the second type of compounds can be an N-oxide or N,N'-dioxide. If the heterocyclic moiety has three atoms of nitrogen, the second type of compounds can be any of an N-oxide, an N,N'-dioxide, an N,N',N"- trioxide. [0078] The benzene-derived moiety can include a substituent sucfr as a pyridyl group connected to the benzene molecule via an oxygen link, or a sulfonyl |group. The pyridyl group connected to the benzene molecule can be further substituted. The siibstituents in the pyridyl group can include the same moieties as described above for the first type of compounds of the present invention. The sulfonyl group connected to the benzene molecule can be also further substituted. The substituents in the sulfonyl group can include the substituted pyridyl group described above for the first type of compounds of the present invention.
[0079] Optionally, the benzene-derived moiety of the compounds of the second type can contain a second substituent, e.g., methyl, halogen or methoxy, which can be located in any position of the benzene ring. Some exemplary benzene-derived moieties that can be included in the second type of compounds can include tert-butyl phenyl, trifluoromethoxyphenyl, methoxyphenyl, dimethylamino, dimethylaminophenyl, aminophenyl, trifiuoroethoxyphenyl, trifluoromethoxychlorophenyl, trifluoromethoxybromophenyl, trifluoroethoxychlorophenyl, chlorophenyl, dichlorophenyl, trifluoromethyl phenyl, trifiuoromethylchloro phenyl, chlorotoluyl, N-phenylacetamide, N,N-alkyl-benzamide, isopropoxyphenyl, alkoxyphenyl, dialkoxyphenyl, acetylphenyl.
[0080] The compounds of the second type include heterocyclic compounds having the general structure (B), or an N-oxide, or N,N'-dioxide, N,N',N"-trioxide, or a pharmaceutically acceptable salt thereof, and can inhibit the activity of a kinase, such as any kinase in the MAPK signaling pathway. The general structure (B) can be represented as follows:
Figure imgf000033_0001
(B)
[0081] In structure (B), each of Z1, Z2 and Z3 can be, independently, N, CH, N=CH, O, S or N-R4, wherein R4 is hydrogen or lower alkyl, with the further proviso that at least one of Z1, Z2 and Z3 is not CH; X can be absent or be NH; Y can be absent or can be one of the following moieties:
Figure imgf000033_0002
[0082] Further, in structure (B), the substitutents R1, R2, and R3 can be as follows:
[0083] R1 can be an unsubstituted or a substituted C3-C12 heteroaryl having 1 -3 heteroatoms such as N, S or O;
[0084] R2 can be any one of hydrogen, halogen, C1-Ci8 alkyl (e.g., methyl), -OH, -NO2, - CN, C1-Ci8 alkoxy (e.g., methoxy), -NHSO2R5, -SO2NHR5, -NHCOR5, -NH2, -NR5R6 , - S(O)R5, -S(O)2R5, -CO2R5, -CONR5R6, and where R5 and R6 are independently selected from hydrogen, a Ci-Cis alkyl and a substituted Ci-Ci2 alkyl; and
[0085] R3 can be hydrogen, a Ci-Cis alkyl, a substituted Ci-Ci2 alkyl, a C1-C12 cycloalkyl, a substituted C1-Ci2 cycloalkyl, a substituted C3-Ci0 cycloalkyl having 0-3 heteroatoms such as N, S, or O, an aryl such as a Cδ-Ci2 aryl, a substituted aryl such as a substituted C6-Ci2 aryl, a heterocycle, a substituted heterocycle, a heteroaryl such as a C3-Ci2heteroaryl having 1-3 heteroatoms such as N, S or O, a substituted heteroaryl such as substituted C3-Ci2 heteroaryl having 1-3 heteroatoms such as N, S or O, a C7-C24 aralkyl, a substituted C7-C24 aralkyl, a C7-C24 alkylaryl, and a substituted C7-C24 alkaryl.
[0086] The substituent Ri can include a substituted pyridyl or a substituted pyrimidyl group. The substituents in the substituted pyridyl or substituted pyrimidyl group can include an amido moiety, an aminoalkyl group (e.g., aminomethyl), or a carboxyl group, or a carboxylate group. The amido moiety attached to the pyridyl/pyrimidyl group can be in turn also substituted by attaching to the nitrogen in the amido moiety a substitutent selected from an alkyl (e.g., methyl), an alkylaminoalkyl (e.g., diethylamino alkyl), a pyridyl, an alkyl pyrrolidine, an alkyl morpholine, and an alkyl piperazine groups.
[0087] Particular, non-limiting examples of Ri that can be used in compounds having the structure (B) include any of the following moieties:
Figure imgf000034_0001
Figure imgf000035_0001
Figure imgf000036_0001
where n can be an integer selected from a group consisting of 0, 1, 2, and 3.
[0088] Some particular non-limiting examples of the substituent R3 that can be used in compounds having the structure (B) include tert-butyl phenyl, trifluoromethoxyphenyl, methoxyphenyl, dimethylaminophenyl, aminophenyl, trifluoroethoxyphenyl, trifluoromethoxychlorophenyl, trifluoromethoxybromophenyl, trifluoroethoxychlorophenyl, chlorophenyl, dichlorophenyl, trifluoromethyl phenyl, trifluoromethylchlorophenyl, chlorotoluyl, N-phenylacetamide, N,N-alkyl-benzamide, isopropoxyphenyl, alkoxyphenyl, dialkoxyphenyl, and acetylphenyl. These and yet other moieties that can be used as the substituent R3 can be illustrated as follows:
Figure imgf000036_0002
Figure imgf000037_0001
Figure imgf000038_0001
Figure imgf000039_0001
where n can be an integer selected from a group consisting of 0, 1, 2, and 3, and R' is hydrogen, a C1-Ci8 alkyl, or a substituted Ci-Ci8 alkyl.
[0089] Some general examples of compounds described by the general structure (B) include a compound having the general structures (V)-(XXVIII):
Figure imgf000040_0001
XVII XVIII XIX
Figure imgf000040_0002
XX XXI XXII
Figure imgf000041_0001
XXIII XXV
Figure imgf000041_0002
[0090] In the case of 1,2,4-triazoles, there exist three tautomeric structures, as shown helow:
1 2 1 2 2 1
Figure imgf000041_0003
4H-l,2,4-triazole lH-l,2,4-triazole lH-l,2,4-triazoIe
[0091] Which tautomeric structure is prevailing depends on the suhstituents on the triazole moiety and on the reaction conditions. As known to those having ordinary skill in the art, typically, lH-l,2,4-triazole is the most common tautomeric form, especially if an amino substituent is attached to the ring. Even though all three tautomeric structures can be present, all the generic structures and all the examples having 1,2,4-triazole moiety are shown herein in one tautomeric form, such as 4H-l,2,4-triazole, for simplicity and for the comparison with its direct analogues, such as examples containing 1,3,4-oxadiazole moiety. Using only 4H- tautomeric form to draw the structures for the sake of simplicity, does not imply that the compounds of the examples (30)-(74) shown below exist in that particular tautomeric form.
[0092] Some examples of particular compounds described by the general structure (B) include compounds having formulae (34)-(83):
Figure imgf000042_0001
(34) (35)
Figure imgf000042_0002
(38) (39)
Figure imgf000042_0003
(40) (41)
Figure imgf000042_0004
(42) (43)
Figure imgf000042_0005
(45)
(44)
Figure imgf000042_0006
(46) (47)
Figure imgf000043_0001
(48) (49)
Figure imgf000043_0002
(50) (51)
Figure imgf000043_0003
(52) (53)
Figure imgf000043_0004
(54) (55)
Figure imgf000043_0005
Figure imgf000044_0001
(60) (61)
Figure imgf000044_0002
(62) (63)
Figure imgf000044_0003
(64) (65)
Figure imgf000044_0004
(66) (67)
Figure imgf000044_0005
(68) (69)
Figure imgf000044_0006
(70) (71)
Figure imgf000044_0007
(72) (73)
Figure imgf000045_0001
(74) (75)
Figure imgf000045_0002
(78) (79)
Figure imgf000045_0003
(80) (81)
Figure imgf000045_0004
[0093] Appropriately substituted 1,2,4-triazoles of the type (V) described above and illustrated by the general structure (B) can be synthesized using one of several reaction schemes, for example as shown in Schemes V, VI and VII below. The appropriate method can be chosen based on the required substitution, availability of the starting materials and the ease of synthesis.
Figure imgf000046_0001
Scheme VI
Figure imgf000047_0001
NaSCN
Figure imgf000047_0002
Figure imgf000047_0004
Figure imgf000047_0003
Figure imgf000047_0005
Scheme VII
[0094] 1,2,4-triazoles of the type (VI) described above, can be made as shown in Scheme VIII or by an alternative route as outlined in Scheme IX.
Figure imgf000047_0006
Figure imgf000047_0007
Scheme VIII NaSCN
Figure imgf000048_0001
Figure imgf000048_0002
Figure imgf000048_0003
Figure imgf000048_0004
Figure imgf000048_0005
Scheme IX
[0095] Appropriately substituted 2-amino- 1 ,3 ,4-oxadoazoles of type (VIII), can be synthesized using one of several reaction schemes, for example, as shown by Schemes X, XI and XII.
Figure imgf000048_0006
Scheme X
Figure imgf000049_0001
Figure imgf000049_0002
Scheme XI
Figure imgf000049_0003
Figure imgf000049_0004
Scheme XII [0096] Appropriately substituted 2-amino- 1 ,3 ,4-oxadiazoles of type (IX), can be synthesized using one of several reaction schemes, for example, as shown by Schemes XIII and XIV.
Figure imgf000050_0001
Scheme XIII
Figure imgf000050_0002
Figure imgf000050_0003
Figure imgf000050_0004
Figure imgf000050_0005
Scheme XIV
[0097] Appropriately substituted 2-amino-l,3,4-thiadiazoles of type (XI), can be prepared by a method which is outlined in Scheme XV.
Figure imgf000051_0001
Figure imgf000051_0002
Scheme XV
[0098] Appropriately substituted 2-amino-l,3,4-thiadiazoles of type (XII), can be prepared by a method which is outlined in Scheme XVI.
Figure imgf000051_0003
Figure imgf000051_0004
Figure imgf000052_0001
AcOH, microwave
Figure imgf000052_0002
Figure imgf000052_0003
Scheme XVI
[0099] Appropriately substituted 3-amino-l,2,4-triazines of type (XXIII)-(XXV), can be prepared by a method which is outlined in Scheme XVII.
Figure imgf000052_0004
Figure imgf000052_0005
R1-HaI
Figure imgf000052_0006
xxm
Figure imgf000052_0007
Figure imgf000053_0001
XXV
Scheme XVII
Appropriately substituted 3-amino-l,2,4-triazines of type (XXVI)-(XXVIII), can be prepared by a method which is outlined in Scheme XVIII.
Figure imgf000053_0002
Figure imgf000053_0003
Figure imgf000053_0004
1'" IUi. Ii ••' 'Ul' i,..,il PU(I ,,.,Ji
Figure imgf000054_0001
Scheme XVm
[0100] The compounds and methods of the present invention, either when administered alone or in combination with other agents (e.g., chemotherapeutic agents or protein therapeutic agents described below) are useful in treating a variety of disorders, including, but not limited to, for example, cancer, eye disease, inflammation, psoriasis, and a viral infection. The kinds of cancer that can be treated include, but are not limited to, an alimentary/gastrointestinal tract cancer, colon cancer, liver cancer, skin cancer, breast cancer, ovarian cancer, prostate cancer, lymphoma, leukemia, kidney cancer, lung cancer, muscle cancer, bone cancer, bladder cancer or brain cancer.
[0101] Embodiments of the present invention also provide articles of manufacture that can include a packaging material and a pharmaceutical composition contained within the packaging material. The packaging material can comprise a label which indicates that the pharmaceutical composition can be used for treatment of one or more disorders identified above.
[0102] The pharmaceutical composition can include a compound according to the present invention. In addition to a compound of the present invention, the pharmaceutical may also contain other therapeutic agents, and may be formulated, for example, by employing conventional solid or liquid vehicles or diluents, as well as pharmaceutical additives of a type appropriate to the mode of desired administration (for example, excipients, binders, preservatives, stabilizers, flavors, etc.) according to techniques known in the art of pharmaceutical formulation. [0103] Thus, in one embodiment, the invention provides a pharmaceutical composition including a therapeutic agent and a compound of the invention. The compound is present in a concentration effective to treat cancer.
[0104] The compounds of the invention may be formulated into therapeutic compositions as natural or salt forms. Pharmaceutically acceptable non-toxic salts include the base addition salts (formed with free carboxyl or other anionic groups) which may be derived from inorganic bases such as, for example, sodium, potassium, ammonium, calcium, or ferric hydroxides, and such organic bases as isopropylamine, trimethylamine, 2-ethylamino- ethanol, histidine, procaine, and the like. Such salts may also be formed as acid addition salts with any free cationic groups and will generally be formed with inorganic acids such as, for example, hydrochloric, sulfuric, or phosphoric acids, or organic acids such as acetic, citric, p- toluenesulfonic, methanesulfonic acid, oxalic, tartaric, mandelic, and the like.
[0105] Salts of the invention can include amine salts formed by the protonation of an amino group with inorganic acids such as hydrochloric acid, hydrobromic acid, hydroiodic acid, sulfuric acid, phosphoric acid, and the like. Salts of the invention can also include amine salts formed by the protonation of an amino group with suitable organic acids, such as p-toluenesulfonic acid, acetic acid, methanesulfonic acid and the like. Additional excipients which are contemplated for use in the practice of the present invention are those available to those of ordinary skill in the art, for example, those found in the United States Pharmacopeia Vol. XXII and National Formulary Vol. XVII, U.S. Pharmacopeia Convention, Inc., Rockville, MD (1989), the relevant contents of which is incorporated herein by reference. In addition, polymorphs of the invention compounds are included in the present invention.
[0106] Pharmaceutical compositions of the invention may be administered by any suitable means, for example, orally, such as in the form of tablets, capsules, granules or powders; sublingually; buccally; parenterally, such as by subcutaneous, intravenous, intramuscular, intrathecal, or intracisternal injection or infusion techniques (e.g., as sterile injectable aqueous or non-aqueous solutions or suspensions); nasally such as by inhalation spray; topically, such as in the form of a cream or ointment; or rectally such as in the form of suppositories; in dosage unit formulations containing non-toxic, pharmaceutically acceptable vehicles or diluents. The present compounds may, for example, be administered in a form suitable for immediate release or extended release. Immediate release or extended release may be achieved by the use of suitable pharmaceutical compositions comprising the present compounds, or, particularly in the case of extended release, by the use of devices such as subcutaneous implants or osmotic pumps. The present compounds may also be administered liposomally.
[0107] In addition to primates, such as humans, a variety of other mammals can be treated according to the method of the present invention. For instance, mammals including, but not limited to, cows, sheep, goats, horses, dogs, cats, guinea pigs, rats or other bovine, ovine, equine, canine, feline, rodent or murine species can be treated. However, the method can also be practiced in other species, such as avian species (e.g., chickens).
[0108] The pharmaceutical compositions for the administration of the compounds of this embodiment, either alone or in combination with other therapeutic agents, may conveniently be presented in dosage unit form and may be prepared by any of the methods well known in the art of pharmacy. All methods include bringing the active ingredient into association with the carrier which constitutes one or more accessory ingredients. In general, the pharmaceutical compositions are prepared by uniformly and intimately bringing the active ingredient into association with a liquid carrier or a finely divided solid carrier or both, and then, if necessary, shaping the product into the desired formulation. In the pharmaceutical composition the active object compound is included in an amount sufficient to produce the desired effect upon the process or condition of diseases. The pharmaceutical compositions containing the active ingredient may be in a form suitable for oral use, for example, as tablets, troches, lozenges, aqueous or oily suspensions, dispersible powders or granules, emulsions, hard or soft capsules, or syrups or elixirs.
[0109] Compositions intended for oral use may be prepared according to any method known to the art for the manufacture of pharmaceutical compositions and such compositions may contain one or more agents selected from the group consisting of sweetening agents, flavoring agents, coloring agents and preserving agents in order to provide pharmaceutically elegant and palatable preparations. Tablets contain the active ingredient in admixture with non-toxic pharmaceutically acceptable excipients which are suitable for the manufacture of tablets. These excipients may be for example, inert diluents, such as calcium carbonate, sodium carbonate, lactose, calcium phosphate or sodium phosphate; granulating and disintegrating agents, for example, corn starch, or alginic acid; binding agents, for example starch, gelatin or acacia, and lubricating agents, for example magnesium stearate, stearic acid or talc. The tablets may be uncoated or they may be coated by known techniques to delay disintegration and absorption in the gastrointestinal tract and thereby provide a sustained action over a longer period. For example, a time delay material such as glyceryl monostearate Ji-" IL. i'i / U '.::::» n.j ;;;;;ι> >• .».* ««.• ■> «.«.. » or glyceryl distearate may be employed. They may also be coated to form osmotic therapeutic tablets for control release.
[0110] Formulations for oral use may also be presented as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent, for example, calcium carbonate, calcium phosphate or kaolin, or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium, for example peanut oil, liquid paraffin, or olive oil.
[0111] Aqueous suspensions contain the active materials in admixture with excipients suitable for the manufacture of aqueous suspensions. Such excipients are suspending agents, for example sodium carboxymethylcellulose, methylcellulose, hydroxy- propylmethylcellulose, sodium alginate, polyvinyl-pyrrolidone, gum tragacanth and gum acacia; dispersing or wetting agents may be a naturally-occurring phosphatide, for example lecithin, or condensation products of an alkylene oxide with fatty acids, for example polyoxyethylene stearate, or condensation products of ethylene oxide with long chain aliphatic alcohols, for example heptadecaethyleneoxycetanol, or condensation products of ethylene oxide with partial esters derived from fatty acids and a hexitol such as polyoxyethylene sorbitol monooleate, or condensation products of ethylene oxide with partial esters derived from fatty acids and hexitol anhydrides, for example polyethylene sorbitan monooleate. Also useful as a solubilizer is polyethylene glycol, for example. The aqueous suspensions may also contain one or more preservatives, for example ethyl, or /ϊ-propyl, p- hydroxybenzoate, one or more coloring agents, one or more flavoring agents, and one or more sweetening agents, such as sucrose or saccharin.
[0112] Oily suspensions may be formulated by suspending the active ingredient in a vegetable oil, for example arachis oil, olive oil, sesame oil or coconut oil, or in a mineral oil such as liquid paraffin. The oily suspensions may contain a thickening agent, for example beeswax, hard paraffin or cetyl alcohol. Sweetening agents such as those set forth above, and flavoring agents may be added to provide a palatable oral preparation. These compositions may be preserved by the addition of an anti-oxidant such as ascorbic acid.
[0113] Dispersible powders and granules suitable for preparation of an aqueous suspension by the addition of water provide the active ingredient in admixture with a dispersing or wetting agent, suspending agent and one or more preservatives. Suitable dispersing or wetting agents and suspending agents are exemplified by those already mentioned above. Additional excipients, for example sweetening, flavoring and coloring agents, may also be present.
[0114] Syrups and elixirs may be formulated with sweetening agents, for example glycerol, propylene glycol, sorbitol or sucrose. Such formulations may also contain a demulcent, a preservative and flavoring and coloring agents.
[0115] The pharmaceutical compositions may be in the form of a sterile injectable aqueous or oleagenous suspension. This suspension may be formulated according to the known art using those suitable dispersing or wetting agents and suspending agents which have been mentioned above. The sterile injectable preparation may also be a sterile injectable solution or suspension in a parenterally-acceptable diluent or solvent or cosolvent or complexing agent or dispersing agent or excipient or combination thereof, for example 1,3-butanediol, polyethylene glycols, polypropylene glycols, ethanol or other alcohols, povidones, various brands of TWEEN surfactant, sodium dodecyl sulfate, sodium deoxycholate, dimethylacetamide, polysorbates, poloxamers, cyclodextrins, lipids, and excipients such as inorganic salts (e.g., sodium chloride), buffering agents (e.g., sodium citrate, sodium phosphate), and sugars (e.g., saccharose and dextrose). Among the acceptable vehicles and solvents that may be employed are water, dextrose solutions, Ringer's solutions and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose any bland fixed oil may be employed including synthetic mono- or diglycerides. In addition, fatty acids such as oleic acid find use in the preparation of injectables.
[0116] Depending on the condition being treated, these pharmaceutical compositions may be formulated and administered systemically or locally. Techniques for formulation and administration may be found in the latest edition of "Remington's Pharmaceutical Sciences" (Mack Publishing Co, Easton Pa.). Suitable routes may, for example, include oral or transniucosal administration; as well as parenteral delivery, including intramuscular, subcutaneous, intramedullary, intrathecal, intraventricular, intravenous, intraperitoneal, or intranasal administration. For injection, the pharmaceutical compositions of the invention may be formulated in aqueous solutions, preferably in physiologically compatible buffers such as Hanks' solution, Ringer's solution, or physiologically buffered saline. For tissue or cellular administration, penetrants appropriate to the particular barrier to be permeated are used in the formulation. Such penetrants are generally known in the art. Pharmaceutical formulations for parenteral administration include aqueous solutions of the active compounds If" «... ii in water-soluble form. Additionally, suspensions of the active compounds may be prepared as appropriate oily injection suspensions. Suitable lipophilic solvents or vehicles include fatty oils such as sesame oil, or synthetic fatty acid esters, such as ethyl oleate or triglycerides, or liposomes. Aqueous injection suspensions may contain substances that increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, or dextran. Optionally, the suspension may also contain suitable stabilizers or agents that increase the solubility of the compounds to allow for the preparation of highly concentrated solutions.
[0117] The compounds of the present invention may also be administered in the form of suppositories for rectal administration of the drug. These compositions can be prepared by mixing the drug with a suitable non-irritating excipient which is solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum to release the drug. Such materials are cocoa butter and polyethylene glycols.
[0118] For topical use, creams, ointments, jellies, solutions or suspensions, etc., containing the compounds of the present invention are employed. (For purposes of this application, topical application shall include mouthwashes and gargles).
[0119] In one embodiment, the invention compounds are administered in combination with an anti-inflammatory agent, antihistamines, chemotherapeutic agent, immunornodulator, therapeutic antibody or a protein kinase inhibitor, e.g., a tyrosine kinase inhibitor, to a subject in need of such treatment. While not wanting to be limiting, chemotherapeutic agents include antimetabolites, such as methotrexate, DNA cross-linking agents, such as cisplatin/carboplatin; alkylating agents, such as canbusil; topoisomerase I inhibitors such as dactinomicin; microtubule inhibitors such as taxol (paclitaxol), and the like. Other chemotherapeutic agents include, for example, a vinca alkaloid, mitomycin-type antibiotic, bleomycin-type antibiotic, antifolate, colchicine, demecoline, etoposide, taxane, anthracycline antibiotic, doxorubicin, daunorubicin, carminomycin, epirubicin, idarubicin, mithoxanthrone, 4-dimethoxy-daunomycin, 11-deoxydaunorubicin, 13-deoxydaunorubicin, adriamycin- 14-benzoate, adriamycin- 14-octanoate, adriamycin- 14-naphthaleneacetate, amsacrine, carmustine, cyclophosphamide, cytarabine, etoposide, lovastatin, melphalan, topetecan, oxalaplatin, chlorambucil, methtrexate, lomustine, thioguanine, asparaginase, vinblastine, vindesine, tamoxifen, or mechlorethamine. While not wanting to be limiting, therapeutic antibodies include antibodies directed against the HER2 protein, such as trastuzumab; antibodies directed against growth factors or growth factor receptors, such as bevacizumab, which targets vascular endothelial growth factor, and OSI-774, which targets epidermal growth factor; antibodies targeting integrin receptors, such as Vitaxin (also known as MEDI-522), and the like. Classes of anticancer agents suitable for use in compositions and methods of the present invention include, but are not limited to: 1) alkaloids, including, microtubule inhibitors (e.g., Vincristine, Vinblastine, and Vindesine, etc.), microtubule stabilizers (e.g., Paclitaxel [Taxol], and Docetaxel, Taxotere, etc.), and chromatin function inhibitors, including, topoisomerase inhibitors, such as, epipodophyllotoxins (e.g., Etoposide [VP-16], and Teniposide [VM-26], etc.), and agents that target topoisomerase I (e.g., Camptothecin and Isirinotecan [CPT-Il], etc.); 2) covalent DNA-binding agents [alkylating agents], including, nitrogen mustards (e.g., Mechlorethamine, Chlorambucil, Cyclophosphamide, Ifosphamide, and Busulfan [Myleran], etc.), nitrosoureas (e.g., Carmustine, Loniustine, and Semustine, etc.), and other alkylating agents (e.g., Dacarbazine, Hydroxymethylmelamine, Thiotepa, and Mitocycin, etc.); 3) noncovalent DNA-binding agents [antitumor antibiotics], including, nucleic acid inhibitors (e.g., Dactinomycin [Actinomycin D], etc.), anthracyclines (e.g., Daunorubicin [Daunomycin, and Cerubidine], Doxorubicin [Adriamycin], and Idarubicin [Idamycin], etc.), anthracenediones (e.g., anthracycline analogues, such as, [Mitoxantrone], etc.), bleomycins (Blenoxane), etc., and plicamycin (Mithramycin), etc.; 4) antimetabolites, including, antifolates (e.g., Methotrexate, Folex, and Mexate, etc.), purine antimetabolites (e.g., 6-Mercaptopurine [6-MP, Purinethol], 6-Thioguanine [6-TG], Azathioprine, Acyclovir, Ganciclovir, Chlorodeoxyadenosine, 2- Chlorodeoxyadenosine [CdA], and 2'-Deoxycoformycin [Pentostatin], etc.), pyrimidine antagonists (e.g., fluoropyrimidines [e.g., 5-fluorouracil (Adrucil), 5-fluorodeoxyuridine (FdUrd) (Floxuridine)] etc.), and cytosine arabinosides (e.g., Cytosar [ara-C] and Fludarabine, etc.); 5) enzymes, including, L-asparaginase; 6) hormones, including, glucocorticoids, such as, antiestrogens (e.g., Tamoxifen, etc.), nonsteroidal antiandrogens (e.g., Flutamide, etc.), and aromatase inhibitors (e.g., anastrozole [Arimidex], etc.); 7) platinum compounds (e.g., Cisplatin and Carboplatin, etc.); 8) monoclonal antibodies conjugated with anticancer drugs, toxins, and/or radionuclides, etc.; 9) biological response modifiers (e.g., interferons [e.g., IFN-.alpha., etc.] and interleukins [e.g., IL-2, etc.], etc.); 10) adoptive immunotherapy; 11) hematopoietic growth factors; 12) agents that induce tumor cell differentiation (e.g., all-trans-retinoic acid, etc.); 13) gene therapy techniques; 14) antisense therapy techniques; 15) tumor vaccines; 16) therapies directed against tumor metastases (e.g., Batimistat, etc.); and 17) inhibitors of angiogenesis.
[0120] The pharmaceutical composition and method of the present invention may further comprise other therapeutically active compounds as noted herein which are usually applied in 005/030614
60
IH1 IL, It / U ϊa iu ::::iκ> ,;jι iunut J- u the treatment of the above mentioned pathological conditions. Examples of other therapeutic agents include the following: cyclosporins (e.g., cyclosporin A), CTLA4-Ig, antibodies such as ICAM-3, anti-IL-2 receptor (Anti-Tac), anti-CD45RB, anti-CD2, anti-CD3 (OKT-3), anti- CD4, anti-CD80, anti-CD86, agents blocking the interaction between CD40 and gp39, such as antibodies specific for CD40 and/or gp39 (i.e., CD154), fusion proteins constructed from CD40 and gp39 (CD40Ig and CD8gp39), inhibitors, such as nuclear translocation inhibitors, of NF-kappa B function, such as deoxyspergualin (DSG), cholesterol biosynthesis inhibitors such as HMG CoA reductase inhibitors (lovastatin and simvastatin), non-steroidal antiinflammatory drugs (NS AIDs) such as ibuprofen and cyclooxygenase inhibitors such as rofecoxib, steroids such as prednisone or dexamethasone, gold compounds, antiproliferative agents such as methotrexate, FK506 (tacrolimus, Prograf), mycophenolate mofetil, cytotoxic drugs such as azathioprine and cyclophosphamide, TNF-a inhibitors such as tenidap, anti- TNF antibodies or soluble TNF receptor, and rapamycin (sirolimus or Rapamune) or derivatives thereof.
[0121] Other agents that may be administered in combination with invention compounds include protein therapeutic agents such as cytokines, immunomodulatory agents and antibodies. As used herein the term "cytokine" encompasses chemokines, interleukins, lymphokines, monokines, colony stimulating factors, and receptor associated proteins, and functional fragments thereof. As used herein, the term "functional fragment" refers to a polypeptide or peptide which possesses biological function or activity that is identified through a defined functional assay.
[0122] The cytokines include endothelial monocyte activating polypeptide II (EMAP-II), granulocyte-macrophage-CSF (GM-CSF), granulocyte-CSF (G-CSF), macrophage-CSF (M- CSF), IL-I, IL-2, IL-3, IL-4, IL-5, IL-6, IL-12, and IL-13, interferons, and the like and which is associated with a particular biologic, morphologic, or phenotypic alteration in a cell or cell mechanism.
[0123] When other therapeutic agents are employed in combination with the compounds of the present invention they may be used for example in amounts as noted in the Physician Desk Reference (PDR) or as otherwise determined by one having ordinary skill in the art.
[0124] In the treatment or prevention of conditions which involve cellular proliferation, an appropriate dosage level can generally be between about 0.01 and about 1000 mg per 1 kg of patient body weight per day which can be administered in single or multiple doses. For example, the dosage level can be between about 0.01 and about 250 mg/kg per day; more narrowly, between about 0.5 and about 100 mg/kg per day. A suitable dosage level can be between about 0.01 and about 250 mg/kg per day, between about 0.05 and about 100 mg/kg per day, or between about 0.1 and about 50 mg/kg per day, or about 1.0 mg/kg per day. For example, within this range the dosage can be between about 0.05 and about 0.5 mg/kg per day, or between about 0.5 and about 5 mg/kg per day, or between about 5 and about 50 mg/kg per day. For oral administration, the compositions can be provided in the form of tablets containing between about 1.0 and about 1,000 mg of the active ingredient, for example, about 1.0, about 5.0, about 10.0, about 15.0, about 20.0, about 25.0, about 50.0, about 75.0, about 100.0, about 150.0, about 200.0, about 250.0, about 300.0, about 400.0, about 500.0, about 600.0, about 750.0, about 800.0, about 900.0, and about 1,000.0 mg of the active ingredient for the symptomatic adjustment of the dosage to the patient to be treated. The compounds can be administered on a regimen of 1 to 4 times per day, such as once or twice per day. There may be a period of no administration followed by another regimen of administration.
[0125] It will be understood, however, that the specific dose level and frequency of dosage for any particular patient may be varied and will depend upon a variety of factors including the activity of the specific compound employed, the metabolic stability and length of action o of that compound, the age, body weight, general health, sex, diet, mode and time of administration, rate of excretion, drug combination, the severity of the particular condition, and the host undergoing therapy.
[0126] Compounds of the present invention can be used, alone or in combination with an effective amount of a therapeutic antibody or chemically attached to a tumor tissue targeting antibody (or therapeutic fragment thereof), a chemotherapeutic or an immunotoxic agent, for treatment of tumors. Illustrative examples of chemotherapeutic agents that can be used for this purpose include doxorubicin, docetaxel, or taxol. It should be further understood that the invention includes combination therapy including a compound of the invention, including but not limited to vasculostatic agents, such as tyrosine, serine or threonine kinase inhibitors, for example, Src-family inhibitors, and any chemotherapeutic agent or therapeutic antibody.
[0127] The present invention also provides screening assays using appropriate cells which express any kinases within the MAPK pathway. Such cells include cells from mammals, yeast, Drosophila or E. coli. For example, cells which express the Raf polypeptide or any kinase downstream of Raf such as MEK or ERKl /2 or respond to Raf polypeptide or MAPK It"'' <U» Sf „ ' 'U' /3 ILB .....S . -■•« 1I-H ' ■••>>" - pathway polypeptides are contacted with a test compound to observe binding, or stimulation or inhibition of a functional response. The cells which are contacted with the candidate compound are compared with the same cells which are not contacted for Raf polypeptide or MAPK pathway polypeptide activity
[0128] This invention contemplates the treatment and/or amelioration of such diseases by administering a MAPK pathway polypeptides inhibiting amount of a compound. Without wishing to be bound by any particular theory of the functioning of the MAPK pathway polypeptides, it is believed that among the useful inhibitors of MAPK pathway polypeptides function are those compounds which inhibit the kinase activity of the MAPK pathway polypeptides. Other sites of inhibition are, of course, possible owing to its position in a signal transduction cascade. Inhibitors of protein-protein interactions between, for example Raf polypeptide or MAPK pathway polypeptides and other factors could lead to the development of pharmaceutical agents for the modulation of Raf polypeptide or MAPK pathway polypeptides activity.
[0129] Targeting an allosteric site of the protein is a very promising approach for pharmaceutical intervention. Further, the traditional approach of inhibiting various protein kinases includes targeting the ATP binding site. The invention is not meant to be limited by any particular mechanism of inhibition. The assays of the invention may test binding of a candidate compound wherein adherence to the cells bearing the Raf polypeptide or MAPK pathway polypeptides is detected by means of a label directly or indirectly associated with the candidate compound or in an assay involving competition with a labeled competitor. Further, these assays may test whether the candidate compound results in a signal generated by activation of MAPK pathway polypeptides, using detection systems appropriate to the cells bearing the Raf polypeptide or MAPK pathway polypeptides. Inhibitors of activation are generally assayed in the presence of a known agonist and the effect on activation by the agonist by the presence of the candidate compound is observed. Standard methods for conducting such screening assays are well understood in the art and are also illustrated in the examples below (e.g., direct and raf 1 -MEKl assays or MAPK pathway cellular assays).
[0130] The following examples are provided to further illustrate the advantages and features of the present invention, but are not intended to limit the scope of the invention. P1 IL. B ■' U ai U ia .- „,..(. .uu .w ....... ..
Example 1 -General Methods
[0131] Example 1 describes general synthetic procedures that were used to make the compounds described in the subsequent examples. AU solvents were used without further purification. Reactions can be usually conducted without an inert gas atmosphere unless specified otherwise. The reported yields are based on unoptimized conditions and single test runs. The yields can be optimized by changing the reaction conditions, such as solvent, use of base or acid, temperature, use of catalyst and the time of the reaction. Microwave reactions were run in Emrys™ Process vials (2-5 mL) using Initiator module (Biotage/Personal chemistry). All 1H NMR were run on a 500 MHz Bruker NMR or Bruker Avance 400 MHz NMR. Chemical shifts are reported in delta (δ) units, parts per million (ppm) downfield from tetramethylsilane. Coupling constants are reported in hertz (Hz). A Waters LC/MS system is used in identity and purity analysis. This system includes a 2795 separation module, a 996 photodiode array detector and a ZQ2000 mass spectrometer. A Zorbax SB column (150 x 4.6 mm 3.5μ, Agilent Technologies) was used for the LC. Column temperature was 4O0C. Compounds were separated using gradient elution with mobile phases of water (0.05% TFA (A)) and acetonitrile (0.05% TFA (B)). Flow rate was lmL/min. The gradient program used in separation was 0-15 min: 5-60 % B; 15-15.5 min: 60-100 % B; 15.5-17 min: 100 % B.
Example 2. Synthesis of 3-amino-benzori,2,4~|triazine-7-ol-l-oxide
Figure imgf000064_0001
[0132] 7.7 g (0.05 mol) of 4-amino-3-nitrophenol was dissolved in 20 mL of glacial acetic acid and the resulting bright-red solution was heated to approximately 1000C in 500 mL round-bottom flask equipped with a long condenser. To this solution was added a solution of 16.81 g (8.0 equivalent, 0.4 mol) of cyanamide in 20 mL of concentrated hydrochloric acid. In approximately 5-10 min the reaction mixture started to boil vigorously, so the heating was removed and it was stirred without heating until boiling subsided. Then the heating was reapplied and the reaction mixture was refluxed for 48 hrs. Then 150 mL of 30 % NaOH was added and the resulting dark-reddish solution was refluxed for additional 3 hrs. Then it was cooled down to room temperature and dark-red slurry was formed. The red precipitate was filtered, re-dissolved in 200 mL of water and IN HCl was added in portions with stirring until P C I / IfJ ?"» U !!» .•' .»..'.i «.,ii iui .au is pH reached 5-4. The solution changed color from dark-red into light-yellow and a light- yellow fine precipitate was formed. The precipitate was filtered, washed twice with 50 mL of water, twice with 50 mL of acetonitrile and finally twice with 50 mL of diethyl ether and dried in vacuum to give 4.6 g of a bright-yellow solid. Yield: 51.7 %.
[0133] 1H NMR (DMSOd6): δ 6.96 (s, 2H), 7.35-7.38 (m, 2H), 7.45-7.47 (m, IH), 10.36 (s, IH).
Example 3. Synthesis of 3-amino-benzoFl ,2,41triazine-7-ol
Figure imgf000065_0001
[0134] 4.6 g (25.82 mmol) of 3-amino-benzo[l,2,4]triazine-7-ol-l -oxide was dissolved in 200 mL of 1 : 1 mixture of dimethylformamide and methanol. 0.5 g of 10% Pd/C was added to this solution and H2 gas was bubbled through the solution for 3 hours. The progress of the reaction was monitored by TLC, using a 9: 1 mixture of dichloromethane/methanol as an eluent and a UV lamp. The starting material is highly fluorescent under UV, while the product is not. When the reaction was complete, the resulting dark solution was filtered through a short pad of silica gel and solvent was removed in vacuum to produce a dirty- brown solid. 40 mL of ethyl acetate and 40 mL of methanol were added to the solid and the resulting suspension was heated to reflux for about 10 min. Then the suspension was allowed to cool down to ambient temperature. The solid was collected by filtration, washed with 40 mL of ethyl acetate, 40 mL of diethyl ether and dried in vacuum to yield 3.2 g of the product in a form of a greenish solid. Yield: 76%.
[0135] 1H NMR (DMSO-d6): δ 7.18 (s, 1 H), 7.36 (d, J= 2.6 Hz, 1 H), 7.40-7.42 (dd, Jj = 9.1 Hz, J2 = 2.6 Hz, 1 H), 7.45-7.46 (d, J= 9.1 Hz, IH).
Example 4. Synthesis of 4-chloro-pyridine-2-carboxylic acid methyl ester hydrochloride salt
Figure imgf000065_0002
[0136] 2.4 mL (0.031 mol, 0.16 equivalent) of anhydrous N,N-dimethylformamide was added dropwise to 72 mL (1.23 mol, 5.0 equivalent) of thionyl chloride in a temperature F" C If /' LI "3 Ul 3 / J U U J. -1 range of 40-500C under argon blanket. The solution was stirred at this temperature for 10 min, then 24.0 g (0.195 mol, 1.0 equivalent) of picolinic acid was added slowly in portions. The reaction mixture was heated at 70-750C with a reflux condenser under argon for 30 hours. Evolution of SO2 gas was observed. The reaction mixture changed colors from green to orange, then to purple over 2 hours, then resulted in an orange solution with a yellow precipitate. It was cooled down to ambient temperature and 150 mL of anhydrous toluene was added. The suspension was concentrated to about 50 mL total on rotovap. This process was repeated three times.
[0137] The resulting orange suspension was cooled down to - 2O0C and 200 mL of methanol was added. The reaction mixture was left to stir at ambient temperature for 18 hours. Then the clear-yellow solution was transferred into a round-bottom flask and solvent was removed in vacuum. The resulting yellow solid was dissolved with heating to 5O0C in 50 mL of methanol, upon cooling 300 mL of diethyl ether were added. The solution was left to stand at O0C for 18 hours. The white precipitate that formed was collected by filtration, washed extensively with diethyl ether and dried in vacuum to yield 29.05 g of the product as a white fluffy solid. Yield: 71.5 %.
[0138] 1H NMR (DMSOd6): δ 3.88 (s, 3H), 7.81-7.83 (dd, J1 = 1.9 Hz, J2 = 5.4 Hz, IH), 8.06-8.07 (d, J= 1.9 Hz, IH), 8.68-8.69 (d, J= 5.4 Hz, IH).
Example 5. Synthesis of 4-chloro-pyridme-2-carboxyric acid methyl amide
Figure imgf000066_0001
[0139] A suspension of 17.8 g (0.103 mol, 1 eq) of 4-chloro-pyridine-2-carboxylic acid methyl ester hydrochloride in 15 mL of methanol was cooled to O0C and slowly treated with a 2.0 M solution of methylamine in tetrahydrofuran at a rate that kept internal temperature below 50C. The reaction mixture was stirred at O0C for 2 hours, then slowly allowed to warm up to ambient temperature and stirred for 18 hours. Solvent was removed in vacuum, approx. 200 mL of ethyl acetate was added and the resulting suspension was filtered. The precipitate was washed with 100 mL of ethyl acetate. The combined ethyl acetate solutions were K IU Ii / U' ;"3 tun si •■* « "">' ••■-* ••■' washed three times with 100 niL of brine and dried over sodium sulfate. Solvent was removed in vacuum to yield 14.16 g of the product as orange oil. Yield: 80.5 %.
[0140] 1H NMR (DMSOd6): δ 2.81-2.82 (d, J= 4.8 Hz, 3H), 7.73-7.75 (dd, J1 = 2.1 Hz, J2 = 5.4 Hz, IH), 8.00-8.01 (d, J= 2.1 Hz, IH), 8.60-8.61 (d, J= 5.4 Hz, IH), 8.84 (q, J= 4.8 Hz, IH).
Example 6. Synthesis of 4-("3-amino-benzori.2.41triazin-7-yloxy)-pyridine-2-carboxylic acid methylamide
Figure imgf000067_0001
[0141] 3.2 g (19.73 mmol) of 3-amino-benzo[l,2,4]triazine-7-ol was dissolved in 80 niL of anhydrous dimethylformamide under argon atmosphere. 2.44 g (21.71 mmol, 1.1 equivalent) of solid potassium terf-butoxide was added to the solution. The resulting dark- red mixture was heated to about 1000C and stirred at that temperature for 15 min. A solution of 3.7 g (21.71 mmol, 1.1 equivalent) of 4-chloro-pyridine-2-carboxylic acid methylamide in 10 mL of anhydrous dimethylformamide was added, followed by 3.28 g (23.68 mmol, 1.2 equivalent) of anhydrous K2CO3. The reaction mixture was heated at 14O0C for 30 hrs. The progress of the reaction was monitored by LC/MS. Then it was allowed to cool down to ambient temperature. The resulting dark-brown slurry was poured into 500 mL of water and 100 mL of ethyl acetate. The formed precipitate was collected by filtration, washed with 50 mL of water, 50 mL of methanol, 50 mL of diethyl ether and dried in vacuum to produce 3.22 g of the product as a dirty-yellow solid. The filtrate was extracted four times with 100 mL of ethyl acetate. The combined extracts were washed 3 times with 100 mL of water, then with brine and dried over anhydrous sodium sulfate. Solvent was removed in vacuum to yield additional 1.2 g of the product in a form of a yellow solid. Yield: 75% combined.
[0142] 1H NMR (DMSOd6): δ 2.78-2.79 (d, J= 4.8 Hz, 3H), 7.25-7.27 (dd, Ji = 2.6 Hz, J2 = 5.6 Hz, 1 H), 7.50-7.51 (d, J= 2.6 Hz, IH), 7.66-7.68 (d, J= 9.2 Hz, IH), 7.71-7.73 (dd, J1 = 2.7 Hz, J2 = 9.2 Hz, 1 H), 7.71 (s, 2H), 8.03-8.05 (d, J= 2.7 Hz, IH), 8.54-8.55 (d, J = 5.6 Hz, IH), 8.78-8.80 (q, J= 4.8 Hz, IH). I"-" C T / 11 'b U " "ft - ' »":ι- IU11U1 ■"»•' " Example 7. Synthesis of 4-[3-f4-chloro-3-trifluoromethyl-phenylamino)-benzori,2,41triazin-
7-yloxyl-pyridme-2-carboxyrie acid methylamide trifluoroacetate salt
Figure imgf000068_0001
CF3COOH
[0143] To a vial with 2 niL of anhydrous dimethylformamide under argon atmosphere were added 15.4 mg (0.0168 mmol, 0.05 equivalent) of fr-zs(dibenzyllideneatone) dipalladium(O), 21.0 mg (0.033 mmol, 0.1 equivalent) of BINAP, 220.0 mg (0.675 mmol, 2.0 equivalent) of anhydrous cesium carbonate, 175.1 mg (0.675 mmol, 2.0 equivalent) of 5- bromo-2-chlorobenzotrifluoride, and 100 mg (0.337 mmol, 1.0 equivalent) of 4-(3-amino- benzo[l,2,4]triazin-7-yloxy)-pyridine-carboxylic acid methylamide, in that particular order. Argon gas was bubbled through the mixture for 5 min. Then the vial was capped and the reaction mixture was heated to 12O0C with stirring under argon atmosphere for 18 hours. At this point LC/MS indicated about 40 % conversion to the product. As pointed out in the previous examples, the longer reaction times result in the formation of the by-product and partial decomposition. So, the reaction mixture was allowed to cool down to ambient temperature, filtered through 0.3 μm syringe filter and purified by reverse-phase prep-HPLC using acetonitrile/water mixture with 0.1 % of TFA.
[0144] ESI-MS: [M+H]+, 475, 477. 1H NMR (DMSOd6): δ 2.79-2.90 (d, J= 4.8 Hz, 3H), 7.31-7.33 (dd, J1 = 2.4 Hz, J2 = 5.4 Hz, IH), 7.55-7.55 (d, J= 2.4 Hz, IH), 7.73-7.75 (d, J= 8.9 Hz, IH), 7.89-7.91 (dd, J1 = 2.5 Hz, J2 = 9.2 Hz, IH), 7.94-7.95 (d, J= 9.2 Hz, IH), 8.24 (d, J= 2.5 Hz, 2H), 8.25-8.27 (dd, J1 = 2.6 Hz, J2 = 8.9 Hz, IH), 8.55 (d, J= 2.6 Hz, IH), 8.60 (d, J= 5.4 Hz, IH), 8.81-8.82 (q, J= 4.8 Hz, IH), 11.36 (s, IH).
Example 8. Synthesis of 4-r3-(4-chloro-phenylaminoVbenzo[l,2,41triazin-7-yloxy1-pyridine- 2-carboxylic acid methylamide trifluoroacetate salt
Figure imgf000068_0002
CF3COOH 614
68
P' C "I" /' O Ϊ3 U !'3 -'' '-:;;ι' 1U! '" ••■"" " '»■■ [0145] To a vial with 2 niL of anhydrous dimethylforaiamide under argon atmosphere were added 15.4 mg (0.0168 mmol, 0.05 equivalent) of £rø(dibenzyllideneatone) dipalladium(O), 21.0 mg (0.033 mmol, 0.1 equivalent) of BESfAP, 220.0 mg (0.675 mmol, 2.0 equivalent) of anhydrous cesium carbonate, 161.0 mg (0.675 mmol, 2.0 equivalent) of 1- chloro-4-iodobenzene, and 100 mg (0.337 mmol, 1.0 equivalent) of 4-(3-amino- benzo[l,2,4]triazin-7-yloxy)-pyridine-carboxylic acid methylamide, in that particular order. Argon gas was bubbled through the mixture for 5 min. Then the vial was capped and the reaction mixture was heated to 12O0C with stirring under argon atmosphere for 18 hours. At this point LC/MS indicated about 30 % conversion to the product. As it was observed from previous examples, the longer reaction times result in the formation of the by-product and partial decomposition. The reaction mixture was allowed to cool down to ambient temperature, filtered through, 0.22 μ syringe filter and purified by reverse-phase preperative HPLC using acetonitrile/water mixture with 0.1 % of TFA.
[0146] ESI-MS: [M+H]+, 407, 409. 1H NMR (DMSOd6): δ 2.79-2.80 (d, J= 4.88 Hz, 3H), 7.30-7.32 (dd, J1 = 2.6 Hz, J2 = 5.6 Hz, IH), 7.44-7.46 (d, J = 6.8 Hz, 2H), 7.54-7.55 (d, J= 2.6 Hz, IH), 7.84-7.87 (dd, J1 = 2.6 Hz, J2 = 9.05 Hz, IH), 7.92-7.94 (d, J= 9.05 Hz, IH), 8.00-8.01 (d, J= 6.8 Hz, 2H), 8.19-8.20 (d, J= 2.6 Hz, IH), 8.58-8.59 (d, J= 5.6 Hz, IH), 8.81-8.82 (q, J= 4.88 Hz, IH), 11.08 (s, IH).
Example 9. Synthesis of 4-r3-(3-Mfluromethyl-benzenesulfonylaminoVbenzo[l,2,41triazin- 7-yloxyl-pyridine-2-carboxylic acid methyl amide trifluoroacetate salt
Figure imgf000069_0001
[0147] 100 mg (0.337 mmol, 1.0 equivalent) of 4-(3-amino-benzo[l,2,4]triazin-7-yloxy)- pyridine-carboxylic acid methylamide were dissolved in 2 mL of anhydrous dimethylformamide with heating to about 1000C. 45.4 mg (0.405 mmol, 1.2 equivalent) of solid tert-BuOK was added to the solution. The resulting dark-red solution was stirred at 1000C for 30 min, then it was allowed to cool down to ambient temperature. 100 mg (0.405 mmol, 1.2 equivalent) of 3-trifluoromethyl-benzenesulfonyl chloride was added to the mixture via a syringe. It was allowed to stir at ambient temperature for 2 hours. The reaction P1 C 1 / IJ hi U ii / -3 U O ,.JL -ir mixture was filtered through 0.22 μ syringe filter and purified by reverse-phase preperative HPLC using acetonitrile/water mixture with 0.1 % of TFA.
[0148] ESI-MS: [M+H]+, 505, 506, 507. 1H NMR (DMSOd6): δ 2.78-2.79 (d, J= 4.8 Hz, 3H), 7.29-7.31 (dd, J1 = 2.6 Hz, J2 = 5.7 Hz, IH), 7.56 (d, J= 2.6 Hz, IH), 7.87-7.90 (t, J= 7.8 Hz, IH), 7.93-7.94 (d, J= 9.2 Hz, IH), 7.97-7.99 (dd, J1 = 2.6 Hz, J2 = 9.2 Hz, IH), 8.06- 8.08 (d, J= 7.8 Hz, IH), 8.21 (d, J= 2.5 Hz, IH), 8.43-8.46 (m, 2H), 8.57-8.58 (d, J= 5.7 Hz, IH), 8.80-8.81 (q, J= 4.8 Hz, IH).
Example 10. Synthesis of 4-r3-(3-trifluromethyl-benzoylamino)-benzo[l,2,41triazin-7- yloxyl-pyridine-2-carboxylic acid methyl amide trifluoroacetate salt
Figure imgf000070_0001
[0149] 100 mg (0.337 mmol, 1.0 equivalent) of 4-(3-amino-benzo[l,2,4]triazin-7-yloxy)- pyridine-carboxylic acid methylamide were dissolved in 2 mL of anhydrous dimethylformamide with heating to about 1000C. 45.4 mg (0.405 mmol, 1.2 equivalent) of solid tert-BuOK was added to the solution. The resulting dark-red solution was stirred at 1000C for 30 min, then it was allowed to cool down to ambient temperature. 84.5 mg (0.405 mmol, 1.2 equivalent) of 3-trifluoromethyl-benzoyl chloride was added to the mixture via a syringe. It was allowed to stir at ambient temperature for 2 hours. The reaction mixture was filtered through 0.22 μ syringe filter and purified by reverse-phase prep-HPLC using acetonitrile/water mixture with 0.1 % of TFA.
[0150] ESI-MS: [M+H]+, 469, 470. 1H NMR (DMSOd6): δ 2.80-2.81 (d, J= 4.88 Hz, 3H), 7.37-7.39 (dd, J1 = 2.6 Hz, J2 = 5.7 Hz, IH), 7.64 (d, J= 2.6 Hz, IH), 7.81-7.84 (t, J= 7.8 Hz, IH), 8.03-8.06 (m, 2H), 8.15-8.17 (d, J= 9.2 Hz, IH), 8.33-8.34 (d, J= 2.6 Hz, IH), 8.36-8.38 (d, J= 8.0 Hz, IH), 8.45 (s, IH), 8.62-8.63 (d, J= 5.7 Hz, IH), 8.84-8.85 (q, J = 4.88 Hz, IH), 12.29 (s, IH). Example 11. Synthesis of 4-r4-rtrifluromethoxy-benzoylamino)-benzori,2,41triazin-7- yloxyi-Pyridine-2-carboxylic acid methyl amide trifluρrρacetate salt
Figure imgf000071_0001
CF3COOH
[0151] 100 mg (0.337 mmol, 1.0 equivalent) of 4-(3-amino-benzo[l,2,4]triazin-7-yloxy)- pyridine-carboxylic acid methylamide were dissolved in 2 mL of anhydrous DMF with heating to about 100 0C. 45.4 mg (0.405 mmol, 1.2 equivalent) of solid 7-BuOK was added to the solution. The resulting dark-red solution was stirred at 100 0C for 30 min, then it was allowed to cool down to ambient temperature. 64 μL (91.0 mg, 0.405 mmol, 1.2 equivalent) of 4-trifluoromethoxy-benzoyl chloride was added to the mixture via a syringe. It was allowed to stir at ambient temperature for 2 hours. The reaction mixture was filtered through 0.22 μ syringe filter and purified by reverse-phase prep-HPLC using acetonitrile/water mixture with 0.1 % of TFA as a solvent system.
[0152] ESI-MS: [M+H]+, 485, 486. 1H NMR (DMSO-d6): δ 2.80-2.81 (d, J = 4.8 Hz, 3H), 7.37-7.38 (dd, J1 = 2.6 Hz, J2 = 5.6 Hz, IH), 7.55-7.57 (d, J= 8.8 Hz, 2H), 7.63-7.64 (d, J= 2.6 Hz, IH), 8.03-8.05 (dd, J1 = 2.7 Hz, J2= 9.1 Hz, IH), 8.14-8.15 (d, J= 9.1 Hz, IH), 8.20-8.22 (d, J= 8.8 Hz, 2H), 8.32-8.33 (d, J=2.7 Hz, IH), 8.83-8.84 (q, J= 4.8 Hz, IH), 12.12 (s, IH).
Example 12. Synthesis of 4-r3-(trifluromethoxy-benzoylamino)-benzori,2,41triazin-7- yloxyl-pyridine-2-carboxylic acid methyl amide trifluoroacetate salt
Figure imgf000071_0002
[0153] 100 mg (0.337 mmol, 1.0 equivalent) of 4-(3-amino-benzo[l,2,4]triazm-7-yloxy)- pyridine-carboxylic acid methylamide were dissolved in 2 mL of anhydrous DMF with heating to about 1000C. 45.4 mg (0.405 mmol, 1.2 equivalent) of solid 7-BuOK was added to the solution. The resulting dark-red solution was stirred at 100 0C for 30 min, then it was allowed to cool down to ambient temperature. 64 μL (91.0 mg, 0.405 mmol, 1.2 equivalent) of 3-trifluoromethoxy-benzoyl chloride was added to the mixture via a syringe. It was allowed to stir at ambient temperature for 2 hours. The reaction mixture was filtered through II"1" If..,.. H ..'' 1U1 wti IUB .....is ■■■ i' «»•» '•'"" •
0.22 μ syringe filter and purified by reverse-phase prep-HPLC using acetonitrile/water mixture with 0.1 % of TFA as a solvent system.
[0154] ESI-MS: [M+H]+, 485, 486. 1H NMR (DMSOd6): δ 2.80-2.81 (d, J= 4.8 Hz, 3H), 7.37-7.39 (dd, J1 = 2.5 Hz, J2 = 5.6 Hz, IH), 7.63-7.64 (d, J= 2.6 Hz, IH), 7.67-7.74 (m, 2H), 8.03-8.05 (dd, J1 = 2.8 Hz, J2= 9.36 Hz, IH), 8.05 (m, IH), 8.12-8.14 (m, IH), 8.14-8.16 (d, J=9.36 Hz, IH), 8.33 (d, J= 2.8 Hz, IH), 8.62-8.63 (d, J= 5.7 Hz, IH), 8.83-8.84 (q, J= 4.8 Hz, IH), 12.21 (s, IH).
Example 13. Synthesis of 4-r3-(chloro-benzenesulfonylamino)-benzorL2,41triazin-7-yloxyl- pyridine-2-carboxylic acid methyl amide trifluoroacetate salt
Figure imgf000072_0001
[0155] 100 mg (0.337 mmol, 1.0 equivalent) of 4-(3-ammo-benzo[l,2,4]triazin-7-yloxy)- pyridine-carboxylic acid methylamide were dissolved in 2 mL of anhydrous DMF with heating to about 100 0C. 45.4 mg (0.405 mmol, 1.2 equivalent) of solid *-BuOK was added to the solution. The resulting dark-red solution was stirred at 100 0C for 30 min, then it was allowed to cool down to ambient temperature. 85.8 mg (0.405 mmol, 1.2 equivalent) of 3- chloro-benzene sulfonyl chloride was added to the mixture via a syringe. It was allowed to stir at ambient temperature for 2 hours. The reaction mixture was filtered through 0.22 μ syringe filter and purified by reverse-phase prep-HPLC using acetonitrile/water mixture with 0.1 % of TF A.
[0156] ESI-MS: [M+H]+, 471, 473, 474. 1H NMR (DMSOd6): δ 2.78-2.79 (d, J= 4.9 Hz, 3H), 7.30-7.32 (dd, J1 = 2.6 Hz, J2 = 5.6 Hz, IH), 7.57 (d, J= 2.6 Hz, IH), 7.65-7.68 (t, J = 9.0 Hz, IH), 7.75-7.77 (m, IH), 7.98-7.99 (m, 2H), 8.10-8.12 (d, J= 7.8 Hz, IH), 8.15 (t, J= 1.8 Hz, IH), 8.22 (d, J= 2.5 Hz, IH), 8.57-8.58 (d, J= 5.4 Hz, IH), 8.80-8.83 (q, J= 4.9 Hz, IH). Ii1"" ILi- it / 5I..." 2i o ;;;;;» .- ....JJ tun M,»U „_. » Example 14. Synthesis of 4-[2-('tri£luoromethyl-benzenesulfonylamino')-benzo[1.2,41tria2m-
7-yloxyl -Pyridine-2-carboxylic acid methyl amide trifluoroacetate salt
Figure imgf000073_0001
CF3COOH
[0157] 100 mg (0.337 mmol, 1.0 equivalent) of 4-(3-amino-benzo[l,2,4]triazin-7-yloxy)- pyridine-carboxylic acid methylamide were dissolved in 2 niL of anhydrous DMF with heating to about 100 0C. 45.4 mg (0.405 mmol, 1.2 equivalent) of solid £-BuOK was added to the solution. The resulting dark-red solution was stirred at 100 0C for 30 min, then it was allowed to cool down to ambient temperature. 100 mg (0.405 mmol, 1.2 equivalent) of 2- trifluoromethyl-benzenesulfonyl chloride was added to the mixture via a syringe. It was allowed to stir at ambient temperature for 2 hours. The reaction mixture was filtered through 0.22 μ syringe filter and purified by reverse-phase prep-HPLC using acetonitrile/water mixture with 0.1 % of TFA.
[0158] ESI-MS: [M+H]+, 505, 506, 507. 1H NMR (DMSOd6): δ 2.78-2.79 (d, J= 4.8 Hz, 3H), 7.29-7.31 (dd, J1 = 2.6 Hz, J2 = 5.7 Hz, IH), 7.56 (d, J= 2.6 Hz, IH), 7.87-7.99 (m, 5H), 8.21 (d, J= 2.6 Hz, IH), 8.56-8.58 (d, IH), 8.59-8.60 (d, J= 8.0 Hz, IH), 8.80-8.81 (q, J = 4.8 Hz, IH).
Example 15. Synthesis of 4-r2-chloro-5-(trifluoromethyl-benzenesulfonylamino)- benzori,2,41triazin-7-yloxyl-ρyridine-2-carboxylic acid methyl amide trifluoroacetate salt
Figure imgf000073_0002
[0159] The experimental procedure that was used was the same as described in Example 9.
[0160] ESI-MS: [M+H]+, 539, 541, 542. 1H NMR (DMSOd6): δ (ppm) 2.78-2.79 (d, J= 4.9 Hz, 3H), 7.27-7.29 (dd, J1 = 2.6 Hz, J2 = 5.7 Hz, IH), 7.55 (d, J= 2.6 Hz, IH), 7.63-7.65 (d, J= 9.1 Hz, IH), 7.86-7.88 (d, J= 8.4 Hz, IH), 7.91-7.94 (dd, J1 = 2.6 Hz, J2 = 9.1 Hz, IH), 8.04-8.06 (dd, , J1 = 2.0 Hz, J2 = 8.4 Hz, IH), 8.17-8.18 (d, J= 2.6 Hz, IH), 8.56-8.57 (d, J= 5.7 Hz, IH), 8.60-8.61 (d, J= 2.0 Hz, IH), 8.80-8.81 (q, J= 4.9 Hz, IH). it"" IU Si !i..,lf Si U !ZB ^' ...-J-1 1I"* "».» ..Ji»
Example 16. Synthesis of 443-cmoro-6-methoxy-benzenesulfonylaminoV benzori,2,41triazin-7-vloxvl-pyridine-2-carboxvlic acid methyl amide trifluoroacetate salt
Figure imgf000074_0001
[0161] The experimental procedure that was used was the same as described in Example
9.
[0162] ESI-MS: [M+H]+, 501, 503, 504. 1H NMR (DMSOd6): δ (ppm) 2.78-2.79 (d, J= 4.9 Hz, 3H), 3.85 (s, 3H), 7.20-7.22 (d, J= 8.9 Hz, IH), 7.29-7.31 (dd, J1 = 2.6 Hz, J2 = 5.7 Hz, IH), 7.55 (d, J= 2.6 Hz, IH), 7.67-7.70 (dd, J1 = 2.8 Hz, J2 = 8.9 Hz, IH), 7.80-7.81 (d, J= 9.2 Hz, IH), 7.92-7.95 (dd, J1 = 2.8 Hz, J2 = 9.2 Hz, IH), 8.05-8.06 (d, J= 2.8 Hz, IH), 8.19-8.20 (d, J= 2.6 Hz, IH), 8.57-8.58 (d, J= 5.6 Hz, IH), 8.80-8.81 (q, J= 4.9 Hz, IH), 13.00 (br.s. IH).
Example 17. Synthesis of 4-[3-(5-chloro-thiophene-2-sulfonylamino)-benzo[l,2,41triazin-7- yloxyl-ρyridine-2-carboxylic acid methyl amide trifluoroacetate salt
Figure imgf000074_0002
CF3COOH
[0163] The experimental procedure that was used was the same as described in Example
9.
[0164] ESI-MS: [M+H]+, 477, 479. 1H NMR (DMSOd6): δ (ppm) 2.79-2.80 (d, J= 4.8 Hz, 3H), 7.26-7.27 (d, J= 4.1 Hz, IH), 7.32-7.34 (dd, J1 = 2.6 Hz, J2 = 5.6 Hz, IH), 7.59 (d, J= 2.6 Hz, IH), 7.88-7.89 (d, J= 4.1 Hz, IH), 7.99-8.02 (dd, J1 = 2.7 Hz, J2 = 9.1 Hz, IH)5 8.12-8.14 (d, J= 9.1 Hz, IH), 8.26 (d, J= 2.7 Hz, IH), 8.59 (d, J= 5.6 Hz, IH), 8.82-8.83 (q, J= 4.8 Hz, IH), 13.00 (br.s. IH). if" K is ..' 'U' .::;:u tut :z$ ; %..» lu- _.u» ■■■ r
Example 18. Synthesis of 4-r2-chloro-3-trifluromethyl-benzoylaminoVbenzori,2.41triazin-7- yloxyl-τ)yridine-2-carboxylic acid methyl amide trifluoroacetate salt
Figure imgf000075_0001
[0165] The experimental procedure that was used was the same as described in Example 10.
[0166] ESI-MS: [M+H]+, 503, 505, 506. 1H NMR (DMSOd6): δ (ppm) 2.80-2.81 (d, J= 4.8 Hz, 3H), 7.36-7.37 (dd, J1 = 2.6 Hz5J2 = 5.6 Hz, IH), 7.61 (d, J= 2.6 Hz, IH), 7.82-7.83 (d, J= 8.4 Hz, IH), 7.90-7.92 (dd, J1 = 2.1 Hz, J2 = 8.4 Hz, IH), 8.00-8.03 (m, 2H), 8.09 (m, IH), 8.29 (m, IH), 8.61-8.62 (d, J = 5.6 Hz, IH), 8.83-8.84 (q, J= 4.8 Hz, IH), 12.41 (s, IH).
Example 19. Synthesis of 4-r2-chloro-3-trifluromethyl-benzoylamino)-benzorL2,41triazin-7- yloxyl-pyridine-2-carboxvlic acid methyl amide trifluoroacetate salt
Figure imgf000075_0002
[0167] The experimental procedure that was used was the same as described in Example
10.
[0168] ESI-MS: [M+H]+, 435, 437. 1H NMR (DMSOd6): δ (ppm) 2.80-2.81 (d, J= 4.8 Hz, 3H), 7.37-7.39 (dd, J1 = 2.6 Hz, J2 = 5.6 Hz, IH), 7.59-7.62 (t, J= 7.8 Hz, IH), 7.63-7.64 (d, J= 2.6 Hz, IH), 7.72-7.74 (m, IH), 8.03-8.05 (m, 2H), 8.13 (m, IH), 8.15 (d, J= 9.1 Hz, IH), 8.33 (d, J = 2.6 Hz, IH), 8.62-8.63 (d, J= 5.6 Hz, IH), 8.83-8.84 (q, J= 4.8 Hz, IH), 12.12 (s, IH). 11 ILt !i ..' i|...|i z'ii tut ::;:;» „> u «uι iu» -«.. -ir
Example 20. Synthesis of 4-r2,4-dichloro-benzoylaminoVbenzori,2,41triazin-7-yloxyl- pyridine-2-carboxylic acid methyl amide trifluoroacetate salt
Figure imgf000076_0001
CF3COOH
[0169] The experimental procedure that was used was the same as described in Example
10.
[0170] ESI-MS: [M+H]+, 469, 471, 472. 1H NMR (DMSOd6): δ (ppm) 2.80-2.81 (d, J= 4.8 Hz, 3H), 7.35-7.37 (dd, J1 = 2.6 Hz, J2 = 5.6 Hz, IH), 7.55-7.57 (dd, Ji = IS Hz, J2 = 8.3 Hz, IH), 7.61 (d, J= 2.6 Hz, IH), 7.68-7.69 (d, J= 8.3 Hz, IH), 7.76-7.77 (d, J= 1.9 Hz, IH), 8.01 (m, 2H), 8.28-8.29 (m, IH), 8.61 (d, J = 5.6 Hz, IH), 8.83-8.84 (q, J = 4.8 Hz, IH)5 12.28 (s, IH).
Example 21. Synthesis of 4-[2-fluoro-3-chloro-5-trifluoromethyl-benzoylamino)- benzoT l,2,41triazin-7-yloxyl-pyridine-2-carboxylic acid methyl amide trifluoroacetate salt
Figure imgf000076_0002
[0171] The experimental procedure that was used was the same as described in Example
10.
[0172] ESI-MS: [M+H]+, 521, 523, 524. 1H NMR (DMSOd6): δ (ppm) 2.80-2.81 (d, J = 4.8 Hz, 3H), 7.36-7.38 (dd, J1 = 2.6 Hz, J2 = 5.6 Hz, IH), 7.62 (d, J= 2.6 Hz, IH), 8.02-8.05 (dd, J1 = 2.6 Hz, J2 = 9.2 Hz, IH), 8.07-8.09 (d, J= 9.2 Hz, IH), 8.14-8.15 (dd, IH), 8.31- 8.32 (d, J= 2.6 Hz, IH), 8.33-8.34 (dd, J1 = 2.1 Hz, J2 = 6.4 Hz, IH), 8.62 (d, J= 5.6 Hz, IH), 8.83-8.84 (q, J= 4.8 Hz, IH), 12.45 (s, IH). IV" '4,..I- U ..' IU' .,-.!' H-Il „...11 .»' ..».!> il-.it i|,j! ...U.. Ii
Example 22. Synthesis of 4-r3-f4-chloro-3-trifluoromethyl-benzoylaminoV benzori,2,41triazm-7-yloxyl-Pyridine-2-carboxylic acid methyl amide trifluoroacetate salt
Figure imgf000077_0001
[0173] The experimental procedure that was used was the same as described in Example 10.
[0174] ESI-MS: [MR-H]+, 503, 505, 506. 1H NMR (DMSO-d6): δ (ppm) 2.80-2.81 (d, J= 4.8 Hz, 3H), 7.37-7.39 (dd, J1 = 2.6 Hz, J2 = 5.6 Hz, IH), 7.63-7.64 (d, J= 2.6 Hz, IH), 7.95- 7.97 (d, J= 8.4 Hz, IH), 8.04-8.06 (dd, J1 = 2.7 Hz, J2 = 9.3 Hz, IH), 8.15-8.17 (d, J= 9.3 Hz, IH), 8.33-8.34 (d, J= 2.7 Hz, IH), 8.34-8.36 (dd, J1 = Ll Hz, J2 = 8.4 Hz, IH), 8.54- 8.56 (d, J= 2.1 Hz, IH), 8.62-8.63 (d, J= 5.6 Hz, IH), 8.83-8.84 (q, J= 4.8 Hz, IH), 12.35 (s, IH).
Example 23. Synthesis of 4-r3-(2-chloro-3-trifluoromethyl-benzoylamino)- benzori,2,41triazin-7-vloxv1-pyridine-2-carboxvlic acid methyl amide trifluoroacetate salt
Figure imgf000077_0002
[0175] The experimental procedure that was used was the same as described in Example 10.
[0176] ESI-MS: [M+H]+, 503, 505, 506. 1H NMR (DMSOd6): δ (ppm) 2.80-2.81 (d, J = 4.8 Hz, 3H), 7.35-7.36 (dd, J7 = 2.6 Hz, J2 = 5.5 Hz, IH), 7.60-7.61 (d, J= 2.6 Hz, IH), 7.66- 7.69 (t, J= 7.8 Hz, IH), 7.92-7.94 (m, 2H), 7.99-8.02 (m, 2H), 8.29 (d, J= 2.8 Hz, IH), 8.61- 8.62 (d, J= 5.6 Hz, IH), 8.83-8.84 (q, J= 4.8 Hz, IH), 12.43 (s, IH). Example 24. Synthesis of 4-r3-(3-trifluoromethoxy-benzoγlaminoVbenzo[l,2,41triazin-7- vloxvl-Pvridine-2-carboxyric acid methyl amide trifluoroacetate salt
Figure imgf000078_0001
CF3COOH
[0177] The experimental procedure that was used was the same as described in Example 10.
[0178] ESI-MS: [M+H]+, 485, 486, 487. 1H NMR (DMSOd6): δ (ppm) 2.80-2.81 (d, J = 4.8 Hz, 3H), 7.35-7.37 (dd, J1 = 2.6 Hz, J2 = 5.6 Hz, IH), 7.48-7.50 (d, J= 8.3 Hz, IH), 7.53- 7.56 (dt, Ji= 0.9 Hz, J2 = 7.6 Hz, IH), 7.62 (d, J= 2.6 Hz, IH), 7.66-7.68 (dt, Ji= 1.7 Hz, J2 = 8.0 Hz, IH), 7.78-7.79 (dt, Ji = 1.7 Hz, J2 = 7.6 Hz, IH), 7.99-8.02 (m, 2H), 8.29 (d, J= 2.6 Hz, IH), 8.61 (d, J= 5.6 Hz, IH), 8.83-8.84 (q, J= 4.8 Hz, IH), 12.22 (s, IH).
Example 25. Synthesis of 4-i3-r(5-Methyl-isoxazole-3-carbonyl)-amino]- benzori,2,4]triazin-7-yloxyl-pyridine-2-carboxylic acid methyl amide trifluoroacetate salt
Figure imgf000078_0002
CF3COOH
[0179] The experimental procedure that was used was the same as described in Example 10. 1H NMR (DMSOd6): δ (ppm) 2.53 (s, 3H), 2.80-2.81 (d, J= 4.8 Hz, 3H), 6.79 (s, IH), 7.37-7.38 (dd, J1 = 2.5 Hz, J2 = 5.5 Hz, IH), 7.63-7.64 (d, J= 2.4 Hz, IH), 8.04-8.06 (dd, Ji= 2.5 Hz, J2 = 9.2 Hz, IH), 8.15-8.17 (d, J= 9.2 Hz, IH), 8.32-8.33 (d, J= 2.5 Hz, IH), 8.61- 8.62 (d, J= 5.5 Hz, IH), 8.83-8.84 (q, J= 4.8 Hz, IH), 12.04 (s, IH).
Example 26 Testing of Inhibition of Raf Kinase In Vitro
[0180] The ability of compounds of general structure (A) of the present invention to inhibit the kinase activity of Raf 1 was evaluated using two methods: a direct and a Rafl- MEKl assay. In the direct assay, kinase reactions were conducted in 96-well plates by combining recombinant human rafl (29.4 U/well, Upstate, Lake Placid, NY), ATP (3 μM), myelin basic protein substrate (MBP, lmg/ml, Upstate, Lake Placid, NY), and test agents (at concentrations ranging from about 1 nM/1 to about 10 μM), in the presence of kinase reaction buffer. The Rafl -MEKl assay utilized 2.9U/well rafl and 0.25 ug/well inactive MEKl (MEKl inactive, Upstate, Lake Placid, NY) and 3uM ATP. After reacting for 60 minutes at 30 0C, residual ATP was measured using a luciferase-based assay (KinaseGlo, Promega Corp.) as a measure of kinase activity. Data from four wells were then averaged and used to determine IC5O values for the test compounds (Prism software package, GraphPad Software, San Diego CA).
[0181] The test results were as follows: a known Raf inhibitor, compound A, displayed an IC50 of 16 nM; a known Raf inhibitor, compound B, displayed an IC50 of 43 nM; and an invention compound C, showed an IC50 of 76 nM. Other invention compounds exemplified in Figure 1, displayed an IC50 below 100 μM.
Example 27. Testing of Inhibition of MAPK Pathway in Cellular Assay
[0182] Western Blot: Early passage primary human umbilical vein endothelial cells (HUVECs) were maintained in EGM-2 containing SingleQuots (Cambrex, East Rutherford, NJ), 10% FBS, 1OmM HEPES, and 50 μg/ml gentamicin. Prior to treatment of the cells with inhibitor, the HUVECs were starved for 18h by replacing serum-containing complete media with serum-free and SingleQuot-free media. The starved cells were pre-treated with inhibitors for 60 min at various concentrations (0-20 μM). Next the HUVECs were treated with 50 ng/ml VEGF or FGF (Peprotech, Rocky Hill, NJ) for 6 min and the cells were immediately washed with ice-cold PBS. Cells were lysed with ice-cold RIPA buffer containing 10OmM Tris pH 7.5, 150 mM NaCl, 1 niM EDTA, 1% deoxycholate, 1% Triton X-100, 0.1% SDS, 2 mM PMSF, one Complete-Mini protease inhibitor tablet (Roche, Indianapolis, IN; 1 tablet/ 7 ml of lysis buffer) and the phophatase inhibitors NaF (500 mM) and orthovanadate (1 mM). The cells were scraped and lysates transferred and centrifuged at 15,000 g for 10 min. Supernatants were transferred to new tubes and protein concentration was quantitated using the BCA protein reagent (Pierce, Rockford, IL). Cell lysates containing 20 μg of total protein were separated by 10% SDS-PAGE, transferred to nitrocellulose, and blocked in 5% milk in TBST. Anti phospho-ERK Thr 202/Tyr 204 (Cell Signaling, Beverly, MA), anti-phospho-MEK Ser217/221 (Cell Signaling), and c-Raf (BD Biosciences Pharmingen, San Diego, CA) used as primary antibodies were detected with horseradish peroxidase-conjugated goat anti-mouse or rabbit secondary antibodies and bands were visualized using the SuperSignal West Pico chemiluminescence reagent system (Pierce) and Kodak X-ray film (Rochester, NY).
[0183] Bay 43-9006 (Raf/FGF inhibitor) showed reduction of expression of p-MEK and p-ERK with IC50 between 200 and 300 nM when tested in this assay. UO 126 (MEK inhibitor) showed reduction in p-Erk levels with IC50 between 200 and 300 nM, while p- MEK levels were unaffected. The results are shown in Table 1. As can be seen, compounds of the invention showed reduction in p-MEK and p-ERK levels with IC50 between 400 nM and 20 μM.
Example 28. Cell Viability Assay
[0184] XTT assay: HUVECs were seeded at 10,000 cells/well of a tissue culture treated 96-well plate treated with collagen type I and grown overnight in the complete EGM-2 media as described above. The following morning, the inhibitors were serial diluted with DMSO and added to the cells with a final DMSO concentration of 1%. After 24-48 hours cell viability was measured with an XTT assay (Sigma, St. Louis, MO). The cells were also photographed to compare morphological differences to the XTT trends observed. Determination of the IC50 values was performed with quantitative software (Prism software package, GraphPad Software, San Diego CA). Several inhibitors blocked cell proliferation and induced apoptosis at concentrations below 1 μM and experiments were repeated three times to confirm the observations. The compounds of the invention displayed IC50 between 100 nM and 40 uM in this assay (Table 1).
Figure imgf000081_0001
at S uM
active at 5 uM
active at 5 uM
at 5 uM
Figure imgf000082_0001
Example 29. Synthesis of S-methyl N-r4-chloro-3 rtrifluoromethvDphenyllisothiourea hydroiodide
Figure imgf000082_0002
[0185] 4-Chloro-3-trifluoromethyl-phenylthiourea (5.0 g, 19.63 mmol) was dissolved in ca. 80 mL of anhydrous MeOH and methyl iodide (2.93 g, 20.61 mmol) was added via a syringe. The reaction mixture was refluxed for 12 hours. Then it was cooled down to ambient temperature and solvent was removed in vacuo to give colorless oil (7.85 g), which was taken to the next step without further purifications.
[0186] S-methyl N-[4-trifluoromethoxyphenyl]isothiourea hydroiodide, S-methyl N-[4- hydroxy-phenyl]isothiourea hydroiodide, S-methyl N-[3-hydroxy-phenyl]isothiourea hydroiodide and S-methyl N-[3-(trifluoromethyl)phenyl]isothiourea hydroiodide_were prepared according to the method of this example.
[0187] In the case of 1,2,4-triazoles three tautomeric structures can be present, as shown below:
Figure imgf000083_0001
4H-l,2,4-triazoIe lH-l,2,4-triazole lH-l,2,4-triazole
[0188] Even though all three tautomeric structures can exist, all the generic structures and all the examples having 1,2,4-triazole moiety are shown only in one tautomeric form, such as 4H-l,2,4-triazole for simplicity and for the comparison with its direct analogues, such as examples containing 1,3,4-oxadiazole moiety. The prevailing tautomeric structure depends on the substituents on the triazole moiety and on the reaction conditions. As has been shown in the literature, IH- 1,2,4-triazole is usually the most common tautomeric form, especially if an amino substituent is attached to the ring. Using only 4H-tautomeric form to draw the structures for the sake of simplicity, does not imply that the compounds of the examples that follow necessarily exist in that particular tautomeric form. Using this approach, the IUPAC names for the examples below are provided for 4H-tautomeric form only, however it is understood, that upon the elucidation of the exact tautomeric structure the numbering of the substituents may differ from the one that is provided.
Example 30. Synthesis of 4-r5-(4-chloro-3-trifluoromethyl-phenylamino)- 4HT 1.2,41triazol-3-vll-phenol
Figure imgf000083_0002
[0189] 4-hydroxybenzoic acid hydrazide (3.0 g, 19.66 mmol) and S-methyl N-[4-chloro- 3-(trifluoromethyl)phenyl]isothiourea hydroiodide (7.8 g, 19.66 mmol) were suspended in 100 mL of anhydrous pyridine. The reaction mixture was refiuxed for 18 hours under Ar atmosphere. Then it was cooled down to ambient temperature and pyridine was removed in vacuo. The resulting yellow oil was re-dissolved in a small amount of ethyl acetate, loaded on a short pad of silica gel and eluted with 5:1 hexane/ethyl acetate, then with 100 % ethyl acetate to collect the product. The product was re-purified by a second silica gel column chromatography, using ISCO system (80 g pre-packed column, 25 min run, 20% to 50 % EtOAc gradient in hexane). Solvent was removed in vacuo to give the title product as a white solid (2.83 g). Yield 40.4 %. [0190] ESI-MS: [M+H]+ 355.1, 356.8,. 1H NMR (DMSO-d6): δ 6.89-6.91 (d, J= 8.7 Hz, 2H), 7.53-7.55 (d, J= 8.8 Hz, IH), 7.70-7.72 (dd, J1 = 8.8 Hz, J2 = 2.6 Hz, IH), 7.77-7.79 (d, J= 8.7 Hz, 2H), 8.24-8.25 (d, J= 2.6 Hz, IH), 9.81 (s, IH), 9.98 (s, IH), 13.62 (s, IH).
Example 31. Synthesis of 4-(4-r5-("4-chloro-3-trifluoromethγl-ρhenylamino)-4H- rL2.41triazol-3-yl]-ρhenoxy>-pyridine-2-carboxylic acid methylamide trifluoroacetic acid salt
Figure imgf000084_0001
CF3COOH
[0191] ESI-MS: [M+H]+,490, 491. 1H NMR (DMSO-d6): δ 2.78-2.79 (d, J= 4.8 Hz, 3H), 7.24-7.25 (m, IH), 7.40-7.42 (d, J= 8.7 Hz, 2H), 7.48 (s, IH), 7.56-7.58 (d, J= 8.8 Hz, IH), 7.79-7.82 (dd, Jj = 2.6 Hz, J2 = 8.8 Hz, IH), 8.07-8.09 ( d, J= 8.7 Hz, 2H), 8.24-8.25 (d , J= 2.6 Hz, IH), 8.55-8.56 (m, IH), 8.80-8.81 (m, IH), 9.95 (s, IH).
Example 32. Synthesis of (4-chloro-3-trifluorometh.yl-τ)henyl)-|5-r4-(pyridin-3- yloxy)-phenyll-4H-ri,2,41triazol-3-yU -amine trifluoroacetic acid salt
Figure imgf000084_0002
CF3COOH
[0192] 4-[5-(4-chloro-3-trifluoromethyl-phenylamino)-4H[l,2,4]triazol-3-yl]-phenol (127.8 mg, 0.36 mmol) was dissolved in 3 niL of anhydrous DMF in a 5 mL microwave vial (Personal Chemistry). Solid potassium bis(trimethylsilyl)amide (144.0 mg, 0.72 mmol) was added and the reaction mixture was stirred with heating at 80 0C for 15 min, then 3- bromopyridine (68.3 mg, 0.432 mmol) was added, followed by anhydrous K2CO3 (50.0 mg, 0.36 mmol). Then the vial was capped and microwaved at 250 0C for 30 min. Then the reaction mixture was diluted with ca. ImL of MeOH, filtered through 0.22 urn syringe filter and purified by reverse-phase preparative HPLC in acetonitrile/water system with 0.01% TFA. The product was isolated as a TFA salt (15.1 mg).
[0193] ESI-MS: [M+H]+, 432, 433. 1H NMR (DMSO-d6): δ 7.19-7.21 (d, J= 8.8 Hz, 2H), 7.49-7.51 (dd, J1 = 4.5 Hz, J2 = 8.6 Hz, IH), 7.53-7.55 (d, J= 8.8 Hz, IH), 7.58-7.60 (m, IH), 7.74-7.76 (m, IH), 7.95-7.98 (d , J= 8.8 Hz, 2H), 8.21-8.21 (d , J= 2.6 Hz, IH), 8.44 (m, IH), 8.48 (m, IH), 9.88 (s, IH).
Example 33. Synthesis of methyl 4-(ρyridine-3-vloxy)benzoate
Figure imgf000085_0001
[0194] 3-hydroxypyridine (6.17 g, 64.87 mmol) was dissolved in 100 mL of anhydrous DMF under argon atmosphere. Solid K2CO3 (8.96 g, 64.87 mmol) was added, followed by neat methyl 4-fluorobenzoate (10.0 g, 64.87 mmol). The reaction mixture was heated at 135 0C for 10 hrs. The absence of the starting material was confirmed by LC/MS. The reaction mixture was cooled down to ambient temperature and poured into ca. 500 mL OfH2O. The resulting solution was extracted 3 times with ca. 150 mL of EtOAc (during the extraction small volumes of MeOH, Et2O and brine were added to facilitate the separation). Combined EtOAc layers, were washed twice with sat. NaHCO3, twice with brine and dried over anhydrous Na2SO4. Solvent was removed in vacuo to give dark-red oil, which was purified by silica gel chromatography using 1:1 mixture of EtOAc/Hexane as eluent to give the title product (4.8 g, 32.3 % yield) as yellow solid.
[0195] ESI-MS: [M+H]+, 230, 231. 1HNMR (DMSO-d6): δ 3.83 (s, 3H), 7.09-7.12 (d, J = 8.8 Hz, 2H), 7.48-7.51 (dd, J1 = 8.4 Hz, J2 = 4.9 Hz, IH), 7.58-7.61 (dq, J1 = 8.4 Hz, J2 = 1.4 Hz, IH), 7.96-7.99 (d, J= 8.8 Hz, 2H), 8.46-8.47 (m, 2H).
Example 34. Synthesis of 4-(pyridine-3-yloxy)benzohydrazide
Figure imgf000085_0002
[0196] Methyl 4-(pyridine-3-yloxy)benzoate (4.8 g, 20.94 mmol) was dissolved in ca. 150 mL of EtOH and anhydrous hydrazine (4.08 g , 4.0 mL) was added via a syringe. The resulting yellow solution was refluxed for 24 hrs. Then solvent was removed in vacuo to give the title product (4.8 g, 100 % yield) as yellow viscous oil, which upon standing slowly solidified. [0197] ESI-MS: [M+H]+, 230, 231. 1H NMR (DMSO-d6): δ 4.13 (br s., 2H), 7.06-7.09 (d, J= 8.7 Hz, 2H), 7.45-7.47 (dd, J1 = 8.4 Hz, J2 = 4.9 Hz, IH), 7.51-7.54 (dq, J1 = 8.4 Hz, J2 = 1.4 Hz, IH), 7.85-7.88 (d, J= 8.8 Hz, 2H), 8.42-8.43 (m, 2H)5 9.74 (s, IH).
Example 35. Synthesis of ("4-chloro-3-trifluorometliyl-plienyl)-|5-r4-rpyridin-3- vloxv)-phenvll-4H-ri.2.41triazol-3-yl>-amine
Figure imgf000086_0001
[0198] 4-(pyridine-3-yloxy)benzohydrazide (2.33 g, 10.2 mmol) was dissolved in ca. 70 HiL of anhydrous pyridine and S-methyl N-[4-chloro-3-(trifluoromethyl)phenyl] isothiourea hydroiodide (4.04 g, 10.2 mmol) was added. The reaction mixture was refluxed for 18 hrs under Ar. The pyridine was removed in vacuo and the resulting residue was purified by silica gel chromatography using EtOAc as eluent to give the title product (0.56 g) as a white solid.
[0199] ESI-MS: [M+H]+, 432, 433. 1H NMR (DMSOd6): δ 7.20-7.23 (d, J= 8.8 Hz, 2H), 7.46-7.49 (dd, J1 = 8.4 Hz, J2 = 4.5 Hz, IH), 7.54-7.57 (m, 2H), 7.75-7.77 (dd, J1 = 8.8 Hz, J2 = 2.6 Hz, IH), 7.97-7.99 (d , J= 8.8 Hz, 2H), 8.23-8.24 (d , J= 2.6 Hz, IH), 8.43 (m, IH), 8.46 (d, J= 2.6 Hz, IH), 9.88 (s, IH), 13.91 (s, IH).
Example 36. Synthesis of 6-{4-[5-(4-chloro-3-trifluoromethyl-phenylaminoV4H- ri,2,41triazol-3-yl1-phenoxy}-pyrimidine-2,4-diamine
Figure imgf000086_0002
[0200] 4-[5-(4-chloro-3-trifluoromethyl-phenylamino)-4Η[l,2,4]triazol-3-yl]-phenol (1.3 g, 3.66 mmol) was dissolved in 18 mL of anhydrous dioxane in a 10-20 niL microwave vial (Personal Chemistry). Solid Cs2CO3 (1.19 g, 3.66 mmol, 1.0 eq) was added and the reaction mixture was stirred with heating at 80 0C for 10 min, then 4-chloro-2,4-diaminopyrimidine (0.530 g, 3.66 mmol) was added. The vial was capped and microwaved at 200 0C for 25 min. Then the reaction mixture was diluted with ca. 10 mL of MeOH, transferred into a round- bottom flask and concentrated in vacuo to ca. 20 mL. The resulting reddish solution was loaded on a short pad of silica gel and eluted first with 100 % ethyl acetate to remove the unreacted starting material and then with 20 % MeOH in EtOAc to elute the product. Tlαe product was further purified by ISCO system (80 g pre-packed column, 40 min method, 0% to 10 % MeOH gradient in ethyl acetate). Solvent was removed in vacuo to give the title product as an off-white solid (0.785 g). Yield 46.3 %.
[0201] ESI-MS: [M+H]+, 463, 464, 465. 1H NMR (DMSO-d6): δ 5.15 (s, IH), 6.03 (s, 2H), 6.31 (s, 2H), 7.25-7.27 (d, J= 8.6 Hz, 2H), 7.55-7.57 (d, J= 8.8 Hz, IH), 7.77-7.78 (m, IH), 7.96-7.97 (d , J= 8.8 Hz, 2H), 8.25-8.26 (d , J= 2.6 Hz, IH), 9.91 (s, IH), 13.91 (s, IH). Anal. Calcd for (C19H14ClF3N8O x 0.4 EtOAc): C, 49.68; H, 3.48; N, 22.50, Found: C, 49.61; H, 3.55; N, 22.90.
Example 37. Synthesis of 6-{4-r5-(4-chloro-3-trifluoromethyl-phenylamino)-4H- rL2.41triazol-3-yll-phenoxy|-pyrimidine-2.4-diamine methanesulfonic acid salt
Figure imgf000087_0001
MeSO3H
[0202] 6-{4-[5-(4-Chloro-3-trifluoromethyl-phenylamino)-4H-[l,2,4]triazol-3-yl]- phenoxy}-pyrimidine-2,4-diamine x 0.4 EtOAc complex (470.0 mg, 0.943 mmol) was dissolved in ca. 50 niL of anhydrous methanol and methasulfonic acid (0.0612 mL, 0.943 mmol, 1.0 eq.) was added. The resulting solution was stirred for 30 min. Solvent was removed in vacuo and the resulting light-yellow foam was dried at 70 0C in high vacuum for 3 hrs to give the title compound as an off-white solid (527.3 mg). Yield 100%.
[0203] ESI-MS: [M+Η]+, 463, 464. 1H NMR (DMSO-d6): δ 2.36 (s, 3H), 5.40 (s, IH), 7.39-7.40 (d, J= 8.6 Hz, 2H), 7.57-7.58 (d, J= 8.8 Hz, IH), 7.78 (m, IH), 7.80 (br.s., 4H), 8.02-8.03 (d , J= 8.8 Hz, 2H), 8.25-8.26 (d , J= 2.6 Hz, IH), 9.94 (s, IH), 13.98 (br..s. IH). Anal. Calcd for (Ci9H14ClF3N8O x 1 CH3SO3H): C, 42.98; H, 3.25; N, 20.05, F^und: C, 42.93; H, 3.62; N, 20.12. Example 38. Synthesis of methyl 4-r(2.6-diammopvrimidin-4-yl)oxv1benzoate
Figure imgf000088_0001
[0204] Methyl 4-hydroxybenzoate (1.52 g, 10.0 mmol) was dissolved in 18 niL of anhydrous dioxane in 10-20 niL microwave vial (Personal Chemistry) and solid Cs2CO3 was added to this solution. The suspension was stirred at ambient temperature for 10 min, then A- chloro-2,6-diaminopyrimidine (1.45 g, 10.0 mmol) was added. The vial was capped and microwaved at 2000C for 40 min. Then MeOH was added to dissolve the formed suspension to produce a clear amber solution. The solution was transferred into a round-bottom flask and concentrated down to ca. 20 mL. This solution was purified by silica gel chromatography using 100 % ethyl acetate as eluent. The product was additionally re- crystallized from ca. 50 mL of 4:1 mixture of EtOAc/MeOH. The product was filtered, washed with 40 mL of EtOAc, 40 mL of anhydrous Et2O and dried in vacuo to give the title product as a white solid (0.812 g). Yield 31.2%.
[0205] ESI-MS: [M+H]+, 261.01. 1HNMR (DMSO-d6): δ 3.84 (s, 3H), 5.19 (s, IH), 6.06 (br. s, 2H), 6.37 (br.s, 2H), 7.19-7.21 (d, J= 8.7 Hz, 2H), 7.95-7.97 (d, J= 8.7 Hz, 2H). 13C NMR (DMSO-d6) 52.1, 78.3, 121.0, 125.3, 130.9, 157.8, 163.2, 165.7, 166.6, 169.2.
Example 39. Synthesis of 4-r(2,6-diaminoρyrimidin-4-yl')oxylbenzohvdrazide
Figure imgf000088_0002
[0206] Methyl 4-[(2,6-diammopyrimidin-4-yl)oxy]benzoate (2.74 g, 10.52 mmol) was suspended in ca. 180 mL of anhydrous methanol and anhydrous hydrazine (1.021 g, 1.0 mL, 31.85 mmol, 3.03 eq) was added to this suspension. The reaction mixture was refluxed for 3 hours, then MeOH was very slowly distilled off till the total volume reached ca. 30 mL. This solution was allowed to stand at ambient temperature for 48 hrs. A white precipitate slowly crystallized out. It was collected, washed with 40 mL of EtOAc, 40 mL of anhydrous Et2O and dried in vacuo to give the title product as a fine white powder (2.02 g). Yield 73.7 %.
[0207] ESI-MS: [M+H]+, 261.12. 1HNMR (DMSO-d6): δ 4.54 (br.s., 2H), 5.13 (s, IH), 6.01 (br. s, 2H), 6.30 (br.s, 2H), 7.13-7.14 (d, J= 8.7 Hz, 2H), 7.83-7.84 (d, J= 8.7 Hz, 2H), 9.75 (s, IH). 13C NMR (DMSO-d6) 77.9, 120.9, 128.5, 129.3, 155.8, 163.3, 165.4, 166.6, 169.7.
Example 40. Synthesis of 6-{4-[5-("4-chloro-3-trifluoromethyl-phenylamino)-4H- r 1 ,2,4~ltriazol-3 -yl] -phenoxy} -pyrimidine-2 ,4-diamine
Figure imgf000089_0001
[0208] 4-[(2,6-diaminopyrimidin-4-yl)oxy]benzohydrazide (1.48 g, 5.68 mmol) and S- methyl N-[4-chloro-3-(trifluoromethyl)phenyl]isothiourea hydroiodide (2.33 g, 5.89 mmol) were suspended in 30 mL of anhydrous pyridine. The reaction mixture was refluxed for 18 hours under Ar atmosphere. The formed yellow solution was cooled down to ambient temperature and pyridine was removed in vacuo. The resulting yellow foamy solid was re- dissolved in 50 mL of 5: 1 EtOAc/MeOΗ; ca. 15 g of silica gel was added and solvent was removed in vacuo. The impregnated silica gel was packed into ISCO column and the product was purified using ISCO system (80 g pre-packed column, 50 min run, 0% to 10 % gradient of solvent B in solvent A [Solvent A - 4 mL of MeOH, 4mL of Et3N in 4 L of CH2Cl2; Solvent B - 4 mL Of Et3N in 4 L of MeOH]). Solvent was removed in vacuo to give the title product as a white solid (1.33 g). Yield 50.5 %.
[0209] ESI-MS: [M+H]+, 463, 464. 1H NMR (DMSO-d6): δ 5.15 (s, IH), 6.03 (s, 2H), 6.31 (s, 2H), 7.25-7.27 (d, J = 8.6 Hz, 2H), 7.55-7.57 (d, J= 8.8 Hz, IH), 7.77-7.78 (m, IH), 7.96-7.97 (d , J= 8.8 Hz, 2H), 8.25-8.26 (d , J= 2.6 Hz, IH), 9.91 (s, IH), 13.91 (s, IH). Anal. Calcd for (C19H14ClF3N8O x 0.4 EtOAc): C, 49.68; H, 3.48; N, 22.50, Found: C, 49.61; H, 3.55; N, 22.90. Example 41. Synthesis of 6-i4-r5-(4-chloro-3-trifluoromethyl-phenylaminoV4H- ri.2.41triazol-3-yl1-ρhenoxvl-ρvridazin-3-vlamine trifluoroacetic acid salt
Figure imgf000090_0001
CF3COOH
[0210] 4-[5-(4-chloro-3-trifluoromethyl-phenylamino)-4H[l,2,4]triazol-3-yl]-phenol (120.0 mg, 0.338 mmol) was dissolved in 3 niL of anhydrous DMF in a 5 mL microwave vial (Personal Chemistry). Solid potassium bis(trimethylsilyl)amide (81.0 mg, 0.406 mmol) was added and the reaction mixture was stirred with heating at 80 0C for 15 min, then 3-amino-6- chloro-pyridazine (48.2 mg, 0.372 mmol) was added, followed by anhydrous K2CO3 (46.7 mg, 0.338 mmol). Then the vial was capped and microwaved at 200 0C for 30 min. After reaction was complete, the reaction mixture was diluted with ca. ImL of MeOH, filtered through 0.22 um syringe filter and purified by reverse-phase preparative HPLC in acetonitrile/water system with 0.01% TFA. The product was isolated as a TFA salt (18.2 mg).
[0211] ESI-MS: [M+H]+, 448, 449. 1H NMR (DMSO-d6): δ 7.42-7.43 (d, J= 8.7 Hz, 2H), 7.54-7.56 (d, J= 9.7 Hz, IH), 7.56-7.58 (d, J= 8.8 Hz, IH), 7.76-7.79 (m, IH), 7.78- 7.80 (d, J= 9.7 Hz, IH), 8.01-8.04 (d , J= 8.8 Hz, 2H), 8.25-8.26 (d , J= 2.6 Hz, IH), 8.49 (br.s., 2H), 9.94 (s, IH).
Example 42. Synthesis of 4-r5-(4-trifluoromethoxy-phenylamino)- 4HT1.2.41triazol-3-yll-phenol
Figure imgf000090_0002
[0212] 4-hydroxybenzoic acid hydrazide (0.643 g, 4.23 mmol) and S-methyl N-[4- (trifluoromethoxy)phenyl]ώo^7zoM?-βα hydroiodide (1.6 g, 4.23 mmol) were suspended in 10 mL of anhydrous pyridine. The reaction mixture was refiuxed for 24 hours, during which time it changed color from yellow into orange-red. Then it was cooled down to ambient temperature and poured with stirring into 150 mL of ice- water. The formed white solid was collected, washed thoroughly with water and dried in air. The resulting residue was purified by silica gel chromatography using Isco column with 10% to 100 % gradient of ethyl acetate in hexane. Solvent was removed in vacuo to give the title product as a pinkish solid (575.2 mg). Yield 40.4 %.
[0213] ESI-MS: [M+H]+ 337, 338. 1H NMR (DMSOd6): δ 6.85-6.87 (d, J= 8.0 Hz, 2H), 7.19-7.20 (d, J= 8.0 Hz, 2H), 7.61-7.63 (d, J= 8.7 Hz, 2H), 7.75-7.77 (d , J= 8.7 Hz, 2H), 9.39 (s, IH), 9.92 (s, IH), 13.42 (s, IH).
Example 43. Synthesis of 4-{4-r5-(4-trifluoromethoxy-phenylammo)-4H- ri,2,41triazol-3-yri-phenoxyl-pyridine-2-carboxylic acid methylamide trifluoroacetic acid salt
Figure imgf000091_0001
[0214] 4-[5-(4-trifluoromethoxy-phenylamino)-4H[l,2,4]triazol-3-yl]-phenol (66.4 mg, 0.197 mmol) was dissolved in 2 mL of anhydrous DMF in a 5 mL microwave vial (Personal Chemistry). Solid potassium bis(trimethylsilyl)amide (39.4 mg, 0.197 mmol) was added and the reaction mixture was stirred with heating at 800C for 15 min, then 4-chloro-2- pyridinecarboxamide (33.7 mg, 0.197 mmol) was added, followed by anhydrous K2CO3 (27.3 mg, 0.197 mmol). Then the vial was capped and microwaved at 2000C for 15 min. Then the reaction mixture was diluted with ca. ImL of MeOH, filtered through 0.22 um syringe filter and purified by reverse-phase preparative HPLC in acetonitrile/water system with 0.01% TFA. The product was isolated as a TFA salt (46.6 mg).
[0215] ESI-MS: [MH-H]+ 471, 472. 1H NMR (DMSO-d6): δ 2.78-2.79 (d, J= 4.8 Hz, 3H), 123-125 ( dd, J1 = 5.6 Hz, J2 = 2.6 Hz, IH), 7.25-7.27 (d, J= 8.6 Hz, 2H), 7.38-7.40 (d, J=
8.6 Hz, 2H), 1 Al-I Al (d, J= 2.6 Hz, IH), 7.66-7.70 (d, J= 8.7 Hz, 2H), 8.08-8.11 (d, J =
8.7 Hz, 2H), 8.55-8.56 (d, J= 5.6 Hz, IH), 8.79-8.82 (t, J= 4.8 Hz, IH), 9.58 (s, IH).
Example 44. Synthesis of {5-r4-("pyridin-4-yloxy)-phenyll-4H-π.2.41triazol-3-vU-(4- trifluoromethoxy-phenvD-amine trifluoroacetic acid salt
Figure imgf000091_0002
CF3COOH [0216] 4-[5-(4-trifluoromethoxy-phenylamino)-4H[l,2,4]triazol-3-yl]-phenol (106.0 mg, 0.315 mmol) was dissolved in 2 mL of anhydrous DMF in a 5 niL microwave vial (Personal Chemistry). Solid potassium bis(trimethylsilyl)amide (157.2 mg, 0.788 mmol) was added and the reaction mixture was stirred with heating at 800C for 15 min, then 4-chloropyridine hydrochloride (56.7 mg, 0.378 mmol) was added, followed by anhydrous K2CO3 (44.0 mg, 0.315 mmol). Then the vial was capped and microwaved at 2500C for 20 min. Then the reaction mixture was diluted with ca. ImL of MeOH, filtered through 0.22 um syringe filter and purified by reverse-phase preparative HPLC in acetonitrile/water system with 0.01% TFA. The product was isolated as a TFA salt (66.5 mg of off-white solid).
[0217] ESI-MS: [M+H]+, 415, 416. 1H NMR (DMSO-d6): δ 7.26-7.27 (d, J= 8.7 Hz, 2H), 7.46-7.48 (m, 2H), 7.46-7.48 (d, J= 7.1 Hz, 2H), 7.67-7.70 (d, J= 8.7 Hz, 2H)5 8.13- 8.16 (d, J= 8.7 Hz, 2H), 8.77-8.78 (d, J= 7.1 Hz, 2H), 9.64 (s, IH).
Example 45. Synthesis of 6-{4-[5-(4-trifluoromethoxy-phenylamino)-4H-π,2,41triazol-3-vn- phenoxyl-pyridazm-3-ylamine trifluoroacetic acid salt
Figure imgf000092_0001
CF3COOH
[0218] 4-[5-(4-trifluoromethoxy-phenylamino)-4H[l ,2,4]triazol-3-yl] -phenol (112.0 mg, 0.33 mmol) was dissolved in 2 mL of anhydrous DMF in a 5 mL microwave vial (Personal Chemistry). Solid potassium bis(trimethylsilyl)amide (132.8 mg, 0.66 mmol) was added and the reaction mixture was stirred with heating at 800C for 15 min, then 3-amino-6- chloropyridazine (47.4 mg, 0.366 mmol) was added, followed by anhydrous K2CO3 (46.0 mg, 0.33 mmol). Then the vial was capped and microwaved at 250 0C for 15 min. Then the reaction mixture was diluted with ca. ImL of MeOH, filtered through 0.22 um syringe filter and purified by reverse-phase preparative HPLC in acetonitrile/water system with 0.01% TFA. The product was isolated as a TFA salt (52.1 mg of brown crystalline solid).
[0219] ESI-MS: [M+H]+,431. 1HNMR (DMSO-d6): δ 7.25-7.26 (d, J= 8.7 Hz, 2H), 7.39-7.41 (d, J= 8.7 Hz, 2H), 7.52-7.54 (d, J= 9.7 Hz, IH), 7.67-7.68 (d, J= 8.7 Hz, 2H), 7.76-7.78 (d, J= 9.7 Hz, IH), 8.03-8.06 (d, J= 8.7 Hz, 2H), 9.58 (s, IH). Example 46. Synthesis of 6-{4-r5-("4-trifluoromethoxy-ρhenylaminoV4H-ri.2.41triazol-3- yl1-phenoxyl-pyrimidine-2,4-diamine trifluoroacetic acid salt
Figure imgf000093_0001
CF3COOH
[0220] 4-[5-(4-tTrifluoromethoxy-phenylamino)-4H[l ,2,4]triazol-3-yl]-phenol (112.0 mg, 0.33 mmol) was dissolved in 2 mL of anhydrous DMF in a 5 mL microwave vial (Personal Chemistry). Solid potassium bis(trimethylsilyl)amide (132.8 mg, 0.66 mmol) was added and the reaction mixture was stirred with heating at 800C for 15 min, then 4-chloro-2,6-diamino- pyrimidine (53.0 mg, 0.366 mmol) was added, followed by anhydrous K2CO3 (46.0 mg, 0.33 mmol). Then the vial was capped and microwaved at 250 0C for 15 min. Then the reaction mixture was diluted with ca. ImL of MeOH, filtered through 0.22 um syringe filter and purified by reverse-phase preparative HPLC in acetonitrile/water system with 0.01% TFA. The product was isolated as a TFA salt (51.1 mg of beige solid).
[0221] ESI-MS: [M+H]+, 445, 446. 1H NMR (DMSO-d6): δ 5.36 (s, IH), 7.25-7.27 (br. d, J= 8.0 Hz, 2H), 7.35-7.36 (br. d, J= 8.0 Hz, 2H), 7.67-7.68 (d, J= 8.7 Hz, 2H), 7.67 (br.s., 4H), 8.02-8.04 (d, J= 8.7 Hz, 2H), 9.57 (br. s, IH).
Example 47. Synthesis of 4-(5-jr3-(trifluromethvDphenyllammo>-4H-L2,4-triazol-3-vD- phenol
Figure imgf000093_0002
[0222] S-methyl N-[3-(trifluoromethyl)phenyl]isothiourea hydroiodide (9.09 g, 25.11 mmol) and 4-hydroxybenzoic acid hydrazide (3.82 g, 25.11 mmol) were suspended in ca. 50 mL of anhydrous pyridine under Ar. The mixture was brought to reflux and refluxed under Ar for 12 hrs. Then the dark-yellow solution was cooled down to ambient temperature and pyridine was removed in vacuo. The resulting reddish-yellow solid was re-dissolved in ca. 50 mL of 4:1 mixture of EtOAc/MeOΗ, ca. 20 g of silica gel was added and solvent was removed in vacuo. The impregnated silica gel was loaded into 25 g ISCO sample cartridge and the product was purified using ISCO system (solid method, 80 g column, 45 min, 0% to 50% EtOAc gradient in hexane). Solvent was removed in vacuo to give the title product as a white solid (3.83 g). Yield 47.6%.
[0223] ESI-MS: [M+H]+, 321.09. 1H NMR (DMSOd6): δ 6.89-6.90 (d, J= 8.6 Hz, 2H), 7.09-7.11 (d, J= 7.3 Hz, IH), 7.42-7.46 (t, J= 7.9 Hz, IH), 7.73-7.74 (d, J= 7.0 Hz, IH), 7.77-7.79 (d, J= 8.6 Hz, 2H), 8.09 ( s, IH), 9.65 (s, IH), 9.97 (br. s., IH).
Example 48. Synthesis of 6-[4-("5-(r3-(trifluoromethyl)phenyl1ammo|-4H- ri,2,4]triazol-3-viyphenoxy1-pyrimidine-2,4-diamine
Figure imgf000094_0001
[0224] 4-(5-{[3-(trifluromethyl)phenyl]amino}-4H-l,2,4-triazol-3-yl)phenol (160 mg, 0.5 mmol) was dissolved in 3 mL of anhydrous dioxane in 2-5 niL microwave vial (Personal Chemistry). Solid Cs2CO3 (163.0 mg, 0.5 mmol) was added, followed by 4-chloro-2,6,- diaminopyrimidine (79.5 mg, 0.55 mmol). The vial was capped and micro waved at 200 0C for 20 min. Then ca. 3 mL of MeOH was added to dissolve the formed suspension, the solution was transferred into a round-bottom flask and solvent was removed in vacuo. The residue was re-dissolved in 3 mL of DMF, filtered through 0.22 u syringe filter and purified by reverse phase preparative ΗPLC using acetontrile/water system with 0.01% of TFA.
[0225] The fractions containing the product were collected and partitioned between EtOAc and saturated aqueous NaHCO3. Ethyl acetate layer was washed with brine, dried over anhydrous sodium sulfate and filtered. Solvent was removed in vacuo to give the title compound as an off-white solid (92.2 mg).
[0226] ESI-MS: [M+H]+, 429.08. 1H NMR (DMSO-d6): δ 5.15 (s, IH), 6.02 (s, 2H), 6.30 (s, 2H), 7.12 (m, IH), 7.25-7.27 (d, J= 7.4 Hz, 2H), 7.45-7.47 (m, IH), 7.75-7.76 (m, IH), 7.95-7.97 (U1 J= 8.6 Hz, 2H), 8.10 (s, IH), 9.73 (s, IH), 13.84 (s, IH).
Example 49. Synthesis of 5-r4-(pyrimidm-5-yloxy)phenyl1-N-r3- (trifluoromethyl)phenyll-4H-l,2,4-triazol-3-amine
Figure imgf000094_0002
[0227] 4-(5-{[3-(trifluromethyl)phenyl]ammo}-4H-l,2,4-triazol-3-yl)plienol (100 mg, 0.31 mmol) was dissolved in 3 mL of anhydrous dioxane in 2-5 niL microwave vial (Personal Chemistry). Solid Cs2CO3 (203.4 mg, 0.62 mmol) was added, followed by 5-bromo- pyrimidine (100 mg, 0.62 mmol). Then 1 mL of anhydrous DMF was added, the vial was capped and microwaved at 250 0C for 30 min. Then ca. 3 mL of MeOH was added to dissolve the formed suspension, the solution was transferred into a round-bottom flask and solvent was removed in vacuo. The residue was re-dissolved in 3 mL of DMF, filtered through 0.22 u syringe filter and purified by reverse phase preparative ΗPLC using acetontrile/water system with 0.01% of TFA. The fractions containing the product were collected and partitioned between EtOAc and saturated aqueous NaHCO3. Ethyl acetate layer was washed with brine, dried over anhydrous sodium sulfate and filtered. Solvent was removed in vacuo to give the title compound as a light-brown foamy solid (34.7 mg).
[0228] ESI-MS: [M+H]+, 399.06. 1H NMR (DMSO-d6): δ 7.12 (m, IH), 7.30-7.32 (d, J= 7.4 Hz, 2H), 7.46 (m, IH), 7.76 (m, IH), 8.00-8.02 (d, J= 8.6 Hz, 2H), 8.09 (s, IH), 8.73 (s, 2H), 9.06 (s, IH), 9.73 (s, IH), 13.95 (s, IH).
Example 50. Synthesis of 5-r4-(pyridin-3-yloxy)phenyll-N-[3- (trifluoromethyl)phenyll-4H-r 1 ,2,4"|triazol-3-amine
Figure imgf000095_0001
[0229] 4-(5-{[3-(trifluromethyl)phenyl]amino}-4H-l,2,4-triazol-3-yl)phenol (100 mg, 0.31 mmol) was dissolved in 2 mL of anhydrous DMF in 2-5 mL microwave vial (Personal Chemistry). Solid Cs2CO3 (203.4 mg, 0.62 mmol) was added, followed by 3-bromopyridme (74.0 mg, 0.468 mmol). The vial was capped and microwaved at 250 0C for 30 min. Then ca. 1 mL of MeOH was added to dissolve the formed suspension. The resulting reddish-brown solution was filtered through 0.22 u syringe filter and purified by reverse phase preparative ΗPLC using acetontrile/water system with 0.01% of TFA. The fractions, containing the product, were collected and partitioned between EtOAc and saturated aqueous NaHCO3. Ethyl acetate layer was washed with brine, dried over anhydrous sodium sulfate and filtered. Solvent was removed in vacuo to give the title compound as a yellow solid (22.4 mg).
[0230] ESI-MS: [M+H]+, 398.11. 1HNMR (DMSO-d6): δ 7.09 (d, J= 7.6 Hz, IH), 7.20- 7.22 (d, J= 8.6 Hz, 2H), 7.46-7.48 (m, 2H), 7.52-7.54 (m, IH), 7.75-7.76 (d, J= 8.2 Hz, IH), 7.99-8.02 (d, J= 8.6 Hz, 2H), 8.09 (s, IH), 8.42-8.43 (d, J= 4.5 Hz, IH), 8.45-8.46 (d, J= 2.6 Hz, IH), 9.73 (s, IH), 13.85 (s, IH).
Example 51. Synthesis of 4-Methoxy-6-r4-(5-ir3-("trifluoromethyl)phenyllamino>- 4H- 1 ,2,4-triazol-3 -vr)phenoxvlpvrimidin-2-amine
Figure imgf000096_0001
[0231] 4-(5-{[3-(trifluromethyl)phenyl]amino}-4H-l,2,4-triazol-3-yl)plienol (100 mg, 0.31 mmol) was dissolved in 3 mL of anhydrous dioxane in 2-5 niL microwave vial (Personal Chemistry). Solid Cs2CO3 (101.7 mg, 0.31 mmol) was added, followed by 2-amino-4- chloro-6-methoxypyrimidine (55.0 mg, 0.34 mmol). The vial was capped and microwaved at 200 0C for 15 min. Then ca. 3 mL of MeOH was added to dissolve the formed suspension. The resulting reddish-brown solution was transferred into a round-bottom flask and solvent was removed in vacuo. The residue was re-dissolved in 3 mL of DMF, filtered through 0.22 u syringe filter and purified by reverse phase preparative ΗPLC using acetontrile/water system with 0.01% of TFA. The fractions, containing the product, were collected and partitioned between EtOAc and saturated aqueous NaHCO3. Ethyl acetate layer was washed with brine, dried over anhydrous sodium sulfate and filtered. Solvent was removed in vacuo to give the title compound as an off-white solid (55.6 mg).
[0232] ESI-MS: [M+H]+, 443.87. 1H NMR (DMSO-d6): δ 3.80 (s, 3H), 5.53 (s, IH), 6.70 (s, 2H), 7.11-7.12 (d, J= 7.4 Hz, IH), 7.30-7.32 (d, J= 8.4 Hz, 2H), 7.44-7.47 (t, J= 7.6 Hz, IH), 7.74-7.76 (d, J= 8.4 Hz, IH), 7.97-7.98 (d, J= 8.4 Hz, 2H), 8.10 (s, IH), 9.73 (s, IH), 13.88 (s, IH).
Example 52. Synthesis of 6-r4-(5-{r3-(trifluoromethyl)phenyllamino}-4H- ri.2,41triazol-3-yl)-phenoxvl-pyrimidin-4-amine
Figure imgf000096_0002
[0233] 4-(5-{[3-(trifluromethyl)phenyl]amino}-4H-l,2,4-triazol-3-yl)phenol (100 mg, 0.31 mmol) was dissolved in 3 mL of anhydrous dioxane in 2-5 mL microwave vial (Personal Chemistry). Solid Cs2CO3 (101.7 mg, 0.31 tnmol) was added, followed by 4-amino-6- chloro-pyrimidine (48.5 mg, 0.37 mmol). The vial was capped and microwaved at 200 0C for 5 min. Then ca. 3 niL of MeOH was added to dissolve the formed suspension. The resulting reddish-brown solution was transferred into a round-bottom flask and solvent was removed in vacuo. The residue was re-dissolved in 3 mL of DMF, filtered through 0.22 u syringe filter and purified by reverse phase preparative HPLC using acetontrile/water system with 0.01% of TFA. The fractions, containing the product, were collected and partitioned between EtOAc and saturated aqueous NaHCO3. Ethyl acetate layer was washed with brine, dried over anhydrous sodium sulfate and filtered. Solvent was removed in vacuo to give the title compound as a white solid (76.6 mg).
[0234] ESI-MS: [M+H]+, 461.0. 1H NMR (DMSO-d6): δ 2.31 (s, 3H), 5.49 (s, IH), 6.93 (s, 2H), 7.14 (m, IH), 7.33-7.35 (d, J= 7.5 Hz, 2H), 7.45-7.48 (t, J= 7.6 Hz, IH), 7.75 (m, IH), 8.00-8.02 (d, J= 8.6 Hz, 2H), 8.10 (s, IH), 9.75 (s, IH), 13.88 (s, IH).
Example 53. Synthesis of 2-fMethylthioV6-r4-f5-(r3- (trifluoromethyl)phenyllamino|-4H-l,2,4-triazol-3-yl)-phenoxyl-pyrimidin-4-amine
Figure imgf000097_0001
[0235] 4-(5-{[3-(trifluromethyl)phenyl]amino}-4H-l,2,4-triazol-3-yl)phenol (100 mg, 0.31 mmol) was dissolved in 3 mL of anhydrous dioxane in 2-5 mL microwave vial (Personal Chemistry). Solid Cs2CO3 (101.7 mg, 0.31 mmol) was added, followed by 4-amino-6- chloro-2-(methylthio)-pyrimidine (60.3 mg, 0.34 mmol). The vial was capped and microwaved at 200 0C for 10 min. Then ca. 3 mL of MeOH was added to dissolve the formed suspension. The resulting reddish-brown solution was transferred into a round-bottom flask and solvent was removed in vacuo. The residue was re-dissolved in 3 mL of DMF, filtered through 0.22 u syringe filter and purified by reverse phase preparative ΗPLC using acetontrile/water system with 0.01% of TFA. The fractions, containing the product, were collected and partitioned between EtOAc and saturated aqueous NaHCO3. Ethyl acetate layer was washed with brine, dried over anhydrous sodium sulfate and filtered. Solvent was removed in vacuo to give the title compound as a white solid (39.5 mg). [0236] ESI-MS: [M+H]+, 461.0. 1H NMR (DMSO-d6): δ 3.80 (s, 3H), 5.53 (s, IH), 6.70 (s, 2H), 6.30 (s, 2H), (d, J= 8.6 Hz, 2H),
Example 54. Synthesis of 4-[5-(4-trifluoromethoxy-phenylV4H-[L2,41triazol-3-ylaminol- phenol
Figure imgf000098_0001
[0237] 4-trifluoromethoxybenzoic acid hydrazide (1.1 g, 5.0 mmol) and S-methyl N-[4- hydroxy-phenyl]isothiourea hydroiodide (1.55 g, 5.0 mmol) were suspended in 10 niL of anhydrous pyridine. The reaction mixture was refluxed for 24 hours, during which time it changed color from yellow into orange-red. Then it was cooled down to ambient temperature and poured with stirring into 150 mL of ice-water. The aqueous layer was decanted and the resulting residue was purified by silica gel chromatography using 1 : 1 mixture of ethyl acetate/hexane. Solvent was removed in vacuo to give the title product as a pinkish-grey solid (684.0 mg). 40.6 % yield.
[0238] ESI-MS: [M+Η]+, 337, 338. 1H NMR (DMSOd6): δ 7.68-6.71 (d, J= 8.8 Hz, 2H), 7.32-7.35 (d, J= 8.8 Hz, 2H), 7.47-7.49 (d, J= 8.8 Hz, 2H), 8.04-8.07 (d, J= 8.8 Hz, 2H), 9.05 (br. s, IH).
Example 55. Synthesis of 4-{4-r5-(4-trifluoromethoxy-ρhenyl)-4H-π,2,41triazol-3- ylamino]-phenoxyl-pyridine-2-carboxyh'c acid methylamide trifluoroacetic acid salt
Figure imgf000098_0002
CF3COOH
[0239] 4-[5-(4-trifluoromethoxy-phenyl)-4H[l,2,4]triazol-3-ylammo]-phenol (134.5 mg, 0.4 mmol) was dissolved in 2 mL of anhydrous DMF in a 5 mL microwave vial (Personal Chemistry). Solid potassium bis(trimethylsilyl)amide (120.0 mg, 0.6 mmol) was added and the reaction mixture was stirred with heating at 800C for 15 min, then 4-chloro-2-pyridine- carboxamide (68.2 mg, 0.4 mmol) was added, followed by anhydrous K2CO3 (62.0 mg, 0.44 mmol). Then the vial was capped and microwaved at 150 0C for 30 min. Then the reaction mixture was diluted with ca. ImL of MeOH, filtered through 0.22 um syringe filter and purified by reverse-phase preparative HPLC in acetonitrile/water system with 0.01% TFA. The product was isolated as a TFA salt (31.7 mg of white solid).
[0240] ESI-MS: [MH-H]+ 471, 472. 1H NMR (DMSOd6): δ 2.77-2.78 (d, J= 4.8 Hz, 3H), 7.13-7.15 (d, J= 8.3 Hz, 2H), 7.13 (m, IH), 7.40 (br. s, IH), 7.51-7.53 (d, J= 8.3 Hz, 2H), 7.70-7.73 (d, J= 8.8 Hz5 2H), 8.09-8.11 (d, J= 8.8 Hz, 2H), 8.48-8.49 (d, J= 5.3 Hz, IH), 8.77-8.78 (q, J= 4.8 Hz, IH), 9.56 (s, IH).
Example 56. Synthesis of 3-^5-(4-chloro-3-trifluoromethyl-phenylaminoV 4H^l .2,41triazol-3-vll-ρhenol
Figure imgf000099_0001
[0241] 3-hydroxybenzoic acid hydrazide (2.98 g, 19.58 mmol) and S-methyl N-[4-chloro- 3-(trifluoromethyl)phenyl]isothiourea hydroiodide (7.78 g, 19.63mmol) were suspended in 40 mL of anhydrous pyridine. The reaction mixture was refluxed for 18 hours, during which time it changed color from yellow into dark-red. Then it was cooled down to ambient temperature and poured with stirring into 250 mL of ice-water. The aqueous solution was decanted and the oily residue was purified by silica gel chromatography on Isco column using 0 => 50 % gradient of ethyl acetate in hexane. Solvent was removed in vacuo to give the title product as a white solid (2.176 g). 31.3 % yield.
[0242] ESI-MS: [M+Η]+ 355.1, 356.8,. 1H NMR (DMSOd6): δ 6.88-6.90 (dq, J1 = 7.9 Hz, J2= 0.9 Hz, IH), 7.31-7.34 (t, J= 7.9 Hz, IH), 7.35-7.39 (m, 2H), 7.54-7.56 (d, J= 8.8 Hz, IH), 7.74-7.76 (dd, J1 = 8.8 Hz, J2= 2.7 Hz, IH), 8.23-8.24 (d, J= 2.7 Hz, IH), 9.79 (s, IH), 9.87 (s, IH), 13.86 (s, IH). Example 57. Synthesis of 4-{3-r5-(4-chloro-3-trifluoromethyl-t)henylaminoV4H- ri,2.41triazol-3-yll-phenoxy|-pyridine-2-carboxylic acid methylamide trifluoroacetic acid salt
Figure imgf000100_0001
CF3COOH
[0243] 3-[5-(4-chloro-3-trifluorometliyl-phenylaniino)-4H[l,2,4]triazol-3-yl]-plienol (100 mg, 0.282 mniol) was dissolved in 2 mL of anhydrous DMF in a 5 mL microwave vial (Personal Chemistry). Solid potassium bis(trimethylsilyl)amide (140.6 mg, 0.705 mmol) was added and the reaction mixture was stirred with heating at 80 0C for 15 min, then 4-chloro-2- pyridine-carboxamide (52.9 mg, 0.31 mmol) was added, followed by anhydrous K2CO3 (19.5 mg, 0.141 mmol). Then the vial was capped and microwaved at 250 0C for 20 min. Then the reaction mixture was diluted with ca. ImL of MeOH, filtered through 0.22 um syringe filter and purified by reverse-phase preparative ΗPLC in acetonitrile/water system with 0.01% TFA. The product was isolated as a TFA salt (25.0 mg of white solid).
[0244] ESI-MS: [M+Η]+489, 490, 491. 1H NMR (DMSOd6): δ 2.78-2.79 (d, J= 4.8 Hz, 3H), 7.25-7.26 (dd, J1 = 2.5 Hz, J2= 5.5 Hz, IH), 7.38 (m, IH), 7.48-7.49 (d, J= 2.5 Hz, IH), 7.55-7.57 (d, J= 8.8 Hz, IH), 7.67-7.70 (t, J= 7.8 Hz, IH), 7.73 (br. s., IH), 7.78 (br. s., IH), 7.91-7.93 (d, J= 7.8 Hz, IH), 8.20-8.21 (d, J= 2.6 Hz, IH), 8.56-8.57 (d, J= 5.5 Hz, IH), 8.80-8.81 (q, J= 4.8 Hz, IH), 9.93 (s, IH).
Example 58. Synthesis of 6-{3-r5-(4-chloro-3-trifluoromethyl-ρhenylamino)-4H- ri,2,41triazol-3-yll-phenoxy}-pyrimidine-2,4-diamme trifluoroacetic acid salt
Figure imgf000100_0002
CF3COOH
[0245] 3-[5-(4-chloro-3-trifluoromethyl-phenylamino)-4H[l,2,4]triazol-3-yl]-phenol (100 mg, 0.282 mmol) was dissolved in 2 mL of anhydrous DMF in a 5 mL microwave vial. Solid US2005/030614
100
potassium bis(trimethylsilyl)amide (140.6 mg, 0.705 mmol) was added and the reaction mixture was stirred with heating at 80 0C for 15 min, then 4-chloro-2,6-diamino-pyrimidine (44.8 mg, 0.31 mmol) was added, followed by anhydrous K2CO3 (19.5 mg, 0.141 mmol). Then the vial was capped and microwaved at 2500C for 20 min. Then the reaction mixture was diluted with ca. ImL of MeOH, filtered through 0.22 um syringe filter and purified by reverse-phase preparative HPLC in acetonitrile/water system with 0.01% TFA. The product was isolated as a TFA salt (37.8 mg of beige solid).
[0246] ESI-MS: [M+H]+ 464, 465. 1H NMR (DMSO-d6): δ 5.40 (s, IH), 7.33-7.35 (d, J= 7.6 Hz, IH), 7.57-7.59 (d, J= 8.8 Hz, IH), 7.62-7.65 (t, J= 7.8 Hz, IH), 7.73 (br. s., IH), 7.80 (br. m., IH), 7.88-7.89 (d, J= 7.8 Hz, IH), 8.21-8.22 (d, J= 2.6 Hz, IH), 9.96 (s, IH).
Example 59. Synthesis of (4-chloro-3-trifluoromethyl-phenyl)-{5-F3-(pyridin-4- yloxy)-phenyll-4H-ri,2,41triazol-3-yl| -amine trifluoroacetic acid salt
Figure imgf000101_0001
CF3COOH
[0247] 3-[5-(4-chloro-3-trifluoromethyl-phenylamino)-4Hi;i,2,4]triazol-3-yl]-phenol (100 mg, 0.282 mmol) was dissolved in 2 mL of anhydrous DMF in a 5 mL microwave vial. Solid potassium bis(trimethylsilyl)amide (140.6 mg, 0.705 mmol) was added and the reaction mixture was stirred with heating at 80 0C for 15 min, then 4-chloro-pyridine hydrochloride (46.5 mg, 0.31 mmol) was added, followed by anhydrous K2CO3 (19.5 mg, 0.141 mmol). Then the vial was capped and microwaved at 2500C for 20 min. Then the reaction mixture was diluted with ca. ImL of MeOH, filtered through 0.22 um syringe filter and purified by reverse-phase preparative ΗPLC in acetonitrile/water system with 0.01% TFA. The product was isolated as a TFA salt (34.6 mg of beige solid).
[0248] ESI-MS: [M+Η]+ 432, 433. 1H NMR (DMSO-d6): δ 7.43-7.45 (br. s. IH), 7.46- 7.47 (d, J= 7.0 Hz, 2H), 7.56-7.58 (d, J= 8.8 Hz, IH), 7.72-7.75 (t, J= 7.8 Hz, IH), 7.80 (br. s., IH), 7.81 (br. s, IH), 7.98-8.00 (d,J= 7.8 Hz, IH), 8.21-8.22 (d, J= 2.6 Hz, 1H8.76- 8.78 (d, J= 7.0 Hz, IH), 9.98 (s, IH). Example 60. Synthesis of 6-{3-r5-("4-chloro-3-trifluoromethyl-phenylaminoV4H- ri,2,41triazol-3-vll-phenoxy>-pvridazin-3-vlamine trifluoroacetic acid salt
Figure imgf000102_0001
CF3COOH
[0249] 3-[5-(4-chloro-3-trifluoromethyl-phenylamino)-4H[l,2,4]triazol-3-yl]-plienol (100 mg, 0.282 mmol) was dissolved in 2 niL of anhydrous DMF in a 5 mL microwave vial. Solid potassium bis(trimethylsilyl)amide (140.6 mg, 0.705 mmol) was added and the reaction mixture was stirred with heating at 800C for 15 min, then 3-amino-6-chloro-pyridazine (40.2 mg, 0.31 mmol) was added, followed by anhydrous K2CO3 (19.5 mg, 0.141 mmol). Then the vial was capped and microwaved at 2500C for 20 min. Then the reaction mixture was diluted with ca. ImL of MeOH, filtered through 0.22 um syringe filter and purified by reverse-phase preparative ΗPLC in acetonitrile/water system with 0.01% TFA. The product was isolated as a TFA salt (35.5 mg of light-brown solid).
[0250] ESI-MS: [M+Η]+ 448, 449. 1H NMR (DMSOd6): δ 7.38-7.39 (br. s. IH), 7.55- 7.57 (d, J= 9.6 Hz, IH), 7.57-7.58 (d, J= 8.6 Hz, IH), 7.62-7.65 (t, J= 7.8 Hz, IH), 7.78 (br. s., IH), 7.80 (br. s, IH), 7.82-7.84 (d, J= 9.6 Hz, IH), 7.86-7.88 (d, J= 7.8 Hz, IH), 8.22-8.23 (d, J= 2.6 Hz, IH), 8.52 (br.s, 2H), 9.94 (s, IH).
Example 61. Synthesis of 4-(5-{[4-chloro-3-(trifluoromethyl)-phenyllamino>-l,3,4- oxadiazol-2-yl)phenol
Figure imgf000102_0002
[0251] Mercury (II) oxide (yellow) (4.55 g, 21.0 mmol) was suspended in 60 mL of anhydrous MeOH under Ar. A bright-orange suspension was formed. To this suspension was added 4-hydroxybenzoic acid hydrazide (3.20 g, 21.0 mmol) and 4-chloro-3- trifluoromethyl-phenylisothiocyanate (5.0 g, 21.0 mmol). The reaction mixture was refluxed for 2 hours. The solvent was removed in vacuo. The black residue was redissolved in 100 mL of EtOAc and the resulting black suspension was filtered through a short pad of silica gel. The filtrate was mixed with 10 g of dry silica gel and solvent was removed in vacuo. The impregnated silica gel was loaded on silica gel column and the product was separated using a gradient of hexane: ethyl acetate mixture starting from 50:50 ratio and finishing at 0: 100. All fractions containing the product were combined; solvent was removed in vacuo to give a grey solid. The solid was heated in 50 mL of 4:1 mixture of EtOAc/MeOH. The formed suspension was cooled down to ambient temperature and filtered to give the title product as a white crystalline solid (4.54 g, 60.7 % yield).
[0252] ESI-MS: [M+H]+ 356.0. 1H NMR (DMSOd6): δ 6.92-6.95 (d, J= 8.8 Hz, 2H), 7.69-7.70 (d, J= 8.8 Hz, IH), 7.71-7.73 (d, J= 8.8 Hz, 2H), 7.82-7.84 (dd, J/ = 8.8 Hz, J2 = 2.6 Hz, IH), 8.16-8.17 (d, J= 2.6 Hz, IH), 10.21 (br s., IH), 11.11 (br s., IH).
Example 62. Synthesis of 6-r4-(5-{r4-Chloro-3-trifluoromethyl-phenyl1aminol-l,3,4- oxadiazol-2-yl)-phenoxy1-pvrimidine-2,4-diamine
Figure imgf000103_0001
[0253] 4-(5- {[4-chloro-3-(trifluoromethyl)-phenyl]amino} -1 ,3 ,4-oxadiazol-2-yl)phenol (1.067 g, 3.0 mmol) was dissolved in ca. 70 mL of anhydrous DMF under argon. Solid potassium bis(trimethylsilyl)amide (0.718 g, 3.6 mmol) was added and the resulting yellow solution was heated at 700C for 1.5 hours. Then solid K2CO3 (0.414 g, 3.0 mmol) was added, followed by 2,6-diamino-4-chloropyrimidine (0.520 g, 3.6 mmol). The reaction mixture was left to reflux under argon for 30 hours. Then it was allowed to cool down to ambient temperature and poured into ca. 500 mL of water. The aqueous mixture was extracted 5 times with 100 mL of EtOAc. Combined EtOAc extracts were washed three times with 100 mL of brine, and dried over anhydrous Na2SO4. Solvent was removed in vacuo to a give a reddish-yellow residue, which was purified by silica gel chromatography using EtOAc as an eluent. Fractions, containing the product, were collected; solvent was removed in vacuo to give the product as a reddish-yellow solid. The solid was re-crystallized from 10 mL of EtOAc, collected, washed thoroughly with diethyl ether and dried in vacuo to give the title compound (0.527 g, 38 % yield) as beige solid.
[0254] ESI-MS: [M+H]+464, 465. 1H NMR (DMSO-d6): δ 5.20 (s, IH), 6.05 (br s., 2H), 6.34 (br.s., 2H), 7.27-7.30 (d, J= 8.7 Hz, 2H), 7.71-7.72 (d, J= 8.8 Hz, IH), 7.84-7.88 (dd, J1 = 8.8 Hz, J2 = 2.6 Hz, IH), 7.89-7.91 (d, J= 8.7 Hz, 2H), 8.18-8.19 (d, J= 2.6 Hz, IH)5 11.26 (s, IH). Anal. Calcd for C19H13ClF3N7O2: C, 49.20; H, 2.83; N, 21.14, Found: C, 49.08; H, 3.21; N, 20.95.
Example 63. Synthesis of 6-[4-(5-{[4-chloro-3-trifluoromgthyl-phenvnamino>-1.3.4- oxadiazol-2-yl)-phenoxy]-pyrimidine-2,4-diamine trifluoroacetic acid salt
Figure imgf000104_0001
CF3COOH
[0255] 4-(5-{[4-chloro-3-(trifluoromethyl)-phenyl]amino}-l,3,4-oxadiazol-2-yl)phenol (100 mg, 0.281 mmoD was dissolved in 2.5 mL of anhydrous DMF in a 5 mL microwave vial. Solid potassium bis(trimethylsilyl)amide (140.6 mg, 0.7O3 mmol) was added and the reaction mixture was stirred with heating at 80 0C for 15 min, then 6-chloro-2,4-diamino- pyrimidine (81.3 mg, 0.562 mmol) was added, followed by anhydrous K2CO3 (19.5 mg, 0.141 mmol). Then the vial was capped and microwaved at 200 0C for 15 min. Then the reaction mixture was diluted with ca. 1 mL of MeOH, filtered through 0.22 um syringe filter and purified by reverse-phase preparative HPLC in acetonitrile/water system with 0.01% TFA. The product was isolated as a TFA salt (75.8 mg of beige solid).
[0256] ESI-MS: [M+H]+464, 465. 1HNMR (DMSO-d6): δ 5.41 (s, IH), 7.39-7.42 (d, J= 8.7 Hz, 2H), 7.63 (br s., 4H), 7.72-7.74 (d, J= 8.8 Hz, IH), 7.85-7.87 (dd, J1 = 8.8 Hz, J2 = 2.7 Hz, IH), 7.95-7.97 (d, J= 8.7 Hz, 2H), 8.20 (d, J= 2.7 Hz, IH), 11.29 (s, IH).
Example 64. Synthesis of N-[4-chloro-3-ftrifluoromethyl')phenyl]-5-r4-('pyridin-3- yloxy)phenyl]-l,3.4-oxadiazol-2-amine trifluoroacetate salt
Figure imgf000104_0002
CF3COOH
[0257] 4-(5-{[4-chloro-3-(trifluoromethyl)-phenyl]amino}-l,3,4-oxadiazol-2-yl)phenol (100 mg, 0.281 mmol) was dissolved in ca. 2 mL of anhydrous DMF under argon. Solid potassium bis(trimethylsilyl)amide (140.2 mg, 0.702 mmol) was added and the resulting yellow solution was heated at 8O0C for 15 min. Then solid K2CO3 (19.4 mg, 0.140 mmol) was added, followed by 3-bromopyridine (89.0 mg, 0.562 mmol). The reaction mixture was microwaved at 250 0C for 10 min. Then it was diluted with 1 niL of MeOH, filtered and purified by preparative reverse-phase chromatography using acetonitrile/water with 0.1 % TFA gradient. The major peak having the mass of the product was collected; solvent was removed in vacuo to give the title product as a brown oil (20.6 mg).
[0258] ESI-MS: [M+H]+ 433.5, 434.3. 1H NMR (DMSOd6): δ 7.24-7.26 (d, J= 8.8 Hz, 2H), 7.57-7.59 (dd, J1 = 8.4 Hz, J2 = 4.7 Hz, IH), 7.68-7.71 (dq, J1 = 8.4 Hz, J2 = 1.4 Hz, IH), 7.71-7.73 (d, J= 8.8 Hz, 1H), 7.84-7.86 (dd, J1 = 8.8 Hz, J2 = 2.7 Hz, IH), 7.92-7.94 (d, J= 8.8 Hz, 2H), 8.17-8.18 (d, J= 2.7 Hz, IH), 8.50 (br d, J= 4.0 Hz, IH), 8.54 (br s, IH), 11.24 (s, IH).
Example 65. Synthesis of N-r4-chloro-3-(trifluoromethyl')phenyll-5-[4-(pyridin-4- yloxy)phenyll-l,3,4-oxadiazol-2-amine trifluoroacetate salt
Figure imgf000105_0001
[0259] 4-(5-{ [4-chloro-3 -(trifluoromethyl)-phenyl] amino} - 1 ,3 ,4-oxadiazol-2-yl)phenol (100 mg, 0.281 mmol) was dissolved in ca. 2 mL of anhydrous DMF under argon. Solid potassium bis(trimethylsilyl)amide (225 mg, 1.12 mmol) was added and the resulting yellow solution was heated at 800C for 15 min. Then solid K2CO3 (38.8 mg, 0.281 mmol) was added, followed by 4-chloropyridine hydrochloride (84.3 mg, 0.562 mmol). The reaction mixture was microwaved at 200 0C for 25 min (Initiator, Biotage). Then it was diluted with 1 mL of MeOH, filtered through 0.22 urn syringe filter and purified by preparative reverse- phase chromatography using acetonitrile/water gradient with 0.1 % TFA. The major peak having the mass of the product was collected; solvent was removed in vacuo to give the title product as a white fluffy solid (85.6 mg).
[0260] ESI-MS: [M+H]+ 435.3. 1H NMR (DMSO-d6): δ 7.37-7.38 (d, J= 4.8 Hz, 2H), 7.48-7.50 (d, J= 8.8 Hz, 2H), 7.72-7.74 (d, J= 8.8 Hz, IH), 7.85-7.88 (dd, J1 = 8.8 Hz, J2 = 2.7 Hz, IH), 8.03-8.06 (d, J= 8.8 Hz, IH), 8.19-8.20 (d, J= 2.7 Hz, IH), 8.74 (br s., 2H), 11.31 (s, IH). Example 66. Synthesis of N-F4-chloro-3-('trifluorometliyl')ρhenyll-5-r4-(pyrimidin-5- yloxy)phenyll-l,3,4-oxadiazol-2-amine trifluoroacetate salt
Figure imgf000106_0001
CF3COOH
[0261] 4-(5-{[4-chloro-3-(trifluoromethyl)-phenyl]amino}-l,3;,4-oxadiazol-2-yl)phenol (100 mg, 0.281 mmol) was dissolved in ca. 2 mL of anhydrous DMF under argon. Solid potassium bis(trimethylsilyl)amide (140.2 mg, 0.702 mmol) was added and the resulting yellow solution was heated at 8O0C for 15 min. Then solid K2CO3 (19.4 mg, 0.140 mmol) was added, followed by 3-bromopyrimidine (89.4 mg, 0.562 mmol). The reaction mixture was microwaved at 200 0C for 15 min. Then it was diluted with 1 mL of MeOH, filtered through 0.22 um syringe filter and purified by preparative reverse-phase chromatography using acetonitrile/water gradient with 0.1 % TFA. The major peak having the mass of the product was collected; solvent was removed in vacuo to give the title product as a white fluffy solid (73.0 mg of white crystalline solid).
[0262] ESI-MS: [M+H]+ 434.3, 435.3. IH NMR (DMSO-d6): δ 7.31-7.33 (d, J= 8.8 Hz, 2H), 7.71-7.73 (d, J= 8.8 Hz, IH), 7.84-7.86 (dd, J1 = 8.8 Hz, J2 = 2.7 Hz, IH), 7.93-7.95 (d, J= 8.8 Hz, 2H), 8.17-8.18 (d, J= 2.7 Hz, IH), 8.77 (s, 2H), 9.09 (s, IH), 11.25 (s, IH).
Example 67. Synthesis of 4-r4-(5-{r4-chloro-3-(trifluoromethyl)phenyllamino|-l,3,4- oxadiazol-2-yl)ρhenoxvl -N-methvlpyridine-2-carboxamide trifluoroacetate salt
Figure imgf000106_0002
[0263] 4-(5-{[4-chloro-3-(trifluoromethyl)-phenyl]amino}-l,3,4-oxadiazol-2-yl)phenol (100 mg, 0.281 mmol) was dissolved in ca. 2 mL of anhydrous DMF under argon. Solid potassium bis(trimethylsilyl)amide (140.2 mg, 0.702 mmol) was added and the resulting yellow solution was heated at 8O0C for 15 min. Then solid K2CO3 (19.4 mg, 0.140 mmol) was added, followed by 4-chloro-2-pyridine-carboxamide (52.7 mg, 0.309 mmol). The reaction mixture was microwaved at 200 C for 15 min. Then it was diluted with 1 rnL of MeOH, filtered through 0.22 um syringe filter and purified by preparative reverse-phase chromatography using acetonitrile/water gradient with 0.1 % TFA. The major peak having the mass of the product was collected; solvent was removed in vacuo to give the title product as a white solid (67.5 mg).
[0264] ESI-MS: [M+H]+ 490.4, 491.3 . 1H NMR (DMSO-d6): δ 2.79-2.80 (d, J= 4.9 Hz, 3H), 7.26-7.28 (dd, J1 = 5.6 Hz, J2 = 2.6 Hz, IH), 7.43-7.44 (d, J= 6.8 Hz, 2H), 7.49 (d, J= 2.6 Hz, IH), 7.72-7.74 (d, J= 8.8 Hz, IH), 7.85-7.86 (dd, J1 = 8.8 Hz, J2 = 2.6 Hz, IH), 8.01- 8.03 (d, J= 6.8 Hz, 2H), 8.19 (d, J= 2.6 Hz, IH), 8.57-8.58 (d, J= 5.7 Hz, IH), 8.79-8.81 (q, J= 4.9 Hz, IH), 11.28 (s, IH).
Example 68. Synthesis of 4-[5-(4-chloro-3-(trifluoromethyl)-phenylV4H-l,2,4-triazol-3- ylaminolphenol
Figure imgf000107_0001
[0265] 4-chloro-3-trifluoromethylbenzoic acid hydrazide (2.89 g, 12.1 mmol) and S- methyl N-(4-hydroxyphenyl)isothiourea hydroiodide (3.75 g, 12.1 mmol) were suspended in 40 mL of anhydrous pyridine. The reaction mixture was refluxed for 18 hours, during which time it changed color from yellow into dark-red. Then it was cooled down to ambient temperature and poured with stirring into 250 mL of ice-water. The aqueous solution was decanted and the oily residue was purified by silica gel chromatography using 1:1 mixture of ethyl acetate/hexane. Solvent was removed in vacuo to give the title product as a white solid (1.95 g). Yield 45.5 %.
Example 69. Synthesis of 6-r4-({5-r4-chloro-3-(trifluoromethyl)phenyl-4H-l,2,4-triazol-3- yllamino)phenoxylpyrimidine-2,4-diamine trifluoroacetate salt
Figure imgf000107_0002
CF3COOH [0266] 4-[5-(4-chloro-3-(trifluoromethyl)-phenyl)-4H-l ,2,4-triazol-3-ylamino]phenol (100 mg, 0.282 mmol) was dissolved in 2 niL of anhydrous DMF. Solid potassium bis(trimethylsilyl)amide (140.6 mg, 0.705 mmol) was added and the resulting solution was heated at 800C for 15 min. Then solid K2CO3 (20 mg, 0.141 mmol) was added, followed by 4-chloro-2,6-diamino-pyrimidine (61.1 mg, 0.422 mmol). The reaction mixture was microwaved at 200 0C for 20 min. Then it was diluted with 1 mL of MeOH, filtered through 0.22 um syringe filter and purified by preparative reverse-phase chromatography using acetonitrile/water gradient with 0.1 % TFA. The major peak having the mass of the product was collected, solvent was removed in vacuo to give trifluoroacetate salt of the product as a white solid (28.6 mg).
[0267] ESI-MS: [M+H]+ 463.4, 464.4. IH NMR (DMSO-d6): δ 5.24 (s, IH), 7.14-7.16 (d, J= 8.8 Hz, 2H), 7.65-7.66 (d, J= 8.8 Hz, 2H), 7.81 (br s., 4H), 7.88-7.90 (d, J= 8.4 Hz, IH), 8.23-8.25 (dd, J1 = 8.4 Hz, J2 = 1.7 Hz, IH), 8.37 (s, IH), 9.66 (br s., IH).
Example 70. Synthesis of 5-|"4-chloro-3-(trifluoromethyl)phenyll-N-r4-(pyridine-4- yloxy)phenyl1-4H-l,2,4-triazol-3-amme trifluoroacetate salt
Figure imgf000108_0001
CF3COOH
[0268] 4-[5-(4-chloro-3-(trifluoromethyl)-phenyl)-4H-l ,2,4-triazol-3-ylamino]phenol (100 mg, 0.282 mmol) was dissolved in 2 mL of anhydrous DMF. Solid potassium bis(trimethylsilyl)amide (196.2 mg, 0.983 mmol) was added and the resulting solution was heated at 800C for 15 min. Then solid K2CO3 (20 mg, 0.141 mmol) was added, followed by 4-chloropyridine hydrochloride (63.2 mg, 0.421 mmol). The reaction mixture was microwaved at 220 0C for 30 min. Then it was diluted with 1 mL of MeOH, filtered through 0.22 um syringe filter and purified by preparative reverse-phase chromatography using acetonitrile/water gradient with 0.1 % TFA. The major peak having the mass of the product was collected, solvent was removed in vacuo to give trifluoroacetate salt of the product as a light-brown solid (23.5 mg).
[0269] ESI-MS: [M+Η]+ 432, 433 . 1H NMR (DMSO-d6): δ 7.23-7.25 (d, J= 8.8 Hz, 2H), 7.39-7.40 (d, J= 7.2 Hz, 2H), 7.74-7.77 (d, J= 8.8 Hz, 2H), 7.89-7.90 (m, IH), 8.24-8.26 F
(dd, J1 = 8.4 Hz, J2 = 2.0 Hz, IH), 8.38 (s, IH), 8.73-8.74 (d, J= 7.2 Hz, 2H), 9.75 (br s., IH).
Example 71. Synthesis of 5-[4-chloro-3-ftrifluoromethγl)phenyll-N-r4-(τ)yrimidin-5- vloxy)phenvll-4H-l ,2.4-triazol-3 -amine trifluoroacetate salt
Figure imgf000109_0001
[0270] 4-[5-(4-chloro-3-(trifluoromethyl)-phenyl)-4H-l,2,4-triazol-3-ylamino]phenol (100 mg, 0.282 mmol) was dissolved in 2 mL of anhydrous DMF. Solid potassium bis(trimethylsilyl)amide (84.1 mg, 0.421 mmol) was added and the resulting solution was heated at 800C for 15 min. Then solid K2CO3 (20 mg, 0.141 mmol) was added, followed by 5-bromopyrimidine (67.0 mg, 0.421 mmol). The reaction mixture was microwaved at 220 0C for 20 min. Then it was diluted with 1 mL of MeOH, filtered through 0.22 um syringe filter and purified by preparative reverse-phase chromatography using acetonitrile/water gradient with 0.1 % TFA. The major peak having the mass of the product was collected; solvent was removed in vacuo to give trifluoroacetate salt of the product as a light-brown solid (25.0 mg).
[0271] ESI-MS: [M+Η]+ 432.9, 435. 1H NMR (DMSOd6): δ 7.14-7.16 (d, J= 8.9 Hz5 2H), 7.63-7.67 (d, J= 8.9 Hz, 2H), 7.87-7.89 (d, J= 8.4 Hz, IH), 8.22-8.24 (dd, J1 = 8.4 Hz, J2 = 2.0 Hz, IH), 8.36 (s, IH), 8.54 (s, 2H), 8.93 (s, IH), 9.57 (br s., IH).
Example 72. Synthesis of 6-[4-d5-r4-chloro-3-(trifluoromethyl)ρhenyll-4H-12,4-triazol-3- vUammo)phenoxylpyridazin-3 -amine trifluoroacetate salt
Figure imgf000109_0002
[0272] 4-[5-(4-chloro-3-(trifluoromethyl)-phenyl)-4H-l ,2,4-triazol-3-ylamino]phenol (100 mg, 0.282 mmol) was dissolved in 2 mL of anhydrous DMF. Solid potassium bis(trimethylsilyl)amide (140.6 mg, 0.705 mmol) was added and the resulting solution was heated at 8O0C for 15 min. Then solid K2CO3 (20 mg, 0.141 mmol) was added, followed by 3-amino-6-chloropyridazine (54.6 mg, 0.421 mmol). The reaction mixture was microwaved at 220 0C for 40 min. Then it was diluted with 1 mL of MeOH, filtered through 0.22 um syringe filter and purified hy preparative reverse-phase chromatography using acetonitrile/water gradient with 0.1 % TFA. The major peak having the mass of the product was collected; solvent was removed in vacuo to give trifluoroacetate salt of the product as a light-brown solid (23.1 mg).
[0273] ESI-MS: [M+H]+ 448, 449. 1H NMR (DMSO-d6): δ 7.16-7.18 (d, J= 8.9 Hz, 2H), 7.51-7.53 (d, J= 9.7 Hz, IH), 7.62-7.66 (d, J= 8.9 Hz, 2H), 7.73-7.75 (d, J= 9.7 Hz, IH), 7.88 (d, J = 8.4 Hz, IH), 8.23-8.25 (dd, J1 = 8.4 Hz, J2 = 2.0 Hz, IH), 8.36 (s, IH), 8.48 (br s., 2H), 9.62 (br s., IH).
Example 73. Synthesis of 4-r4-("(5-r4-chloro-3-(trifluoromethyl)phenyll-4H-l,2,4-triazol-3- yl>amino)phenoxyl-N-methylt)yridine-2-carboxamide trifluoroacetate salt
Figure imgf000110_0001
CF3COOH
[0274] 4-[5-(4-chloro-3-(trifluoromethyl)-phenyl)-4H-l ,2,4-triazol-3-ylamino]phenol (100 mg, 0.282 mmol) was dissolved in 2 mL of anhydrous DMF. Solid potassium bis(trirnethylsilyl)amide (140.6 mg, 0.705 mmol) was added and the resulting solution was heated at 800C for 15 min. Then solid K2CO3 (20 mg, 0.141 mmol) was added, followed by 4-chloro-2-pyridine-carboxamide (71.9 mg, 0.421 mmol). The reaction mixture was microwaved at 220 0C for 20 min. Then it was diluted with 1 mL of MeOH, filtered through 0.22 um syringe filter and purified by preparative reverse-phase chromatography using acetonitrile/water gradient with 0.1 % TFA. The major peak having the mass of the product .was collected; solvent was removed in vacuo to give trifluoroacetate salt of the product as yellow solid (25.4 mg).
[0275] ESI-MS: [M+Η]+ 489, 490 . 1H NMR (DMSO-d6): δ 2.77-2.78 (d, J= 4.8 Hz, 3H), 7.14-7.15 (dd, J1 = 5.6 Hz, J2 = 2.5 Hz, IH), 7.15-7.17 (d, J= 9.0 Hz, 2H), 7.40 (d, J= 2.5 Hz, IH), 7.68-7.71 (d, J= 9.0 Hz, 2H), 7.87-7.89 (d, J= 8.4 Hz, IH), 8.23-8.25 (dd, J1 = 8.4 Hz, J2 = 2.0 Hz, IH), 8.37 (s, IH), 8.49-8.50 (d, J= 5.6 Hz, IH), 8.75-8.78 (q, J= 4.8 Hz, IH), 9.65 (br s., IH). Example 74. Synthesis of 5-[4-chloro-3-ftrifluorometh.yl)phenyll-N-[4-("pyridine-3- yloxy)phenyl]-4H-l,2,4-triazol-3-amine trifluoroacetate salt
Figure imgf000111_0001
[0276] 4-[5-(4-chloro-3-(trifluoromethyl)-phenyl)-4H-l ,2,4-triazol-3-ylamino]phenol (200 mg, 0.562 mmol) was dissolved in 2 niL of anhydrous DMF. Solid potassium bis(trimethylsilyl)amide (280.3 mg, 1.405 mmol) was added and the resulting solution was heated at 800C for 15 min. Then solid K2CO3 (40 mg, 0.282 mmol) was added, followed by 3-bromopyridine (177.6 mg, 1.12 mmol). The reaction mixture was microwaved at 250 0C for 20 min. Then it was diluted with 1 mL of MeOH, filtered through 0.22 um syringe filter and purified by preparative reverse-phase chromatography using acetonitrile/water gradient with 0.1 % TFA. The major peak having the mass of the product was collected, solvent was removed in vacuo to give trifluoroacetate salt of the product as a yellow solid (17.0 mg).
[0277] ESI-MS: [M+Η]+ 432, 433 . 1H NMR (DMSOd6): δ 7.09-7.11 (d, J= 9.0 Hz, 2H), 7.50-7.52 (m, 2H), 7.63-7.66 (d, J- 9.0 Hz, 2H), 7.87-7.89 (d, J= 8.4 Hz, IH), 8.22-8.25 (dd, Ji = 8.4 Hz, J2 = 2.0 Hz, IH), 8.37-8.38 (m, 2H), 8.43 (m, IH), 9.56 (br s., IH).
Example 75. Synthesis of 4-(5-{r3-(Trifluoromethyl)phenyllamino|-l,3,4-oxadiazol-2- vpphenol
Figure imgf000111_0002
[0278] Mercury (II) oxide yellow (5.33 g, 24.60 mmol) was suspended in ca. 70 mL of anhydrous methanol. 4-Hydroxybenzoic acid hydrazide (3.74 g, 24.60 mmol) was added to this bright-orange suspension, followed by 3-triflurormethylphenylisothiocyanate (5.0 g, 24.60 mmol). The reaction mixture was brought to reflux and refluxed for 2 hours. The reaction mixture turned pitch-black in color and formed black precipitate. Then it was cooled down to ambient temperature and filtered through a short pad of Celite, then through a short pad of silica gel. Then methanol was removed in vacuo and the resulting grey precipitate was re-crystallized from ca. 100 mL of EtOAc. The formed white crystalline solid was filtered, washed with a small amount of EtOAc and dried in vacuo to give the title product as white crystals (7.182 g). Yield 71.3%.
[0279] ESI-MS: [M+H]+ 322.0. 1H NMR (DMSOd6): δ 6.92-6.95 (d, J= 8.7 Hz, 2H), 7.33-7.35 (d, J = 8.3 Hz, IH), 7.57-7.60 (t, J= 8.0 Hz, IH), 7.72-7.75 (d, J= 8.7 Hz, 2H), 7.80-7.82 (dd, J1 = 8.0 Hz, J2 = 1.8 Hz, IH), 8.06 (s, IH), 10.21 (s, IH), 10.99 (br s., IH). 13C NMR (DMSO-d6) 112.9, 114.5, 116.1, 117.9, 120.6, 127.6, 129.7, 130.0, 130.3, 139.6, 158.3, 158.9, 160.1.
Example 76. Synthesis of 6-r4-(5-{r3-(trifluoromethyl)phenyl1amino|-l,3,4-oxadiazol-2- yl)phenoxvlpyrimidine-2,4-diamine
Figure imgf000112_0001
[0280] 4-(5-{[3-(trifluoromethyl)-phenyl]amino}-l,3,4-oxadiazol-2-yl)phenol (160.6 mg, 0.5 mmol) was dissolved in 3 niL of anhydrous DMF in a 2-5 mL microwave vial (Personal Chemistry). Solid potassium bis(trimethylsilyl)amide (119.7 mg, 0.6 mmol) was added and the reaction mixture was stirred with heating at 800C for 10 min, then 6-chloro-2,4~diamino- pyrimidine (86.7 mg, 0.6 mmol) was added, followed by anhydrous K2CO3 (69.1 mg, 0.5 mmol). Then the vial was capped and microwaved at 200 0C for 20 min. Then the reaction mixture was diluted with ca. 1 mL of MeOH, filtered through 0.22 um syringe filter and purified by reverse-phase preparative HPLC in acetonitrile/water system with 0.01% TFA. Fractions, containing the product, were partitioned between ca. 40 mL of EtOAc and ca. 40 mL of saturated NaHCO3. EtOAc layer was washed with brine, dried over anhydrous Na2SO4 and filtered. Solvent was removed in vacuo to give the title product as a beige solid (81.0 mg). Yield 37.7%.
[0281] ESI-MS: [M+H]+ 430.29. 1H NMR (DMSO-d6): δ 5.19 (s, 1H),6.O4 (s, 2H), 6.33 (s, 2H), 7.27-7.30 (d, J= 8.7 Hz, 2H), 7.36-7.37 (d, J= 7.9 Hz, IH), 7.59-7.63 (t, J= 8.0 Hz, IH), 7.82-7.83 (dd, J1 = 8.0 Hz, J2 = 1.8 Hz, IH), 7.89-7.92 (d, J= 8.7 Hz, 2H), 8.08 (s, IH). 11.11 (s, IH). Example 77. Synthesis of 5-[4-(Pyrimidin-5-yloxy')phenyll-N-r3-(trifluoroinethyπphenyll-
1 ,3 ,4-oxadiazol-2-amine
Figure imgf000113_0001
[0282] 4-(5-{[3-(trifluoromethyl)-phenyl]amino}-l,3,4-oxadiazol-2-yl)phenol (160.6 mg, 0.5 mmol) was dissolved in 3 niL of anhydrous DMF in a 2-5 niL microwave vial (Personal Chemistry). Solid potassium bis(trimethylsilyl)amide (149.6 mg, 0.75 mmol) was added and the reaction mixture was stirred with heating at 800C for 10 min, then 5-bromopyrimidine (119.2 mg, 0.75 mmol) was added, followed by anhydrous K2CO3 (69.1 mg, 0.5 mmol). Then the vial was capped and microwaved at 200 0C for 20 min. Then the reaction mixture was diluted with ca. 1 mL of MeOH, filtered through 0.22 um syringe filter and purified by reverse-phase preparative HPLC in acetonitrile/water system with 0.01% TFA. Fractions, containing the product, were partitioned between ca. 40 mL of EtOAc and ca. 40 mL of saturated NaHCO3. EtOAc layer was washed with brine, dried over anhydrous Na2SO4 and filtered. Solvent was removed in vacuo to give the title product as a beige solid (87.0 mg). Yield 43.5%.
[0283] ESI-MS: [M+H]+ 400.16. 1HNMR (DMSO^6): δ 7.31-7.33 (d, J= 8.7 Hz, 2H), 7.35-7.37 (d, J= 7.9 Hz, IH), 7.59-7.62 (t, J= 8.0 Hz, IH), 7.81-7.83 (dd, J1 = 8.0 Hz, J2 = 1.8 Hz, IH), 7.93-7.95 (d, J= 8.7 Hz, 2H), 8.07 (s, IH), 8.76 (s, 2H), 9.08 (s, IH), 11.11 (s, IH).
Example 78. Synthesis of 4-(5-r(4-chlorophenyl)amino1-l,3,4-oxadiazol-2-yliphenol
Figure imgf000113_0002
[0284] Mercury (II) oxide yellow (6.38 g, 29.47 mmol) was suspended in ca. 70 mL of anhydrous methanol. 4-Hydroxybenzoic acid hydrazide (4.48 g, 29.47 mmol) was added to this bright-orange suspension, followed by 4-chlorophenylisothiocyanate (5.0 g, 29.47 mmol). The reaction mixture was brought to reflux and refluxed for 2 hours. The reaction mixture turned pitch-black in color and formed black precipitate. Then it was cooled down to ambient temperature and filtered through a short pad of Celite, then through a short pad of silica gel. Then methanol was removed in vacuo and the resulting grey precipitate was re- crystallized from ca. 40 niL of EtOAc. The formed white precipitate was filtered, washed with a small amount of EtOAc and dried in vacuo to give the title product as a white powder.
[0285] ESI-MS: [M+H]+ 287.94. 1H NMR (DMSO-d6): δ 6.91-6.94 (d, J= 8.7 Hz, 2H), 7.39-7.41 (d, J= 8.9 Hz, 2H), 7.61-7.63 (d, J= 8.9 Hz, 2H), 7.71-7.74 (d, J= 8.7 Hz, 2H), 10.19 (s, IH), 10.73 (s, IH). 13C NMR (DMSO-d6) 114.6, 116.1, 118.5, 125.3, 127.5, 128.9, 137.8, 158.1, 159.1, 160.0.
Example 79. Synthesis of 6-(4-{5-[('4-chlorophenyl)aminol-l,3.4-oxadiazol-2-yl|phenoxy>) ρyrimidine-2.4-diamine
Figure imgf000114_0001
[0286] 4-{5-[(4-chlorophenyl)amino]-l,3,4-oxadiazol-2-yl}phenol (144.0 mg, 0.5 mmol) was dissolved in 3 mL of anhydrous DMF in a 2-5 mL microwave vial (Personal Chemistry). Solid potassium bis(trimethylsilyl)amide (100.0 mg, 0.5 mmol) was added and the reaction mixture was stirred with heating at 800C for 10 min, then 6-chloro-2,4-diamino-pyrirnidine (72.3 mg, 0.5 mmol) was added, followed by anhydrous K2CO3 (34.5 mg, 0.25 mmol). Then the vial was capped and microwaved at 200 0C for 15 min. Then the reaction mixture was diluted with ca. 1 mL of MeOH, filtered through 0.22 um syringe filter and purified by reverse-phase preparative HPLC in acetonitrile/water system with 0.01% TFA. Fractions, containing the product, were partitioned between ca. 40 mL of EtOAc and ca. 40 mL of saturated NaHCO3. EtOAc layer was washed with brine, dried over anhydrous Na2SO4 and filtered. Solvent was removed in vacuo to give the title product as a beige solid (24.5 mg).
[0287] ESI-MS: [M+H]+ 396.25 . 1H NMR (DMSO-d6): δ 5.19 (s, IH), 6.04 (s, 2H), 6.33 (s, 2H), 7.26-7.29 (d, J= 8.7 Hz, 2H), 7.41-7.43 (d, J= 8.9 Hz, 2H), 7.63-7.65 (d, J= 8.9 Hz, 2H), 7.88-7.90 (d, J= 8.7 Hz, 2H), 10.85 (s, IH).
Example 80. Synthesis of 5-r4-(Pyrimidin-5-yloxy)phenyl1-N-r4-chloro-phenyll-l,3,4- oxadiazol-2-amine
Figure imgf000114_0002
[0288] 4-{5-[(4-chlorophenyl)amino]-l ,3,4-oxadiazol-2-yl}phenol (144.0 mg, 0.5 mmol) was dissolved in 3 mL of anhydrous DMF in a 2-5 niL microwave vial (Personal Chemistry). Solid potassium bis(trimethylsilyl)amide (100.0 mg, 0.5 mmol) was added and the reaction mixture was stirred with heating at 80 0C for 10 min, then 5-bromopyrimidine (79.5 mg, 0.5 mmol) was added, followed by anhydrous K2CO3 (34.5 mg, 0.25 mmol). Then the vial was capped and microwaved at 200 0C for 15 min. Then the reaction mixture was diluted with ca. 1 mL of MeOH, filtered through 0.22 um syringe filter and purified by reverse-phase preparative HPLC in acetonitrile/water system with 0.01% TFA. Fractions, containing the product, were partitioned between ca. 40 mL of EtOAc and ca. 40 mL of saturated NaHCO3. EtOAc layer was washed with brine, dried over anhydrous Na2SO4 and filtered. Solvent was removed in vacuo to give the title product as a beige solid (61.6 mg).
[0289] ESI-MS: [M+H]+ 366.24. 1H NMR (DMSO-d6): δ 7.30-7.32 (d, J= 8.7 Hz, 2H), 7.41-7.43 (d, J= 8.9 Hz, 2H), 7.63-7.65 (d, J= 8.9 Hz, 2H), 7.92-7.94 (d, J= 8.7 Hz, 2H), 8.76 (s, 2H), 9.08 (s, IH), 10.85 (s, IH).
Example 81. Synthesis of 4-(5-{r2-chloro-5-(trifluoromethyl)phenyllamino>-L3,4- oxadiazol-2-yl)phenol
Figure imgf000115_0001
[0290] Mercury (II) oxide yellow (1.82 g, 8.41 mmol) was suspended in ca. 50 mL of anhydrous methanol. 4-Hydroxybenzoic acid hydrazide (1.28 g, 8.41 mmol) was added to this bright-orange suspension, followed by 2-chloro-5-trifluoromethyl-phenylisothiocyanate (2.0 g, 8.41 mmol). The reaction mixture was brought to reflux and refluxed for 2 hours. The reaction mixture turned pitch-black in color and formed black precipitate. Then it was cooled down to ambient temperature and filtered through a short pad of Celite, then through a short pad of silica gel. Then methanol was removed in vacuo and the resulting grey solid was re-crystallized from ca. 20 mL of EtOAc. The formed white precipitate was filtered, washed with anhydrous Et2O and dried in vacuo to give the title product as a white solid (2.638 g). Yield 88.1%
[0291] ESI-MS: [M+H]+ 356.22. 1H NMR (DMSO-d6): δ 6.93-6.96 (d, J= 8.7 Hz, 2H), 7.41-7.43 (dd, J7 = 8.3 Hz, J2 = 1.7 Hz, IH), 7.72-7.75 (m, 3H), 8.61 (s, IH), 10.23 (s, IH), 10.30 (s, IH). Example 82. Synthesis of 6-(4-(5-rr2-chloro-5-trifluoromethyl-phenyl)aminol-1.3.4- oxadiazol-2-vUphenoxy) pyrimidme-2.4-diamine
Figure imgf000116_0001
[0292] 4-(5-{[2-chloro-5-(trifluoromethyl)phenyl]amino}-l,3,4-oxadiazol-2-yl)phenol (177.85 mg, 0.5 mmol) was dissolved in 3 mL of anhydrous DMF in a 2-5 mL microwave vial (Personal Chemistry). Solid potassium bis(trimethylsilyl)amide (200.0 mg, 1.0 mmol) was added and the reaction mixture was stirred with heating at 800C for 10 min, then 6- chloro-2,4-diamino-pyrimidine (86.7 mg, 0.6 mmol) was added, followed by anhydrous K2CO3 (69.1 mg, 0.5 mmol). Then the vial was capped and microwaved at 180 0C for 30 min. Then the reaction mixture was diluted with ca. 1 mL of MeOH, filtered through 0.22 um syringe filter and purified by reverse-phase preparative HPLC in acetonitrile/water system with 0.01% TFA. Fractions, containing the product, were partitioned between ca. 50 mL of EtOAc and ca. 50 mL of saturated aqueous NaHCO3. EtOAc layer was washed with brine, dried over anhydrous Na2SO4 and filtered. Solvent was removed in vacuo to give the title product as an off-white solid (131.1 mg). Yield 56.5%.
[0293] ESI-MS: [M+H]+ 464.21. 1HNMR (DMSO-d6): δ 5.20 (s, IH), 6.05 (s, 2H), 6.34 (s, 2H), 7.28-7.31 (d, J= 8.7 Hz, 2H), 7.46-7.48 (dd, J1 = 8.3 Hz, J2 = 1.8 Hz, IH), 7.76-7.78 (d, J= 8.3 Hz, IH), 7.90-7.92 (d, J= 8.7 Hz, 2H), 8.62 (s, IH), 10.47 (s, IH).
Example 83. Synthesis of 5-r4-(Pyrimidm-5-yloxy)phenyll-N-[2-chloro-5-(trifluoromethyl)- phenyll -1,3 ,4-oxadiazol-2-amine
Figure imgf000116_0002
[0294] 4-(5- { [2-chloro-5 -(trifluoromethyl)phenyl] amino } - 1 ,3 ,4-oxadiazol-2-yl)phenol (177.85 mg, 0.5 mmol) was dissolved in 3 mL of anhydrous DMF in a 2-5 mL microwave vial (Personal Chemistry). Solid potassium bis(trimethylsilyl)amide (200.0 mg, 1.0 mmol) was added and the reaction mixture was stirred with heating at 800C for 10 min, then 5- bromopyrimidine (95.4 mg, 0.6 mmol) was added, followed by anhydrous K2CO3 (69.1 mg, 0.5 mmol). Then the vial was capped and microwaved at 180 0C for 40 min. Then the reaction mixture was diluted with ca. 1 niL of MeOH, filtered through 0.22 um syringe filter and purified by reverse-phase preparative HPLC in acetonitrile/water system with 0.01% TFA. Fractions, containing the product, were partitioned between ca. 50 mL of EtOAc and> ca. 50 mL of saturated aqueous NaHCO3. EtOAc layer was washed with brine, dried over anhydrous Na2SO4 and filtered. Solvent was removed in vacuo to give the title product as a beige solid (129.1 mg). Yield 59.5%.
[0295] ESI-MS: [MH-H]+ 434.20. 1H NMR (DMSO-d6): δ 7.32-7.35 (d, J= 8.7 Hz, 2H), 7.46-7.48 (dd, J1 = 8.3 Hz, J2 = 1.8 Hz, IH), 7.76-7.78 (d, J= 8.3 Hz, IH), 7.94-7.96 (d, J = 8.7 Hz, 2H), 8.61 (s, IH), 8.77 (s, 2H), 9.09 (s, IH), 10.47 (s, IH).
Example 84 . Synthesis of 4- (5-[(3-chlorophenyl)amino~l-l,3,4-oxadiazol-2-vU phenol
Figure imgf000117_0001
[0296] Mercury (II) oxide yellow (6.38 g, 29.47 mmol) was suspended in ca. 100 mL of anhydrous methanol. 4-Hydroxybenzoic acid hydrazide (4.48 g, 29.47 mmol) was added to this bright-orange suspension, followed by 3-chlorophenylisothiocyanate (5.0 g, 29.47 mmol). The reaction mixture was brought to reflux and refluxed for 2 hours. The reaction mixture turned pitch-black in color and formed black precipitate. Then it was cooled down to ambient temperature and filtered through a short pad of Celite. Then it was purified by silica gel chromatography using 0% to 20 % methanol gradient in EtOAc. Solvent was removed in vacuo and the resulting grey precipitate was re-crystallized from ca. 50 mL of EtOAc. The formed white crystalline solid was filtered, washed with a small amount of EtOAc, anhydrous Et2O and dried in vacuo to give the title product as a white powder (7.606 g). Yield 89.7%.
[0297] ESI-MS: [M+H]+ 288.26. 1H NMR (DMSO-d6): δ 6.92-6.94 (d, J= 8.7 Hz, 2H), 7.00-7.02 (dd, J, = 7.9 Hz, J2 = 1.8 Hz, IH), 7.33-7.36 (t, J= 8.1 Hz, IH), 7.47-7.49 (dd, J1 = 8.1 Hz, J2 = 1.8 Hz, IH), 7.72-7.74 (d, J= 8.7 Hz, 2H), 7.76-1.7% (t, J= 2.1 Hz, IH), 10.19 (s, IH), 10.81 (s, IH). 13C NMR (DMSO-d6) 114.6, 115.5, 116.1, 116.4, 121.3, 127.6, 130.6, 133.5, 140.3, 158.3, 159.0, 160.1. Example 85. Synthesis of 6-(4-{5-rθ-chloroρhenvDamino1-l,3Λ-oxadiazol-2-yl}Ohenoχy)
Pvrimidine-2.4-diamine
Figure imgf000118_0001
[0298] 4-{5-[(3-chlorophenyl)amino]-l,3,4-oxadiazol-2-yl}phenol (143.8 mg, 0.5 mmol) was dissolved in 3 mL of anhydrous DMF in a 2-5 mL microwave vial (Personal Chemistry). Solid potassium bis(trimethylsilyl)amide (200.0 mg, 1.0 mmol) was added and the reaction mixture was stirred with heating at 80 0C for 10 min, then 6-chloro-2,4-diamino-pyrimidine (86.7 mg, 0.6 mmol) was added, followed by anhydrous K2CO3 (69.1 mg, 0.5 mmol). Then the vial was capped and microwaved at 1800C for 40 min. Then the reaction mixture was diluted with ca. 1 mL of MeOH, filtered through 0.22 um syringe filter and purified by reverse-phase preparative HPLC in acetonitrile/water system with 0.01% TFA. Fractions, containing the product, were partitioned between ca. 50 mL of EtOAc and ca. 50 mL of saturated aqueous NaHCO3. EtOAc layer was washed with brine, dried over anhydrous Na2SO4 and filtered. Solvent was removed in vacuo to give the title product as a light-yellow solid (62.0 mg). Yield 31.3%.
[0299] ESI-MS: [M+H]+ 396.22. 1H NMR (DMSO-d6): δ 5.19 (s, IH), 6.04 (s, 2H), 6.33 (s, 2H), 7.06-7.08 (m, IH), 7.27-7.29 (d, J= 8.7 Hz, 2H), 7.37-7.41 (t, J= 8.1 Hz, IH), 7.50- 7.52 (dd, J7 = 8.1 Hz5 J2 = 1.8 Hz, IH), 7.78-7.79 (t, J= 2.1 Hz, IH), 7.89-7.90 (d, J= 8.7 Hz5 2H), 10.95 (s, IH).
Example 86. Synthesis of 5-r4-(Pyrimidin-5-yloxy)phenyll-N-[3-chloro-phenyl1-l,3,4- oxadiazol-2-amine
Figure imgf000118_0002
[0300] 4-{5-[(3-chlorophenyl)amino]-l,3,4-oxadiazol-2-yl}phenol (143.8 mg, 0.5 mmol) was dissolved in 3 mL of anhydrous DMF in a 2-5 mL microwave vial (Personal Chemistry). Solid potassium bis(trimethylsilyl)amide (200.0 mg, 1.0 mmol) was added and the reaction mixture was stirred with heating at 80 0C for 10 min, then 5-bromopyrimidine (95.4 mg, 0.6 mmol) was added, followed by anhydrous K2CO3 (69.1 mg, 0.5 mmol). Then the vial was capped and microwaved at 180 0C for 30 min. Then the reaction mixture was diluted with ca. 1 rnL of MeOH, filtered through 0.22 um syringe filter and purified by reverse-phase preparative HPLC in acetonitrile/water system with 0.01% TFA. Fractions, containing the product, were partitioned between ca. 50 niL of EtOAc and ca. 50 mL of saturated aqueous NaHCO3. EtOAc layer was washed with brine, dried over anhydrous Na2SO4 and filtered. Solvent was removed in vacuo to give the title product as a light-yellow solid (74.7 mg). Yield 40.8%.
[0301] ESI-MS: [M+H]+ 366.23 . 1H NMR (DMSO-d6): δ 7.06-7.08 (m, IH), 7.31-7.33 (d, J= 8.7 Hz, 2H), 7.37-7.41 (t, J= 8.1 Hz, IH), 7.50-7.52 (dd, J1 = 8.1 Hz, J2 = 1.8 Hz, IH), 7.77-7.79 (t, J= 2.0 Hz, IH), 7.93-7.94 (d, J= 8.7 Hz, 2H), 8.77 (s, 2H), 9.09 (s, IH), 10.96 (s, IH).
Example 87. Synthesis of 5-r4-(Pyridin-3-yloxy)phenyl1-l,3,4-oxadiazol-2-amme hydrobromide salt
Figure imgf000119_0001
[0302] 4-(pyridine-3-yloxy)benzohydrazide (3.7 g, 16.14 mmol) was dissolved in 100 mL of anhydrous THF and 3.0 M solution of cyanogen bromide (5.38 mL, 16.14 mmol) was added via syringe. Within 5-10 min of stirring an orange precipitate started to form. The reaction mixture was brought to reflux and refluxed for 1 hr. Then it was cooled down to ambient temperature and filtered. The collected orange precipitate was washed with ca. 100 mL of THF, ca. 100 mL of EtOAc, anhydrous Et20 and dried in vacuo to give the title product as an orange solid (4.40 g). Yield 81.4%.
[0303] ESI-MS: [M+H]+ 255.05 . 1H NMR (DMSO-d6): δ 7.27-7.29 (d, J= 8.8 Hz, 2H), 7.75-7.78 (dd, J1 = 8.5 Hz, J2 = 5.0 Hz, IH), 7.85-7.87 (d, J= 8.8 Hz, 2H), 7.92-7.94 (m, IH), 8.59-8.60 (dd, J1 = 5.0 Hz, J2 = 1.0 Hz, IH), 8.69-8.70 (d, J= 2.7 Hz, IH). Example 88. Synthesis of N-f5-[4-fτ3yridm-3-yloxy')phenyll-L3,4-oxadiazol-2-vU-4-
(1rifluoromethoxv)benzamide
Figure imgf000120_0001
[0304] 5-[4-(Pyridin-3-yloxy)phenyl]-l,3,4-oxadiazol-2~amine hydrobromide salt (167.5 mg, 0.5 mmol) was suspended in 2 niL of anhydrous pyridine. 4-Trifluoromethoxybenzoyl chloride (167.2 mg, 117 uL, 0.75 mmol) was added directly into the solution. The reaction mixture formed an orange-red solution with a small amount of precipitate. It was left to stir for 6 hours. Then it was diluted with ca. 1 mL of MeOH, filtered through 0.22 u syringe filter and purified by reverse phase preparative HPLC using acetonitrile/water mixture containing 0.01% of TFA. Fractions, containing the product, were combined and partitioned between ca. 40 mL of EtOAc and ca. 40 mL of saturated aqueous NaHCO3. The EtOAc layer was washed with brine, dried over anhydrous sodium sulfate and filtered. Solvent was removed in vacuo to give the title product as a yellow solid (89.0 mg). Yield 40.2%.
[0305] ESI-MS: [M+H]+ 442.82. 1H NMR (DMSO-d6): δ 7.20-7.22 (d, J= 8.8 Hz, 2H), 7.44_7.46 (d, J= 8.6 Hz, 2H), 7.48-7.51 (dd, J1 = 8.4 Hz, J2 = 4.6 Hz, IH), 7.59-7.62 (ddd, J1 = 8.4 Hz, J2 = 2.8 Hz, J3 = 1.2 Hz, IH), 7.94-7.96 (d, J= 8.8 Hz, 2H), 8.17-8.19 (d, J= 8.6 Hz, 2H), 8.45-8.46 (dd, J1 = 4.6 Hz, J2 = 1.2 Hz, IH), 8.48-8.49 (d, J= 2.8 Hz, IH).
Example 89. Synthesis of N-i5-r4-foyridm-3-yloxy)phenyl'|-l,3,4-oxadiazol-2-vU-3-
(trifluoromethyl)benzamide
Figure imgf000120_0002
[0306] 5-[4-(Pyridin-3-yloxy)phenyl]-l,3,4-oxadiazol-2-amine hydrobromide salt (167.5 mg, 0.5 mmol) was suspended in 2 mL of anhydrous pyridine. 3-Trifluoromethylbenzoyl chloride (156.4 mg, 0.75 mmol) was added to the solution. The reaction mixture formed an orange-red solution with a small amount of white precipitate. It was left to stir for 3 hours. Then it was diluted with ca. 1 mL of MeOH, filtered through 0.22 u syringe filter and purified by reverse phase preparative HPLC using acetonitrile/water mixture containing 0.01% of TFA. Fractions, containing the product, were combined and partitioned between ca. 40 mL of EtOAc and ca. 40 mL of saturated aqueous NaHCO3. The EtOAc layer was washed with brine, dried over anhydrous sodium sulfate and filtered. Solvent was removed in vacuo to give the title product as a yellow solid (50.0 mg). Yield 23.4%.
[0307] ESI-MS: [M+H]+ 426.94. 1H NMR (DMSO-d6): δ 7.21-7.23 (d, J= 8.8 Hz, 2H), 7.49-7.51 (dd, J1 = 8.4 Hz, J2 = 4.6 Hz, IH), 7.60-7.62 (ddd, J1 = 8.4 Hz, J2 = 2.8 Hz, J3 = 1.2 Hz, IH), 7.75-7.76 (t, J= 7.7 Hz, IH), 7.95-7.98 (m, 3H), 8.33-8.34 (d, J= 7.7 Hz, IH), 8.39 (s, IH), 8.46-8.47 (dd, J1 = 4.6 Hz, J2 = 1.2 Hz, IH), 8.48-8.49 (d, J= 2.8 Hz, IH).
Example 90. Synthesis of 4-Bromo-N-l5-r4-("r)yridin-3-yloxy)phenyll-l,3,4-oxadiazol-2-yll- benzamide
Figure imgf000121_0001
[0308] 5-[4-(Pyridin-3-yloxy)phenyl]-l,3,4-oxadiazol-2 -amine hydrobromide salt (167.5 mg, 0.5 mmol) was suspended in 2 mL of anhydrous pyridine. 4-Bromobenzoyl chloride (164.6 mg, 0.75 mmol) was added to the solution. The reaction mixture formed an orange- red solution with a small amount of white precipitate. It was left to stir for 3 hours. Then it was diluted with ca. 1 mL of MeOH, filtered through 0.22 u syringe filter and purified by reverse phase preparative HPLC using acetonitrile/water mixture containing 0.01% of TFA. Fractions, containing the product, were combined and partitioned between ca. 40 mL of EtOAc and ca. 40 mL of saturated aqueous NaHCO3. The EtOAc layer was washed with brine, dried over anhydrous sodium sulfate and filtered. Solvent was removed in vacuo to give the title product as a yellow solid (46.5 mg). Yield 21.2%.
[0309] ESI-MS: [M+H]+ 438.84. 1H NMR (DMSO-d6): δ 7.20-7.22 (d, J= 8.8 Hz, 2H), 7.49-7.51 (dd, J1 = 8.4 Hz, J2 = 4.6 Hz, IH), 7.60-7.62 (ddd, J1 = 8.4 Hz, J2 = 2.8 Hz, J3 = 1.2 Hz, IH), 7.68-7.70 (d, J= 8.4 Hz, 2H), 7.95-7.97 (d, J= 8.8 Hz, 2H), 8.00-8.02 (d, J= 8.4 Hz, 2H), 8.45-8.46 (dd, J1 = 4.6 Hz, J2 = 1.2 Hz, IH), 8.48-8.49 (d, J= 2.8 Hz, IH). Example 91. Synthesis of N-{5-r4-fayridin-3-yloxy)phenyll-l,3,4-oxadiazol-2-vU-3-
(trifluoromethoxyVbenzamide
Figure imgf000122_0001
[0310] 5-[4-(Pyridin-3-yloxy)phenyl]-l,3,4-oxadiazol-2-amine hydrobromide salt (167.5 mg, 0.5 mmol) was suspended in 2 niL of anhydrous pyridine. Neat 3- trifluoromethoxybenzoyl chloride (117 uL) was added directly into the solution. The reaction mixture formed an orange-red solution with a small amount of white precipitate. It was left to stir for 3 hours. Then it was diluted with ca. 1 mL of MeOH, filtered through 0.22 u syringe filter and purified by reverse phase preparative HPLC using acetonitrile/water mixture containing 0.01% of TFA. Fractions, containing the product, were combined and partitioned between ca. 40 mL of EtOAc and ca. 40 mL of saturated aqueous NaHCO3. The EtOAc layer was washed with brine, dried over anhydrous sodium sulfate and filtered. Solvent was removed in vacuo to give the title product as a yellow solid (38.8 mg). Yield 17.5%.
[0311] ESI-MS: [M+H]+ 442.85. 1H NMR (DMSO-d6): δ 7.22-7.24 (d, J= 8.8 Hz, 2H), 7.49-7.51 (dd, J1 = 8.4 Hz, J2 = 4.6 Hz, IH), 7.60-7.62 (ddd, J1 = 8.4 Hz, J2 = 2.8 Hz, J3 = 1.2 , Hz, IH), 7.61-7.63 (m, IH), 7.68.-7.71 (t, J= 7.7 Hz, IH), 7.97-7.98 (m, 3H), 8.08-8.10 (d, J = 7.7 Hz, IH), 8.46-8.47 (dd, J7 = 4.6 Hz, J2 = 1.2 Hz, IH), 8.48-8.49 (d, J= 2.8 Hz, IH).
Example 92. Synthesis of 4-methoxy-N-{5-r4-(pyridin-3-yloxy)phenyll-L3.4-oxadiazol-2- vl|-3-(trifluoromethyl)-benzamide
Figure imgf000122_0002
[0312] 5-[4-(Pyridin-3-yloxy)phenyl]-l,3,4-oxadiazol-2-amine hydrobromide salt (167.5 mg, 0.5 mmol) was suspended in 2 mL of anhydrous pyridine. Neat 4-methoxy-3- trifluoromethylbenzoyl chloride (179.0 mg, 0.75 mmol) was added directly into the solution. The reaction mixture formed an orange-red solution with a small amount of white precipitate. It was left to stir for 3 hours. Then it was diluted with ca. 1 mL of MeOH, filtered through I 0.22 u syringe filter and purified by reverse phase preparative HPLC using acetonitrile/water mixture containing 0.01% of TFA. Fractions, containing the product, were combined and partitioned between ca. 40 mL of EtOAc and ca. 40 mL of saturated aqueous NaHCO3. The EtOAc layer was washed with brine, dried over anhydrous sodium sulfate and filtered. Solvent was removed in vacuo to give the title product as a yellow solid (37.0 mg). Yield 16.2%.
[0313] ESI-MS: [M+H]+ 456.85. 1H NMR (DMSO-d6): δ 3.97 (s, 3H), 7.21-7.23 (d, J= 8.8 Hz, 2H), 7.38-7.40 (d, J= 9.1 Hz, IH), 7.49-7.51 (dd, J1 = 8.4 Hz, J2 = 4.6 Hz, IH), 7.60- 7.62 (ddd, Ji = 8.4 Hz, J2 = 2.8 Hz, J3 = 1.2 Hz, IH), 7.95-7.97 (d, J= 8.8 Hz, 2H), 8.34-8.35 (m, 2H), 8.46-8.47 (dd, J1 = 4.6 Hz, J2 = 1.2 Hz, IH), 8.48-8.49 (d, J= 2.8 Hz, IH).
Example 93. Synthesis of 2,2-Difluoro-N-{5-r4-(pyridm-3-yloxy)phenyri-l,3,4-oxadiazol-2- yll-l,3-benzodioxole-5-carboxamide
Figure imgf000123_0001
[0314] 5-[4-(Pyridin-3-yloxy)phenyl]-l,3,4-oxadiazol-2-amine hydrobromide salt (167.5 mg, 0.5 mmol) was suspended in 2 mL of anhydrous pyridine. Neat 2,2-difluoro-l,3- benzodioxole-5-carbonyl chloride (165.4 mg, 0.75 mmol) was added directly into the solution. The reaction mixture formed an orange-red solution with a small amount of white precipitate. It was left to stir for 3 hours. Then it was diluted with ca. 1 mL of MeOH, filtered through 0.22 u syringe filter and purified by reverse phase preparative HPLC using acetonitrile/water mixture containing 0.01% of TFA. Fractions, containing the product, were combined and partitioned between ca. 40 mL of EtOAc and ca. 40 mL of saturated aqueous NaHCO3. The EtOAc layer was washed with brine, dried over anhydrous sodium sulfate and filtered. Solvent was removed in vacuo to give the title product as a yellow solid (27.0 mg). Yield 12.3%.
[0315] ESI-MS: [M+H]+ 456.85. 1H NMR (DMSO-d6): δ 7.19-7.21 (d, J= 8.8 Hz, 2H), 7.47-7.51 (m, 2H), 7.58-7.60 (ddd, J1 = 8.4 Hz, J2 = 2.8 Hz, J3 = 1.2 Hz, IH), 7.93-7.95 (d, J = 8.8 Hz, 2H), 7.99-8.01 (m, 2H), 8.45-8.46 (dd, J1 = 4.6 Hz, J2 = 1.2 Hz, IH), 8.48-8.49 (d, J= 2.8 Hz, IH). Example 94. Synthesis of 3-Chloro-2-fluoro-N-l5-14-('pyridin-3-yloxy')phenγll-l,3,4- oxadiazol-2-yl)-5-(rtfluoromethyl>benzamide
Figure imgf000124_0001
[0316] 5-[4-(Pyridin-3-yloxy)phenyl]-l,3,4-oxadiazol-2-amine hydrobromide salt (167.5 mg, 0.5 mmol) was suspended in 2 niL of anhydrous pyridine. Neat 3-chloro-2-fluoro-5~ trifluoromethylbenzoyl chloride (250 uL) was added directly into the solution. The reaction mixture formed a red solution with a small amount of white precipitate. It was left to stir for 18 hours. Then it was diluted with ca. 1 mL of MeOH, filtered through 0.22 u syringe filter and purified by reverse phase preparative HPLC using acetonitrile/water mixture containing 0.01% of TFA. Fractions, containing the product, were combined and partitioned between ca. 40 mL of EtOAc and ca. 40 mL of saturated aqueous NaHCO3. The EtOAc layer was washed with brine, dried over anhydrous sodium sulfate and filtered. Solvent was removed in vacuo to give the title product as a yellow solid (83.4 mg). Yield 34.8%.
[0317] ESI-MS: [M+H]+ 480.71. 1H NMR (DMSO-d6): δ 7.18-7.20 (d, J= 8.8 Hz, 2H), 7.47-7.50 (dd, J1 = 8.4 Hz, J2 = 4.6 Hz, IH), 7.58-7.60 (ddd, J1 = 8.4 Hz, J2 = 2.8 Hz, J3 = 1.2 Hz, IH), 7.92-7.94 (d, J= 8.8 Hz, 2H), 8.13-8.16 (m, 2H), 8.44-8.45 (dd, J1 = 4.6 Hz, J2 = 1.2 Hz, IH), 8.47-8.48 (d, J= 2.8 Hz, IH).
Example 95. Synthesis of 4-Fluoro-N-{5-r4-(pyridin-3-yloxy)ϋhenyll-L3,4-oxadiazol-2-yl}-
3 -(trifluoromethvD-benzamide
Figure imgf000124_0002
[0318] 5-[4-(Pyridin-3-yloxy)phenyl]-l,3,4-oxadiazol-2-amine hydrobromide salt (167.5 mg, 0.5 mmol) was suspended in 2 mL of anhydrous pyridine. Neat 4-fluoro-3- trifluoromethylbenzoyl chloride (200 uL) was added directly into the solution. It was left to stir for 18 hours. The reaction mixture formed a red solution with a yellow precipitate. Then it was diluted with ca. 1 mL of MeOH, filtered through 0.22 u syringe filter and purified by reverse phase preparative HPLC using acetonitrile/water mixture containing 0.01% of TFA. Fractions, containing the product, were combined and partitioned between ca. 40 mL of EtOAc and ca. 40 mL of saturated aqueous NaHCO3. The EtOAc layer was washed with brine, dried over anhydrous sodium sulfate and filtered. Solvent was removed in vacuo to give the title product as a yellow solid (105.1 mg). Yield 47.3%.
[0319] ESI-MS: [M+H]+ 444.79. 1HNMR (DMSO-d6): δ 7.18-7.20 (d, J= 8.8 Hz, 2H), 7.47-7.50 (dd, J1 = 8.4 Hz, J2 = 4.6 Hz, IH), 7.53-7.60 (m, 2H), 7.93-7.94 (d, J= 8.8 Hz, 2H), 8.40-8.43 (m, IH), 8.43-8.45 (dd, J1 = 4.6 Hz, J2 = 1.2 Hz, IH), 8.47-8.48 (d, J= 2.8 Hz, IH).
Example 96. Synthesis of N-i5-[4-(pyridin-3-yloxy)phenyl1-l,3,4-oxadiazol-2-yl|-2-
(trifluoromethoxvVbenzamide
Figure imgf000125_0001
[0320] 5-[4-(Pyridin-3-yloxy)phenyl]-l,3,4-oxadiazol-2-amine hydrobromide salt (167.5 mg, 0.5 mmol) was suspended in 2 mL of anhydrous pyridine. Neat 2- trifluoromethoxybenzoyl chloride (100 uL) was added directly into the solution. It was left to stir for 18 hours. The reaction mixture formed an orange solution with a yellow precipitate. Then it was diluted with ca. 1 mL of MeOH, filtered through 0.22 u syringe filter and purified by reverse phase preparative HPLC using acetonitrile/water mixture containing 0.01% of TFA. Fractions, containing the product, were combined and partitioned between ca. 40 mL of EtOAc and ca. 40 mL of saturated aqueous NaHCO3. The EtOAc layer was washed with brine, dried over anhydrous sodium sulfate and filtered. Solvent was removed in vacuo to give the title product as a bright-yellow solid (20.2 mg). Yield 9.1%.
[0321] ESI-MS: [M+H]+ 443.04. 1H NMR (DMSO-d6): δ 7.22-7.24 (d, J= 8.8 Hz, 2H), 7.48-7.54 (m, 4H), 7.60-7.63 (ddd, J1 = 8.4 Hz, J2 = 2.8 Hz, J3 = 1.2 Hz, IH), 7.69-7.71 (t, J = 7.8 Hz, IH), 7.80-7.81 (d, J= 7.5 Hz, IH), 7.94-7.96 (d, J= 8.8 Hz, 2H), 8.47-8.49 (m, 2H). Example 97. Synthesis of N-amino-N'-^-chloro-S-trifluoronietliyl-plienyl-guanidine hydroiodide
Figure imgf000126_0001
[0322] A mixture of 2.54 g 4-chloro-3-trifluoromethyl-phenylthiourea, 0.62 ml of iodomethane in 50 mL of anhydrous EtOH was refluxed for 1 hr to give l-[4-chloro-3- (trifluoromethyl)phenyl]-S-methylisothiourea hydroiodide. Then it was cooled down to ambient temperature and treated with 0.35 g of 98 % hydrazine, heated gently with stirring until the initial vigorous evolution of MeSH subsided and then refluxed for additional 1 hour.
Example 98. Synthesis of 4-(3-(r4-chloro-3-("trifluoromethyl)phenyllamino>-l,2,4-triazin-5- ypphenol
Figure imgf000126_0002
[0323] N-amino-N'- [4-chloro-3 -(trifluoromethyl)phenyl] -guanidine hydroiodide can be reacted with about 1.0-1.5 equivalents of 4-hydroxy-phenylglyoxale in 1:1 mixture of methanol/water to yield the title product. The product can be isolated by precipitation or by extraction with a number of solvents, such as ethyl acetate, methylene chloride or diethyl ether or by silica gel column chromatography.
Example 99. Synthesis of 4-r4-(3-{r4-chloro-3-(trifluoromethyl)phenyl1amino|- r 1.Σ^Itriazin-S-ylVphenoxyi-N-methylpyridine^-carboxamide
Figure imgf000126_0003
[0324] 7.23 g (19.73 mmol) of 4-(3- {[4-chloro-3-(trifluoromethyl)phenyl]amino} -1 ,2,4- triazin-5-yl)phenol can be dissolved in 80 mL of anhydrous DMF under argon atmosphere. 2.44 g (21.71 mmol, 1.1 equivalent) of solid potassium tert-butoxide can be added to the solution. The resulting mixture can be heated to about 1000C and stirred at that temperature for 15 min. Then a solution of 3.7 g (21.71 mmol, 1.1 equivalent) of 4-chloro-pyridine-2- carboxylic acid methylamide in 10 mL of anhydrous DMF can be added, followed by 3.28 g (23.68 mmol, 1.2 equivalent) of anhydrous K2CO3. The reaction mixture can be heated at L 140 0C for 30 hrs. The progress of the reaction can be monitored by LC/MS. Then it can be allowed to cool down to ambient temperature. The resulting mixture can be poured into 500 mL of water and 100 mL of ethyl acetate. The aqueous layer can be extracted with a number of solvents, such as ethyl acetate, methylene chloride or ether. The combined extracts can be washed 3 times with 100 mL of water, then with brine and dried over anhydrous sodium sulfate. Solvent can be removed in vacuum to yield crude 4-[4-(3-phenylamino- [l,2,4]triazin-6-yl)-phenoxy]-pyridine-2-carboxylic acid methylamide. The product can be than purified using silica gel column chromatography. Those having ordinary skill in the art can determine which solvent system can be used as an eluents in the chromatographic purification.
Example 100. Synthesis of 4-r3-ethvlsulfanyl-[l,2,41triazin-5-vlVphenol
Figure imgf000127_0001
[0325] 1.0-1.5 equivalent of 4-hydroxyphenylglyoxale can be reacted with 1-amino-S- ethylisothiourea hydrobromide in 1:1 mixture of methanol/water with 1.0-2.0 equivalent of K2CO3 to yield 4-(3-ethylsulfanyl-[l,2,4]triazin-5-yl)-phenol. The product can be isolated by a number of methods known to one skilled in the art, such as precipitation or by extraction with a number of solvents, such as ethyl acetate, methylene chloride or diethyl ether or by silica gel column chromatography.
Example 101. Synthesis of 4-("3-amino-ri.2.41triazin-5-ylVphenol
Figure imgf000127_0002
[0326] 1.0 equivalent of 4-(3-ethylsulfanyl-[l,2,4]triazin-6-yl)-phenol can be reacted with 1-5 equivalent of ammonia in dioxane to give 4-(3-amino-[l,2,4]triazin-5-yi)-phenol. The product can be isolated by a number of methods known to one skilled in the art, such as precipitation or by extraction with a number of solvents, such as ethyl acetate, methylene chloride or diethyl ether or by silica gel column chromatography. Example 102. Synthesis of 4-r4-(3-ammo-[L2,41triazin-5-ylVphenoxyl-N-methvfoyridine-2- carboxamide
Figure imgf000128_0001
[0327] 3.71 g (19.73 mmol) of 4-(3-amino-[l,2,4]triazin-5-yl)-phenol can be dissolved in 80 niL of anhydrous DMF under argon atmosphere. 2.44 g (21.71 mmol, 1.1 equivalent) of solid potassium tert-butoxide can be added to the solution. The resulting mixture can be heated to about 100 0C and stirred at that temperature for 15 min. Then a solution of 3.7 g (21.71 mmol, 1.1 equivalent) of 4-chloro-pyridine-2-carboxylic acid methylamide in 10 mL of anhydrous DMF can be added, followed by 3.28 g (23.68 mmol, 1.2 eq.) of anhydrous K2CO3. The reaction mixture can be heated at 140 0C for 30 hrs. The progress of the reaction can be monitored by LC/MS. Then it can be allowed to cool down to ambient temperature. The resulting mixture can be poured into 500 mL of water and 100 mL of ethyl acetate. The aqueous layer can be extracted with a number of solvents, such as ethyl acetate, methylene chloride or ether. The combined extracts can be washed 3 times with 100 mL of water, then with brine and dried over anhydrous sodium sulfate. Solvent can be removed in vacuum to give crude 4-[4-(3-amino-[l,2,4]triazin-5-yl)-phenoxy]-N-methylpyridine-2- carboxamide. The product can be than purified using silica gel column chromatography. Those having ordinary skill in the art can determine which solvent system can be used as an eluents in the chromatographic purification.
Example 103. Synthesis of 4-[4-(3-ir4-chloro-3-(trifluoromethyl)benzoyl1amino>- [L2.41triazin-5-yl)-phenoxyl-N-methylpyridine-2-carboxamide
Figure imgf000128_0002
[0328] 108.6 mg (0.337 mmol, 1.0 eq) of 4-[4-(3-amino-[l,2,4]triazin-5-yl)~ρhenoxy]-N- methylpyridine-2-carboxamide can be dissolved in 2 mL of anhydrous DMF with heating to about 1000C. 45.4 mg (0.405 mmol, 1.2 equivalent) of solid ^-BuOK can be added to the solution, followed by 0.405 mmol (1.2 equivalent) of 4-chloro-3-trifluoromethylbenzoyl chloride. It can be allowed to stir at ambient temperature for 1-2 hours. The product can be isolated by a number of methods known to those skilled in the art, such as precipitation or by extraction with a number of solvents, such as ethyl acetate, methylene chloride or diethyl ether or by silica gel column chromatography, or by reverse-phase prep-HPLC chromatography.
Example 104. Synthesis of 4-{4-r3-("{r4-chloro-3-(trifluoromethyl)phenyllsulfonyl>ammoV r 1 ,2,41triazin-5-vlVphenoxv> -N-methylpyridine-2-carboxamide
Figure imgf000129_0001
[0329] 108.6 mg (0.337 mmol, 1.0 equivalent) of 4-[4-(3-amino-[l,2,4]triazin-5-yl)- phenoxy] -N-methylpyridine-2-carboxamide can be dissolved in 2 niL of anhydrous pyridine with heating to about 100 0C. 0.405 mmol (1.2 equivalent) bf 4-chloro-3-trifluoromethyl- benzenesulfonyl chloride can be added. The reaction mixture can be allowed to stir at ambient temperature for 1-2 hours. The product can be isolated by a number of methods known to one skilled in the art, such as precipitation or by extraction with a number of solvents, such as ethyl acetate, methylene chloride or diethyl ether or by silica gel column chromatography, or by reverse-phase preparative HPLC.
Example 105. Testing of Inhibition of MAPK Pathway in Cellular Assay
[0330] Some compounds described by the general structure (B) were tested for inhibition of MAPK pathway in cellular assay. Western Blot: Early passage primary human umbilical vein endothelial cells (HUVECs) were maintained in EGM-2 containing SingleQuots (Cambrex, East Rutherford, NJ), 10% FBS, 1OmM HEPES, and 50 μg/ml gentamicin. Prior to treatment of the cells with inhibitor, the HUVECs were starved for 18h by replacing serum-containing complete media with serum-free and SingleQuot-free media. The starved cells were pre-treated with inhibitors for 60 min at various concentrations (0-20 μM). Next the HUVECs were treated with 50 ng/ml VEGF or FGF (Peprotech, Rocky Hill, NJ) for 6 min and the cells were immediately washed with ice-cold PBS. Cells were lysed with ice- cold RIPA buffer containing 10OmM Tris pH 7.5, 150 mM NaCl, 1 mM EDTA, 1% deoxycholate, 1% Triton X-100, 0.1% SDS, 2 mM PMSF, one Complete-Mini protease inhibitor tablet (Roche, Indianapolis, IN; 1 tablet/ 7 ml of lysis buffer) and the phophatase inhibitors NaF (500 mM) and orthovanadate (1 mM). The cells were scraped and lysates transferred and centrifuged at 15,000 g for 10 min. Supernatants were transferred to new tubes and protein concentration was quantitated using the BCA protein reagent (Pierce, Rockford, IL). Cell lysates containing 20 μg of total protein were separated by 10% SDS- PAGE, transferred to nitrocellulose, and blocked in 5% milk in TBST. Anti phospho-ERK Thr 202/Tyr 204 (Cell Signaling, Beverly, MA), anti-phospho-MEK Ser217/221 (Cell Signaling), and c-Raf (BD Biosciences Pharmingen, San Diego, CA) used as primary antibodies were detected with horseradish peroxidase-conjugated goat anti-mouse or rabbit secondary antibodies and bands were visualized using the SuperSignal West Pico chemiluminescence reagent system (Pierce) and Kodak X-ray film (Rochester, NY).
[0331] Bay 43-9006 (Raf/FGF inhibitor) showed reduction of expression of p-MEK and p-ERK with IC50 between 200 and 300 nM when tested in this assay. U0126 (MEK inhibitor) showed reduction in p-Erk levels with IC50 between 200 and 300 nM, while p- MEK levels were unaffected. The results are shown in Table 1. As can be seen, compounds of the invention showed reduction in p-MEK and p-ERK levels with IC50 between 400 nM and 20 μM.
Example 106. Cell Viability Assay
[0332] Some compounds described by the general structure (B) were tested for cell viability. XTT assay: HUVECs were seeded at 10,000 cells/well of a tissue culture treated 96-well plate treated with collagen type I and grown overnight in the complete EGM-2 media as described above. The following morning, the inhibitors were serial diluted with DMSO and added to the cells with a final DMSO concentration of 1%. After 72 hours cell viability was measured with an XTT assay (Sigma, St. Louis, MO). The cells were also photographed to compare morphological differences to the XTT trends observed. Determination of the IC50 values was performed with quantitative software (Prism software package, GraphPad Software, San Diego CA). Several inhibitors blocked cell proliferation and induced apoptosis at concentrations below 1 μM and experiments were repeated three times to confirm the observations. The compounds of the invention displayed IC50 between 100 nM and 40 uM in this assay (Table 2). Table 2. Test Results for Examples 105 and 106
Figure imgf000131_0001
Figure imgf000132_0001
Figure imgf000133_0001
L.
Figure imgf000134_0001
[0333] Although the invention has been described with reference to the above examples, it will be understood that modifications and variations are encompassed within the spirit and scope of the invention. Accordingly, the invention is limited only by the following claims.

Claims

iu ft y 1W • ■■ •••»» - • >• »».
WHAT IS CLAIMED IS:
1. A compound having the structure (A) or an N-oxide, N,N'-dioxide, N,N',N"- trioxide, or a pharmaceutically acceptable salt thereof:
Figure imgf000135_0001
(A) wherein:
Y is absent or is a moiety selected from a group consisting of:
Figure imgf000135_0002
Ri is a substituent selected from a group consisting of an aryl, a substituted aryl, a heterocycle, a heteroaryl, a substituted heterocycle, and a substituted heteroaryl;
R2 is a substituent selected from a group consisting of hydrogen, hydroxyl, Ci-C6 alkyl, C1-C6 alkylamino, halogen, Ci-Ce alkoxy, -NO2, -NH2, and -C≡N; and
R3 is a substituent selected from a group consisting of an aryl, a substituted aryl, a heterocycle, a heteroaryl, a substituted heterocycle, and a substituted heteroaryl.
2. The compound of claim 1, wherein:
Ri is selected from a group consisting Of C6-Ci2 aryl; C3-Ci2 heteroaryl having 1-3 heteroatoms selected from N, S and O, substituted C3-CI0 cycloalkyl having 0-3 heteroatoms selected from N, S, and O, substituted C6-Ci2 aryl, substituted C3-Ci2 heteroaryl having 1-3 heteroatoms selected from N, S and O, C7-C24 aralkyl; C7-C24 alkylaryl; substituted C7-C24 aralkyl; and substituted C7-C24 alkaryl;
R3 is selected from a group consisting Of C6-Ci2 aryl; C3-Ci2 heteroaryl having 1-3 heteroatoms selected from N, S and O, substituted C3-CiO cycloalkyl having 0-3 heteroatoms selected from N, S, and O, substituted C6-Ci2 aryl, substituted C3-Ci2 heteroaryl having 1-3 heteroatoms selected from N, S and O, C7-C24 aralkyl; C7-C24 alkylaryl; substituted C7-C24 aralkyl; and substituted C7-C24 alkaryl.
3. The compound of claim 1, wherein the compound having the structure (A) is selected from the group of compounds having the structures (Al), (A2), (A3), and (A4):
Figure imgf000136_0001
4. The compound of claim 3, wherein the compound has the structure (Al) and wherein:
Ri is selected from a group consisting of
Figure imgf000137_0001
Figure imgf000137_0002
Figure imgf000137_0003
Figure imgf000137_0004
Figure imgf000138_0001
Figure imgf000139_0001
R3 is selected from a group consisting of:
Figure imgf000140_0001
140
Figure imgf000141_0001
141
Figure imgf000142_0001
Figure imgf000143_0001
n is an integer selected from a group consisting of 0, 1, 2, and 3.
5. The compound of claim 3, wherein the compound having the structure (Al) is selected from a group consisting of compounds having structures (1) and (2):
Figure imgf000143_0002
(1)
Figure imgf000143_0003
(2)
6. The compound of claim 3, wherein the compound having structure (Al) is selected from a group consisting of compounds having the structures (3), (4), (5) and (6):
Figure imgf000143_0004
(3) (4)
Figure imgf000143_0005
7. The compound of claim 3, wherein the compound having structure (Al) is selected from a group consisting of compounds having the structures (7)-(16):
Figure imgf000144_0001
(7) (8)
Figure imgf000144_0002
(10) (H)
Figure imgf000144_0003
(14) (15)
Figure imgf000144_0004
(16) The compound of claim 7, wherein the compound has the formula:
Figure imgf000144_0005
9. The compound of claim 7, wherein the compound has the formula:
Figure imgf000145_0001
10. The compound of claim 7, wherein the compound has the formula:
Figure imgf000145_0002
11. The compound of claim 7, wherein the compound has the formula:
Figure imgf000145_0003
YL. The compound of claim 7, wherein the compound has the formula:
Figure imgf000145_0004
13. The compound of claim 7, wherein the compound has the formula:
Figure imgf000145_0005
14. The compound of claim 7, wherein the compound has the formula:
Figure imgf000145_0006
15. The compound of claim 7, wherein the compound has the formula:
Figure imgf000145_0007
16. The compound of claim 7, wherein the compound has the formula:
Figure imgf000146_0001
17. The compound of claim 7, wherein the compound has the formula:
Figure imgf000146_0002
18. A compound having the structure (B) or an N-oxide, N,N'-dioxide, N5N' ,N"- trioxide, or a pharmaceutically acceptable salt thereof:
Figure imgf000146_0003
(B)
wherein:
each Of Z1, Z2 and Z3 is independently selected from a group consisting of N, CH, N=CH, O, S and N-R4, wherein R4 is hydrogen or lower alkyl, with the further proviso that at least one Of Z1, Z2 and Z3 is not CH;
X is absent or is NH; and
Y is absent or is selected from a group consisting of the following moieties:
Figure imgf000146_0004
R1 is an unsubstituted or a substituted C3-C12 heteroaryl having 1-3 heteroatoms;
R2 is selected from a group consisting of of hydrogen, halogen, C1-C18 alkyl, -OH, -NO2, -CN, C1-C18 alkoxy, -NHSO2R5, -SO2NHR5, -NHCOR5, -NH2, -NR5R6 , -S(O)R5, -S(O)2R5, -CO2R5, -CONR5R6, wherein R5 and R6 are independently selected from a group consisting of hydrogen, a C1-C18 alkyl, and a substituted C1-C12 alkyl; and R3 is selected from a group consisting of hydrogen, a Ci-C18 alkyl, a substituted C1- Ci2 alkyl, a Ci-Ci2 cycloalkyl, a substituted Ci-Ci2 cycloalkyl, a substituted C3-Ci0 cycloalkyl having 0-3 heteroatoms, an C6-C12 aryl, a substituted C6-Ci2 aryl, a heterocycle, a substituted heterocycle, a C3-Ci2 heteroaryl having 1-3 heteroatoms, a substituted C3-Ci2 heteroaryl having 1-3 heteroatoms, a C7-C24 aralkyl, a substituted C7-C24 aralkyl, a C7-C24 alkylaryl, and a substituted C7-C24 alkaryl.
19. The compound of claim 18, wherein Ri is selected from the group consisting of:
Figure imgf000147_0001
Figure imgf000148_0001
R3 is selected from the group consisting of:
Figure imgf000149_0001
Figure imgf000150_0001
Figure imgf000151_0001
n is an integer selected from a group consisting of 0, 1, 2, and 3, and
R' is selected from a group consisting of hydrogen, a C1-Ci8 alkyl, and a substituted Ci-Ci8 alkyl.
20. The compound of claim 18, wherein the compound having structure (B) is selected from a group consisting of compounds having formulae (17)-(35):
Figure imgf000151_0002
(17)
Figure imgf000152_0001
(18)
Figure imgf000152_0002
(19)
Figure imgf000152_0003
(20)
Figure imgf000152_0004
(22)
Figure imgf000152_0005
(23)
Figure imgf000152_0006
(24)
Figure imgf000153_0001
(25)
Figure imgf000153_0002
(26)
Figure imgf000153_0003
(27)
Figure imgf000153_0004
(28)
Figure imgf000153_0005
(29)
Figure imgf000153_0006
(30)
Figure imgf000154_0001
(31)
Figure imgf000154_0002
(32)
Figure imgf000154_0003
(33)
Figure imgf000154_0004
(34)
Figure imgf000154_0005
(35)
21. The compound of claim 20, wherein the compound has the formula:
Figure imgf000154_0006
22. The compound of claim 20, wherein the compound has the formula:
Figure imgf000154_0007
23. The compound of claim 20, wherein the compound has the formula:
Figure imgf000155_0001
24. The compound of claim 20, wherein the compound has the formula:
Figure imgf000155_0002
25. The compound of claim 20, wherein the compound has the formula:
Figure imgf000155_0003
26. The compound of claim 20, wherein the compound has the formula:
Figure imgf000155_0004
27. The compound of claim 20, wherein the compound has the formula:
Figure imgf000155_0005
28. The compound of claim 20, wherein the compound has the formula:
Figure imgf000155_0006
29. The compound of claim 20, wherein the compound has the formula:
Figure imgf000155_0007
30. The compound of claim 20, wherein the compound has the formula:
Figure imgf000156_0001
31. The compound of claim 20, wherein the compound has the formula:
Figure imgf000156_0002
32. The compound of claim 20, wherein the compound has the formula:
Figure imgf000156_0003
33. The compound of claim 20, wherein the compound has the formula:
Figure imgf000156_0004
34. The compound of claim 20, wherein the compound has the formula:
Figure imgf000156_0005
35. The compound of claim 20, wherein the compound has the formula:
Figure imgf000156_0006
36. The compound of claim 20, wherein the compound has the formula:
Figure imgf000157_0001
37. The compound of claim 20, wherein the compound has the formula:
Figure imgf000157_0002
38. The compound of claim 20, wherein the compound has the formula:
Figure imgf000157_0003
39. The compound of claim 20, wherein the compound has the formula:
Figure imgf000157_0004
40. A compound comprising a derivative of benzotriazine, the compound including a benzotriazine moiety having at least a first substituent attached to the benzene ring of benzotriazine and a second substitutent attached to the triazine ring of the benzotriazine, wherein:
(a) the first substitutent includes a substituted phenyl, substituted pyridyl or substituted pyrimidyl group; and
(b) the second substitutent is selected from a secondary amino group, a substituted amide group, and a substituted sulfonylamino group.
41. The compound of claim 40, wherein the substitutent in the substituted pyridyl group comprises an amido moiety, an aminoalkyl group, carboxyl group or a carboxylate group. 42. The compound of claim 41 , wherein the nitrogen in the amido moiety additionally carries a third substitutent selected from an alkyl, an alkylaminoalkyl, a pyridyl, an alkyl pyrrolidine, an alkyl morpholine, and an alkyl piperazine groups.
43. The compound of claim 40, wherein the second substitutent includes a group derived from a compound selected from benzene, pyridine, thiophene, and isoxazole.
44. The compound of claim 43, wherein the group derived from benzene is selected from tert-butyl phenyl, trifluoromethoxyphenyl, methoxyphenyl, dimethylamino, dimethylaminophenyl, aminophenyl, trifluoroethoxyphenyl, trifluoromethoxychlorophenyl, trifluoromethoxybromophenyl, trifluoroethoxychlorophenyl, chlorophenyl, dichlorophenyl, trifluoromethyl phenyl, trifluoromethylchloro phenyl, chlorotoluyl, N-phenylacetamide, N9N- alkyl-benzamide, isopropoxyphenyl, alkoxyphenyl, dialkoxyphenyl, acetylphenyl.
45. A compound comprising a benzene derived moiety bridged to a heterocyclic moiety, wherein:
(a) the benzene-derived moiety includes a molecule of benzene having a substitutent selected from:
a pyridyl group connected to the benzene molecule via an oxygen link, and
a sulfonyl group; and
(b) the heterocyclic moiety is selected from triazine, triazole, oxadiazole, pyridazine, pyrimidine, pyridine, oxazole, pyrazol, pyrrole, imidazole, thiadiazole.
46. The compound of claim,45, wherein the pyridyl group includes a first substitutent comprising an amido moiety, an alkylamino group, or a carboxyl group.
47. The compound of claim 46, wherein the nitrogen in the amido moiety additionally carries a second substituent selected from an alkyl, an alkylamino alkyl, a pyridyl, an alkyl pyrrolidine, an alkyl morpholine, and an alkyl piperazine groups.
48. The compound of claim 45, wherein the heterocyclic moiety optionally includes a third substituent connected to the heterocyclic moiety, wherein the third substituent is selected from a secondary amino group, a substituted amide group, and a substituted sulfonylamino group. 49. The compound of claim 48, wherein the third substitutent includes a group derived from a compound selected from benzene, thiophene, and isoxazole.
50. The compound of claim 49, wherein the group derived from benzene is selected from tert-butyl phenyl, trifluoromethoxyphenyl, methoxyphenyl, dimethylamino, dimethylaminophenyl, aminophenyl, trifluoroethoxyphenyl, trifluoromethoxychlorophenyl, trifluoromethoxybromophenyl, trifluoroethoxychlorophenyl, chlorophenyl, dichlorophenyl, trifluoromethyl phenyl, trifluoromethylchloro phenyl, chlorotoluyl, N-phenylacetamide, N,N- alkyl-benzamide, isopropoxyphenyl, alkoxyphenyl,dialkoxyphenyl, acetylphenyl.
51. The compound of claim 45, wherein the bridge between the benzene derived moiety and the heterocyclic moiety includes a single bond or a nitrogen atom.
52. A method for treating a disorder comprising administering to a subject in need thereof an effective amount of a compound, wherein the compound is set forth in Structures (A), (B) or a combination thereof.
53. The method of claim 52, wherein the disorder is selected from a group consisting of cancer, eye disease, inflammation, psoriasis, and a viral infection.
54. The method of claim 53, wherein the cancer is an alimentary/gastrointestinal tract cancer, colon cancer, liver cancer, skin cancer, breast cancer, ovarian cancer, prostate cancer, lymphoma, leukemia, kidney cancer, lung cancer, muscle cancer, bone cancer, bladder cancer or brain cancer.
55. The method of claim 52, wherein the disorder is associated with a kinase.
56. The method of claim 55, wherein disorder is associated with a MAPK kinase pathway.
57. A method for treating a disorder, comprising administering to a subject in need thereof an effective amount of a compound having the structure (A):
Figure imgf000159_0001
(A) wherein:
Y is absent or is a moiety selected from a group consisting of:
Figure imgf000160_0001
R1 is a substituent selected from a group consisting of an aryl, a substituted aryl, a heterocycle, a heteroaryl, a substituted heterocycle, and a substituted heteroaryl;
R2 is a substituent selected from a group consisting of hydrogen, hydroxyl, C1-C6 alkyl, C1-C6 alkylamino, halogen, Ci-C6 alkoxy, -NO2, -NH2, and -C≡N; and
R3 is a substituent selected from a group consisting of an aryl, a substituted aryl, a heterocycle, a heteroaryl, a substituted heterocycle, and a substituted heteroaryl.
58. The method of claim 57, wherein:
Ri is selected from a group consisting of C6-Ci2 aryl; C3-Ci2heteroaryl having 1-3 heteroatoms selected from N, S and O, substituted C3-CiO cycloalkyl having 0-3 heteroatoms selected from N, S, and O, substituted C6-Ci2 aryl, substituted C3-Ci2 heteroaryl having 1-3 heteroatoms selected from N, S and O, C7-C24 aralkyl; C7-C24 alkylaryl; substituted C7-C24 aralkyl; and substituted C7-C24 alkaryl;
R3 is selected from a group consisting of C6-Ci2 aryl; C3-Ci2 heteroaryl having 1-3 heteroatoms selected from N, S and O, substituted C3-CiO cycloalkyl having 0-3 heteroatoms selected from N, S, and O, substituted C6-Ci2 aryl, substituted C3-Ci2 heteroaryl having 1-3 heteroatoms selected from N, S and O, C7-C24 aralkyl; C7-C24 alkylaryl; substituted C7-C24 aralkyl; and substituted C7-C24 alkaryl.
59. The method of claim 57, wherein the compound having the structure (A) is selected from the group of compounds having the structures (Al), (A2), (A3), and (A4):
Figure imgf000160_0002
Figure imgf000161_0001
(A4) 60. The method of claim 57, wherein: R1 is selected from a group consisting of
Figure imgf000161_0002
Figure imgf000162_0001
Figure imgf000163_0001
R3 is selected from a group consisting of:
Figure imgf000163_0002
Figure imgf000164_0001
164
Figure imgf000165_0001
ı65
Figure imgf000166_0001
Figure imgf000167_0001
n is an integer selected from a group consisting of 0, 1, 2, and 3.
61. The method of claim 59, wherein the compound having the structure (Al) is selected from a group consisting of compounds having structures (1) and (2):
Figure imgf000167_0002
(1)
Figure imgf000167_0003
(2)
62. The method of claim 59, wherein the compound having structure (Al) is selected from a group consisting of compounds having the structures (3), (4), (5) and (6):
Figure imgf000167_0004
(3) (4)
Figure imgf000167_0005
(5) (6) 14
167
63. The method of claim 62, wherein the compound having structure (Al) is selected from a group consisting of compounds having the structures (7)-(16):
Figure imgf000168_0001
(7) (8)
Figure imgf000168_0002
(14) (15)
Figure imgf000168_0003
(16)
64. A method for treating a disorder, comprising administering to a subject in need thereof an effective amount of a compound having the structure (B):
Figure imgf000169_0001
(B)
wherein:
each Of Z1, Z2 and Z3 is independently selected from a group consisting of N, CH, N=CH, O, S and N-R4, wherein R4 is hydrogen or lower alkyl, with the further proviso that at least one of Z1, Z2 and Z3 is not CH;
X is absent or is NH; and
Y is absent or is selected from a group consisting of the following moieties:
Figure imgf000169_0002
R1 is an unsubstituted or a substituted C3-Ci2heteroaryl having 1-3 heteroatoms;
R2 is selected from a group consisting of of hydrogen, halogen, C1-C18 alkyl, -OH, -NO2, -CN, C1-C18 alkoxy, -NHSO2R5, -SO2NHR5, -NHCOR5, -NH2, -NR5R6 , -S(O)R5, -S(O)2R5, -CO2R5, -CONR5R6, wherein R5 and R6 are independently selected from a group consisting of hydrogen, a C1-Ci8 alkyl, and a substituted CpC12 alkyl; and
R3 is selected from a group consisting of hydrogen, a C1-C18 alkyl, a substituted C1- Ci2 alkyl, a C1-Ci2 cycloalkyl, a substituted Ci-Ci2 cycloalkyl, a substituted C3-CiO cycloalkyl having 0-3 heteroatoms, an C6-Ci2 aryl, a substituted C6-Ci2 aryl, a heterocycle, a substituted heterocycle, a C3-C12 heteroaryl having 1-3 heteroatoms, a substituted C3-Ci2 heteroaryl having 1-3 heteroatoms, a C7-C24 aralkyl, a substituted C7-C24 aralkyl, a C7-C24 alkylaryl, and a substituted C7-C24 alkaryl.
65 The method of claim 64, wherein Ri is selected from the group consisting of:
Figure imgf000170_0001
Figure imgf000171_0001
Figure imgf000172_0001
Figure imgf000173_0001
Figure imgf000174_0001
n is an integer selected from a group consisting of 0, 1, 2, and 3, and
R' is selected from a group consisting of hydrogen, a C1-C18 alkyl, and a substituted C1-C18 alkyl.
66. The method of claim 64, wherein the compound having structure (B) is selected from a group consisting of compounds having formulae (17)-(35):
Figure imgf000174_0002
(17)
Figure imgf000175_0001
(18)
Figure imgf000175_0002
(19)
Figure imgf000175_0003
(20)
Figure imgf000175_0004
(22)
Figure imgf000175_0005
(23)
Figure imgf000175_0006
(24)
Figure imgf000176_0001
(25)
Figure imgf000176_0002
(26)
Figure imgf000176_0003
(27)
Figure imgf000176_0004
(28)
Figure imgf000176_0005
(29)
Figure imgf000176_0006
(30)
Figure imgf000177_0001
(31)
Figure imgf000177_0002
(32)
Figure imgf000177_0003
(33)
Figure imgf000177_0004
(34)
Figure imgf000177_0005
(35)
67. The method of claim 57 or 64, wherein the disorder is selected from a group consisting of cancer, eye disease, inflammation, psoriasis, and a viral infection.
68. The method of claim 67, wherein the cancer is an alimentary/gastrointestinal tract cancer, colon cancer, liver cancer, skin cancer, breast cancer, ovarian cancer, prostate US2005/030614
177
cancer, lymphoma, leukemia, kidney cancer, lung cancer, muscle cancer, bone cancer, bladder cancer or brain cancer.
69. A pharmaceutical composition comprising a compound set forth in structures (A), (B) or any combination thereof, in a pharmaceutically acceptable carrier.
70. An article of manufacture comprising packaging material and a pharmaceutical composition contained within the packaging material, wherein the packaging material comprises a label which indicates that the pharmaceutical composition can be used for treatment of disorders and wherein said pharmaceutical composition comprises a compound set forth in structures (A), (B) or any combination thereof.
71. An article of manufacture comprising packaging material and a pharmaceutical composition contained within said packaging material, wherein said packaging material comprises a label which indicates that said pharmaceutical composition can be used for treatment of cancer and wherein said pharmaceutical composition comprises a compound set forth in structures (A), (B) or any combination thereof.
72. A method of treating a disorder, comprising the administration of a therapeutically effective amount of at least one compound set forth in structures (A), (B) or any combination thereof, or pharmaceutically acceptable salts, hydrates, solvates, crystal forms and individual diastereomers thereof, to a subject in need of such treatment.
73. The method of claim 72, wherein the disorder is selected from a group consisting of cancer, eye disease, inflammation, psoriasis, and a viral infection.
74. The method of claim 73, wherein the cancer is an alimentary/gastrointestinal tract cancer, colon cancer, liver cancer, skin cancer, breast cancer, ovarian cancer, prostate cancer, lymphoma, leukemia, kidney cancer, lung cancer, muscle cancer, bone cancer, bladder cancer or brain cancer.
75. A method of treating a disorder comprising the administration of a therapeutically effective amount of at least one compound as set forth in structures (A), (B) or any combination thereof, or pharmaceutically acceptable salts, hydrates, solvates, crystal forms and individual diastereomers thereof, in combination with a chemotherapeutic agent, immunomodulatory agent, therapeutic antibody or a protein kinase inhibitor, to a subject in need of such treatment. 76. A method of treating a subject having cancer comprising administering to the subject a therapeutically effective amount of a compound as set forth in structures (A), (B) or any combination thereof, thereby treating the subject.
77. A process for making a pharmaceutical composition comprising combining a combination of a compound set forth in structures (A), (B) or any combination thereof or its pharmaceutically acceptable salts, hydrates, solvates, crystal forms salts and individual diastereomers thereof, and a pharmaceutically acceptable carrier.
78. A pharmaceutical composition comprising a compound as set forth in Structure (Al) in a pharmaceutically acceptable carrier.
79. A pharmaceutical composition comprising a compound as set forth in Structure (A2) in a pharmaceutically acceptable carrier.
80. A pharmaceutical composition comprising a compound as set forth in Structure (A3) in a pharmaceutically acceptable carrier.
81. A pharmaceutical composition comprising a compound as set forth in Structure (A4) in a pharmaceutically acceptable carrier.
82. A pharmaceutical composition comprising a compound as set forth in Structure (B) in a pharmaceutically acceptable carrier.
83. A method for treating cancer or a tumor in a subject, comprising administering to a subject in need thereof an effective amount of a therapeutic antibody, chemotherapeutic agent or imrnunotoxic agents, in combination with a compound set forth in Structures (A), (B) or any combination thereof, thereby treating the cancer or tumor in the subject.
84. A pharmaceutical composition comprising a therapeutic agent and at least one compound as set forth in Structures (Al), (A2), (A3), (A4) or any combination thereof, in a concentration effective to treat cancer in a subject.
85. The composition of claim 84, wherein the cancer is an alimentary/gastrointestinal tract cancer, colon cancer, liver cancer, skin cancer, breast cancer, ovarian cancer, prostate cancer, lymphoma, leukemia, kidney cancer, lung cancer, muscle cancer, bone cancer, bladder cancer or brain cancer. 86. The composition of claim 85, wherein the cancer is colon cancer or lung cancer.
87. The method of claim 84, wherein the therapeutic agent is an antimetabolite; a DNA cross-linking agent; alkylating agent; topoisomerase I inhibitor; microtubule inhibitors, a vinca alkaloid, mitomycin-type antibiotic, and a bleomycin-type antibiotic.
88. The method of claim 84, wherein the therapeutic agent is methotrexate, cisplatin/carboplatin; canbusil; dactinomicin; taxol (paclitaxel), antifolate, colchicine, demecoline, etoposide, taxane/taxol, docetaxel, doxorubicin, anthracycline antibiotic, doxorubicin, daunorubicin, carminomycin, epirubicin, idarubicin, mithoxanthrone, 4- dimethoxy-daunomycin, 11-deoxydaunorubicin, 13-deoxydaunorubicin, adriamycin-14- benzoate, adriamycin-14-octanoate or adriamycin-14-naphthaleneacetate, irinotecan, topotecan, gemcitabine, 5-fluorouracil, leucovorin carboplatin, cisplatin, taxanes, tezacitabine, cyclophosphamide, vinca alkaloids, imatinib, anthracyclines, rituximab, trastuzumab.
89. The method of claim 88, wherein the therapeutic agent is doxorubicin, docetaxol, or taxol.
90. The method of claim 84, wherein the therapeutic agent is trastuzumab, bevacizumab, OSI-774, or Vitaxin.
PCT/US2005/030614 2004-08-25 2005-08-24 Heterocyclic compounds and methods of use WO2006024034A1 (en)

Priority Applications (8)

Application Number Priority Date Filing Date Title
MX2007002208A MX2007002208A (en) 2004-08-25 2005-08-24 Heterocyclic compounds and methods of use.
CA002578283A CA2578283A1 (en) 2004-08-25 2005-08-24 Heterocyclic compounds and methods of use
EP05792421A EP1799656A4 (en) 2004-08-25 2005-08-24 Heterocyclic compounds and methods of use
JP2007530214A JP5275628B2 (en) 2004-08-25 2005-08-24 Heterocyclic compounds and methods of use
AU2005276974A AU2005276974B2 (en) 2004-08-25 2005-08-24 Heterocyclic compounds and methods of use
RU2007110934/04A RU2468021C2 (en) 2004-08-25 2005-08-24 Heterocyclic compounds and their application
NZ553492A NZ553492A (en) 2004-08-25 2005-08-24 Triazole derivatives and methods of use
IL181524A IL181524A (en) 2004-08-25 2007-02-22 Heterocyclic compounds, pharmaceutical compositions containing them and their use in the manufacture of medicaments

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US60429804P 2004-08-25 2004-08-25
US60/604,298 2004-08-25
US69616805P 2005-07-01 2005-07-01
US60/696,168 2005-07-01

Publications (1)

Publication Number Publication Date
WO2006024034A1 true WO2006024034A1 (en) 2006-03-02

Family

ID=35967888

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2005/030614 WO2006024034A1 (en) 2004-08-25 2005-08-24 Heterocyclic compounds and methods of use

Country Status (11)

Country Link
US (4) US7652051B2 (en)
EP (2) EP2532653A1 (en)
JP (2) JP5275628B2 (en)
AU (1) AU2005276974B2 (en)
CA (1) CA2578283A1 (en)
IL (1) IL181524A (en)
MX (1) MX2007002208A (en)
NZ (2) NZ588896A (en)
PL (1) PL382308A1 (en)
RU (1) RU2468021C2 (en)
WO (1) WO2006024034A1 (en)

Cited By (23)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008059259A2 (en) * 2006-11-16 2008-05-22 Sentinel Oncology Limited Anticancer compounds with a benzo ( 1, 2, 4 ) triazin-3yl-amine core
EP2141164A1 (en) * 2008-07-01 2010-01-06 Mutabilis New 1,2,4-triazine derivatives and biological applications thereof
WO2010020305A1 (en) 2008-08-18 2010-02-25 Merck Patent Gmbh Oxadiazole derivatives for treating diabetes
DE102008038222A1 (en) 2008-08-18 2010-02-25 Merck Patent Gmbh Indazol-5-carboxylic acid derivatives
WO2010068738A1 (en) 2008-12-10 2010-06-17 Dana-Farber Cancer Institute, Inc. Mek mutations conferring resistance to mek inhibitors
WO2010076238A1 (en) 2008-12-29 2010-07-08 Fovea Pharmaceuticals Sa Substituted quinazoline compounds
WO2010092041A1 (en) 2009-02-13 2010-08-19 Fovea Pharmaceuticals Sa [1, 2, 4] triazolo [1, 5 -a] pyridines as kinase inhibitors
WO2011106298A1 (en) 2010-02-25 2011-09-01 Dana-Farber Cancer Institute, Inc. Braf mutations conferring resistance to braf inhibitors
WO2011119894A2 (en) 2010-03-24 2011-09-29 Kinagen, Inc Heterocyclic compounds useful for kinase inhibition
US8084618B2 (en) 2004-08-25 2011-12-27 Targegen, Inc. Heterocyclic compounds and methods of use
WO2011161159A1 (en) 2010-06-22 2011-12-29 Fovea Pharmaceuticals Heterocyclic compounds, their preparation and their therapeutic application
US8110687B2 (en) 2005-12-08 2012-02-07 Millennium Pharmaceuticals, Inc. Bicyclic compounds with kinase inhibitory activity
EP2534145A2 (en) * 2010-02-08 2012-12-19 Kinagen, Inc. Therapeutic methods and compositions involving allosteric kinase inhibition
US8481536B2 (en) 2004-04-08 2013-07-09 Targegen, Inc. Benzotriazine inhibitors of kinases
WO2013169858A1 (en) 2012-05-08 2013-11-14 The Broad Institute, Inc. Diagnostic and treatment methods in patients having or at risk of developing resistance to cancer therapy
WO2014181287A1 (en) * 2013-05-09 2014-11-13 Piramal Enterprises Limited Heterocyclyl compounds and uses thereof
WO2019164794A1 (en) * 2018-02-20 2019-08-29 Agios Pharmaceuticals, Inc. Methods of use for trisubstituted benzotriazole derivatives
US11078540B2 (en) 2010-03-09 2021-08-03 Dana-Farber Cancer Institute, Inc. Methods of diagnosing and treating cancer in patients having or developing resistance to a first cancer therapy
US11147801B2 (en) 2017-04-24 2021-10-19 Aurigene Discovery Technologies Limited Methods of use for trisubstituted benzotriazole derivatives as dihydroorotate oxygenase inhibitors
US11180473B2 (en) 2020-03-27 2021-11-23 Landos Biopharma, Inc. PLXDC2 ligands
US11247987B2 (en) 2017-10-06 2022-02-15 Forma Therapeutics, Inc. Inhibiting ubiquitin specific peptidase 30
US11535618B2 (en) 2018-10-05 2022-12-27 Forma Therapeutics, Inc. Fused pyrrolines which act as ubiquitin-specific protease 30 (USP30) inhibitors
EP3958679A4 (en) * 2019-04-26 2023-08-16 Purdue Research Foundation Cf3-, ocf3-, scf3- and sf5-containing antibacterial agents

Families Citing this family (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20050056227A (en) * 2002-10-03 2005-06-14 탈자진 인코포레이티드 Vasculostatic agents and methods of use thereof
US20050282814A1 (en) * 2002-10-03 2005-12-22 Targegen, Inc. Vasculostatic agents and methods of use thereof
EP1893216A4 (en) * 2005-06-08 2012-08-08 Targegen Inc Methods and compositions for the treatment of ocular disorders
US8133900B2 (en) * 2005-11-01 2012-03-13 Targegen, Inc. Use of bi-aryl meta-pyrimidine inhibitors of kinases
RU2448959C2 (en) * 2005-11-01 2012-04-27 Таргеджен, Инк. Bi-aryl-metha-pyrimidine kinase inhibitors
US8604042B2 (en) * 2005-11-01 2013-12-10 Targegen, Inc. Bi-aryl meta-pyrimidine inhibitors of kinases
US20070161645A1 (en) * 2005-11-02 2007-07-12 Targegen, Inc. Thiazole inhibitors targeting resistant kinase mutations
US7919108B2 (en) * 2006-03-10 2011-04-05 Cook Incorporated Taxane coatings for implantable medical devices
US20080286325A1 (en) * 2006-01-05 2008-11-20 Med Institute, Inc. Cyclodextrin elution media for medical device coatings comprising a taxane therapeutic agent
US7875284B2 (en) * 2006-03-10 2011-01-25 Cook Incorporated Methods of manufacturing and modifying taxane coatings for implantable medical devices
US7691858B2 (en) * 2006-04-25 2010-04-06 Targegen, Inc. Kinase inhibitors and methods of use thereof
US8642067B2 (en) 2007-04-02 2014-02-04 Allergen, Inc. Methods and compositions for intraocular administration to treat ocular conditions
CA2723358A1 (en) * 2008-05-05 2009-11-12 Allison B. Reiss Method for improving cardiovascular risk profile of cox inhibitors
AU2010363329A1 (en) 2010-11-07 2013-05-09 Targegen, Inc. Compositions and methods for treating myelofibrosis
EP2739143B1 (en) * 2011-08-05 2018-07-11 Gary A. Flynn Preparation and methods of use for ortho-aryl 5- membered heteroaryl-carboxamide containing multi-targeted kinase inhibitors
US11629143B2 (en) 2020-10-01 2023-04-18 Vibliome Therapeutics, Llc HipK4 inhibitors and uses thereof

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4160833A (en) * 1973-02-02 1979-07-10 Ciba-Geigy Corporation 1,2,4-Benzotriazine-1,4-di-N-oxide derivatives
US4309211A (en) * 1979-08-31 1982-01-05 Ici Australia Limited Herbicidal 1,2,4-benzotriazines
US5849738A (en) * 1986-09-25 1998-12-15 Sri International 1,2,4-benzotriazine oxides as radiosensitizers and selective cytotoxic agents

Family Cites Families (154)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2003199A (en) 1930-05-31 1935-05-28 Johnson Frank James Automatic coal stoker
US2003149A (en) 1931-05-22 1935-05-28 Autographic Register Co Manifolding
US2003065A (en) 1931-06-20 1935-05-28 John R Ditmars Composition for coating sheets, fibrous stocks, and the like
US2004102A (en) 1932-02-24 1935-06-11 Daniel A Dickey Hollow steel propeller construction
US2004138A (en) 1932-11-30 1935-06-11 Byers A M Co Method of making wrought iron pipe
US2002165A (en) 1933-07-08 1935-05-21 Charles A Winslow Air cleaner
US2003187A (en) 1933-10-02 1935-05-28 Frederick H Shaw Automobile radio device
US2003166A (en) 1933-10-26 1935-05-28 Zancan Ottavio Front drive for motor cars
US2001051A (en) 1933-12-01 1935-05-14 Angelina Mariani Tamperproof meter box with cutout control and fuse drawer for electric meters
US2004092A (en) 1933-12-15 1935-06-11 John L Chaney Device for indicating the temperature of liquids
US2003060A (en) 1934-04-02 1935-05-28 Ernest L Heckert Thermostatic controlling device
US2667486A (en) 1951-05-24 1954-01-26 Research Corp 2,4-diamino pteridine and derivatives
IL26578A (en) 1965-10-04 1970-11-30 Merck & Co Inc Pteridine compounds and their preparation
DE2255947A1 (en) 1972-11-15 1974-05-22 Bayer Ag SUBSTITUTED 3-AMINO-BENZO-1,2,4TRIAZINE-DI-N-OXIDES (1,4), METHOD FOR MANUFACTURING AND USING THEM AS ANTIMICROBIAL AGENTS
FR2275461A1 (en) 1974-06-18 1976-01-16 Labaz NEW STABILIZERS FOR POLYMERS AND COPOLYMERS OF VINYL CHLORIDE
US4309112A (en) * 1979-05-15 1982-01-05 Sherwood Medical Industries Inc. Rate measurement analyzer
EP0059524A1 (en) 1981-02-09 1982-09-08 Smith and Nephew Associated Companies p.l.c. Pharmaceutical composition containing aminopteridines or aminopyrimido(4,5-d)pyrimidines
DE3205638A1 (en) 1982-02-17 1983-08-25 Hoechst Ag, 6230 Frankfurt Trisubstituted pyrimidine-5-carboxylic acids and their derivatives, processes for their preparation, and their use as pesticides
US4490289A (en) 1982-09-16 1984-12-25 Hoffmann-La Roche Inc. Homogeneous human interleukin 2
US5214059A (en) 1989-07-03 1993-05-25 Hoechst-Roussel Pharmaceuticals Incorporated 2-(aminoaryl) indoles and indolines as topical antiinflammatory agents for the treatment of skin disorders
US5665543A (en) 1989-07-18 1997-09-09 Oncogene Science, Inc. Method of discovering chemicals capable of functioning as gene expression modulators
US5776502A (en) 1989-07-18 1998-07-07 Oncogene Science, Inc. Methods of transcriptionally modulating gene expression
US5062685A (en) * 1989-10-11 1991-11-05 Corning Incorporated Coated optical fibers and cables and method
GB9003553D0 (en) 1990-02-16 1990-04-11 Ici Plc Herbicidal compositions
MY107955A (en) 1990-07-27 1996-07-15 Ici Plc Fungicides.
GB9016800D0 (en) 1990-07-31 1990-09-12 Shell Int Research Tetrahydropyrimidine derivatives
DE4025891A1 (en) 1990-08-16 1992-02-20 Bayer Ag PYRIMIDYL-SUBSTITUTED ACRYLIC ACID ESTERS
JPH05345780A (en) 1991-12-24 1993-12-27 Kumiai Chem Ind Co Ltd Pyrimidine or triazine derivative and herbicide
HUT63941A (en) 1992-05-15 1993-11-29 Hoechst Ag Process for producing 4-alkyl-substituted pyrimidine-5-carboxanilide derivatives, and fungicidal compositions comprising same
US5482951A (en) * 1992-05-29 1996-01-09 Kumiai Chemical Industry Co., Ltd. Triazole derivatives as well as insecticide and acaricide
US5763441A (en) 1992-11-13 1998-06-09 Sugen, Inc. Compounds for the treatment of disorders related to vasculogenesis and/or angiogenesis
DE4338704A1 (en) 1993-11-12 1995-05-18 Hoechst Ag Stabilized oligonucleotides and their use
US5530000A (en) 1993-12-22 1996-06-25 Ortho Pharmaceutical Corporation Substituted pyrimidinylaminothiazole derivatives useful as platelet aggreggation inhibitors
GB9506466D0 (en) 1994-08-26 1995-05-17 Prolifix Ltd Cell cycle regulated repressor and dna element
US5597826A (en) 1994-09-14 1997-01-28 Pfizer Inc. Compositions containing sertraline and a 5-HT1D receptor agonist or antagonist
DE19502912A1 (en) 1995-01-31 1996-08-01 Hoechst Ag G-Cap Stabilized Oligonucleotides
US6326487B1 (en) 1995-06-05 2001-12-04 Aventis Pharma Deutschland Gmbh 3 modified oligonucleotide derivatives
US5827850A (en) 1995-09-25 1998-10-27 Sanofi Pharmaceuticals, Inc. 1,2,4-benzotriazine oxides formulations
CA2232989C (en) 1995-09-25 2008-01-29 Sanofi Pharmaceuticals, Inc. 1,2,4-benzotriazine oxides formulations
EP0939632B1 (en) 1996-02-23 2005-10-05 Eli Lilly And Company NON-PEPTIDYL VASOPRESSIN V1a ANTAGONISTS
DE59707681D1 (en) 1996-10-28 2002-08-14 Rolic Ag Zug Crosslinkable, photoactive silane derivatives
EP1707631A3 (en) 1996-11-20 2006-12-27 Introgen Therapeutics, Inc. An improved method for the production and purification of adenoviral vectors
JP3734903B2 (en) 1996-11-21 2006-01-11 富士写真フイルム株式会社 Development processing method
JP3720931B2 (en) 1996-11-26 2005-11-30 富士写真フイルム株式会社 Processing method of silver halide photographic light-sensitive material
US5935383A (en) 1996-12-04 1999-08-10 Kimberly-Clark Worldwide, Inc. Method for improved wet strength paper
DE59807348D1 (en) 1997-02-05 2003-04-10 Rolic Ag Zug Photocrosslinkable silane derivatives
US6070126A (en) 1997-06-13 2000-05-30 William J. Kokolus Immunobiologically-active linear peptides and method of identification
CA2295106C (en) 1997-06-24 2007-03-13 Nikken Chemicals Co., Ltd. 3-anilino-2-cycloalkenone derivatives
US6635626B1 (en) 1997-08-25 2003-10-21 Bristol-Myers Squibb Co. Imidazoquinoxaline protein tyrosine kinase inhibitors
US6685938B1 (en) 1998-05-29 2004-02-03 The Scripps Research Institute Methods and compositions useful for modulation of angiogenesis and vascular permeability using SRC or Yes tyrosine kinases
US6136971A (en) 1998-07-17 2000-10-24 Roche Colorado Corporation Preparation of sulfonamides
US6378526B1 (en) 1998-08-03 2002-04-30 Insite Vision, Incorporated Methods of ophthalmic administration
US6297258B1 (en) 1998-09-29 2001-10-02 American Cyanamid Company Substituted 3-cyanoquinolines
US6288082B1 (en) 1998-09-29 2001-09-11 American Cyanamid Company Substituted 3-cyanoquinolines
AU764479B2 (en) * 1998-10-29 2003-08-21 Bristol-Myers Squibb Company Compounds derived from an amine nucleus that are inhibitors of IMPDH enzyme
NZ513432A (en) * 1999-01-13 2004-02-27 Warner Lambert Co 1-heterocycle substituted diarylamines
FR2792314B1 (en) * 1999-04-15 2001-06-01 Adir NOVEL AMINOTRIAZOLE COMPOUNDS, PROCESS FOR THEIR PREPARATION AND THE PHARMACEUTICAL COMPOSITIONS CONTAINING THE SAME
EP1193255A4 (en) 1999-07-01 2002-08-07 Ajinomoto Kk Heterocyclic compounds and medicinal use thereof
TWI262914B (en) 1999-07-02 2006-10-01 Agouron Pharma Compounds and pharmaceutical compositions for inhibiting protein kinases
TR200200099T2 (en) 1999-07-23 2002-06-21 Shionogi &Co., Ltd. Th2 differentiation inhibitors
US6127382A (en) 1999-08-16 2000-10-03 Allergan Sales, Inc. Amines substituted with a tetrahydroquinolinyl group an aryl or heteroaryl group and an alkyl group, having retinoid-like biological activity
US6093838A (en) 1999-08-16 2000-07-25 Allergan Sales, Inc. Amines substituted with a dihydro-benzofuranyl or with a dihydro-isobenzofuranyl group, an aryl or heteroaryl group and an alkyl group, having retinoid-like biological activity
JP2001089412A (en) 1999-09-22 2001-04-03 Otsuka Pharmaceut Co Ltd Benzene derivative or its pharmaceutically acceptable salt
US6506769B2 (en) 1999-10-06 2003-01-14 Boehringer Ingelheim Pharmaceuticals, Inc. Heterocyclic compounds useful as inhibitors of tyrosine kinases
AU7683500A (en) 1999-10-12 2001-04-23 Takeda Chemical Industries Ltd. Pyrimidine-5-carboxamide compounds, process for producing the same and use thereof
US6638929B2 (en) 1999-12-29 2003-10-28 Wyeth Tricyclic protein kinase inhibitors
US6153752A (en) 2000-01-28 2000-11-28 Creanova, Inc. Process for preparing heterocycles
US20020165244A1 (en) 2000-01-31 2002-11-07 Yuhong Zhou Mucin synthesis inhibitors
GB0004887D0 (en) 2000-03-01 2000-04-19 Astrazeneca Uk Ltd Chemical compounds
US6605615B2 (en) 2000-03-01 2003-08-12 Tularik Inc. Hydrazones and analogs as cholesterol lowering agents
JP2001247411A (en) 2000-03-09 2001-09-11 Tomono Agrica Co Ltd Pest-controlling agent
US6608048B2 (en) 2000-03-28 2003-08-19 Wyeth Holdings Tricyclic protein kinase inhibitors
US20030149061A1 (en) 2000-04-04 2003-08-07 Yoshitaka Nishihara Oily compositions containing highly fat-soluble drugs
US6471968B1 (en) 2000-05-12 2002-10-29 Regents Of The University Of Michigan Multifunctional nanodevice platform
DE10024622A1 (en) 2000-05-18 2001-11-22 Piesteritz Stickstoff New N-(2-pyrimidinyl)-(thio)phosphoric acid triamide compounds are urease inhibitors useful for preventing hydrolysis of urea fertilizers or for reducing release of ammonia in animal stalls
EP1170353B1 (en) 2000-07-06 2005-11-02 Fuji Photo Film Co., Ltd. Liquid crystal composition comprising liquid crystal molecules and aligment promoter
CA2417635C (en) 2000-08-11 2008-02-05 Boehringer Ingelheim Pharmaceuticals, Inc. Heterocyclic compounds useful as inhibitors of tyrosine kinases
US20020137755A1 (en) 2000-12-04 2002-09-26 Bilodeau Mark T. Tyrosine kinase inhibitors
JP2002221770A (en) 2001-01-24 2002-08-09 Fuji Photo Film Co Ltd Silver halide photographic sensitive material and method of processing the same
WO2002064211A1 (en) * 2001-02-09 2002-08-22 Merck & Co., Inc. Thrombin inhibitors
US6949544B2 (en) * 2001-03-29 2005-09-27 Vertex Pharmaceuticals Incorporated Inhibitors of c-Jun N-terminal kinases (JNK) and other protein kinases
EP1401833A2 (en) 2001-05-28 2004-03-31 Aventis Pharma S.A. Chemical derivatives and the use thereof as an anti-elomerase agent
KR100874791B1 (en) 2001-05-29 2008-12-18 바이엘 쉐링 파마 악티엔게젤샤프트 CDV-inhibited pyrimidine, preparation method thereof and use as medicament
WO2002096905A1 (en) * 2001-06-01 2002-12-05 Vertex Pharmaceuticals Incorporated Thiazole compounds useful as inhibitors of protein kinases
JP4342937B2 (en) 2001-07-03 2009-10-14 バーテックス ファーマシューティカルズ インコーポレイテッド Isoxazole pyrimidines as inhibitors of Src and Lck protein kinases
EP1453516A2 (en) 2001-10-17 2004-09-08 Boehringer Ingelheim Pharma GmbH & Co.KG Novel tri-substituted pyrimidines, method for production and use thereof as medicament
US20030187026A1 (en) 2001-12-13 2003-10-02 Qun Li Kinase inhibitors
US20030166932A1 (en) 2002-01-04 2003-09-04 Beard Richard L. Amines substituted with a dihydronaphthalenyl, chromenyl, or thiochromenyl group, an aryl or heteroaryl group and an alkyl group, having retinoid-like biological activity
MXPA04008458A (en) 2002-03-01 2004-12-06 Smithkline Beecham Corp Diamino-pyrimidines and their use as angiogenesis inhibitors.
GB0206215D0 (en) * 2002-03-15 2002-05-01 Novartis Ag Organic compounds
IL164302A0 (en) * 2002-03-29 2005-12-18 Chiron Corp Substituted benzazoles and use thereof as raf kinase inhibitors
MY141867A (en) 2002-06-20 2010-07-16 Vertex Pharma Substituted pyrimidines useful as protein kinase inhibitors
AU2003247959B2 (en) 2002-07-09 2010-07-29 Vertex Pharmaceuticals Incorporated Imidazoles, oxazoles and thiazoles with protein kinase inhibiting activities
CN103169708B (en) 2002-07-29 2018-02-02 里格尔药品股份有限公司 The method that autoimmune disease is treated or prevented with 2,4 pyrimidinediamine compounds
MXPA05001277A (en) 2002-08-02 2005-10-06 Ab Science 2-(3-aminoaryl)amino-4-aryl-thiazoles and their use as c-kit inhibitors.
MXPA05001804A (en) 2002-08-14 2005-05-27 Vertex Pharma Protein kinase inhibitors and uses thereof.
UY27939A1 (en) 2002-08-21 2004-03-31 Glaxo Group Ltd COMPOUNDS
US7230101B1 (en) 2002-08-28 2007-06-12 Gpc Biotech, Inc. Synthesis of methotrexate-containing heterodimeric molecules
DE10240262A1 (en) 2002-08-31 2004-03-11 Clariant Gmbh Production of aryllithium-electrophile reaction products of interest for the pharmaceutical and agrochemical industries comprises using an organolithium compound prepared by reacting an aryl halide with lithium
DE10240261A1 (en) 2002-08-31 2004-03-11 Clariant Gmbh Production of aryllithium-electrophile reaction products of interest for the pharmaceutical and agrochemical industries comprises using an organolithium compound prepared by reacting an aryl halide with lithium
US20050282814A1 (en) 2002-10-03 2005-12-22 Targegen, Inc. Vasculostatic agents and methods of use thereof
KR20050056227A (en) 2002-10-03 2005-06-14 탈자진 인코포레이티드 Vasculostatic agents and methods of use thereof
CA2500368A1 (en) 2002-10-04 2004-04-22 Caritas St. Elizabeth's Medical Center Of Boston, Inc. Inhibition of src for treatment of reperfusion injury related to revascularization
EP1551813A4 (en) * 2002-10-10 2007-07-11 Smithkline Beecham Corp Chemical compounds
AU2003301662A1 (en) 2002-10-21 2004-05-13 Bristol-Myers Squibb Company Quinazolinones and derivatives thereof as factor xa inhibitors
US7262200B2 (en) 2002-10-25 2007-08-28 Vertex Pharmaceuticals Incorporated Indazolinone compositions useful as kinase inhibitors
DE10250708A1 (en) 2002-10-31 2004-05-19 Boehringer Ingelheim Pharma Gmbh & Co. Kg New alkyne compounds having MCH antagonist activity and medicaments containing these compounds
ATE481387T1 (en) 2002-11-19 2010-10-15 Memory Pharm Corp PYRIDINE N-OXIDE COMPOUNDS ALSPHOSPHODIESTERASE-4 INHIBITORS
AU2003280188A1 (en) 2002-12-06 2004-06-30 Warner-Lambert Company Llc Benzoxazin-3-ones and derivatives thereof as inhibitors of pi3k
JP3837670B2 (en) 2002-12-12 2006-10-25 富士通株式会社 Data relay apparatus, associative memory device, and associative memory device utilization information retrieval method
MXPA05006727A (en) 2002-12-20 2005-09-08 Pharmacia Corp Heteroarylalkanoic acids as integrin receptor antagonists.
UA80767C2 (en) 2002-12-20 2007-10-25 Pfizer Prod Inc Pyrimidine derivatives for the treatment of abnormal cell growth
CA2510050A1 (en) 2002-12-20 2004-07-22 Pharmacia Corporation The r-isomer of beta amino acid compounds as integrin receptor antagonists derivatives
WO2004058782A1 (en) 2002-12-24 2004-07-15 Astrazeneca Ab Therapeutic quinazoline derivatives
US7144911B2 (en) 2002-12-31 2006-12-05 Deciphera Pharmaceuticals Llc Anti-inflammatory medicaments
CL2004000192A1 (en) 2003-02-07 2005-03-18 Janssen Pharmaceutica Nv USE OF COMPOUNDS DERIVED FROM 2-AMINOPIRIMIDINE TO PREPARE A MEDICINAL PRODUCT FOR THE PREVENTION OF HIV INFECTION SEXUAL RELATIONS OR AN INTIMATE COUPLE CONTACT.
CN1747941A (en) 2003-02-07 2006-03-15 詹森药业有限公司 HIV inhibiting 1,2,4-triazines
US20040248850A1 (en) 2003-02-11 2004-12-09 Kemia, Inc. Compounds for the treatment of HIV infection
CL2004000306A1 (en) 2003-02-20 2005-04-08 Tibotec Pharm Ltd COMPOUNDS DERIVED FROM PYRIMIDINE REPLACED WITH INDANO; PROCESS FOR PREPARATION; PHARMACEUTICAL COMPOSITION THAT UNDERSTANDS IT; PHARMACEUTICAL COMBINATION; AND ITS USE FOR THE TREATMENT OR PROFILAXIS OF AN INFECTIOUS DISEASE.
HUE035379T2 (en) 2003-04-24 2018-05-02 Coopervision Int Holding Co Lp Hydrogel contact lenses and package systems and production methods for same
ES2380206T3 (en) 2003-08-15 2012-05-09 Novartis Ag 2,4-Pyrimidinediamines useful for the treatment of neoplastic diseases, inflammatory and immune system disorders
JP2007505858A (en) 2003-09-18 2007-03-15 ノバルティス アクチエンゲゼルシャフト 2,4-Di (phenylamino) pyrimidine useful for the treatment of proliferative disorders
CA2537812C (en) * 2003-09-26 2013-01-22 Exelixis, Inc. C-met modulators and method of use
WO2005035541A1 (en) 2003-10-07 2005-04-21 Amedis Pharmaceuticals Ltd. Silicon compounds and their use
DE10356579A1 (en) 2003-12-04 2005-07-07 Merck Patent Gmbh amine derivatives
KR20070026390A (en) 2004-01-23 2007-03-08 암젠 인코포레이션 Compounds and methods of use
AU2005231507B2 (en) * 2004-04-08 2012-03-01 Targegen, Inc. Benzotriazine inhibitors of kinases
JPWO2005108370A1 (en) 2004-04-16 2008-03-21 味の素株式会社 Benzene compounds
RU2468021C2 (en) 2004-08-25 2012-11-27 Таргеджен, Инк. Heterocyclic compounds and their application
US7210697B2 (en) 2004-12-16 2007-05-01 Tricam International, Inc. Convertible handle
KR100919905B1 (en) 2005-01-26 2009-10-06 아이알엠 엘엘씨 Compounds and compositions as protein kinase inhibitors
GB0501999D0 (en) 2005-02-01 2005-03-09 Sentinel Oncology Ltd Pharmaceutical compounds
WO2006101977A2 (en) 2005-03-16 2006-09-28 Targegen, Inc. Pyrimidine compounds and methods of use
US20070032493A1 (en) 2005-05-26 2007-02-08 Synta Pharmaceuticals Corp. Method for treating B cell regulated autoimmune disorders
WO2006128129A2 (en) 2005-05-26 2006-11-30 Synta Pharmaceuticals Corp. Method for treating cancer
EP1893216A4 (en) * 2005-06-08 2012-08-08 Targegen Inc Methods and compositions for the treatment of ocular disorders
WO2007008541A2 (en) 2005-07-08 2007-01-18 Kalypsys, Inc. Cellular cholesterol absorption modifiers
CA2618513A1 (en) 2005-08-11 2007-02-22 Ariad Pharmaceuticals, Inc. Unsaturated heterocyclic derivatives
TW200745066A (en) 2005-09-16 2007-12-16 Torrent Pharmaceuticals Ltd Novel PTP1B inhibitors
US20070072682A1 (en) 2005-09-29 2007-03-29 Crawford James T Iii Head to head electronic poker game assembly and method of operation
US8133900B2 (en) 2005-11-01 2012-03-13 Targegen, Inc. Use of bi-aryl meta-pyrimidine inhibitors of kinases
RU2448959C2 (en) 2005-11-01 2012-04-27 Таргеджен, Инк. Bi-aryl-metha-pyrimidine kinase inhibitors
US8604042B2 (en) 2005-11-01 2013-12-10 Targegen, Inc. Bi-aryl meta-pyrimidine inhibitors of kinases
US20070161645A1 (en) * 2005-11-02 2007-07-12 Targegen, Inc. Thiazole inhibitors targeting resistant kinase mutations
US7604309B2 (en) * 2006-01-19 2009-10-20 King Slide Works Co., Ltd. Auto release retaining mechanism of a slide
US7691858B2 (en) * 2006-04-25 2010-04-06 Targegen, Inc. Kinase inhibitors and methods of use thereof
WO2007127479A2 (en) * 2006-04-28 2007-11-08 Redpoint Bio Corporation Triaryl substituted imidazole derivatives and taste-inhibiting uses thereof
US8030487B2 (en) 2006-07-07 2011-10-04 Targegen, Inc. 2-amino—5-substituted pyrimidine inhibitors
WO2009026345A1 (en) 2007-08-20 2009-02-26 Targegen Inc. Thiazolidinone compounds, and methods of making and using same
WO2009046416A1 (en) 2007-10-05 2009-04-09 Targegen Inc. Anilinopyrimidines as jak kinase inhibitors
WO2009049028A1 (en) 2007-10-09 2009-04-16 Targegen Inc. Pyrrolopyrimidine compounds and their use as janus kinase modulators
WO2009055674A1 (en) 2007-10-26 2009-04-30 Targegen Inc. Pyrrolopyrimidine alkynyl compounds and methods of making and using same
WO2009100305A1 (en) 2008-02-08 2009-08-13 Targegen Inc. Pteridine derivatives for treating respiratory disease
CA2732791A1 (en) 2008-08-05 2010-02-11 Targegen, Inc. Methods of treating thalassemia

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4160833A (en) * 1973-02-02 1979-07-10 Ciba-Geigy Corporation 1,2,4-Benzotriazine-1,4-di-N-oxide derivatives
US4309211A (en) * 1979-08-31 1982-01-05 Ici Australia Limited Herbicidal 1,2,4-benzotriazines
US5849738A (en) * 1986-09-25 1998-12-15 Sri International 1,2,4-benzotriazine oxides as radiosensitizers and selective cytotoxic agents

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See also references of EP1799656A4 *

Cited By (49)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8481536B2 (en) 2004-04-08 2013-07-09 Targegen, Inc. Benzotriazine inhibitors of kinases
US8084618B2 (en) 2004-08-25 2011-12-27 Targegen, Inc. Heterocyclic compounds and methods of use
US8372971B2 (en) 2004-08-25 2013-02-12 Targegen, Inc. Heterocyclic compounds and methods of use
US8110687B2 (en) 2005-12-08 2012-02-07 Millennium Pharmaceuticals, Inc. Bicyclic compounds with kinase inhibitory activity
WO2008059259A3 (en) * 2006-11-16 2008-10-02 Sentinel Oncology Ltd Anticancer compounds with a benzo ( 1, 2, 4 ) triazin-3yl-amine core
WO2008059259A2 (en) * 2006-11-16 2008-05-22 Sentinel Oncology Limited Anticancer compounds with a benzo ( 1, 2, 4 ) triazin-3yl-amine core
EP2141164A1 (en) * 2008-07-01 2010-01-06 Mutabilis New 1,2,4-triazine derivatives and biological applications thereof
WO2010001220A1 (en) * 2008-07-01 2010-01-07 Mutabilis Sa New 1,2,4-triazine derivatives and biological applications thereof
DE102008038222A1 (en) 2008-08-18 2010-02-25 Merck Patent Gmbh Indazol-5-carboxylic acid derivatives
WO2010020305A1 (en) 2008-08-18 2010-02-25 Merck Patent Gmbh Oxadiazole derivatives for treating diabetes
DE102008038220A1 (en) 2008-08-18 2010-02-25 Merck Patent Gmbh oxadiazole
US8377975B2 (en) 2008-08-18 2013-02-19 Merck Patent Gesellschaft Mit Beschrankter Haftung Oxadiazole derivatives for the treatment of diabetes
AU2009284453B2 (en) * 2008-08-18 2014-02-20 Merck Patent Gmbh Oxadiazole derivatives for treating diabetes
WO2010068738A1 (en) 2008-12-10 2010-06-17 Dana-Farber Cancer Institute, Inc. Mek mutations conferring resistance to mek inhibitors
US9084781B2 (en) 2008-12-10 2015-07-21 Novartis Ag MEK mutations conferring resistance to MEK inhibitors
WO2010076238A1 (en) 2008-12-29 2010-07-08 Fovea Pharmaceuticals Sa Substituted quinazoline compounds
WO2010092041A1 (en) 2009-02-13 2010-08-19 Fovea Pharmaceuticals Sa [1, 2, 4] triazolo [1, 5 -a] pyridines as kinase inhibitors
EP2534145A2 (en) * 2010-02-08 2012-12-19 Kinagen, Inc. Therapeutic methods and compositions involving allosteric kinase inhibition
US9260417B2 (en) * 2010-02-08 2016-02-16 Amitech Therapeutic Solutions, Inc. Therapeutic methods and compositions involving allosteric kinase inhibition
EP2534145A4 (en) * 2010-02-08 2013-09-11 Kinagen Inc Therapeutic methods and compositions involving allosteric kinase inhibition
US20140296268A1 (en) * 2010-02-08 2014-10-02 Eric A. Murphy Therapeutic methods and compositions involving allosteric kinase inhibition
US9279144B2 (en) 2010-02-25 2016-03-08 Dana-Farber Cancer Institute, Inc. Screening method for BRAF inhibitors
EP3028699A1 (en) 2010-02-25 2016-06-08 Dana-Farber Cancer Institute, Inc. Braf mutations conferring resistance to braf inhibitors
WO2011106298A1 (en) 2010-02-25 2011-09-01 Dana-Farber Cancer Institute, Inc. Braf mutations conferring resistance to braf inhibitors
US11078540B2 (en) 2010-03-09 2021-08-03 Dana-Farber Cancer Institute, Inc. Methods of diagnosing and treating cancer in patients having or developing resistance to a first cancer therapy
EP2550266A4 (en) * 2010-03-24 2013-11-27 Amitech Therapeutics Solutions Inc Heterocyclic compounds useful for kinase inhibition
AU2016231623B2 (en) * 2010-03-24 2018-03-22 Amitech Therapeutic Solutions, Inc. Heterocyclic compounds useful for kinase inhibition
WO2011119894A2 (en) 2010-03-24 2011-09-29 Kinagen, Inc Heterocyclic compounds useful for kinase inhibition
US8957216B2 (en) 2010-03-24 2015-02-17 Amitech Therapeutic Solutions, Inc. Heterocyclic compounds useful for kinase inhibition
EP2550266A2 (en) * 2010-03-24 2013-01-30 Amitech Therapeutics Solutions, Inc. Heterocyclic compounds useful for kinase inhibition
US9212151B2 (en) 2010-03-24 2015-12-15 Amitech Therapeutic Solutions, Inc. Heterocyclic compounds useful for kinase inhibition
AU2011232372A1 (en) * 2010-03-24 2012-11-08 Amitech Therapeutic Solutions, Inc. Heterocyclic compounds useful for kinase inhibition
WO2011119894A3 (en) * 2010-03-24 2012-02-23 Kinagen, Inc Heterocyclic compounds useful for kinase inhibition
US10214513B2 (en) 2010-03-24 2019-02-26 Amitech Therapeutic Solutions, Inc. Heterocyclic compounds useful for kinase inhibition
AU2011232372B2 (en) * 2010-03-24 2016-06-30 Amitech Therapeutic Solutions, Inc. Heterocyclic compounds useful for kinase inhibition
RU2640862C2 (en) * 2010-03-24 2018-01-18 Амитек Терапетикс Солюшинс, Инк. Heterocyclic compounds, effective for kinase inhibition
KR101844088B1 (en) * 2010-03-24 2018-03-30 아미텍 테러퓨틱 솔루션즈 인크 Heterocyclic compounds useful for kinase inhibition
WO2011161159A1 (en) 2010-06-22 2011-12-29 Fovea Pharmaceuticals Heterocyclic compounds, their preparation and their therapeutic application
WO2013169858A1 (en) 2012-05-08 2013-11-14 The Broad Institute, Inc. Diagnostic and treatment methods in patients having or at risk of developing resistance to cancer therapy
WO2014181287A1 (en) * 2013-05-09 2014-11-13 Piramal Enterprises Limited Heterocyclyl compounds and uses thereof
US11147801B2 (en) 2017-04-24 2021-10-19 Aurigene Discovery Technologies Limited Methods of use for trisubstituted benzotriazole derivatives as dihydroorotate oxygenase inhibitors
US11247987B2 (en) 2017-10-06 2022-02-15 Forma Therapeutics, Inc. Inhibiting ubiquitin specific peptidase 30
WO2019164794A1 (en) * 2018-02-20 2019-08-29 Agios Pharmaceuticals, Inc. Methods of use for trisubstituted benzotriazole derivatives
US11717512B2 (en) 2018-02-20 2023-08-08 Servier Pharmaceuticals Llc Methods of use for trisubstituted benzotriazole derivatives
US11535618B2 (en) 2018-10-05 2022-12-27 Forma Therapeutics, Inc. Fused pyrrolines which act as ubiquitin-specific protease 30 (USP30) inhibitors
US11814386B2 (en) 2018-10-05 2023-11-14 Forma Therapeutics, Inc. Fused pyrrolines which act as ubiquitin-specific protease 30 (USP30) inhibitors
EP3958679A4 (en) * 2019-04-26 2023-08-16 Purdue Research Foundation Cf3-, ocf3-, scf3- and sf5-containing antibacterial agents
US11180473B2 (en) 2020-03-27 2021-11-23 Landos Biopharma, Inc. PLXDC2 ligands
US11597717B2 (en) 2020-03-27 2023-03-07 Landos Biopharma, Inc. Substituted imidazoles as PLXDC2 ligands

Also Published As

Publication number Publication date
IL181524A0 (en) 2007-07-04
RU2007110934A (en) 2008-10-10
NZ553492A (en) 2010-11-26
JP5275628B2 (en) 2013-08-28
AU2005276974A1 (en) 2006-03-02
US8084618B2 (en) 2011-12-27
US20100330069A1 (en) 2010-12-30
US20120220584A1 (en) 2012-08-30
IL181524A (en) 2013-04-30
US7652051B2 (en) 2010-01-26
PL382308A1 (en) 2007-08-20
MX2007002208A (en) 2007-05-08
EP2532653A1 (en) 2012-12-12
EP1799656A1 (en) 2007-06-27
NZ588896A (en) 2012-05-25
RU2468021C2 (en) 2012-11-27
US8372971B2 (en) 2013-02-12
JP2008510839A (en) 2008-04-10
AU2005276974B2 (en) 2012-08-02
EP1799656A4 (en) 2009-09-02
JP2012167111A (en) 2012-09-06
US20130243754A1 (en) 2013-09-19
US20060079526A1 (en) 2006-04-13
CA2578283A1 (en) 2006-03-02

Similar Documents

Publication Publication Date Title
US7652051B2 (en) Heterocyclic compounds and methods of use
US20070161645A1 (en) Thiazole inhibitors targeting resistant kinase mutations
US8846704B2 (en) Thiopyrimidine-based compounds and uses thereof
JP4658037B2 (en) Novel cyanopyridine derivatives useful for the treatment of cancer and other disorders
US10059694B2 (en) 2-aminopyrimidine compound and pharmaceutical composition and use thereof
KR20070113288A (en) Pyrimidine compounds and methods of use
HRP20080087T5 (en) Trisubstituted aryl and heteroaryl derivatives as modulators of metabolism and the prophylaxis and treatment of disorders related thereto
EA011402B1 (en) Nitrogen-containing heterocyclic compounds and pharmaceutical use thereof
WO2012000304A1 (en) Heterocyclic alkynyl benzene compounds and medical compositions and uses thereof
JP2013216698A (en) Heterocyclic compound and use thereof
JP2013075904A (en) Aminopyrimidine useful as kinase inhibitor
WO2011080568A2 (en) Novel pyrimidine compounds as mtor and p13k inhibitors
CN101654452A (en) Heterocyclic compounds and methods of use.
US11407757B2 (en) Heterocycle compounds as Tyro3, Axl and MerTK (TAM) family of receptor tyrosine kinase inhibitors
US10183013B2 (en) Cyclic sulfonamide containing derivatives as inhibitors of hedgehog signaling pathway
CA2715069A1 (en) Novel heterocycles
AU2012244331A1 (en) Heterocyclic compounds and methods of use
KR20170030550A (en) Quinoxaline compounds, method for preparing the same and use thereof

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BW BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KM KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NA NG NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SM SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): GM KE LS MW MZ NA SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LT LU LV MC NL PL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 2578283

Country of ref document: CA

Ref document number: 181524

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: MX/a/2007/002208

Country of ref document: MX

Ref document number: 382308

Country of ref document: PL

WWE Wipo information: entry into national phase

Ref document number: 12007500472

Country of ref document: PH

Ref document number: 2007530214

Country of ref document: JP

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2005276974

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 553492

Country of ref document: NZ

WWE Wipo information: entry into national phase

Ref document number: 2041/DELNP/2007

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: 2005792421

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2007110934

Country of ref document: RU

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2005276974

Country of ref document: AU

Date of ref document: 20050824

Kind code of ref document: A

WWP Wipo information: published in national office

Ref document number: 2005276974

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 200580035918.9

Country of ref document: CN

WWP Wipo information: published in national office

Ref document number: 2005792421

Country of ref document: EP