WO1999016755A1 - Novel angiogenesis inhibitors - Google Patents

Novel angiogenesis inhibitors Download PDF

Info

Publication number
WO1999016755A1
WO1999016755A1 PCT/US1998/019789 US9819789W WO9916755A1 WO 1999016755 A1 WO1999016755 A1 WO 1999016755A1 US 9819789 W US9819789 W US 9819789W WO 9916755 A1 WO9916755 A1 WO 9916755A1
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
aryl
heteroaryl
heterocyclyl
phenyl
Prior art date
Application number
PCT/US1998/019789
Other languages
French (fr)
Inventor
Mark T. Bilodeau
Randall W. Hungate
April M. Cunningham
Timothy J. Koester
Original Assignee
Merck & Co., 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
Priority claimed from GBGB9810544.8A external-priority patent/GB9810544D0/en
Application filed by Merck & Co., Inc. filed Critical Merck & Co., Inc.
Priority to CA002303830A priority Critical patent/CA2303830A1/en
Priority to EP98948427A priority patent/EP1017682A4/en
Priority to AU95003/98A priority patent/AU744939B2/en
Priority to JP2000513841A priority patent/JP2001518470A/en
Publication of WO1999016755A1 publication Critical patent/WO1999016755A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D235/00Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, condensed with other rings
    • C07D235/02Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, condensed with other rings condensed with carbocyclic rings or ring systems
    • C07D235/04Benzimidazoles; Hydrogenated benzimidazoles
    • C07D235/06Benzimidazoles; Hydrogenated benzimidazoles with only hydrogen atoms, hydrocarbon or substituted hydrocarbon radicals, directly attached in position 2
    • 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
    • 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
    • A61P35/00Antineoplastic agents
    • 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
    • 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
    • C07D401/04Heterocyclic 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 directly linked by a ring-member-to-ring-member bond
    • 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
    • C07D409/02Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings
    • C07D409/04Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
    • C07D417/04Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems

Definitions

  • Tyrosine kinases are a class of enzymes that catalyze the transfer of the terminal phosphate of adenosine triphospate to tyrosine residues in protein substrates. Tyrosine kinases are believed, by way of substrate phosphorylation, to play critical roles in signal transduction for a number of cell functions. Though the exact mechanisms of signal transduction is still unclear, tyrosine kinases have been shown to be important contributing factors in cell proliferation, carcinogenesis and cell differentiation. Accordingly, inhibitors of these tyrosine kinases are useful for the prevention and treatment chemotherapy of proliferative diseases dependent on these enzymes.
  • Neoangiogenesis occurs in conjunction with tumor growth and in certain diseases of the eye. It is characterized by excessive activity of vascular endothelial growth factor.
  • VEGF Vascular endothelial growth factor binds the high affinity membrane-spanning tyrosine kinase receptors KDR and Flt-1.
  • KDR mediates the mitogenic function of VEGF
  • Flt-1 appears to modulate non-mitogenic functions such as those associated with cellular adhesion. Inhibiting KDR thus modulates the level of mitogenic VEGF activity.
  • VEGF vascular growth in the retina leads to visual degeneration culminating in blindness.
  • Ocular VEGF mRNA and protein are elevated by conditions such as retinal vein occlusion in primates and decreased p0 2 levels in mice that lead to neovascularization.
  • Intraocular injections of anti-VEGF monoclonal antibodies or VEGF receptor immunofusions inhibit ocular neovascularization in both primate and rodent models. Regardless of the cause of induction of VEGF in human diabetic retinopathy, inhibition of ocular VEGF is useful in treating the disease.
  • VEGF vascular endothelial growth factor
  • monoclonal anti-VEGF antibodies inhibit the growth of human tumors in nude mice. Although these same tumor cells continue to express VEGF in culture, the antibodies do not diminish their mitotic rate. Thus tumor-derived VEGF does not function as an autocrine mitogenic factor. Therefore, VEGF contributes to tumor growth in vivo by promoting angiogenesis through its paracrine vascular endothelial cell chemotactic and mitogenic activities.
  • These monoclonal antibodies also inhibit the growth of typically less well vascularized human colon cancers in athymic mice and decrease the number of tumors arising from inoculated cells.
  • VEGF -binding construct of Flk-1 Viral expression of a VEGF -binding construct of Flk-1, the mouse KDR receptor homologue, truncated to eliminate the cytoplasmic tyrosine kinase domains but retaining a membrane anchor, virtually abolishes the growth of a transplantable glioblastoma in mice presumably by the dominant negative mechanism of heterodimer formation with membrane spanning endothelial cell VEGF receptors.
  • Embryonic stem cells which normally grow as solid tumors in nude mice, do not produce detectable tumors if both VEGF alleles are knocked out. Taken together, these data indicate the role of VEGF in the growth of solid tumors.
  • KDR or Flt-1 are implicated in pathological neoangiogenesis, and these are useful in the treatment of diseases in which neoangiogenesis is part of the overall pathology, e.g., diabetic retinal vascularization, as well as various forms of cancer.
  • Cancers which are treatable in accordance with the present invention demonstrate high levels of gene and protein expression.
  • cancers include cancers of the brain, genitourinary tract, lymphatic system, stomach, larynx and lung. These include histiocytic lymphoma, lung adenocarcinoma and small cell lung cancers. Additional examples include cancers in which overexpression or activation of Raf-activating oncogenes (e.g., K-ras, erb-B) is observed. More particularly, such cancers include pancreatic and breast carcinoma.
  • Raf-activating oncogenes e.g., K-ras, erb-B
  • the present invention relates to compounds which inhibit tyrosine kinase enzymes, compositions which contain tyrosine kinase inhibiting compounds and methods of using tyrosine kinase inhibitors to treat tyrosine kinase-dependent diseases/conditions such as neoangiogenesis, cancer, atherosclerosis, diabetic retinopathy or inflammatory diseases, in mammals.
  • X is N or C;
  • R is H, C 0 alkyl, C 3-6 cycloalkyl, C 5 . 10 aryl, halo, OH, C 3 . 10 heterocyclyl, or C 5 . 10 heteroaryl; said alkyl, alkenyl, alkynyl, aryl, heteroaryl and heterocyclyl being optionally substituted with from one to three members selected from R a ;
  • R 2 &R 3 are independently H, C ⁇ _ 6 alkyl, C 5 . 10 aryl, C3-6 cycloalkyl, OH, N0 2 , -NH 2 , or halogen;
  • R 4 is H, C ⁇ J O alkyl, C3-6 cycloalkyl, C,_ 6 alkoxy C 2-10 alkenyl,
  • R 5 is H, or C,. 6 alkyl, OR, halo, NH 2 or N0 2 ;
  • R a is H, C 0 alkyl, halogen, N0 2 , OR, -NR NR 7 R 8 , R 7 R 8j
  • R is H, or C,_ 6 alkyl, C ⁇ -6 alkylR 9 ;
  • R 9 is C 5 . 10 aryl, C 3 . 10 heterocyclyl, or C 5 . 10 heteroaryl;
  • R7&R8 are independently H, C 0 alkyl, C3-6 cycloalkyl, COR, C 5 . 10 aryl, C 3 . 10 heterocyclyl, or C 5 . 10 heteroaryl or NR7R8 can be taken together to form a heterocyclic 5-10 membered saturated or unsaturated ring containing, in addition to the nitrogen atom, one to two additional heteroatoms selected from the group consisting of N, O and S.
  • a pharmaceutical composition which is comprised of a compound represented by the formula I:
  • R l 5 R 2 , R 3 , R 4 and R 5 are described as above or a pharmaceutically acceptable salt or hydrate or prodrug thereof in combination with a carrier.
  • Also included is a method of treating or preventing a tyrosine kinase dependent disease or condition in a mammal which comprises administering to a mammalian patient in need of such treatment a tyrosine kinase dependent disease or condition treating amount of a compound of formula I or a pharmaceutically acceptable salt, hydrate or pro-drug thereof. Also included is a method of treating or preventing cancer in a mammalian patient in need of such treatment which is comprised of admininstering to said patient an anti-cancer effective amount of a compound of formula I or a pharmaceutically acceptable salt, hydrate or pro-drug thereof.
  • Also included in the present invention is a method of treating or preventing diseases in which neoangiogenesis is implicated, which is comprised of administering to a mammalian patient in need of such treatment a compound of formula I or a pharmaceutically acceptable salt, hydrate or pro-drug thereof in an amount which is effective for reducing neoangiogenesis.
  • a method of treating or preventing ocular disease in which neoangiogenesis occurs is included herein, which is comprised of administering to a mammalian patient in need of such treatment a compound of formula I or a pharmaceutically acceptable salt hydrate or pro-drug thereof in an amount which is effective for treating said ocular disease.
  • a method of treating or preventing retinal vascularization is included herein, which is comprised of administering to a mammalian patient in need of such treatment a compound of formula I or a pharmaceutically acceptable salt, hydrate or pro-drug thereof in an amount which is effective for treating retinal vascularization.
  • Diabetic retinopathy is an example of a disease in which neoangiogenesis or retinal vascularization is part of the overall disease etiology.
  • a method of treating or preventing age-related macular degeneration is also included.
  • alkyl refers to a monovalent alkane
  • hydrocarbon (hydrocarbon) derived radical containing from 1 to 10 carbon atoms unless otherwise defined. It may be straight, branched or cyclic. Preferred straight or branched alkyl groups include methyl, ethyl, propyl, isopropyl, butyl and t-butyl. Preferred cycloalkyl groups include cyclopropyl, cyclobutyl, cycloheptyl, cyclopentyl and cyclohexyl.
  • Alkyl also includes a straight or branched alkyl group which contains or is interrupted by a cycloalkylene portion. Examples include the following:
  • alkylene and monovalent alkyl portion(s) of the alkyl group can be attached at any available point of attachment to the cycloalkylene portion.
  • substituted alkyl when substituted alkyl is present, this refers to a straight, branched or cyclic alkyl group as defined above, substituted with 1-3 groups of R a , described herein.
  • alkenyl refers to a hydrocarbon radical straight, branched or cyclic containing from 2 to 10 carbon atoms and at least one carbon to carbon double bond. Preferably one carbon to carbon double bond is present, and up to four non- aromatic (non-resonating) carbon-carbon double bonds may be present.
  • Preferred alkenyl groups include ethenyl, propenyl, butenyl and cyclohexenyl. As described above with respect to alkyl, the straight, branched or cyclic portion of the alkenyl group may contain double bonds and may be substituted with one to three groups of R a , when a substituted alkenyl group is provided.
  • alkynyl refers to a hydrocarbon radical straight, branched or cyclic, containing from 2 to 10 carbon atoms and at least one carbon to carbon triple bond. Up to three carbon- carbon triple bonds may be present.
  • Preferred alkynyl groups include ethynyl, propynyl and butynyl. As described above with respect to alkyl, the straight, branched or cyclic portion of the alkynyl group may contain triple bonds and may be substituted with 1-3 groups of R a , when a substituted alkynyl group is provided.
  • Aryl refers to 5-10 membered aromatic rings e.g., phenyl, substituted phenyl and like groups as well as rings which are fused, e.g., naphthyl and the like.
  • Aryl thus contains at least one ring having at least 5 atoms, with up to two such rings being present, containing up to 10 atoms therein, with alternating (resonating) double bonds between adjacent carbon atoms.
  • the preferred aryl groups are phenyl and naphthyl.
  • Aryl groups may likewise be substituted with 1 -3 groups of R a as defined herein.
  • Preferred substituted aryls include phenyl and naphthyl substituted with one or two groups.
  • heterocycle, heteroaryl or heterocyclic represents a stable 5- to 7- membered mono- or bicyclic or stable 7- to 10-membered bicyclic heterocyclic ring system, any ring of which may be saturated or unsaturated, and which consists of carbon atoms and from one to three heteroatoms selected from the group consisting of N, O and S, and wherein the nitrogen and sulfur heteroatoms may optionally be oxidized, and the nitrogen heteroatom may optionally be quaternized, and including any bicyclic group in which any of the above-defined heterocyclic rings is fused to a benzene ring.
  • the heterocyclic ring may be attached at any heteroatom or carbon atom which results in the creation of a stable structure.
  • heterocycle, heteroaryl or heterocyclic may be substituted with 1-3 groups of R a .
  • heterocyclic elements include piperidinyl, piperazinyl, 2-oxopiperazinyl, 2-oxopiperidinyl, 2- oxopyrrolodinyl, 2-oxoazepinyl, azepinyl, pyrrolyl, 4-piperidonyl, pyrrolidinyl, pyrazolyl, pyrazolidinyl, imidazolyl, imidazolinyl, imidazolidinyl, pyridyl, pyrazinyl, pyrimidinyl, pyridazinyl, oxazolyl, oxazolidinyl, isoxazolyl, isoxazolidinyl, morpholinyl, thiazolyl, thiazolidinyl, isothiazolyl, quinuclidinyl, isothiazolidinyl,
  • alkoxy refers to those groups of the designated length in either a straight or branched configuration and if two or more carbon atoms in length, they may include a double or a triple bond.
  • alkoxy groups are methoxy, ethoxy, propoxy, isopropoxy, butoxy, isobutoxy, tertiary butoxy, pentoxy, isopentoxy, hexoxy, isohexoxy allyloxy, propargyloxy, and the like.
  • halogen is intended to include the halogen atom fluorine, chlorine, bromine and iodine.
  • prodrug refers to compounds which are drug precursors which, following administration and absorption, release the drug in vivo via some metabolic process.
  • exemplary prodrugs include acyl amides of the amino compounds of this inventon such as amides of alkanoic(C,_ 6 )acids, amides of aryl acids (e.g., benzoic acid) and alkane(C]. 6 )dioic acids.
  • Tyrosine kinase dependent diseases or conditions refers to hyperproliferative disorders which are initiated/maintained by aberrant tyrosine kinase enzyme activity. Examples include psoriasis, cancer, immunoregulation (graft rejection), atherosclerosis, rheumatoid arthritis, angiogenesis (e.g. tumor growth, diabetic retinopathy), etc.
  • X is N or C
  • R is H, C M0 alkyl, C 3-6 cycloalkyl, C 5 . 10 aryl, halo, OH, C 3 . 10 heterocyclyl, or C 5 . 10 heteroaryl; said alkyl, alkenyl, alkynyl, aryl, heteroaryl and heterocyclyl being optionally substituted with from one to three members selected from R a ;
  • R 2 &R 3 are independently H, C ⁇ _ 6 alkyl, C 5 . 10 aryl, C3-6 cycloalkyl, OH, N0 2 , -NH 2 , or halogen;
  • R 4 is H, Cj.jn alkyl, C3-6 cycloalkyl, C,. 6 alkoxy C 2 . 10 alkenyl,
  • R 5 is H, or C,_ 6 alkyl, OR, halo, NH 2 or N0 2 ;
  • R is H, C J . JO alkyl, halogen, N0 2 , OR, -NR, NR 7 R 8? R 7 R 8j
  • R is H, or Cj. 6 alkyl, Cj -6 alkylR 9 ;
  • R 9 is C 5 .j 0 aryl, C 3 . 10 heterocyclyl, or C 5 . 10 heteroaryl;
  • R7&R8 are independently H, C 0 alkyl, C3-6 cycloalkyl, COR, C 5 .j 0 aryl, C 3 . 10 heterocyclyl, or C 5 . 10 heteroaryl or NR7R8 can be taken together to form a heterocyclic
  • R is C J . JO alkyl, C 3-6 cycloalkyl, C 5 . 10 aryl, C 5 . 10 heteroaryl, or C 3 . 10 heterocyclyl, said alkyl, aryl, heteroaryl and heterocyclyl being optionally substituted with from one to three members selected from R a and all other variables are as described above.
  • Rj is C J . JO alkyl, C 5 ., 0 aryl, C 3 . 10 heterocyclyl, or C 5 . 10 heteroaryl, said alkyl, aryl, heteroaryl and heterocyclyl being optionally substituted with from one to three members selected from R a and all other variables are as described above.
  • Rj is H, C J . J O alkyl, C 5 .j 0 aryl, C 3 . 10 heterocyclyl, or C 5 .j 0 heteroaryl; said alkyl, aryl, heteroaryl and heterocyclyl being optionally substituted with from one to three members selected from R a ;
  • R 2 &R 3 are independently H, C,_ 6 alkyl, C 3-6 cycloalkyl, OH, or halogen;
  • R 4 is H, Cj., 0 alkyl, C 3 . 6 cycloalkyl, C 5 .j 0 aryl, C 5 ., 0 heteroaryl, C 3 .j 0 heterocyclyl, Cj -6 alkoxyNR 7 R 8 , N0 2 , OH, -NH 2 or said alkyl, aryl, heteroaryl and heterocyclyl being optionally substituted with from one to three members selected from R a ; and all other variables are as described above.
  • R j is C 5 _i 0 aryl, C 3 . ]0 heterocyclyl, or C 5 .j 0 heteroaryl; said aryl, heteroaryl and heterocyclyl being optionally substituted with from one to three members selected from R a ;
  • R 2 &R 3 are independently H or C j _ 6 alkyl
  • R 4 is C J . JO alkyl, C 5 ., 0 aryl, C 5 . 10 heteroaryl, C 3 .j 0 heterocyclyl said alkyl, aryl, heteroaryl and heterocyclyl being optionally substituted with from one to three members selected from R a ; and all other variables are as described above.
  • Examples of the compounds of this invention are: l-phenyl-5-(4-methoxyphenyl)benzimidaole, 1 -phenyl-5-(4-(2-( 1 -piperidinyl)ethoxy)phenyl)benzimidazole, 3-phenyl-6-(4-methoxylphenyl)imidazo[4,5-b]pyridine,
  • the invention described herein includes a pharmaceutical composition which is comprised of a compound of formula I or a pharmaceutically acceptable salt or hydrate thereof in combination with a carrier.
  • pharmaceutically acceptable salts and “hydrates” refer to those salts and hydrated forms of the compound which would be apparent to the pharmaceutical chemist, i.e., those which favorably affect the physical or pharmacokinetic properties of the compound, such as solubility, palatability, absorption, distribution, metabolism and excretion.
  • Other factors, more practical in nature, which are also important in the selection are the cost of the raw materials, ease of crystallization, yield, stability, solubility, hygroscopicity and flowability of the resulting bulk drug.
  • a counterion e.g., an alkali metal cation such as sodium or potassium.
  • suitable counterions include calcium, magnesium, zinc, ammonium, or alkylammonium cations such as tetramethylammonium, tetrabutylammonium, choline, triethylhydroammonium, meglumine, triethanolhydroammonium, etc.
  • An appropriate number of counterions is associated with the molecule to maintain overall charge neutrality.
  • the compound is positively charged, e.g., protonated, an appropriate number of negatively charged counterions is present to maintain overall charge neutrality.
  • salts also include acid addition salts.
  • the compound can be used in the form of salts derived from inorganic or organic acids or bases. Examples include acetate, adipate, alginate, aspartate, benzoate, benzenesulfonate, bisulfate, butyrate, citrate, camphorate, camphorsulfonate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, fumarate, glucoheptanoate, glycerophosphate, hemisulfate, heptanoate, hexanoate, hydrochloride, hydrobromide, hydroiodide, 2- hydroxyethanesulfonate, lactate, maleate, methanesulfonate, 2- naphthalenesulfonate, nicotinate, oxalate, pamoate, pectinate, persulfate,
  • Base salts include ammonium salts, alkali metal salts such as sodium and potassium salts, alkaline earth metal salts such as calcium and magnesium salts, salts with organic bases such as dicyclohexylamine salts, N-methyl-D-glucamine, and salts with amino acids such as arginine, lysine, and so forth.
  • the basic nitrogen-containing groups may be quaternized with such agents as lower alkyl halides, such as methyl, ethyl, propyl, and butyl chloride, bromides and iodides; dialkyl sulfates like dimethyl, diethyl, dibutyl; and diamyl sulfates, long chain halides such as decyl, lauryl, myristyl and stearyl chlorides, bromides and iodides, aralkyl halides like benzyl and phenethyl bromides and others.
  • Other pharmaceutically acceptable salts include the sulfate salt ethanolate and sulfate salts.
  • the compounds of the present invention may have asymmetric centers and occur as racemates, racemic mixtures and as individual diastereomers, or enantiomers with all isomeric forms being included in the present invention.
  • any variable e.g., aryl, heterocyle, Rl, etc
  • its definition on each occcurence is independent of its definition at every other occurrence, unless otherwise stated.
  • the compounds of the invention can be formulated in a pharmaceutical composition by combining the compound with a pharmaceutically acceptable carrier. Examples of such compositions and carriers are set forth below.
  • the compounds may be employed in powder or crystalline form, in solution or in suspension. They may be administered orally, parenterally (intravenously or intramuscularly), topically, transdermally or by inhalation.
  • the carrier employed may be, for example, either a solid or liquid.
  • solid carriers include lactose, terra alba, sucrose, talc, gelatin, agar, pectin, acacia, magnesium stearate, stearic acid and the like.
  • liquid carriers include syrup, peanut oil, olive oil, water and the like.
  • the carrier for oral use may include time delay material well known in the art, such as glyceryl monostearate or glyceryl distearate alone or with a wax.
  • Topical applications may be formulated in carriers such as hydrophobic or hydrophilic bases to form ointments, creams, lotions, in aqueous, oleaginous or alcoholic liquids to form paints or in dry diluents to form powders.
  • Such topical formulations can be used to treat ocular diseases as well as inflammatory diseases such as rheumatoid arthritis, psoriasis, contact dermatitis, delayed hypersensitivity reactions and the like.
  • Examples of oral solid dosage forms include tablets, capsules, troches, lozenges and the like.
  • the size of the dosage form will vary widely, but preferably will be from about 25 mg to about 500mg.
  • Examples of oral liquid dosage forms include solutions, suspensions, syrups, emulsions, soft gelatin capsules and the like.
  • Examples of injectable dosage forms include sterile injectable liquids, e.g., solutions, emulsions and suspensions.
  • Examples of injectable solids would include powders which are reconstituted, dissolved or suspended in a liquid prior to injection.
  • the carrier is typically comprised of sterile water, saline or another injectable liquid, e.g., peanut oil for intramuscular injections.
  • various buffering agents, preservatives and the like can be included.
  • dosages can be varied depending upon the overall condition of the patient, the nature of the illness being treated and other factors.
  • An example of a suitable oral dosage range is from about 0.1 to about 80 mg/kg per day, in single or divided doses.
  • An example of a suitable parenteral dosage range is from about 0.1 to about 80 mg/kg per day, in single or divided dosages, administered by intravenous or intramuscular injection.
  • An example of a topical dosage range is from about 0.1 mg to about 150 mg, applied externally from about one to four times a day.
  • An example of an inhalation dosage range is from about 0.01 mg/kg to about 1 mg/kg per day.
  • the compounds may be administered in conventional dosages as a single agent or in combination with other therapeutically active compounds.
  • the non-limiting examples that follow are illustrations of the compounds of the instant invention and are not meant to limit the invention in any way.
  • Bromoaromatic 4 (0.218 g, 0.744 mmol) and 4- methoxyboronic acid (0.125 g, 0.823 mmol) were dissolved in a mixture of dioxane (4 mL) and water (3 mL). Sodium carbonate (0.60 g, 5.7 mmol) was added and the resulting mixture was degassed and put under argon. Tetrakis(triphenylphosphine)palladium(0) (0.043 g, 0.037 mmol) was added and the reaction was heated to 80 °C. After 14 h the reaction was cooled to ambient temperature, diluted with water and extracted with ethyl actetate (3x).
  • Benzimidazole 7 (0.025g, 0.087 mmol) and N-(2- chloroethyl)piperidine hydrochloride (1 1 mg, 0.059 mmol) were dissolved in anhydrous N,N-dimethylformamide (0.5 mL). Cesuim carbonate (0.085g, 0.26 mmol) was added and the resulting mixture was heated to 50 °C. After 2 h additional and N-(2-chloroethyl)piperidine hydrochloride (11 mg, 0.059 mmol) was added. After 1 h the reaction was allowed to cool, quenched with water and extracted with ethyl acetate (3x).
  • Bromoaromatic 10 (30 mg, 0.102 mmol), 4- methoxyphenylboronic acid (17 mg, 0.112 mmol) was dissolved in 0.75 mL dioxane followed by the addition of 204 ⁇ L of 2M sodium carbonate.
  • the vessel was flushed with argon followed by the addition of tetrakis(triphenylphosphine)palladium(0) (6 mg, 0.005 mmol) and 0.56 mL water.
  • the vessel was flushed again with argon and heated to 80 °C for 2.5 hr.
  • the solution was cooled to room temperature and diluted with water.
  • the product was extracted with ethyl acetate and washed with brine, followed by drying over sodium sulfate.
  • the organic layer was concentrated, and the product dried in vacuo.
  • the crude mixture was purified by flash column chromatography (2.5 x 8 cm silica gel, 8:2 hexane: ethyl
  • Nitroaniline 11 (1.333 g, 4.15 mmol), Zn dust(6.239 g, 95.40 mmol), and 10 mL acetic acid were mixed under argon. The solution was heated to 60 °C for 1 hr until the solution turned light green. The zinc was removed using vacuum filtration with celite and washed with acetic acid. The filtrate was concentrated and 20 mL of trimethylorthoformate was added. The solution was heated to 100 °C for 2 hr followed by cooling to ambient temperature. The solution was concentrated and the crude mixture was purified by flash column chromatography(5 x 16 cm silica gel, 6:4 ethylacetate:hexane) affording 12.
  • the aqueous layer was extracted a second time with dichloromethane w/ 3% 1-butanol.
  • the organic layers were washed with saturated sodium bicarbonate, and dried over sodium sulfate.
  • the organic layers were conentrated at aspirator pressure to remove ethyl acetate and methylene chloride; the 1 -butanol and residual DMF were removed under high pressure.
  • the product was purified using flash column chromatography(silica gel 2.5 x 32.5 cm, 10:1 methylene chloride:methanol). Excess trifluoroacetic acid was added to the product to create the resulting salt, and the mixture was triturated using ether.
  • the TFA salt was dried using phosphorous pentoxide in vacuo to yield 14 (1.10 TFA salt).
  • Bromoaromatic 4 4 (7.1 g, 24.1 mmol) and powdered zinc (36.2 g, 554 mmol, 23 equiv) were stirred in 80 mL glacial acetic acid. The mixture was heated to 60 °C. After lh the reaction was cooled and filtered through a plug of celite and concentrated to dryness. The resulting residue was dissolved in 60 mL of formic acid and heated to 100 °C overnight. The reaction was cooled and concentrated to dryness. Purification by flash column chromatography (6x25 cm silica, 55:45 hexanes/EtOAc) afforded 5.88 g benzimidazole 15 (89% yield).
  • Benzimidazole 15 (2.91 g, 10.7 mmol), diboron pinacol ester (2.97 g, 11.7 mmol) and potassium acetate (3.14 g, 32.0 mmol) were stirred in 20 mL anhydrous DMF under Ar.
  • PdCl 2 (dppf) (0.26 g, 0.32 mmol) was added, solution was degassed and heated to 80 °C. After 20h the reaction was quenched with 125 mL of water and 50 mL of saturated aqueous NaCl and was extracted 3 x with EtOAc. The combined extracts were dried over Na 2 S0 4 , filtered and concentrated to afford 2.77 g of unpurifled boronate.
  • VEGF RECEPTOR KINASE ASSAY VEGF receptor kinase activity is measured by incorporation of radio-labeled phosphate into polyglutamic acid, tyrosine, 4: 1 (pEY) substrate.
  • the phosphorylated pEY product is trapped onto a filter membrane and the incoporation of radio- labeled phosphate quantified by scintillation counting.
  • the intracellular tyrosine kinase domains of human KDR (Terman, B.I. et al. Oncogene (1991) vol. 6, pp. 1677-1683.) and Flt-1 (Shibuya, M. et al. Oncogene (1990) vol. 5, pp. 519- 524) were cloned as glutathione S-transferase (GST) gene fusion proteins. This was accomplished by cloning the cytoplasmic domain of the KDR kinase as an in frame fusion at the carboxy terminus of the GST gene.
  • GST glutathione S-transferase
  • Soluble recombinant GST-kinase domain fusion proteins were expressed in Spodoptera frugiperda (Sf21) insect cells (Invitrogen) using a baculovirus expression vector (pAcG2T, Pharmingen).
  • EDTA 0.5% triton X-100, 10 % glycerol, 10 mg/ml of each leupeptin, pepstatin and aprotinin and ImM phenylmethylsulfonyl fluoride (all Sigma).
  • Dialysis buffer 50 mM Tris pH 7.4, 0.5 M NaCl, 5 mM DTT, 1 mM EDTA, 0.05% triton X-100, 10 % glycerol, 10 mg/ml of each leupeptin, pepstatin and aprotinin and ImM phenylmethylsulfonyl fluoride.
  • Millipore #MAFC NOB GF/C glass fiber 96 well plate.
  • Sf21 cells were infected with recombinant virus at a multiplicity of infection of 5 virus particles/ cell and grown at 27 °C for 48 hours. 2. All steps were performed at 4°C. Infected cells were harvested by centrifugation at 1000 X g and lysed at 4 °C for 30 minutes with 1/10 volume of lysis buffer followed by centrifugation at 100,000Xg for 1 hour. The supernatant was then passed over a glutathione Sepharose column (Pharmacia) equilibrated in lysis buffer and washed with 5 volumes of the same buffer followed by 5 volumes of wash buffer. Recombinant GST- KDR protein was eluted with wash buffer/10 mM reduced glutathione (Sigma) and dialyzed against dialysis buffer.
  • reaction mix containing 5 ⁇ l of 10 X reaction buffer, 5 ⁇ l 25 mM ATP/10 ⁇ Ci [ 33 P]ATP (Amersham), and 5 ⁇ l 10 X substrate.
  • VEGF receptors that mediate mitogenic responses to the growth factor is largely restricted to vascular endothelial cells.
  • Human umbilical vein endothelial cells (HUVECs) in culture proliferate in response to VEGF treatment and can be used as an assay system to quantify the effects of KDR kinase inhibitors on VEGF stimulation.
  • quiescent HUVEC monolayers are treated with vehicle or test compound 2 hours prior to addition of VEGF or basic flbroblast growth factor (bFGF).
  • the mitogenic response to VEGF or bFGF is determined by measuring the incorporation of [ 3 H]thymidine into cellular DNA.
  • HUVECs frozen as primary culture isolates are obtained from Clonetics Corp. Cells are maintained in Endothelial Growth Medium (EGM; Clonetics) and are used for mitogenic assays at passages 3-7.
  • EGM Endothelial Growth Medium
  • HUVEC monolayers maintained in EGM are harvested by trypsinization and plated at a density of 4000 cells per 100 ul
  • Assay Medium per well in 96-well plates. Cells are growth- arrested for 24 hours at 37°C in a humidified atmosphere containing 5% C0 2 .
  • Growth-arrest medium is replaced by 100 ul Assay Medium containing either vehicle (0.25% [v/v] DMSO) or the desired final concentration of test compound. All determinations are performed in triplicate. Cells are then incubated at 37°C/5% C0 2 for 2 hours to allow test compounds to enter cells.
  • the compounds of formula I are inhibitors of VEGF and thus are useful for the inhibition of neoangiogenesis, such as in the treatment of occular disease, e.g., diabetic retinopathy and in the treatment of cancers, e.g., solid tumors.
  • the instant compounds inhibit VEGF-stimulated mitogenesis of human vascular endothelial cells in culture with IC 50 values between 150-650 nM. These compounds also show selectivity over related tyrosine kinases (e.g. FGFR1 and the Src family).

Abstract

The present invention relates to compounds which inhibit tyrosine kinase enzymes, compositions which contain tyrosine kinase inhibiting compounds and methods of using tyrosine kinase inhibitors to treat tyrosine kinase-dependent diseases/conditions such as angiogenesis, cancer, atherosclerosis, diabetic retinopathy or autoimmune diseases, in mammals.

Description

TITLE OF THE INVENTION NOVEL ANGIOGENESIS INHIBITORS
BACKGROUND OF THE INVENTION Tyrosine kinases are a class of enzymes that catalyze the transfer of the terminal phosphate of adenosine triphospate to tyrosine residues in protein substrates. Tyrosine kinases are believed, by way of substrate phosphorylation, to play critical roles in signal transduction for a number of cell functions. Though the exact mechanisms of signal transduction is still unclear, tyrosine kinases have been shown to be important contributing factors in cell proliferation, carcinogenesis and cell differentiation. Accordingly, inhibitors of these tyrosine kinases are useful for the prevention and treatment chemotherapy of proliferative diseases dependent on these enzymes.
For example, a method of treatment described herein relates to neoangiogenesis. Neoangiogenesis occurs in conjunction with tumor growth and in certain diseases of the eye. It is characterized by excessive activity of vascular endothelial growth factor.
Vascular endothelial growth factor (VEGF) binds the high affinity membrane-spanning tyrosine kinase receptors KDR and Flt-1. Cell culture and gene knockout experiments indicate that each receptor contributes to different aspects of angiogenesis. KDR mediates the mitogenic function of VEGF whereas Flt-1 appears to modulate non-mitogenic functions such as those associated with cellular adhesion. Inhibiting KDR thus modulates the level of mitogenic VEGF activity.
Vascular growth in the retina leads to visual degeneration culminating in blindness. VEGF accounts for most of the angiogenic activity produced in or near the retina in diabetic retinopathy. Ocular VEGF mRNA and protein are elevated by conditions such as retinal vein occlusion in primates and decreased p02 levels in mice that lead to neovascularization. Intraocular injections of anti-VEGF monoclonal antibodies or VEGF receptor immunofusions inhibit ocular neovascularization in both primate and rodent models. Regardless of the cause of induction of VEGF in human diabetic retinopathy, inhibition of ocular VEGF is useful in treating the disease.
Expression of VEGF is also significantly increased in hypoxic regions of animal and human tumors adjacent to areas of necrosis. Monoclonal anti-VEGF antibodies inhibit the growth of human tumors in nude mice. Although these same tumor cells continue to express VEGF in culture, the antibodies do not diminish their mitotic rate. Thus tumor-derived VEGF does not function as an autocrine mitogenic factor. Therefore, VEGF contributes to tumor growth in vivo by promoting angiogenesis through its paracrine vascular endothelial cell chemotactic and mitogenic activities. These monoclonal antibodies also inhibit the growth of typically less well vascularized human colon cancers in athymic mice and decrease the number of tumors arising from inoculated cells. Viral expression of a VEGF -binding construct of Flk-1, the mouse KDR receptor homologue, truncated to eliminate the cytoplasmic tyrosine kinase domains but retaining a membrane anchor, virtually abolishes the growth of a transplantable glioblastoma in mice presumably by the dominant negative mechanism of heterodimer formation with membrane spanning endothelial cell VEGF receptors. Embryonic stem cells, which normally grow as solid tumors in nude mice, do not produce detectable tumors if both VEGF alleles are knocked out. Taken together, these data indicate the role of VEGF in the growth of solid tumors. Inhibition of KDR or Flt-1 is implicated in pathological neoangiogenesis, and these are useful in the treatment of diseases in which neoangiogenesis is part of the overall pathology, e.g., diabetic retinal vascularization, as well as various forms of cancer.
Cancers which are treatable in accordance with the present invention demonstrate high levels of gene and protein expression. Examples of such cancers include cancers of the brain, genitourinary tract, lymphatic system, stomach, larynx and lung. These include histiocytic lymphoma, lung adenocarcinoma and small cell lung cancers. Additional examples include cancers in which overexpression or activation of Raf-activating oncogenes (e.g., K-ras, erb-B) is observed. More particularly, such cancers include pancreatic and breast carcinoma.
The present invention relates to compounds which inhibit tyrosine kinase enzymes, compositions which contain tyrosine kinase inhibiting compounds and methods of using tyrosine kinase inhibitors to treat tyrosine kinase-dependent diseases/conditions such as neoangiogenesis, cancer, atherosclerosis, diabetic retinopathy or inflammatory diseases, in mammals.
SUMMARY OF THE INVENTION
A compound is disclosed in accordance with formula
Figure imgf000005_0001
I or a pharmaceutically acceptable salt, hydrate or prodrug thereof,
wherein
X is N or C; R, is H, C 0 alkyl, C3-6 cycloalkyl, C5.10 aryl, halo, OH, C3.10 heterocyclyl, or C5.10 heteroaryl; said alkyl, alkenyl, alkynyl, aryl, heteroaryl and heterocyclyl being optionally substituted with from one to three members selected from Ra;
R2&R3 are independently H, Cι_6 alkyl, C5.10 aryl, C3-6 cycloalkyl, OH, N02, -NH2, or halogen;
R4 is H, C^J O alkyl, C3-6 cycloalkyl, C,_6 alkoxy C2-10 alkenyl,
C2-ιo alkynyl, C5.10 aryl, C3.10 heterocyclyl, Cι-6 alkoxyNR7R8, N02, OH, -NH2 or C5.10 heteroaryl, said alkyl, alkenyl, alkynyl, aryl, heteroaryl and heterocyclyl being optionally substituted with from one to three members selected from Ra;
R5 is H, or C,.6 alkyl, OR, halo, NH2 or N02;
Ra is H, C 0 alkyl, halogen, N02, OR, -NR NR7R8, R7R8j
C5.10 aryl, C5.10 heteroaryl or C3.10 heterocyclyl;
R is H, or C,_6 alkyl, Cι-6 alkylR9;
R9 is C5.10 aryl, C3.10 heterocyclyl, or C5.10 heteroaryl; and
R7&R8 are independently H, C 0 alkyl, C3-6 cycloalkyl, COR, C5.10 aryl, C3.10 heterocyclyl, or C5.10 heteroaryl or NR7R8 can be taken together to form a heterocyclic 5-10 membered saturated or unsaturated ring containing, in addition to the nitrogen atom, one to two additional heteroatoms selected from the group consisting of N, O and S. Also disclosed is a pharmaceutical composition which is comprised of a compound represented by the formula I:
Figure imgf000007_0001
I wherein Rl 5 R2, R3, R4 and R5 are described as above or a pharmaceutically acceptable salt or hydrate or prodrug thereof in combination with a carrier.
Also included is a method of treating or preventing a tyrosine kinase dependent disease or condition in a mammal which comprises administering to a mammalian patient in need of such treatment a tyrosine kinase dependent disease or condition treating amount of a compound of formula I or a pharmaceutically acceptable salt, hydrate or pro-drug thereof. Also included is a method of treating or preventing cancer in a mammalian patient in need of such treatment which is comprised of admininstering to said patient an anti-cancer effective amount of a compound of formula I or a pharmaceutically acceptable salt, hydrate or pro-drug thereof. Also included in the present invention is a method of treating or preventing diseases in which neoangiogenesis is implicated, which is comprised of administering to a mammalian patient in need of such treatment a compound of formula I or a pharmaceutically acceptable salt, hydrate or pro-drug thereof in an amount which is effective for reducing neoangiogenesis.
More particularly, a method of treating or preventing ocular disease in which neoangiogenesis occurs is included herein, which is comprised of administering to a mammalian patient in need of such treatment a compound of formula I or a pharmaceutically acceptable salt hydrate or pro-drug thereof in an amount which is effective for treating said ocular disease.
More particularly, a method of treating or preventing retinal vascularization is included herein, which is comprised of administering to a mammalian patient in need of such treatment a compound of formula I or a pharmaceutically acceptable salt, hydrate or pro-drug thereof in an amount which is effective for treating retinal vascularization. Diabetic retinopathy is an example of a disease in which neoangiogenesis or retinal vascularization is part of the overall disease etiology. Also included is a method of treating or preventing age-related macular degeneration.
These and other aspects of the invention will be apparent from the teachings contained herein.
DETAILED DESCRIPTION OF THE INVENTION
The invention is described herein in detail using the terms defined below unless otherwise specified. The term "alkyl" refers to a monovalent alkane
(hydrocarbon) derived radical containing from 1 to 10 carbon atoms unless otherwise defined. It may be straight, branched or cyclic. Preferred straight or branched alkyl groups include methyl, ethyl, propyl, isopropyl, butyl and t-butyl. Preferred cycloalkyl groups include cyclopropyl, cyclobutyl, cycloheptyl, cyclopentyl and cyclohexyl.
Alkyl also includes a straight or branched alkyl group which contains or is interrupted by a cycloalkylene portion. Examples include the following:
Figure imgf000008_0001
wherein: x plus y = from 0-10; and w plus z = from 0-9. The alkylene and monovalent alkyl portion(s) of the alkyl group can be attached at any available point of attachment to the cycloalkylene portion.
When substituted alkyl is present, this refers to a straight, branched or cyclic alkyl group as defined above, substituted with 1-3 groups of Ra, described herein.
The term "alkenyl" refers to a hydrocarbon radical straight, branched or cyclic containing from 2 to 10 carbon atoms and at least one carbon to carbon double bond. Preferably one carbon to carbon double bond is present, and up to four non- aromatic (non-resonating) carbon-carbon double bonds may be present. Preferred alkenyl groups include ethenyl, propenyl, butenyl and cyclohexenyl. As described above with respect to alkyl, the straight, branched or cyclic portion of the alkenyl group may contain double bonds and may be substituted with one to three groups of Ra, when a substituted alkenyl group is provided.
The term "alkynyl" refers to a hydrocarbon radical straight, branched or cyclic, containing from 2 to 10 carbon atoms and at least one carbon to carbon triple bond. Up to three carbon- carbon triple bonds may be present. Preferred alkynyl groups include ethynyl, propynyl and butynyl. As described above with respect to alkyl, the straight, branched or cyclic portion of the alkynyl group may contain triple bonds and may be substituted with 1-3 groups of Ra, when a substituted alkynyl group is provided. Aryl refers to 5-10 membered aromatic rings e.g., phenyl, substituted phenyl and like groups as well as rings which are fused, e.g., naphthyl and the like. Aryl thus contains at least one ring having at least 5 atoms, with up to two such rings being present, containing up to 10 atoms therein, with alternating (resonating) double bonds between adjacent carbon atoms. The preferred aryl groups are phenyl and naphthyl. Aryl groups may likewise be substituted with 1 -3 groups of Ra as defined herein. Preferred substituted aryls include phenyl and naphthyl substituted with one or two groups. The term heterocycle, heteroaryl or heterocyclic, as used herein except where noted, represents a stable 5- to 7- membered mono- or bicyclic or stable 7- to 10-membered bicyclic heterocyclic ring system, any ring of which may be saturated or unsaturated, and which consists of carbon atoms and from one to three heteroatoms selected from the group consisting of N, O and S, and wherein the nitrogen and sulfur heteroatoms may optionally be oxidized, and the nitrogen heteroatom may optionally be quaternized, and including any bicyclic group in which any of the above-defined heterocyclic rings is fused to a benzene ring. The heterocyclic ring may be attached at any heteroatom or carbon atom which results in the creation of a stable structure. The heterocycle, heteroaryl or heterocyclic may be substituted with 1-3 groups of Ra. Examples of such heterocyclic elements include piperidinyl, piperazinyl, 2-oxopiperazinyl, 2-oxopiperidinyl, 2- oxopyrrolodinyl, 2-oxoazepinyl, azepinyl, pyrrolyl, 4-piperidonyl, pyrrolidinyl, pyrazolyl, pyrazolidinyl, imidazolyl, imidazolinyl, imidazolidinyl, pyridyl, pyrazinyl, pyrimidinyl, pyridazinyl, oxazolyl, oxazolidinyl, isoxazolyl, isoxazolidinyl, morpholinyl, thiazolyl, thiazolidinyl, isothiazolyl, quinuclidinyl, isothiazolidinyl, indolyl, quinolinyl, isoquinolinyl, benzimidazolyl, thiadiazoyl, benzopyranyl, benzothiazolyl, benzoxazolyl, furyl, tetrahydrofuryl, tetrahydropyranyl, thiophenyl, imidazopyridinyl, tetrazolyl, triazinyl, thienyl, benzothienyl, thiamorpholinyl sulfoxide, thiamo holinyl sulfone, and oxadiazolyl. The term "alkoxy" refers to those groups of the designated length in either a straight or branched configuration and if two or more carbon atoms in length, they may include a double or a triple bond. Exemplary of such alkoxy groups are methoxy, ethoxy, propoxy, isopropoxy, butoxy, isobutoxy, tertiary butoxy, pentoxy, isopentoxy, hexoxy, isohexoxy allyloxy, propargyloxy, and the like.
The term "halogen" is intended to include the halogen atom fluorine, chlorine, bromine and iodine. The term "prodrug" refers to compounds which are drug precursors which, following administration and absorption, release the drug in vivo via some metabolic process. Exemplary prodrugs include acyl amides of the amino compounds of this inventon such as amides of alkanoic(C,_6)acids, amides of aryl acids (e.g., benzoic acid) and alkane(C].6)dioic acids.
Tyrosine kinase dependent diseases or conditions refers to hyperproliferative disorders which are initiated/maintained by aberrant tyrosine kinase enzyme activity. Examples include psoriasis, cancer, immunoregulation (graft rejection), atherosclerosis, rheumatoid arthritis, angiogenesis (e.g. tumor growth, diabetic retinopathy), etc.
The compounds of the present invention are in accordance with formula I:
Figure imgf000011_0001
X is N or C;
R, is H, CM0 alkyl, C3-6 cycloalkyl, C5.10 aryl, halo, OH, C3.10 heterocyclyl, or C5.10 heteroaryl; said alkyl, alkenyl, alkynyl, aryl, heteroaryl and heterocyclyl being optionally substituted with from one to three members selected from Ra;
R2&R3 are independently H, Cι_6 alkyl, C5.10 aryl, C3-6 cycloalkyl, OH, N02, -NH2, or halogen; R4 is H, Cj.jn alkyl, C3-6 cycloalkyl, C,.6 alkoxy C2.10 alkenyl,
C2-ιo alkynyl, C5.j0 aryl, C3.,0 heterocyclyl, d-6 alkoxyNR7R8, N02, OH, -NH2 or C5.j0 heteroaryl, said alkyl, alkenyl, alkynyl, aryl, heteroaryl and heterocyclyl being optionally substituted with from one to three members selected from Ra;
R5 is H, or C,_6 alkyl, OR, halo, NH2 or N02;
R is H, CJ.JO alkyl, halogen, N02, OR, -NR, NR7R8? R7R8j
C5.10 aryl, C5.10 heteroaryl or C3.10 heterocyclyl,
R is H, or Cj.6 alkyl, Cj-6 alkylR9;
R9 is C5.j0 aryl, C3.10 heterocyclyl, or C5.10 heteroaryl; and
R7&R8 are independently H, C 0 alkyl, C3-6 cycloalkyl, COR, C5.j0 aryl, C3.10 heterocyclyl, or C5.10 heteroaryl or NR7R8 can be taken together to form a heterocyclic
5-10 membered saturated or unsaturated ring containing, in addition to the nitrogen atom, one to two additional heteroatoms selected from the group consisting of N, O and S. In one aspect, the invention is described wherein X is C and all other variables are as described above.
In another aspect, the invention is described wherein X is N and all other variables are as described above.
In still another aspect, the invention is described wherein R is CJ.JO alkyl, C3-6 cycloalkyl, C5.10 aryl, C5.10 heteroaryl, or C3.10 heterocyclyl, said alkyl, aryl, heteroaryl and heterocyclyl being optionally substituted with from one to three members selected from Ra and all other variables are as described above. In yet another aspect, the invention is described wherein Rj is CJ.JO alkyl, C5.,0 aryl, C3.10 heterocyclyl, or C5.10 heteroaryl, said alkyl, aryl, heteroaryl and heterocyclyl being optionally substituted with from one to three members selected from Ra and all other variables are as described above.
A preferred subset of compounds of the present invention is realized when: Rj is H, CJ.J O alkyl, C5.j0 aryl, C3.10 heterocyclyl, or C5.j0 heteroaryl; said alkyl, aryl, heteroaryl and heterocyclyl being optionally substituted with from one to three members selected from Ra;
R2&R3 are independently H, C,_6 alkyl, C3-6 cycloalkyl, OH, or halogen;
R4 is H, Cj.,0 alkyl, C3.6 cycloalkyl, C5.j0 aryl, C5.,0 heteroaryl, C3.j0 heterocyclyl, Cj-6 alkoxyNR7R8, N02, OH, -NH2 or said alkyl, aryl, heteroaryl and heterocyclyl being optionally substituted with from one to three members selected from Ra; and all other variables are as described above.
Another preferred subset of compounds of the present invention is realized when: Rj is C5_i0 aryl, C3.]0 heterocyclyl, or C5.j0 heteroaryl; said aryl, heteroaryl and heterocyclyl being optionally substituted with from one to three members selected from Ra;
R2&R3 are independently H or Cj_6 alkyl;
R4 is CJ.JO alkyl, C5.,0 aryl, C5.10 heteroaryl, C3.j0 heterocyclyl said alkyl, aryl, heteroaryl and heterocyclyl being optionally substituted with from one to three members selected from Ra; and all other variables are as described above.
Examples of the compounds of this invention are: l-phenyl-5-(4-methoxyphenyl)benzimidaole, 1 -phenyl-5-(4-(2-( 1 -piperidinyl)ethoxy)phenyl)benzimidazole, 3-phenyl-6-(4-methoxylphenyl)imidazo[4,5-b]pyridine,
3-phenyl-6-(4-(2-(l-piperidinyl)ethoxy)phenyl)imidazo[4,5-b]p3τidine, 3-phenyl-6-(4-(2-(l-piperidinyl)ethoxyphenyl)imidazo[4,5-b]pyridine,
3-(2-thiazoyl)-6-(4-(3-(l-piperidinyl)propylphenyl)imidazo[4,5- b]pyridine,
1 -(2-thiazoyl)-5-(4-(3-( 1 -piperidinyl)propyl)phenyl)benzimidazol, l-(3-thiophenyl)-5-(4-(3-(l-piperidinyl)propyl)phenyl)imidazo[4,5- b]pyridine, l-(3-thiophenyl)-5-(4-(3-(l-piperidinyl)propyl)phenyl)benzimidazole,
3-(3-thiophenyl)-6-(4-(3-(l-piperidinyl)propylphenyl)imidazo[4,5- bjpyridine, l-Phenyl-5-[5-(2-piperidin-l-yl-ethoxy)-pyridin-2-yl]-lH-benzimidazole, l-(4-Cyanophenyl)-5-[6-(2-piperidin-l-yl-ethoxy)-pyridin-3-yl]-lH- benzimidazole,
1 -Phenyl-5-[6-(2-piperidin- 1 -yl-ethoxy)-pyridin-3-yl]- 1 H-benzimidazole,
1 -(3-Cyanophenyl)-5-[6-(2-piperidin- 1 -yl-ethoxy)-pyridin-3-yl]- 1H- benzimidazole, l-(3-Thiophene)-5-[6-(2-piperidin-l-yl-ethoxy)-pyridin-3-yl]-lH- benzimidazole,
[5-(l-Phenyl-lH-benzoimidazol-5-yl)-pyridin-2-yl]-(2-piperidin-l-yl- ethyl)-amine,
[5-( 1 -Phenyl- 1 H-benzoimidazol-5 -yl)-pyridin-2-yl]-(2-morpholin- 1 -yl- ethyl)-amine,
4-( 1 -Phenyl- 1 H-benzoimidazol-5 -yl)- 1 -(3-piperidin- 1 -yl-propyl)- 1 H- pyridin-2-one,
4-( 1 -Phenyl- 1 H-benzoimidazol-5-yl)- 1 -(3 -piperidin- 1 -yl-ethyl)- 1 H- pyridin-2-one, l-(3-Pyridyl)-5-(4-(2-(l-piperidinyl)ethoxy)phenyl)benzimidazole,
1 -(4-Pyridyl)-5-(4-(2-( 1 -piperidinyl)ethoxy)phenyl)benzimidazole,
1 -(3-Pyridyl)-5-[6-(2-piperidin- 1 -yl-ethoxy)-pyridin-3-yl]- 1H- benzimidazole, and 1 -(4-Pyridyl)-5-[6-(2-piperidin- 1 -yl-ethoxy)-pyridin-3-yl]- 1H- benzimidazole or a pharmaceutically acceptable salt, hydrate or prodrug thereof.
The invention described herein includes a pharmaceutical composition which is comprised of a compound of formula I or a pharmaceutically acceptable salt or hydrate thereof in combination with a carrier. As used herein the terms "pharmaceutically acceptable salts" and "hydrates" refer to those salts and hydrated forms of the compound which would be apparent to the pharmaceutical chemist, i.e., those which favorably affect the physical or pharmacokinetic properties of the compound, such as solubility, palatability, absorption, distribution, metabolism and excretion. Other factors, more practical in nature, which are also important in the selection, are the cost of the raw materials, ease of crystallization, yield, stability, solubility, hygroscopicity and flowability of the resulting bulk drug.
When a compound of formula I is present as a salt or hydrate which is non-pharmaceutically acceptable, this can be converted to a salt or hydrate form which is pharmaceutically acceptable in accordance with the present invention.
When the compound is negatively charged, it is balanced by a counterion, e.g., an alkali metal cation such as sodium or potassium. Other suitable counterions include calcium, magnesium, zinc, ammonium, or alkylammonium cations such as tetramethylammonium, tetrabutylammonium, choline, triethylhydroammonium, meglumine, triethanolhydroammonium, etc. An appropriate number of counterions is associated with the molecule to maintain overall charge neutrality. Likewise when the compound is positively charged, e.g., protonated, an appropriate number of negatively charged counterions is present to maintain overall charge neutrality.
Pharmaceutically acceptable salts also include acid addition salts. Thus, the compound can be used in the form of salts derived from inorganic or organic acids or bases. Examples include acetate, adipate, alginate, aspartate, benzoate, benzenesulfonate, bisulfate, butyrate, citrate, camphorate, camphorsulfonate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, fumarate, glucoheptanoate, glycerophosphate, hemisulfate, heptanoate, hexanoate, hydrochloride, hydrobromide, hydroiodide, 2- hydroxyethanesulfonate, lactate, maleate, methanesulfonate, 2- naphthalenesulfonate, nicotinate, oxalate, pamoate, pectinate, persulfate, 3-phenylpropionate, picrate, pivalate, propionate, succinate, tartrate, thiocyanate, tosylate and undecanoate. Base salts include ammonium salts, alkali metal salts such as sodium and potassium salts, alkaline earth metal salts such as calcium and magnesium salts, salts with organic bases such as dicyclohexylamine salts, N-methyl-D-glucamine, and salts with amino acids such as arginine, lysine, and so forth. Also, the basic nitrogen-containing groups may be quaternized with such agents as lower alkyl halides, such as methyl, ethyl, propyl, and butyl chloride, bromides and iodides; dialkyl sulfates like dimethyl, diethyl, dibutyl; and diamyl sulfates, long chain halides such as decyl, lauryl, myristyl and stearyl chlorides, bromides and iodides, aralkyl halides like benzyl and phenethyl bromides and others. Other pharmaceutically acceptable salts include the sulfate salt ethanolate and sulfate salts. The compounds of the present invention, may have asymmetric centers and occur as racemates, racemic mixtures and as individual diastereomers, or enantiomers with all isomeric forms being included in the present invention. When any variable (e.g., aryl, heterocyle, Rl, etc)occurs more than one time in any constituent or in Formula I, its definition on each occcurence is independent of its definition at every other occurrence, unless otherwise stated.
The compounds of the invention can be formulated in a pharmaceutical composition by combining the compound with a pharmaceutically acceptable carrier. Examples of such compositions and carriers are set forth below.
The compounds may be employed in powder or crystalline form, in solution or in suspension. They may be administered orally, parenterally (intravenously or intramuscularly), topically, transdermally or by inhalation.
Thus, the carrier employed may be, for example, either a solid or liquid. Examples of solid carriers include lactose, terra alba, sucrose, talc, gelatin, agar, pectin, acacia, magnesium stearate, stearic acid and the like. Examples of liquid carriers include syrup, peanut oil, olive oil, water and the like. Similarly, the carrier for oral use may include time delay material well known in the art, such as glyceryl monostearate or glyceryl distearate alone or with a wax. Topical applications may be formulated in carriers such as hydrophobic or hydrophilic bases to form ointments, creams, lotions, in aqueous, oleaginous or alcoholic liquids to form paints or in dry diluents to form powders. Such topical formulations can be used to treat ocular diseases as well as inflammatory diseases such as rheumatoid arthritis, psoriasis, contact dermatitis, delayed hypersensitivity reactions and the like. Examples of oral solid dosage forms include tablets, capsules, troches, lozenges and the like. The size of the dosage form will vary widely, but preferably will be from about 25 mg to about 500mg. Examples of oral liquid dosage forms include solutions, suspensions, syrups, emulsions, soft gelatin capsules and the like. Examples of injectable dosage forms include sterile injectable liquids, e.g., solutions, emulsions and suspensions. Examples of injectable solids would include powders which are reconstituted, dissolved or suspended in a liquid prior to injection. In injectable compositions, the carrier is typically comprised of sterile water, saline or another injectable liquid, e.g., peanut oil for intramuscular injections. Also, various buffering agents, preservatives and the like can be included. For the methods of treatment disclosed herein, dosages can be varied depending upon the overall condition of the patient, the nature of the illness being treated and other factors. An example of a suitable oral dosage range is from about 0.1 to about 80 mg/kg per day, in single or divided doses. An example of a suitable parenteral dosage range is from about 0.1 to about 80 mg/kg per day, in single or divided dosages, administered by intravenous or intramuscular injection. An example of a topical dosage range is from about 0.1 mg to about 150 mg, applied externally from about one to four times a day. An example of an inhalation dosage range is from about 0.01 mg/kg to about 1 mg/kg per day.
The examples which follow illustrate the compounds that can be synthesized but they are not limited by the compounds in the tables nor by any particular substituents employed in the schemes for illustrative purposes.
The compounds may be administered in conventional dosages as a single agent or in combination with other therapeutically active compounds. The non-limiting examples that follow are illustrations of the compounds of the instant invention and are not meant to limit the invention in any way.
EXAMPLE 1
Figure imgf000018_0001
l-Bromo-4-fluoro-3 -nitrobenzene (3) (1.14 mL, 9.06 mmol) was dissolved in 5 mL of anhydrous l-methyl-2-pyrrolidinone under argon. Aniline (0.870 mL, 9.55 mmol) was added followed by the addition of N, V-diisopropylethylamine (1.90 mL, 10.9 mmol) and the resulting solution was heated to 120 °C. After 14 h additional aniline (0.082 mL, 0.90 mmol) was added and heating was continued for 8 h. The reaction solution was cooled to ambient temperature, diluted with water and extracted with ethyl acetate (3x). The combined extracts was washed with brine, dried over Na2S0 , filtered and concentrated in vacuo to provide 4.
Η NMR (CDC13) δ 9.46 (bs, 1H), 8.35 (d, 1H, J = 2.4 Hz), 7.45-7.40 (m, 3H), 7.29-7.25 (m, 3H), 7.10 (d, 1H, J = 9.2 Hz).
Figure imgf000019_0001
Bromoaromatic 4 (0.218 g, 0.744 mmol) and 4- methoxyboronic acid (0.125 g, 0.823 mmol) were dissolved in a mixture of dioxane (4 mL) and water (3 mL). Sodium carbonate (0.60 g, 5.7 mmol) was added and the resulting mixture was degassed and put under argon. Tetrakis(triphenylphosphine)palladium(0) (0.043 g, 0.037 mmol) was added and the reaction was heated to 80 °C. After 14 h the reaction was cooled to ambient temperature, diluted with water and extracted with ethyl actetate (3x). The combined extracts was dried with Na2S04, filtered and concentrated to dryness. Purification by flash column chromatography (2 x 16 cm silica gel, 6:1 hexane/ethyl acetate) provided 5. H NMR (CDC13) d 9.48 (bs, 1H), 8.40 (d, 1H, J = 2.4 Hz), 7.58 (dd, 1H, J = 2.4, 9.2 Hz), 7.50 (d, 2H, 9.0 Hz), 7.43 (t, 2H, J = 9.0 Hz), 7.32-7.22 (m, 4H), 6.98 (d, 2H, J - 9.0 Hz), 3.83 (s, 3H).
Figure imgf000019_0002
1 -phenyl-5-(4-methoxyphenyl)benzimidaole. Nitroaniline 5 (0.213 g, 0.665 mmol) and palladium on carbon (10%, 100 mg) were stirred in 8 mL 3: 1 EtOH/AcOH. The reaction was put under a balloon of H2. After 2 h the reaction was filtered through a plug of celite and the filtrate was concentrated to dryness. The resulting residue was dissolved in 1.5 mL trimethylorthoformate and heated to 120 ° C for 30 min. The solution was cooled concentrated to dryness and purified by flash column chromatography (2 x 15 cm silica gel, 1 : 1 hexane/ethyl acetate) which provided 6.
Η NMR (CDC13) δ 8.14 (s, IH), 8.04 (d, IH, J = 0.9 Hz), 7.62-7.50 (m, 8H), 7.48 (t, IH, J = 7.1 Hz), 7.01 (d, 2H, J = 8.8 Hz), 3.87 (s, 3H); FAB mass spectrometry [M+H]+ 301.1 ; Anal. Calcd. for C20H16N2O: C, 79.98; H, 5.37; N, 9.33. Found: C, 79.71; H, 5.48; N, 9.21.
Figure imgf000020_0001
An oven dried flask under argon was charged with benzimidazole 6 (0.039g, 0.13 mmol), aluminum chloride (0.175g, 1.31 mmol), and sodium iodide (0.200g, 1.33 mmol). Anhydrous acetonitrile (1 mL) and dichloromethane (0.5 mL) were added and reaction was heated to reflux. After 44 h the reaction was cooled to ambient temperature, quenched with water and extracted 3 x with ethyl acetate. The combined extracts was dried over Na2S0 , filtered and concentrated to dryness. The resulting residue was triturated with ether, filtered and dried to provide phenol 7.
!H NMR (CDC13) δ 9.48 (s, IH), 8.58 (s, IH), 7.93 (s, IH), 7.73-7.71 (m, 2H), 7.67-7.63 (m, 3H), 7.57-7.49 (m, 4H), 6.86 (d, 2H, J - 8.6 Hz).
Figure imgf000021_0001
1 -phenyl-5-(4-(2-( 1 -piperidinyl)ethoxy)phenyl)benzimidazole
Benzimidazole 7 (0.025g, 0.087 mmol) and N-(2- chloroethyl)piperidine hydrochloride (1 1 mg, 0.059 mmol) were dissolved in anhydrous N,N-dimethylformamide (0.5 mL). Cesuim carbonate (0.085g, 0.26 mmol) was added and the resulting mixture was heated to 50 °C. After 2 h additional and N-(2-chloroethyl)piperidine hydrochloride (11 mg, 0.059 mmol) was added. After 1 h the reaction was allowed to cool, quenched with water and extracted with ethyl acetate (3x). The combined extracts was washed with brine, dried over Na2S0 , filtered and concentrated to dryness. Purification by flash column chromatography (2 x 16 cm silica gel, 9: 1 CH2Cl2/MeOH) provided 8 as a colorless oil.
!H NMR (CDC13) δ 8.14 (s, IH), 8.03 (d, IH, J = 0.9 Hz), 7.62-7.50 (m, 8H), 7.48 (t, IH, J = 7.2 Hz), 7.01 (d, 2H, J = 8.8 Hz), 4.21 (bt, 2H, J = 5.3 Hz), 2.87 (bs, 2H), 2.59 (bs, 4H), 1.66 (bs, 4H), 1.48 (bs, 2H); Mass spectrometry [M+H]+ 398.3.
Figure imgf000021_0002
5-Bromo-2-hydroxy-3-nitropyridine (9) (5.736 g, 0.0262 mol) and 15mL thionyl chloride were added under argon. N,N dimethylformamide (1 mL) was then added and the solution was heated to reflux for 1 hr. By the end of the reaction, the bromohydroxynitropyridine was completely dissolved in solution. After cooling to ambient temperature, 5 mL of toluene was added, and the solution was concentrated under vacuum. The product, 5-bromo-2- chloro-3-nitropyridine, was a yellow crystalline solid. The bromochloronitropyridine was dissolved in 15mL of anhydrous 1 -methyl-2-pyrrolidinone. Aniline (3.580 mL, 0.0393 mol) was added followed by the addition of N,N -diisopropylethylamine (13.69 mL, 0.0786 mol) and the solution was heated to 120 °C. After 1.5 hr., the solution was cooled to ambient temperature and diluted with water. The product was extracted using ethyl acetate and washed with brine. The organic layer was then dried over sodium sulfate, filtered, concentrated, and dried in vacuo. The crude mixture was purified using flash column chromatography (7.5 x 16 cm silica gel, 10:1 hexane: ethyl acetate) to afford 10 . H NMR (CDC13) δ 10.04 (bs, IH), 8.65 (dd, IH, J- 2.2 Hz), 8.50(dd, IH, J=2.4 Hz), 7.60 (d, 2H, J=8.6 Hz), 7.40(t, 2H, J =7.5 Hz), 7.21 (t, lH, J=7.3 Hz).
Figure imgf000022_0001
Bromoaromatic 10 (30 mg, 0.102 mmol), 4- methoxyphenylboronic acid (17 mg, 0.112 mmol) was dissolved in 0.75 mL dioxane followed by the addition of 204 μL of 2M sodium carbonate. The vessel was flushed with argon followed by the addition of tetrakis(triphenylphosphine)palladium(0) (6 mg, 0.005 mmol) and 0.56 mL water. The vessel was flushed again with argon and heated to 80 °C for 2.5 hr. The solution was cooled to room temperature and diluted with water. The product was extracted with ethyl acetate and washed with brine, followed by drying over sodium sulfate. The organic layer was concentrated, and the product dried in vacuo. The crude mixture was purified by flash column chromatography (2.5 x 8 cm silica gel, 8:2 hexane: ethyl acetate), affording 11.
'H NMR (CDC13) δ 10.09 (bs, IH), 8.71 (dd, IH, J=2.4 Hz), 8.66 (dd, IH, J =2.4), 7.67 (d, 2H, J=7.9), 7.49 (d, 2H, J =8.8 Hz), 7.41 (t, 2H, J =1.1 Hz), 7.18 (t, IH, 7 =7.3 Hz), 7.00 (d, 2H, J =8.6 Hz), 3.86 (s, 3H).
Figure imgf000023_0001
3-phenyl-6-(4-methoxylphenyl)imidazo[4,5-b]pyridine
Nitroaniline 11 (1.333 g, 4.15 mmol), Zn dust(6.239 g, 95.40 mmol), and 10 mL acetic acid were mixed under argon. The solution was heated to 60 °C for 1 hr until the solution turned light green. The zinc was removed using vacuum filtration with celite and washed with acetic acid. The filtrate was concentrated and 20 mL of trimethylorthoformate was added. The solution was heated to 100 °C for 2 hr followed by cooling to ambient temperature. The solution was concentrated and the crude mixture was purified by flash column chromatography(5 x 16 cm silica gel, 6:4 ethylacetate:hexane) affording 12.
!H NMR (CDC13) δ 8.61 (dd, IH, J =2.0 Hz), 8.32 (s, IH), 8.22 (dd, IH, J =2.0), 7.74 (d, 2H, J =7.9 Hz), 7.55-7.50 (m, 4H), 7.39 (t, IH, J
=7.3), 6.99 (d, 2H, J =8.8 Hz), 3.80 (s, 3H). Mass spectrometry [M+H]+ 302.3.
EXAMPLE 3
Figure imgf000024_0001
To the imidazopyridine 12 (202 mg, 0.670 mmol) was added a mixture of 10 mL hydrobromic acid and 10 mL acetic acid. The solution was stirred a room temperature for 5 min., followed by heating at 100 °C for 17 hr. The solution was cooled to ambient temperature and concentrated. Toluene (15 mL) was added and the solution was concentrated a second time. The concentrate was placed in vacuo over heating at 40 °C for 40 min., followed by further drying in vacuo at ambient temperature. Purification was acheived by reverse phase column chromatography affording 13.
Η NMR (CD3OD) δ 9.45 (s, IH), 8.82 (dd, IH, J =1.8 Hz), 8.37 (dd, IH, J =1.8 Hz), 7.91 (d, 2H, J =7.7 Hz), 7.68 (t, 2H, J =8.1 Hz), 7.63- 7.57 (m, 3H), 6.95 (d, 2H, / =8.6 Hz).
Figure imgf000024_0002
3-phenyl-6-(4-(2-(l-piperidinyl)ethoxy)phenyl)imidazo[4,5-b]pyridine
Cesium carbonate (296 mg, .908 mmol) and l-(2- chloroethyl)piperidine monochlorohyrdide (84 mg, .454 mmol) were added under argon to a flame dried round bottom flask. Imidazopyridine 13 (87 mg, .303 mmol) was dissolved in 1.5 mL of anhydrous N,N dimethyl formamide under argon. The vessel was heated at 50 °C for 16 hr. and cooled to ambient temperature. The solution was diluted to lOOmL with saturated sodium bicarbonate, and the product was extracted using ethyl acetate. The aqueous layer was extracted a second time with dichloromethane w/ 3% 1-butanol. The organic layers were washed with saturated sodium bicarbonate, and dried over sodium sulfate. The organic layers were conentrated at aspirator pressure to remove ethyl acetate and methylene chloride; the 1 -butanol and residual DMF were removed under high pressure. The product was purified using flash column chromatography(silica gel 2.5 x 32.5 cm, 10:1 methylene chloride:methanol). Excess trifluoroacetic acid was added to the product to create the resulting salt, and the mixture was triturated using ether. The TFA salt was dried using phosphorous pentoxide in vacuo to yield 14 (1.10 TFA salt).
Η NMR (CD3OD) δ 8.66 (s, IH), 8.55 (dd, IH, J =2.0 Hz), 8.17 (dd, IH, .7 =2.0 Hz), 7.82 (d, 2H, 7=8.6 Hz), 7.59-7.52 (m, 4H), 7.46 (t, IH, 7 =7.5 Hz), 7.01 (d, 2H, 7 =8.8 Hz), 4.85 (s, 2H), 4.15 (t, 2H, 7 =5.5 Hz), 2.84 (t, 2H, 7 =5.5 Hz), 2.62 (bs, 4H), 1.65 (m, 4H), 1.50 (m, 2H). Anal. Calcd. for C25H26N4O'1.10 TFA: C, 62.35; H, 5.21 ; N, 10.69. Found: C, 62.32; H, 4.93; N, 10.53.
EXAMPLE 4
Figure imgf000025_0001
Bromoaromatic 44 (7.1 g, 24.1 mmol) and powdered zinc (36.2 g, 554 mmol, 23 equiv) were stirred in 80 mL glacial acetic acid. The mixture was heated to 60 °C. After lh the reaction was cooled and filtered through a plug of celite and concentrated to dryness. The resulting residue was dissolved in 60 mL of formic acid and heated to 100 °C overnight. The reaction was cooled and concentrated to dryness. Purification by flash column chromatography (6x25 cm silica, 55:45 hexanes/EtOAc) afforded 5.88 g benzimidazole 15 (89% yield). *H NMR(CDC13) δ 8.18 (s, IH), 8.05 (d, IH, J=1.7 Hz), 7.60 (t, 2H, 7=7.1 Hz), 7.54-7.48 (m, 3H), 7.46 (dd, IH, 7= 1.8, 8.8 Hz), 7.40 (d, IH, 7=8.8 Hz).
Figure imgf000026_0001
16
1 -Piperidineethanol (1.13 mL, 8.51 mmol) was dissolved in 10 mL anhydrous DMF under Ar. The solution was cooled to 0 °C and NaH (225 mg, 9.38 mmol) was added. After 10 min the mixture was allowed to warm to room temperature and 5-bromo-2-fluoropyridine (1.50 g, 8.52 mmol) was added. After lh the reaction was quenched with water and extracted 3x with EtOAc. The combined extracts were dried over Na2S04, filtered and concentrated to afford 2.20 g (91% yield) of the alkoxypyridine 16. H NMR(CDC13) δ 8.17 (d, IH, 7=2.6 Hz), 7.62 (dd, IH, 7=2.6, 8.8 Hz), 6.67 (d, IH, 7=8.8 Hz), 4.40 (t, 2H, 7=6.0 Hz), 2.74 (t, 2H, 7=5.9 Hz), 2.49 (m, 4H), 1.60 (m, 4H), 1.44 (m, 2H).
Figure imgf000026_0002
l-Pheny -5-[6-(2-piperidin-l-yl-ethoxy)-pyridin-3-yl]-lH-benz 6imidazole
Benzimidazole 15 (2.91 g, 10.7 mmol), diboron pinacol ester (2.97 g, 11.7 mmol) and potassium acetate (3.14 g, 32.0 mmol) were stirred in 20 mL anhydrous DMF under Ar. PdCl2(dppf) (0.26 g, 0.32 mmol) was added, solution was degassed and heated to 80 °C. After 20h the reaction was quenched with 125 mL of water and 50 mL of saturated aqueous NaCl and was extracted 3 x with EtOAc. The combined extracts were dried over Na2S04, filtered and concentrated to afford 2.77 g of unpurifled boronate. The unpurified boronate (650 mg, 2.03 mmol), alkoxypyridine 16 (526 mg, 1.85 mmol), 2M Na2C03 (861 mg, 8.12 mmol), and 4 mL dioxane were added to a round bottom flask. After flushing three times with argon, Pd(PPh3)4 (117 mg, .10 mmol) was added, and the vessel was again flushed three times with argon. The vessel was heated to 80 °C under argon. After 22 hr., the reaction was cooled to room temperature followed by quenching with 25mL water. The mixture was extracted with 4x20 mL ethyl acetate, and the combined organic layers were washed with lx20mL brine. The organic layer was dried over sodium sulfate, filtered, and concentrated. Purification was performed using reverse phase column chromatography (Waters 2x40mm C-18 column, H20:acetonitrile mobile phase gradient). The resulting oil was triturated with ether, filtered and washed with ether, affording 16, a white TFA salt(150 mg, 16% yield). Mp: 160.5 - 162 °C. Η NMR(CDC13) δ 8.41 (d, IH, 7 =2.4 Hz), 8.19 (s, IH), 8.01 (d, IH, 7 =1.3 Hz), 7.90 (dd, IH, 7 =11.0 Hz), 7.61 (m, 3H, 7 =13.6), 7.52 (m, 4H, 7 =31.0 Hz), 6.86 (d, IH, 7 =8.4 Hz), 4.79 (t, 2H, 7 =9.9 Hz), 3.76 (bd, 2H, 7 =11.9 Hz), 3.51 (t, 2H, 7 =9.7 Hz), 2.80 (bt, 2H, 7 =23.1 Hz), 2.06 (m, 2H, 7 =26.2 Hz), 1.89 (s, 2H), 1.65 (s, 2H).
The following compounds can be made by literature methods and/or in combination with methods disclosed herein.
Figure imgf000027_0001
Figure imgf000027_0002
Kinase inhibition is demonstrated in accordance with the following protocol.
VEGF RECEPTOR KINASE ASSAY VEGF receptor kinase activity is measured by incorporation of radio-labeled phosphate into polyglutamic acid, tyrosine, 4: 1 (pEY) substrate. The phosphorylated pEY product is trapped onto a filter membrane and the incoporation of radio- labeled phosphate quantified by scintillation counting.
MATERIALS VEGF receptor kinase
The intracellular tyrosine kinase domains of human KDR (Terman, B.I. et al. Oncogene (1991) vol. 6, pp. 1677-1683.) and Flt-1 (Shibuya, M. et al. Oncogene (1990) vol. 5, pp. 519- 524) were cloned as glutathione S-transferase (GST) gene fusion proteins. This was accomplished by cloning the cytoplasmic domain of the KDR kinase as an in frame fusion at the carboxy terminus of the GST gene. Soluble recombinant GST-kinase domain fusion proteins were expressed in Spodoptera frugiperda (Sf21) insect cells (Invitrogen) using a baculovirus expression vector (pAcG2T, Pharmingen).
Lysis buffer 50 niM Tris pH 7.4, 0.5 M NaCl, 5 mM DTT, 1 mM
EDTA, 0.5% triton X-100, 10 % glycerol, 10 mg/ml of each leupeptin, pepstatin and aprotinin and ImM phenylmethylsulfonyl fluoride (all Sigma).
Wash buffer
50 mM Tris pH 7.4, 0.5 M NaCl, 5 mM DTT, 1 mM EDTA, 0.05% triton X-100, 10 % glycerol, 10 mg/ml of each leupeptin, pepstatin and aprotinin and ImM phenylmethylsulfonyl fluoride. Dialysis buffer
50 mM Tris pH 7.4, 0.5 M NaCl, 5 mM DTT, 1 mM EDTA, 0.05% triton X-100, 50 % glycerol, 10 mg/ml of each leupeptin, pepstatin and aprotinin and ImM phenylmethylsuflonyl fluoride
10 X reaction buffer
200 mM Tris, pH 7.4, 1.0 M NaCl, 50 mM MnCl2, 10 mM DTT and 5 mg/ml bovine serum albumin (Sigma).
Enzyme dilution buffer
50 mM Tris, pH 7.4, 0.1 M NaCl, 1 mM DTT, 10 % glycerol, 100 mg/ml BSA.
10 X Substrate
750 μg/ml poly (glutamic acid, tyrosine; 4:1) (Sigma).
Stop solution
30% trichloroacetic acid, 0.2 M sodium pyrophosphate (both Fisher).
Wash solution
15% trichloroacetic acid, 0.2 M sodium pyrophosphate.
Filter plates
Millipore #MAFC NOB, GF/C glass fiber 96 well plate.
METHOD
A. Protein purification
1. Sf21 cells were infected with recombinant virus at a multiplicity of infection of 5 virus particles/ cell and grown at 27 °C for 48 hours. 2. All steps were performed at 4°C. Infected cells were harvested by centrifugation at 1000 X g and lysed at 4 °C for 30 minutes with 1/10 volume of lysis buffer followed by centrifugation at 100,000Xg for 1 hour. The supernatant was then passed over a glutathione Sepharose column (Pharmacia) equilibrated in lysis buffer and washed with 5 volumes of the same buffer followed by 5 volumes of wash buffer. Recombinant GST- KDR protein was eluted with wash buffer/10 mM reduced glutathione (Sigma) and dialyzed against dialysis buffer.
B. VEGF receptor kinase assay
1. Add 5 μl of inhibitor or control to the assay in 50% DMSO.
2. Add 35 μl of reaction mix containing 5 μl of 10 X reaction buffer, 5 μl 25 mM ATP/10 μCi [33P]ATP (Amersham), and 5 μl 10 X substrate.
3. Start the reaction by the addition of 10 μl of KDR (25 nM) in enzyme dilution buffer.
4. Mix and incubate at room temperature for 15 minutes.
5. Stop by the addition of 50 μl stop solution.
6. Incubate for 15 minutes at 4°C.
7. Transfer a 90 μl aliquot to filter plate.
8. Aspirate and wash 3 times with wash solution. 9. Add 30 μl of scintillation cocktail, seal plate and count in a Wallac Microbeta scintillation counter.
Human Umbilical Vein Endothelial Cell Mitogenesis Assay
Expression of VEGF receptors that mediate mitogenic responses to the growth factor is largely restricted to vascular endothelial cells. Human umbilical vein endothelial cells (HUVECs) in culture proliferate in response to VEGF treatment and can be used as an assay system to quantify the effects of KDR kinase inhibitors on VEGF stimulation. In the assay described, quiescent HUVEC monolayers are treated with vehicle or test compound 2 hours prior to addition of VEGF or basic flbroblast growth factor (bFGF). The mitogenic response to VEGF or bFGF is determined by measuring the incorporation of [3H]thymidine into cellular DNA.
Materials
HUVECs
HUVECs frozen as primary culture isolates are obtained from Clonetics Corp. Cells are maintained in Endothelial Growth Medium (EGM; Clonetics) and are used for mitogenic assays at passages 3-7.
Culture Plates NUNCLON 96-well polystyrene tissue culture plates
(NUNC #167008).
Assay Medium
Dulbecco's modification of Eagle's medium containing 1 g/ml glucose (low-glucose DMEM; Mediatech) plus 10%> (v/v) fetal bovine serum (Clonetics).
Test Compounds
Working stocks of test compounds are diluted serially in 100% dimethylsulfoxide (DMSO) to 400-fold greater than their desired final concentrations. Final dilutions to IX concentration are made directly into Assay Medium immediately prior to addition to cells.
IPX Growth factors
Solutions of human VEGF165 (500 ng/ml; R&D Systems) and bFGF (10 ng/ml; R&D Systems) are prepared in Assay Medium. 10X [3HlThymidine
[Methyl-3H]Thymidine (20 Ci/mmol; Dupont-NEN) is diluted to 80 uCi/ml in low-glucose DMEM.
Cell Wash Medium
Hank's balanced salt solution (Mediatech) containing 1 mg/ml bovine serum albumin (Boehringer-Mannheim).
Cell Lysis Solution 1 N NaOH, 2% (w/v) Na2C03.
Method
1. HUVEC monolayers maintained in EGM are harvested by trypsinization and plated at a density of 4000 cells per 100 ul
Assay Medium per well in 96-well plates. Cells are growth- arrested for 24 hours at 37°C in a humidified atmosphere containing 5% C02.
2. Growth-arrest medium is replaced by 100 ul Assay Medium containing either vehicle (0.25% [v/v] DMSO) or the desired final concentration of test compound. All determinations are performed in triplicate. Cells are then incubated at 37°C/5% C02 for 2 hours to allow test compounds to enter cells.
3. After the 2-hour pretreatment period, cells are stimulated by addition of 10 ul/well of either Assay Medium, 1 OX VEGF solution or 10X bFGF solution. Cells are then incubated at 37°C/5% C02.
4. After 24 hours in the presence of growth factors, 10X [3H]Thymidine (10 ul/well) is added. 5. Three days after addition of [3H]thymidine, medium is removed by aspiration, and cells are washed twice with Cell Wash Medium (400 ul/well followed by 200 ul/well). The washed, adherent cells are then solubilized by addition of Cell Lysis Solution (100 ul/well) and warming to 37°C for 30 minutes. Cell ly sates are transferred to 7-ml glass scintillation vials containing 150 ul of water. Scintillation cocktail (5 ml/vial) is added, and cell-associated radioactivity is determined by liquid scintillation spectroscopy.
Based upon the foregoing assays the compounds of formula I are inhibitors of VEGF and thus are useful for the inhibition of neoangiogenesis, such as in the treatment of occular disease, e.g., diabetic retinopathy and in the treatment of cancers, e.g., solid tumors. The instant compounds inhibit VEGF-stimulated mitogenesis of human vascular endothelial cells in culture with IC50 values between 150-650 nM. These compounds also show selectivity over related tyrosine kinases (e.g. FGFR1 and the Src family).

Claims

WHAT IS CLAIMED IS:
1. A compound in accordance with formula I:
Figure imgf000034_0001
I or a pharmaceutically acceptable salt, hydrate or prodrug thereof,
X is N or C;
R, is H, CJ.JO alkyl, C3.6 cycloalkyl, C5.10 aryl, halo, OH, C3.10 heterocyclyl, or C5.10 heteroaryl; said alkyl, alkenyl, alkynyl, aryl, heteroaryl and heterocyclyl being optionally substituted with from one to three members selected from Ra;
R2&R3 are independently H, Cj.6 alkyl, C5.]0 aryl, C3-6 cycloalkyl, OH, N02, -NH2, or halogen;
R4 is H, CJ.JO alkyl, C3-6 cycloalkyl, Cj.6 alkoxy C2.10 alkenyl, C2-10 alkynyl, C5.10 aryl, C3.10 heterocyclyl, Cj-6 alkoxyNR7R8, N02, OH, -NH2 or C5.10 heteroaryl, said alkyl, alkenyl, alkynyl, aryl, heteroaryl and heterocyclyl being optionally substituted with from one to three members selected from Ra; R5 is H, or Cj.6 alkyl, OR, halo, NH2 or N02;
Ra is H, CJ.JO alkyl, halogen, N02, OR, -NR NR7R8, R7R8,
C5.JO aryl, C5.10 heteroaryl or C3.10 heterocyclyl,
R is H, or C,.6 alkyl, C╬╣-6 alkylR9;
R9 is C5.j0 aryl, C3.,0 heterocyclyl, or C5_╬╣0 heteroaryl; and
R7&R8 are independently H, C,.,0 alkyl, C3-6 cycloalkyl, COR, C5.jo aryl, C3.j0 heterocyclyl, or C5.j0 heteroaryl or NR7R8 can be taken together to form a heterocyclic 5-10 membered saturated or unsaturated ring containing, in addition to the nitrogen atom, one to two additional heteroatoms selected from the group consisting of N, O and S.
2. A compound in accordance with claim 1 wherein X is C and all other variables are as described above.
3. A compound in accordance with claim 1 wherein X is N and all other variables are as described above.
4. A compound in accordance with claim 1 wherein R is CJ.JO alkyl, C3-6 cycloalkyl, C5.,0 aryl, C5.10 heteroaryl, or C3.j0 heterocyclyl, said alkyl, aryl, heteroaryl and heterocyclyl being optionally substituted with from one to three members selected from Ra and all other variables are as described above.
5. A compound in accordance with claim 1 wherein
Rj is CJ.JO alkyl, C5.j0 aryl, C3.10 heterocyclyl, or C5.j0 heteroaryl, said alkyl, aryl, heteroaryl and heterocyclyl being optionally substituted with from one to three members selected from Ra and all other variables are as described above.
6. A compound in accordance with claim 1 wherein:
R, is H, CJ.J O alkyl, C5.10 aryl, C3.10 heterocyclyl, or C5.10 heteroaryl; said alkyl, aryl, heteroaryl and heterocyclyl being optionally substituted with from one to three members selected from Ra;
R2&R3 are independently H, C,_6 alkyl, C3-6 cycloalkyl, OH, or halogen;
R4 is H, CJ.JO alkyl, C3-6 cycloalkyl, C5.,0 aryl, C5.10 heteroaryl, C3.10 heterocyclyl, Cj.6 alkoxyNR7R8, N02, OH, -NH2 or said alkyl, aryl, heteroaryl and heterocyclyl being optionally substituted with from one to three members selected from Ra; and all other variables are as described above.
7. A compound in accordance with claim 1 wherein:
Rj is C5_jo aryl, C3.10 heterocyclyl, or C5.10 heteroaryl; said aryl, heteroaryl and heterocyclyl being optionally substituted with from one to three members selected from Ra;
R2&R3 are independently H or Cj.6 alkyl;
R4 is CJ.JO alkyl, C5.10 aryl, C5.10 heteroaryl, C3.10 heterocyclyl said alkyl, aryl, heteroaryl and heterocyclyl being optionally substituted with from one to three members selected from Ra; and all other variables are as described above.
8. A compound in accordance with claim 1 which is: l-phenyl-5-(4-methoxyphenyl)benzimidaole, 1 -phenyl-5-(4-(2-( 1 -piperidinyl)ethoxy)phenyl)benzimidazole, 3-phenyl-6-(4-methoxylphenyl)imidazo[4,5-b]pyridine,
3-phenyl-6-(4-(2-(l-piperidinyl)ethoxy)phenyl)imidazo[4,5-b]pyridine,
3-phenyl-6-(4-(2-(l-piperidinyl)ethoxyphenyl)imidazo[4,5-b]pyridine,
3-(2-thiazoyl)-6-(4-(3-(l-piperidinyl)propylphenyl)imidazo[4,5- b]pyridine, l-(2-thiazoyl)-5-(4-(3-(l-piperidinyl)propyl)phenyl)benzimidazol, l-(3-thiophenyl)-5-(4-(3-(l-piperidinyl)propyl)phenyl)imidazo[4,5- b]pyridine, l-(3-thiophenyl)-5-(4-(3-(l-piperidinyl)propyl)phenyl)benzimidazole,
3-(3-thiophenyl)-6-(4-(3-(l-piperidinyl)propylphenyl)imidazo[4,5- b]pyridine,
1 -Phenyl-5-[5-(2-piperidin- 1 -yl-ethoxy)-pyridin-2-yl]- 1 H-benzimidazole, l-(4-Cyanophenyl)-5-[6-(2-piperidin-l-yl-ethoxy)-pyridin-3-yl]-lH- benzimidazole,
1 -Phenyl-5-[6-(2-piperidin- 1 -yl-ethoxy)-pyridin-3-yl]- 1 H-benzimidazole, l-(3-Cyanophenyl)-5-[6-(2-piperidin-l-yl-ethoxy)-pyridin-3-yl]-lH- benzimidazole, l-(3-Thiophene)-5-[6-(2-piperidin-l-yl-ethoxy)-pyridin-3-yl]-lH- benzimidazole,
[5-(l-Phenyl-lH-benzoimidazol-5-yl)-pyridin-2-yl]-(2-piperidin-l-yl- ethyl)-amine,
[5-(l-Phenyl-lH-benzoimidazol-5-yl)-pyridin-2-yl]-(2-moφholin-l-yl- ethyl)-amine,
4-(l-Phenyl-lH-benzoimidazol-5-yl)-l-(3-piperidin-l-yl-propyl)-lH- pyridin-2-one, 4-( 1 -Phenyl- 1 H-benzoimidazol-5 -yl)- 1 -(3 -piperidin- 1 -y 1-ethyl)- 1 H- pyridin-2-one, l-(3-Pyridyl)-5-(4-(2-(l-piperidinyl)ethoxy)phenyl)benzimidazole,
1 -(4-Pyridyl)-5-(4-(2-( 1 -piperidinyl)ethoxy)phenyl)benzimidazole,
1 -(3-Pyridyl)-5-[6-(2-piperidin- 1 -yl-ethoxy)-pyridin-3-yl]- 1H- benzimidazole, and l-(4-Pyridyl)-5-[6-(2-piperidin-l-yl-ethoxy)-pyridin-3-yl]-lH- benzimidazole or a pharmaceutically acceptable salt, hydrate or prodrug thereof.
9. A pharmaceutical composition which is comprised of a compound in accordance with claim 1 or a pharmaceutically acceptable salt, prodrug or hydrate thereof in combination with a carrier.
10. A method of treating or preventing cancer in a mammalian patient in need of such treatment which is comprised of admininstering to said patient an anti-cancer effective amount of a compound of claim 1.
11. A method of treating or preventing cancer in accordance with claim 8 wherein the cancer comprises cancers of the brain, genitourinary tract, lymphatic system, stomach, larynx and lung.
12. A method in accordance with claim 11 wherein the cancer comprises histiocytic lymphoma, lung adenocarcinoma, small cell lung cancers, pancreatic cancer, gioblastomas and breast carcinoma.
13. A method of treating or preventing a disease in which neoangiogenesis is implicated, which is comprised of administering to a mammalian patient in need of such treatment a compound of claim 1 or a pharmaceutically acceptable salt, prodrug or hydrate thereof in an amount which is effective for reducing neoangiogenesis.
14. A method in accordance with claim 13 wherein the disease is an ocular disease.
15. A method of treating or preventing retinal vascularization which is comprised of administering to a mammalian patient in need of such treatment a compound of claim 1 or a pharmaceutically acceptable salt, prodrug or hydrate thereof in an amount which is effective for treating retinal vascularization.
16. A method of treating or preventing diabetic retinopathy which is comprised of administering to a mammalian patient in need of such treatment a compound of claim 1 or a pharmaceutically acceptable salt, prodrug or hydrate thereof in an amount which is effective for treating diabetic retinopathy.
17. A method of treating or preventing age-related macular degeneration which is comprised of administering to a mammalian patient in need of such treatment a compound of claim 1 or a pharmaceutically acceptable salt, prodrug or hydrate thereof in an amount which is effective for inflammation.
18. A method of treating or preventing inflammatory diseases which is comprised of administering to a mammalian patient in need of such treatment a compound of claim 1 or a pharmaceutically acceptable salt, prodrug or hydrate thereof in an amount which is effective for inflammation.
19. A method according to claim 18 wherein the inflammatory disease comprises rheumatoid arthritis, psoriasis, contact dermatitis and delayed hypertensitivity reactions.
20. A method for inhibiting tyrosine kinase which comprises administering to a mammalian patient in need of such treatment a therapeutically effective amount of a composition of claim 1.
PCT/US1998/019789 1997-09-26 1998-09-22 Novel angiogenesis inhibitors WO1999016755A1 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
CA002303830A CA2303830A1 (en) 1997-09-26 1998-09-22 Novel angiogenesis inhibitors
EP98948427A EP1017682A4 (en) 1997-09-26 1998-09-22 Novel angiogenesis inhibitors
AU95003/98A AU744939B2 (en) 1997-09-26 1998-09-22 Novel angiogenesis inhibitors
JP2000513841A JP2001518470A (en) 1997-09-26 1998-09-22 Novel angiogenesis inhibitors

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US6015197P 1997-09-26 1997-09-26
US60/060,151 1997-09-26
GB9810544.8 1998-05-15
GBGB9810544.8A GB9810544D0 (en) 1998-05-15 1998-05-15 Novel angiogenesis inhibitors

Publications (1)

Publication Number Publication Date
WO1999016755A1 true WO1999016755A1 (en) 1999-04-08

Family

ID=26313694

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1998/019789 WO1999016755A1 (en) 1997-09-26 1998-09-22 Novel angiogenesis inhibitors

Country Status (5)

Country Link
EP (1) EP1017682A4 (en)
JP (1) JP2001518470A (en)
AU (1) AU744939B2 (en)
CA (1) CA2303830A1 (en)
WO (1) WO1999016755A1 (en)

Cited By (137)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001040217A1 (en) * 1999-11-30 2001-06-07 Pfizer Products Inc. Novel benzoimidazole derivatives useful as antiproliferative agents
WO2001057020A1 (en) * 2000-02-01 2001-08-09 Cor Therapeutics, Inc. INDOLE AND BENZIMIDAZOLE INHIBITORS OF FACTOR Xa
WO2001083485A1 (en) * 2000-04-28 2001-11-08 Bayer Aktiengesellschaft Imidazopyrimidine derivatives and triazolopyrimidine derivatives
EP1178791A2 (en) * 1999-05-07 2002-02-13 Johns Hopkins University School of Medicine The use of a protein tyrosine kinase pathway inhibitor in the treatment of ocular disorders
WO2003075921A3 (en) * 2002-03-05 2003-12-04 Transtech Pharma Inc Mono- and bicyclic azole derivatives that inhibit the interaction of ligands with rage
WO2004020431A2 (en) 2002-08-28 2004-03-11 Pfizer Products Inc. Novel benzoimidazole derivatives useful as antiproliferative agents
US6833456B2 (en) 2002-03-01 2004-12-21 Agouron Pharmaceuticals, Inc. Indolyl-urea derivatives of thienopyridines useful as antiangiogenic agents, and methods for their use
US6869962B2 (en) 2002-06-14 2005-03-22 Agouron Pharmaceuticals, Inc. Benzofused heterozryl amide derivatives of thienopyridines useful as therapeutic agents, pharmaceutical compositions including the same, and methods for their use
US6995171B2 (en) 2001-06-21 2006-02-07 Agouron Pharmaceuticals, Inc. Bicyclic pyrimidine and pyrimidine derivatives useful as anticancer agents
US7053107B2 (en) 2002-12-19 2006-05-30 Agouron Pharmaceuticals, Inc. Indazole compounds and pharmaceutical compositions for inhibiting protein kinases, and methods for their use
US7105530B2 (en) 2000-12-21 2006-09-12 Smithkline Beecham Corporation Pyrimidineamines as angiogenesis modulators
WO2007035744A1 (en) 2005-09-20 2007-03-29 Osi Pharmaceuticals, Inc. Biological markers predictive of anti-cancer response to insulin-like growth factor-1 receptor kinase inhibitors
US7208500B2 (en) 2003-08-29 2007-04-24 Agouron Pharmaceuticals, Inc. Thienopyridine-phenylacetamides and their derivatives useful as new anti-angiogenic agents
US7381824B2 (en) 2003-12-23 2008-06-03 Agouron Pharmaceuticals, Inc. Quinoline derivatives
US7446118B2 (en) 2005-11-30 2008-11-04 Roche Palo Alto Llc 3-amino-1-arylpropyl indoles and aza-substituted indoles and uses thereof
WO2009033094A2 (en) 2007-09-07 2009-03-12 Agensys, Inc. Antibodies and related molecules that bind to 24p4c12 proteins
US7531542B2 (en) 2005-05-18 2009-05-12 Wyeth Benzooxazole and benzothiazole antagonists of gonadotropin releasing hormone receptor
US7534796B2 (en) 2005-02-18 2009-05-19 Wyeth Imidazo[4,5-b]pyridine antagonists of gonadotropin releasing hormone receptor
US7538113B2 (en) 2005-02-18 2009-05-26 Wyeth 4-substituted imidazo[4,5-c]pyridine antagonists of gonadotropin releasing hormone receptor
US7560568B2 (en) 2004-01-28 2009-07-14 Smithkline Beecham Corporation Thiazole compounds
US7582634B2 (en) 2005-02-18 2009-09-01 Wyeth 7-substituted imidazo[4,5-c]pyridine antagonists of gonadotropin releasing hormone receptor
US7582636B2 (en) 2005-05-26 2009-09-01 Wyeth Piperazinylimidazopyridine and piperazinyltriazolopyridine antagonists of Gonadotropin Releasing Hormone receptor
US7598399B2 (en) 2005-11-30 2009-10-06 Roche Palo Alto Llc Methods for synthesis of 3-amino-1-arylpropyl indoles
US7622479B2 (en) * 2001-11-26 2009-11-24 Takeda Pharmaceutical Company Limited Bicyclic derivative, its production and use
US7638517B2 (en) 2005-11-30 2009-12-29 Roche Palo Alto Llc 3-Amino-1-arylpropyl azaindoles and uses thereof
US7666879B2 (en) 2005-02-24 2010-02-23 Eli Lilly And Company Imidazo[1,2-A]pyridine compounds as VEGF-R2 inhibitors
WO2010030360A1 (en) * 2008-09-11 2010-03-18 Arena Pharmaceuticals, Inc. 3H-IMIDAZO[4,5-b]PYRIDIN-5-OL DERIVATIVES USEFUL IN THE TREATMENT OF GPR81 RECEPTOR DISORDERS
US7696210B2 (en) 2004-06-17 2010-04-13 Wyeth Gonadotropin releasing hormone receptor antagonists
WO2010045495A2 (en) 2008-10-16 2010-04-22 University Of Pittsburgh-Of The Commonwealth System Of Higher Education Fully human antibodies to high molecular weight-melanoma associated antigen and uses thereof
US7714130B2 (en) 2004-06-17 2010-05-11 Wyeth Processes for preparing gonadotropin releasing hormone receptor antagonists
EP2194067A2 (en) 2001-01-05 2010-06-09 Pfizer Inc. Antibodies to insulin-like growth factor I receptor (IGF-IR)
WO2010090764A1 (en) 2009-02-09 2010-08-12 Supergen, Inc. Pyrrolopyrimidinyl axl kinase inhibitors
WO2010099364A2 (en) 2009-02-27 2010-09-02 Osi Pharmaceuticals, Inc. Methods for the identification of agents that inhibit mesenchymal-like tumor cells or their formation
WO2010099363A1 (en) 2009-02-27 2010-09-02 Osi Pharmaceuticals, Inc. Methods for the identification of agents that inhibit mesenchymal-like tumor cells or their formation
WO2010099137A2 (en) 2009-02-26 2010-09-02 Osi Pharmaceuticals, Inc. In situ methods for monitoring the emt status of tumor cells in vivo
WO2010099139A2 (en) 2009-02-25 2010-09-02 Osi Pharmaceuticals, Inc. Combination anti-cancer therapy
WO2010099138A2 (en) 2009-02-27 2010-09-02 Osi Pharmaceuticals, Inc. Methods for the identification of agents that inhibit mesenchymal-like tumor cells or their formation
WO2010098866A1 (en) 2009-02-27 2010-09-02 Supergen, Inc. Cyclopentathiophene/cyclohexathiophene dna methyltransferase inhibitors
US7825145B2 (en) 2001-06-18 2010-11-02 Biodiem Ltd Antimicrobial and radioprotective compounds
US7825132B2 (en) 2002-08-23 2010-11-02 Novartis Vaccines And Diagnostics, Inc. Inhibition of FGFR3 and treatment of multiple myeloma
US7838527B2 (en) 2002-11-13 2010-11-23 Novartis Vaccines And Diagnostics, Inc. Methods of treating cancer and related methods
EP2258700A1 (en) 2006-05-09 2010-12-08 Pfizer Products Inc. Cycloalkylamino acid derivatives and pharmaceutical compositions thereof
US7858643B2 (en) 2004-08-26 2010-12-28 Agouron Pharmaceuticals, Inc. Enantiomerically pure aminoheteroaryl compounds as protein kinase inhibitors
US7863305B2 (en) 2004-06-01 2011-01-04 Roche Palo Alto Llc 3-amino-1-arylpropyl indoles as monoamine reuptake inhibitors
WO2011008696A2 (en) 2009-07-13 2011-01-20 Genentech, Inc. Diagnostic methods and compositions for treatment of cancer
US7875624B2 (en) 2004-02-20 2011-01-25 Novartis Vaccines And Diagnostics, Inc. Modulating and measuring cellular adhesion
WO2011014726A1 (en) 2009-07-31 2011-02-03 Osi Pharmaceuticals, Inc. Mtor inhibitor and angiogenesis inhibitor combination therapy
EP2292233A2 (en) 1999-11-11 2011-03-09 OSI Pharmaceuticals, Inc. Pharmaceutical uses of N-(3-Ethynylphenyl)-6,7-bis(2-methoxyethoxy)-4-quinazolinamine
WO2011027249A2 (en) 2009-09-01 2011-03-10 Pfizer Inc. Benzimidazole derivatives
WO2011032013A1 (en) 2009-09-11 2011-03-17 Genentech, Inc. Method to identify a patient with an increased likelihood of responding to an anti-cancer agent
WO2011033006A1 (en) 2009-09-17 2011-03-24 F. Hoffmann-La Roche Ag Methods and compositions for diagnostics use in cancer patients
WO2011073521A1 (en) 2009-12-15 2011-06-23 Petri Salven Methods for enriching adult-derived endothelial progenitor cells and uses thereof
EP2343086A2 (en) 2001-11-09 2011-07-13 Pfizer Products Inc. Antibodies to CD40
WO2011098971A1 (en) 2010-02-12 2011-08-18 Pfizer Inc. Salts and polymorphs of 8-fluoro-2-{4-[(methylamino}methyl]phenyl}-1,3,4,5-tetrahydro-6h-azepino[5,4,3-cd]indol-6-one
WO2011109584A2 (en) 2010-03-03 2011-09-09 OSI Pharmaceuticals, LLC Biological markers predictive of anti-cancer response to insulin-like growth factor-1 receptor kinase inhibitors
WO2011109572A2 (en) 2010-03-03 2011-09-09 OSI Pharmaceuticals, LLC Biological markers predictive of anti-cancer response to insulin-like growth factor-1 receptor kinase inhibitors
WO2011153224A2 (en) 2010-06-02 2011-12-08 Genentech, Inc. Diagnostic methods and compositions for treatment of cancer
WO2012010550A1 (en) 2010-07-19 2012-01-26 F. Hoffmann-La Roche Ag Method to identify a patient with an increased likelihood of responding to an anti-cancer therapy
WO2012012750A1 (en) 2010-07-23 2012-01-26 Trustees Of Boston University ANTI-DEsupR INHIBITORS AS THERAPEUTICS FOR INHIBITION OF PATHOLOGICAL ANGIOGENESIS AND TUMOR CELL INVASIVENESS AND FOR MOLECULAR IMAGING AND TARGETED DELIVERY
WO2012010549A1 (en) 2010-07-19 2012-01-26 F. Hoffmann-La Roche Ag Method to identify a patient with an increased likelihood of responding to an anti-cancer therapy
WO2012042421A1 (en) 2010-09-29 2012-04-05 Pfizer Inc. Method of treating abnormal cell growth
EP2444421A1 (en) 2005-04-26 2012-04-25 Pfizer Inc. P-Cadherin antibodies
EP2444099A1 (en) 2005-03-31 2012-04-25 Agensys, Inc. Antibodies and related molecules that bind to 161P2F10B proteins
WO2012052948A1 (en) 2010-10-20 2012-04-26 Pfizer Inc. Pyridine- 2- derivatives as smoothened receptor modulators
EP2447283A2 (en) 2005-09-07 2012-05-02 Amgen Fremont Inc. Human monoclonal antibodies to activin receptor-like kinase-1 (ALK-1)
EP2476667A2 (en) 2003-02-26 2012-07-18 Sugen, Inc. Aminoheteroaryl compounds as protein kinase inhibitors
WO2012116040A1 (en) 2011-02-22 2012-08-30 OSI Pharmaceuticals, LLC Biological markers predictive of anti-cancer response to insulin-like growth factor-1 receptor kinase inhibitors in hepatocellular carcinoma
WO2012142164A1 (en) 2011-04-12 2012-10-18 The United States Of America, As Represented By The Secretary, Department Of Health & Human Services Human monoclonal antibodies that bind insulin-like growth factor (igf) i and ii
WO2012145183A2 (en) 2011-04-19 2012-10-26 Pfizer Inc. Combinations of anti-4-1bb antibodies and adcc-inducing antibodies for the treatment of cancer
WO2012149014A1 (en) 2011-04-25 2012-11-01 OSI Pharmaceuticals, LLC Use of emt gene signatures in cancer drug discovery, diagnostics, and treatment
WO2013042006A1 (en) 2011-09-22 2013-03-28 Pfizer Inc. Pyrrolopyrimidine and purine derivatives
WO2013050725A1 (en) 2011-10-04 2013-04-11 King's College London Ige anti -hmw-maa antibody
WO2013068902A1 (en) 2011-11-08 2013-05-16 Pfizer Inc. Methods of treating inflammatory disorders using anti-m-csf antibodies
WO2013152252A1 (en) 2012-04-06 2013-10-10 OSI Pharmaceuticals, LLC Combination anti-cancer therapy
US8580833B2 (en) 2009-09-30 2013-11-12 Transtech Pharma, Inc. Substituted imidazole derivatives and methods of use thereof
US8614216B2 (en) 2005-05-23 2013-12-24 Novartis Ag Crystalline and other forms of 4-amino-5-fluoro-3-[6-(4-methylpiperazin-1-yl)-1H-benzimidazol-2-yl]-1H-quinolin-2-one lactic acid salts
US8691849B2 (en) 2008-09-02 2014-04-08 Janssen Pharmaceuticals, Inc. 3-azabicyclo[3.1.0]hexyl derivatives as modulators of metabotropic glutamate receptors
US8691813B2 (en) 2008-11-28 2014-04-08 Janssen Pharmaceuticals, Inc. Indole and benzoxazine derivatives as modulators of metabotropic glutamate receptors
WO2014093383A1 (en) 2012-12-14 2014-06-19 Arrien Pharmaceuticals Llc Substituted 1h-pyrrolo [2,3-b] pyridine and 1h-pyrazolo [3, 4-b] pyridine derivatives as salt inducible kinase 2 (sik2) inhibitors
WO2014107209A2 (en) 2013-01-07 2014-07-10 Arog Pharmaceuticals, Llc Crenolanib for treating flt3 mutated proliferative disorders
US8841323B2 (en) 2006-03-15 2014-09-23 Janssen Pharmaceuticals, Inc. 1, 4-disubstituted 3-cyano-pyridone derivatives and their use as positive allosteric modulators of MGLUR2-receptors
US8906939B2 (en) 2007-03-07 2014-12-09 Janssen Pharmaceuticals, Inc. 3-cyano-4-(4-tetrahydropyran-phenyl)-pyridin-2-one derivatives
US8937060B2 (en) 2009-05-12 2015-01-20 Janssen Pharmaceuticals, Inc. 1,2,4-triazolo [4,3-A] pyridine derivatives and their use for the treatment of prevention of neurological and psychiatric disorders
US8946205B2 (en) 2009-05-12 2015-02-03 Janssen Pharmaceuticals, Inc. 1,2,4-triazolo[4,3-a]pyridine derivatives and their use as positive allosteric modulators of mGluR2 receptors
US8993591B2 (en) 2010-11-08 2015-03-31 Janssen Pharmaceuticals, Inc. 1,2,4-triazolo[4,3-a] pyridine derivatives and their use as positive allosteric modulators of MGLUR2 receptors
US9012448B2 (en) 2010-11-08 2015-04-21 Janssen Pharmaceuticals, Inc. 1,2,4-triazolo[4,3-a]pyridine derivatives and their use as positive allosteric modulators of MGLUR2 receptors
WO2015075598A1 (en) 2013-11-21 2015-05-28 Pfizer Inc. 2,6-substituted purine derivatives and their use in the treatment of proliferative disorders
US9067891B2 (en) 2007-03-07 2015-06-30 Janssen Pharmaceuticals, Inc. 1,4-disubstituted 3-cyano-pyridone derivatives and their use as positive allosteric modulators of mGluR2-receptors
US9085577B2 (en) 2009-05-12 2015-07-21 Janssen Pharmaceuticals, Inc. 7-aryl-1,2,4-triazolo[4,3-A]pyridine derivatives and their use as positive allosteric modulators of mGluR2 receptors
US9114138B2 (en) 2007-09-14 2015-08-25 Janssen Pharmaceuticals, Inc. 1′,3′-disubstituted-4-phenyl-3,4,5,6-tetrahydro-2H,1′H-[1,4′] bipyridinyl-2′-ones
WO2015149721A1 (en) 2014-04-04 2015-10-08 Crown Bioscience, Inc.(Taicang) Methods for determining responsiveness to mek/erk inhibitors
WO2015155624A1 (en) 2014-04-10 2015-10-15 Pfizer Inc. Dihydropyrrolopyrimidine derivatives
WO2015166373A1 (en) 2014-04-30 2015-11-05 Pfizer Inc. Cycloalkyl-linked diheterocycle derivatives
WO2016001789A1 (en) 2014-06-30 2016-01-07 Pfizer Inc. Pyrimidine derivatives as pi3k inhibitors for use in the treatment of cancer
WO2016019280A1 (en) 2014-07-31 2016-02-04 The Government Of The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Human monoclonal antibodies against epha4 and their use
US9271967B2 (en) 2010-11-08 2016-03-01 Janssen Pharmaceuticals, Inc. 1,2,4-triazolo[4,3-a]pyridine derivatives and their use as positive allosteric modulators of mGluR2 receptors
WO2016097918A1 (en) 2014-12-18 2016-06-23 Pfizer Inc. Pyrimidine and triazine derivatives and their use as axl inhibitors
US9480683B2 (en) 2012-09-26 2016-11-01 Arog Pharmaceuticals, Inc. Method of inhibiting mutant C-kit
WO2016178876A2 (en) 2015-05-01 2016-11-10 Cocrystal Pharma, Inc. Nucleoside analogs for treatment of the flaviviridae family of viruses and cancer
WO2017009751A1 (en) 2015-07-15 2017-01-19 Pfizer Inc. Pyrimidine derivatives
EP3135692A1 (en) 2010-06-16 2017-03-01 University of Pittsburgh of the Commonwealth System of Higher Education Antibodies to endoplasmin and their use
EP3170840A1 (en) 2003-09-10 2017-05-24 Warner-Lambert Company LLC Antibodies to m-csf
WO2017096165A1 (en) 2015-12-03 2017-06-08 Agios Pharmaceuticals, Inc. Mat2a inhibitors for treating mtap null cancer
US9708315B2 (en) 2013-09-06 2017-07-18 Janssen Pharmaceutica Nv 1,2,4-triazolo[4,3-a]pyridine compounds and their use as positive allosteric modulators of MGLUR2 receptors
EP2935244B1 (en) * 2012-12-21 2018-06-27 Selvita S.A. Novel benzimidazole derivatives as kinase inhibitors
US10106542B2 (en) 2013-06-04 2018-10-23 Janssen Pharmaceutica Nv Substituted 6,7-dihydropyrazolo[1,5-a]pyrazines as negative allosteric modulators of mGluR2 receptors
EP3409278A1 (en) 2011-07-21 2018-12-05 Tolero Pharmaceuticals, Inc. Heterocyclic protein kinase inhibitors
WO2019075367A1 (en) 2017-10-13 2019-04-18 Tolero Pharmaceuticals, Inc. Pkm2 activators in combination with reactive oxygen species for treatment of cancer
EP3344039A4 (en) * 2015-09-03 2019-08-28 Arizona Board of Regents on behalf of the University of Arizona Small molecule inhibitors of dyrk1a and uses thereof
US10422788B2 (en) 2016-12-19 2019-09-24 Tolero Pharmaceuticals, Inc. Profiling peptides and methods for sensitivity profiling
EP3545956A1 (en) 2013-04-18 2019-10-02 Arrien Pharmaceuticals LLC 3,5-(un)substituted-1h-pyrrolo[2,3-b]pyridine, 1h-pyrazolo[3,4-b]pyridine and 5h- pyrrolo[2,3-b]pyrazine dual itk and jak3 kinase inhibitors
US10463658B2 (en) 2013-10-25 2019-11-05 Videra Pharmaceuticals, Llc Method of inhibiting FLT3 kinase
EP3590932A1 (en) 2013-03-14 2020-01-08 Tolero Pharmaceuticals, Inc. Jak2 and alk2 inhibitors and methods for their use
US10537573B2 (en) 2014-01-21 2020-01-21 Janssen Pharmaceutica Nv Combinations comprising positive allosteric modulators or orthosteric agonists of metabotropic glutamatergic receptor subtype 2 and their use
US10562925B2 (en) 2015-05-18 2020-02-18 Tolero Pharmaceuticals, Inc. Alvocidib prodrugs having increased bioavailability
US10568887B2 (en) 2015-08-03 2020-02-25 Tolero Pharmaceuticals, Inc. Combination therapies for treatment of cancer
US10624880B2 (en) 2015-04-20 2020-04-21 Tolero Pharmaceuticals, Inc. Predicting response to alvocidib by mitochondrial profiling
US10766865B2 (en) 2012-10-16 2020-09-08 Sumitomo Dainippon Pharma Oncology, Inc. PKM2 modulators and methods for their use
US10780086B2 (en) 2012-09-21 2020-09-22 Arog Pharmaceuticals, Inc. Method of inhibiting constitutively active phosphorylated FLT3 kinase
WO2020198077A1 (en) 2019-03-22 2020-10-01 Sumitomo Dainippon Pharma Oncology, Inc. Compositions comprising pkm2 modulators and methods of treatment using the same
US10835525B2 (en) 2012-09-26 2020-11-17 Arog Pharmaceuticals, Inc. Method of inhibiting mutant C-KIT
US11034710B2 (en) 2018-12-04 2021-06-15 Sumitomo Dainippon Pharma Oncology, Inc. CDK9 inhibitors and polymorphs thereof for use as agents for treatment of cancer
US11040038B2 (en) 2018-07-26 2021-06-22 Sumitomo Dainippon Pharma Oncology, Inc. Methods for treating diseases associated with abnormal ACVR1 expression and ACVR1 inhibitors for use in the same
US11078541B2 (en) 2016-11-02 2021-08-03 Arog Pharmaceuticals, Inc. Crenolanib for treating FLT3 mutated proliferative disorders associated mutations
WO2021155006A1 (en) 2020-01-31 2021-08-05 Les Laboratoires Servier Sas Inhibitors of cyclin-dependent kinases and uses thereof
US11279694B2 (en) 2016-11-18 2022-03-22 Sumitomo Dainippon Pharma Oncology, Inc. Alvocidib prodrugs and their use as protein kinase inhibitors
US11369606B2 (en) 2014-01-21 2022-06-28 Janssen Pharmaceutica Nv Combinations comprising positive allosteric modulators or orthosteric agonists of metabotropic glutamatergic receptor subtype 2 and their use
US11465998B2 (en) 2019-04-25 2022-10-11 Regents Of The University Of Minnesota Therapeutic compounds and methods of use thereof
US11471451B2 (en) 2019-08-19 2022-10-18 Arog Pharmaceuticals, Inc. Uses of crenolanib
US11471456B2 (en) 2019-02-12 2022-10-18 Sumitomo Pharma Oncology, Inc. Formulations comprising heterocyclic protein kinase inhibitors
US11497756B2 (en) 2017-09-12 2022-11-15 Sumitomo Pharma Oncology, Inc. Treatment regimen for cancers that are insensitive to BCL-2 inhibitors using the MCL-1 inhibitor alvocidib
US11524006B2 (en) 2020-09-17 2022-12-13 Arog Pharmaceuticals, Inc. Crenolanib for treating TRK kinase associated proliferative disorders
US11602533B2 (en) 2018-12-13 2023-03-14 Arog Pharmaceuticals, Inc. Crenolanib combination therapy
US11642340B2 (en) 2012-09-26 2023-05-09 Arog Pharmaceuticals, Inc. Method of inhibiting mutant C-KIT
US11713310B2 (en) 2020-07-20 2023-08-01 Arog Pharmaceuticals, Inc. Crystal forms of crenolanib and methods of use thereof
US11793802B2 (en) 2019-03-20 2023-10-24 Sumitomo Pharma Oncology, Inc. Treatment of acute myeloid leukemia (AML) with venetoclax failure
US11857546B2 (en) 2021-10-22 2024-01-02 Arog Pharmaceuticals, Inc. Crenolanib for treating FLT3 mutated proliferative disorders relapsed/refractory to prior treatment

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20130076046A (en) * 2011-12-28 2013-07-08 한미약품 주식회사 Novel imidazopyridine derivatives as a tyrosine kinase inhibitor

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DK40192D0 (en) * 1992-03-26 1992-03-26 Neurosearch As IMIDAZOLE COMPOUNDS, THEIR PREPARATION AND USE
AU675484B2 (en) * 1993-03-24 1997-02-06 Neurosearch A/S Benzimidazole compounds, their use and preparation
GB9314884D0 (en) * 1993-07-19 1993-09-01 Zeneca Ltd Tricyclic derivatives
CA2267114A1 (en) * 1996-10-21 1998-04-30 Neurosearch A/S 1-phenyl-benzimidazole compounds and their use as gaba-a receptor modulators

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
Chemical Abstracts Service (C A S); 1 January 1900 (1900-01-01), XP002915126, Database accession no. 78-112663 *
See also references of EP1017682A4 *

Cited By (219)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1178791A2 (en) * 1999-05-07 2002-02-13 Johns Hopkins University School of Medicine The use of a protein tyrosine kinase pathway inhibitor in the treatment of ocular disorders
JP4920134B2 (en) * 1999-05-07 2012-04-18 ジョンズ ホプキンス ユニヴァーシティ スクール オヴ メディシン Use of protein tyrosine kinase pathway inhibitors in the treatment of eye disorders
JP2002544159A (en) * 1999-05-07 2002-12-24 ジョンズ ホプキンス ユニヴァーシティ スクール オヴ メディシン Use of protein tyrosine kinase pathway inhibitors in the treatment of ocular disorders
EP3100730A1 (en) 1999-11-11 2016-12-07 OSI Pharmaceuticals, LLC N-(3-ethynylphenyl)-6,7-bis(2-methoxyethoxy)-4-quinazolinamine for use in the treatment of nsclc
EP2292233A2 (en) 1999-11-11 2011-03-09 OSI Pharmaceuticals, Inc. Pharmaceutical uses of N-(3-Ethynylphenyl)-6,7-bis(2-methoxyethoxy)-4-quinazolinamine
AP1895A (en) * 1999-11-30 2008-10-08 Pfizer Prod Inc Novel benzoimidazole derivatives useful as antiproliferative agents.
EA005407B1 (en) * 1999-11-30 2005-02-24 Пфайзер Продактс Инк. Benzoimidazole derivatives useful as antiproliferative agent
CZ300789B6 (en) * 1999-11-30 2009-08-12 Pfizer Products Inc. Benzimidazole compound and pharmaceutical composition containing thereof for the treatment of abnormal cell growth in mammals
JP2007126468A (en) * 1999-11-30 2007-05-24 Pfizer Prod Inc New benzimidazole derivative useful as antiproliferative agent
US7019147B1 (en) 1999-11-30 2006-03-28 Pfizer Inc. Benzimidazole derivatives useful as antiproliferative agents
WO2001040217A1 (en) * 1999-11-30 2001-06-07 Pfizer Products Inc. Novel benzoimidazole derivatives useful as antiproliferative agents
WO2001057020A1 (en) * 2000-02-01 2001-08-09 Cor Therapeutics, Inc. INDOLE AND BENZIMIDAZOLE INHIBITORS OF FACTOR Xa
WO2001083485A1 (en) * 2000-04-28 2001-11-08 Bayer Aktiengesellschaft Imidazopyrimidine derivatives and triazolopyrimidine derivatives
JP2003535067A (en) * 2000-04-28 2003-11-25 バイエル アクチェンゲゼルシャフト Imidazopyrimidine derivatives and triazolopyrimidine derivatives
US6911443B2 (en) 2000-04-28 2005-06-28 Bayer Aktiengesellschaft Imidazopyrimidine derivatives and triazolopyrimidine derivatives
US7858626B2 (en) 2000-12-21 2010-12-28 Glaxosmithkline Llc Pyrimidineamines as angiogenesis modulators
US7105530B2 (en) 2000-12-21 2006-09-12 Smithkline Beecham Corporation Pyrimidineamines as angiogenesis modulators
US7262203B2 (en) 2000-12-21 2007-08-28 Smithkline Beecham Corporation Pyrimidineamines as angiogenesis modulators
US8114885B2 (en) 2000-12-21 2012-02-14 Glaxosmithkline Llc Chemical compounds
EP2796468A2 (en) 2001-01-05 2014-10-29 Pfizer Inc Antibodies to insulin-like growth factor I receptor
EP2194067A2 (en) 2001-01-05 2010-06-09 Pfizer Inc. Antibodies to insulin-like growth factor I receptor (IGF-IR)
US9234041B2 (en) 2001-01-05 2016-01-12 Pfizer Inc. Antibodies to insulin-like growth factor I receptor
US8569363B2 (en) 2001-06-18 2013-10-29 Biodiem Ltd. Antimicrobial and radioprotective compounds
US8158664B2 (en) 2001-06-18 2012-04-17 Biodiem Ltd. Antimicrobial and radioprotective compounds
US7825145B2 (en) 2001-06-18 2010-11-02 Biodiem Ltd Antimicrobial and radioprotective compounds
US9045452B2 (en) 2001-06-18 2015-06-02 Biodiem Ltd. Antimicrobial and radioprotective compounds
US6995171B2 (en) 2001-06-21 2006-02-07 Agouron Pharmaceuticals, Inc. Bicyclic pyrimidine and pyrimidine derivatives useful as anticancer agents
EP2343086A2 (en) 2001-11-09 2011-07-13 Pfizer Products Inc. Antibodies to CD40
US7622479B2 (en) * 2001-11-26 2009-11-24 Takeda Pharmaceutical Company Limited Bicyclic derivative, its production and use
US6833456B2 (en) 2002-03-01 2004-12-21 Agouron Pharmaceuticals, Inc. Indolyl-urea derivatives of thienopyridines useful as antiangiogenic agents, and methods for their use
US7714013B2 (en) 2002-03-05 2010-05-11 Transtech Pharma, Inc. Azole derivatives and fused bicyclic azole derivatives as therapeutic agents
WO2003075921A3 (en) * 2002-03-05 2003-12-04 Transtech Pharma Inc Mono- and bicyclic azole derivatives that inhibit the interaction of ligands with rage
US7361678B2 (en) 2002-03-05 2008-04-22 Transtech Pharma, Inc. Azole derivatives and fused bicyclic azole derivatives as therapeutic agents
US7737285B2 (en) 2002-03-05 2010-06-15 Transtech Pharma, Inc. Azole derivatives and fused bicyclic azole derivatives as therapeutic agents
US7045528B2 (en) 2002-06-14 2006-05-16 Agouron Pharmaceuticals, Inc. Benzofused heterozryl amide derivatives of thienopyridines useful as therapeutic agents, pharmaceutical compositions including the same, and methods for their use
US6869962B2 (en) 2002-06-14 2005-03-22 Agouron Pharmaceuticals, Inc. Benzofused heterozryl amide derivatives of thienopyridines useful as therapeutic agents, pharmaceutical compositions including the same, and methods for their use
US7825132B2 (en) 2002-08-23 2010-11-02 Novartis Vaccines And Diagnostics, Inc. Inhibition of FGFR3 and treatment of multiple myeloma
WO2004020431A2 (en) 2002-08-28 2004-03-11 Pfizer Products Inc. Novel benzoimidazole derivatives useful as antiproliferative agents
US7838527B2 (en) 2002-11-13 2010-11-23 Novartis Vaccines And Diagnostics, Inc. Methods of treating cancer and related methods
US7053107B2 (en) 2002-12-19 2006-05-30 Agouron Pharmaceuticals, Inc. Indazole compounds and pharmaceutical compositions for inhibiting protein kinases, and methods for their use
EP2476667A2 (en) 2003-02-26 2012-07-18 Sugen, Inc. Aminoheteroaryl compounds as protein kinase inhibitors
US7208500B2 (en) 2003-08-29 2007-04-24 Agouron Pharmaceuticals, Inc. Thienopyridine-phenylacetamides and their derivatives useful as new anti-angiogenic agents
US10280219B2 (en) 2003-09-10 2019-05-07 Amgen Fremont Inc. Antibodies to M-CSF
EP3170840A1 (en) 2003-09-10 2017-05-24 Warner-Lambert Company LLC Antibodies to m-csf
US9718883B2 (en) 2003-09-10 2017-08-01 Amgen Fremont Inc. Antibodies to M-CSF
US7923457B2 (en) 2003-12-23 2011-04-12 Agouron Pharmaceuticals Inc. Quinoline derivatives
US7381824B2 (en) 2003-12-23 2008-06-03 Agouron Pharmaceuticals, Inc. Quinoline derivatives
US7560568B2 (en) 2004-01-28 2009-07-14 Smithkline Beecham Corporation Thiazole compounds
US7875624B2 (en) 2004-02-20 2011-01-25 Novartis Vaccines And Diagnostics, Inc. Modulating and measuring cellular adhesion
US7863305B2 (en) 2004-06-01 2011-01-04 Roche Palo Alto Llc 3-amino-1-arylpropyl indoles as monoamine reuptake inhibitors
US7714130B2 (en) 2004-06-17 2010-05-11 Wyeth Processes for preparing gonadotropin releasing hormone receptor antagonists
US7696210B2 (en) 2004-06-17 2010-04-13 Wyeth Gonadotropin releasing hormone receptor antagonists
US8785632B2 (en) 2004-08-26 2014-07-22 Agouron Pharmaceuticals, Inc. Enantiomerically pure aminoheteroaryl compounds as protein kinase inhibitors
US7858643B2 (en) 2004-08-26 2010-12-28 Agouron Pharmaceuticals, Inc. Enantiomerically pure aminoheteroaryl compounds as protein kinase inhibitors
US7538113B2 (en) 2005-02-18 2009-05-26 Wyeth 4-substituted imidazo[4,5-c]pyridine antagonists of gonadotropin releasing hormone receptor
US7534796B2 (en) 2005-02-18 2009-05-19 Wyeth Imidazo[4,5-b]pyridine antagonists of gonadotropin releasing hormone receptor
US7582634B2 (en) 2005-02-18 2009-09-01 Wyeth 7-substituted imidazo[4,5-c]pyridine antagonists of gonadotropin releasing hormone receptor
US7666879B2 (en) 2005-02-24 2010-02-23 Eli Lilly And Company Imidazo[1,2-A]pyridine compounds as VEGF-R2 inhibitors
EP2444099A1 (en) 2005-03-31 2012-04-25 Agensys, Inc. Antibodies and related molecules that bind to 161P2F10B proteins
EP3300739A2 (en) 2005-03-31 2018-04-04 Agensys, Inc. Antibodies and related molecules that bind to 161p2f10b proteins
EP2444419A1 (en) 2005-04-26 2012-04-25 Pfizer Inc. P-Cadherin antibodies
EP2444421A1 (en) 2005-04-26 2012-04-25 Pfizer Inc. P-Cadherin antibodies
EP2444420A1 (en) 2005-04-26 2012-04-25 Pfizer Inc. P-Cadherin antibodies
US7531542B2 (en) 2005-05-18 2009-05-12 Wyeth Benzooxazole and benzothiazole antagonists of gonadotropin releasing hormone receptor
US8614216B2 (en) 2005-05-23 2013-12-24 Novartis Ag Crystalline and other forms of 4-amino-5-fluoro-3-[6-(4-methylpiperazin-1-yl)-1H-benzimidazol-2-yl]-1H-quinolin-2-one lactic acid salts
US7582636B2 (en) 2005-05-26 2009-09-01 Wyeth Piperazinylimidazopyridine and piperazinyltriazolopyridine antagonists of Gonadotropin Releasing Hormone receptor
US9221915B2 (en) 2005-09-07 2015-12-29 Pfizer Inc. Human monoclonal antibodies to activin receptor-like kinase-1
EP2447283A2 (en) 2005-09-07 2012-05-02 Amgen Fremont Inc. Human monoclonal antibodies to activin receptor-like kinase-1 (ALK-1)
EP2960253A1 (en) 2005-09-07 2015-12-30 Amgen Fremont Inc. Human monoclonal antibodies to activin receptor-like kinase-1
EP3381945A1 (en) 2005-09-07 2018-10-03 Amgen Fremont Inc. Human monoclonal antibodies to activin receptor-like kinase-1
EP2372363A1 (en) 2005-09-20 2011-10-05 OSI Pharmaceuticals, Inc. Biological markers predictive of anti-cancer response to insulin-like growth factor-1
WO2007035744A1 (en) 2005-09-20 2007-03-29 Osi Pharmaceuticals, Inc. Biological markers predictive of anti-cancer response to insulin-like growth factor-1 receptor kinase inhibitors
US7598399B2 (en) 2005-11-30 2009-10-06 Roche Palo Alto Llc Methods for synthesis of 3-amino-1-arylpropyl indoles
US7446118B2 (en) 2005-11-30 2008-11-04 Roche Palo Alto Llc 3-amino-1-arylpropyl indoles and aza-substituted indoles and uses thereof
US7803830B2 (en) 2005-11-30 2010-09-28 Roche Palo Alto Llc 3-amino-1-arylpropyl indoles and AZA-substituted indoles and uses thereof
US7638517B2 (en) 2005-11-30 2009-12-29 Roche Palo Alto Llc 3-Amino-1-arylpropyl azaindoles and uses thereof
US9266834B2 (en) 2006-03-15 2016-02-23 Janssen Pharmaceuticals, Inc. 1, 4-disubstituted 3-cyano-pyridone derivatives and their use as positive allosteric modulators of MGLUR2-receptors
US8841323B2 (en) 2006-03-15 2014-09-23 Janssen Pharmaceuticals, Inc. 1, 4-disubstituted 3-cyano-pyridone derivatives and their use as positive allosteric modulators of MGLUR2-receptors
EP2258700A1 (en) 2006-05-09 2010-12-08 Pfizer Products Inc. Cycloalkylamino acid derivatives and pharmaceutical compositions thereof
US9067891B2 (en) 2007-03-07 2015-06-30 Janssen Pharmaceuticals, Inc. 1,4-disubstituted 3-cyano-pyridone derivatives and their use as positive allosteric modulators of mGluR2-receptors
US8906939B2 (en) 2007-03-07 2014-12-09 Janssen Pharmaceuticals, Inc. 3-cyano-4-(4-tetrahydropyran-phenyl)-pyridin-2-one derivatives
WO2009033094A2 (en) 2007-09-07 2009-03-12 Agensys, Inc. Antibodies and related molecules that bind to 24p4c12 proteins
US11071729B2 (en) 2007-09-14 2021-07-27 Addex Pharmaceuticals S.A. 1′,3′-disubstituted-4-phenyl-3,4,5,6-tetrahydro-2H,1′H-[1,4′]bipyridinyl-2′-ones
US9132122B2 (en) 2007-09-14 2015-09-15 Janssen Pharmaceuticals, Inc. 1′,3′-disubstituted-4-phenyl-3,4,5,6-tetrahydro-2H,1′H-[1,4′]bipyridinyl-2′-ones
US9114138B2 (en) 2007-09-14 2015-08-25 Janssen Pharmaceuticals, Inc. 1′,3′-disubstituted-4-phenyl-3,4,5,6-tetrahydro-2H,1′H-[1,4′] bipyridinyl-2′-ones
US8691849B2 (en) 2008-09-02 2014-04-08 Janssen Pharmaceuticals, Inc. 3-azabicyclo[3.1.0]hexyl derivatives as modulators of metabotropic glutamate receptors
WO2010030360A1 (en) * 2008-09-11 2010-03-18 Arena Pharmaceuticals, Inc. 3H-IMIDAZO[4,5-b]PYRIDIN-5-OL DERIVATIVES USEFUL IN THE TREATMENT OF GPR81 RECEPTOR DISORDERS
US8507473B2 (en) 2008-09-11 2013-08-13 Arena Pharmaceuticals, Inc. 3H-imidazo[4,5-b]pyridin-5-ol derivatives useful in the treatment of GPR81 receptor disorders
WO2010045495A2 (en) 2008-10-16 2010-04-22 University Of Pittsburgh-Of The Commonwealth System Of Higher Education Fully human antibodies to high molecular weight-melanoma associated antigen and uses thereof
US8691813B2 (en) 2008-11-28 2014-04-08 Janssen Pharmaceuticals, Inc. Indole and benzoxazine derivatives as modulators of metabotropic glutamate receptors
WO2010090764A1 (en) 2009-02-09 2010-08-12 Supergen, Inc. Pyrrolopyrimidinyl axl kinase inhibitors
WO2010099139A2 (en) 2009-02-25 2010-09-02 Osi Pharmaceuticals, Inc. Combination anti-cancer therapy
WO2010099137A2 (en) 2009-02-26 2010-09-02 Osi Pharmaceuticals, Inc. In situ methods for monitoring the emt status of tumor cells in vivo
WO2010099364A2 (en) 2009-02-27 2010-09-02 Osi Pharmaceuticals, Inc. Methods for the identification of agents that inhibit mesenchymal-like tumor cells or their formation
WO2010099363A1 (en) 2009-02-27 2010-09-02 Osi Pharmaceuticals, Inc. Methods for the identification of agents that inhibit mesenchymal-like tumor cells or their formation
WO2010098866A1 (en) 2009-02-27 2010-09-02 Supergen, Inc. Cyclopentathiophene/cyclohexathiophene dna methyltransferase inhibitors
WO2010099138A2 (en) 2009-02-27 2010-09-02 Osi Pharmaceuticals, Inc. Methods for the identification of agents that inhibit mesenchymal-like tumor cells or their formation
US9737533B2 (en) 2009-05-12 2017-08-22 Janssen Pharmaceuticals. Inc. 1,2,4-triazolo [4,3-A] pyridine derivatives and their use for the treatment of prevention of neurological and psychiatric disorders
US9226930B2 (en) 2009-05-12 2016-01-05 Janssen Pharmaceuticals, Inc. 1,2,4-triazolo [4,3-a] pyridine derivatives and their use for the treatment of prevention of neurological and psychiatric disorders
US10071095B2 (en) 2009-05-12 2018-09-11 Janssen Pharmaceuticals, Inc. 1,2,4-triazolo [4,3-A] pyridine derivatives and their use for the treatment of neurological and psychiatric disorders
US8937060B2 (en) 2009-05-12 2015-01-20 Janssen Pharmaceuticals, Inc. 1,2,4-triazolo [4,3-A] pyridine derivatives and their use for the treatment of prevention of neurological and psychiatric disorders
US8946205B2 (en) 2009-05-12 2015-02-03 Janssen Pharmaceuticals, Inc. 1,2,4-triazolo[4,3-a]pyridine derivatives and their use as positive allosteric modulators of mGluR2 receptors
US9085577B2 (en) 2009-05-12 2015-07-21 Janssen Pharmaceuticals, Inc. 7-aryl-1,2,4-triazolo[4,3-A]pyridine derivatives and their use as positive allosteric modulators of mGluR2 receptors
WO2011008696A2 (en) 2009-07-13 2011-01-20 Genentech, Inc. Diagnostic methods and compositions for treatment of cancer
WO2011014726A1 (en) 2009-07-31 2011-02-03 Osi Pharmaceuticals, Inc. Mtor inhibitor and angiogenesis inhibitor combination therapy
WO2011027249A2 (en) 2009-09-01 2011-03-10 Pfizer Inc. Benzimidazole derivatives
WO2011032013A1 (en) 2009-09-11 2011-03-17 Genentech, Inc. Method to identify a patient with an increased likelihood of responding to an anti-cancer agent
WO2011033006A1 (en) 2009-09-17 2011-03-24 F. Hoffmann-La Roche Ag Methods and compositions for diagnostics use in cancer patients
US10363241B2 (en) 2009-09-30 2019-07-30 Vtv Therapeutics Llc Substituted imidazole derivatives and methods of use thereof
US8580833B2 (en) 2009-09-30 2013-11-12 Transtech Pharma, Inc. Substituted imidazole derivatives and methods of use thereof
US9598375B2 (en) 2009-09-30 2017-03-21 Vtv Therapeutics Llc Substituted imidazole derivatives and methods of use thereof
WO2011073521A1 (en) 2009-12-15 2011-06-23 Petri Salven Methods for enriching adult-derived endothelial progenitor cells and uses thereof
EP3597651A1 (en) 2010-02-12 2020-01-22 Pfizer Inc Salts and polymorphs of 8-fluoro-2-{4- [(methylamino)methyl]phenyl}-1 ,3,4,5-tetrahydro-6h-azepino[5,4,3- cd]indol-6-one
EP4166558A1 (en) 2010-02-12 2023-04-19 Pfizer Inc. Salts and polymorphs of 8-fluoro-2-{4- [(methylamino)methyl]phenyl}-1 ,3,4,5-tetrahydro-6h-azepino[5,4,3- cd]indol-6-one
WO2011098971A1 (en) 2010-02-12 2011-08-18 Pfizer Inc. Salts and polymorphs of 8-fluoro-2-{4-[(methylamino}methyl]phenyl}-1,3,4,5-tetrahydro-6h-azepino[5,4,3-cd]indol-6-one
EP3150610A1 (en) 2010-02-12 2017-04-05 Pfizer Inc Salts and polymorphs of 8-fluoro-2-{4-[(methylamino}methyl]phenyl}-1,3,4,5-tetrahydro-6h-azepino[5,4,3-cd]indol-6-one
WO2011109584A2 (en) 2010-03-03 2011-09-09 OSI Pharmaceuticals, LLC Biological markers predictive of anti-cancer response to insulin-like growth factor-1 receptor kinase inhibitors
WO2011109572A2 (en) 2010-03-03 2011-09-09 OSI Pharmaceuticals, LLC Biological markers predictive of anti-cancer response to insulin-like growth factor-1 receptor kinase inhibitors
WO2011153224A2 (en) 2010-06-02 2011-12-08 Genentech, Inc. Diagnostic methods and compositions for treatment of cancer
EP3135692A1 (en) 2010-06-16 2017-03-01 University of Pittsburgh of the Commonwealth System of Higher Education Antibodies to endoplasmin and their use
WO2012010548A1 (en) 2010-07-19 2012-01-26 F. Hoffmann-La Roche Ag Method to identify a patient with an increased likelihood of responding to an anti-cancer therapy
WO2012010549A1 (en) 2010-07-19 2012-01-26 F. Hoffmann-La Roche Ag Method to identify a patient with an increased likelihood of responding to an anti-cancer therapy
WO2012010550A1 (en) 2010-07-19 2012-01-26 F. Hoffmann-La Roche Ag Method to identify a patient with an increased likelihood of responding to an anti-cancer therapy
WO2012010551A1 (en) 2010-07-19 2012-01-26 F. Hoffmann-La Roche Ag Method to identify a patient with an increased likelihood of responding to an anti-cancer therapy
EP2848939A1 (en) 2010-07-19 2015-03-18 F. Hoffmann-La Roche AG Method to identify a patient with an increased likelihood of responding to an anti-cancer therapy
EP2848940A1 (en) 2010-07-19 2015-03-18 F. Hoffmann-La Roche AG Method to identify a patient with an increased likelihood of responding to an anti-cancer therapy
EP2824457A1 (en) 2010-07-19 2015-01-14 F. Hoffmann-La Roche AG Method to identify a patient with an increased likelihood of responding to an anti-cancer therapy
EP2801826A1 (en) 2010-07-19 2014-11-12 F. Hoffmann-La Roche AG Method to identify a patient with an increased likelihood of responding to an anti-cancer therapy
WO2012010547A1 (en) 2010-07-19 2012-01-26 F. Hoffmann-La Roche Ag Method to identify a patient with an increased likelihood of responding to an anti-cancer therapy
EP2866032A1 (en) 2010-07-19 2015-04-29 F. Hoffmann-La Roche AG Method to identify a patient with an increased likelihood of responding to an anti-cancer therapy
WO2012012750A1 (en) 2010-07-23 2012-01-26 Trustees Of Boston University ANTI-DEsupR INHIBITORS AS THERAPEUTICS FOR INHIBITION OF PATHOLOGICAL ANGIOGENESIS AND TUMOR CELL INVASIVENESS AND FOR MOLECULAR IMAGING AND TARGETED DELIVERY
EP3696195A1 (en) 2010-07-23 2020-08-19 Trustees of Boston University Anti-despr inhibitors as therapeutics for inhibition of pathological angiogenesis and tumor cell invasiveness and for molecular imaging and targeted delivery
WO2012042421A1 (en) 2010-09-29 2012-04-05 Pfizer Inc. Method of treating abnormal cell growth
WO2012052948A1 (en) 2010-10-20 2012-04-26 Pfizer Inc. Pyridine- 2- derivatives as smoothened receptor modulators
US9271967B2 (en) 2010-11-08 2016-03-01 Janssen Pharmaceuticals, Inc. 1,2,4-triazolo[4,3-a]pyridine derivatives and their use as positive allosteric modulators of mGluR2 receptors
US8993591B2 (en) 2010-11-08 2015-03-31 Janssen Pharmaceuticals, Inc. 1,2,4-triazolo[4,3-a] pyridine derivatives and their use as positive allosteric modulators of MGLUR2 receptors
US9012448B2 (en) 2010-11-08 2015-04-21 Janssen Pharmaceuticals, Inc. 1,2,4-triazolo[4,3-a]pyridine derivatives and their use as positive allosteric modulators of MGLUR2 receptors
WO2012116040A1 (en) 2011-02-22 2012-08-30 OSI Pharmaceuticals, LLC Biological markers predictive of anti-cancer response to insulin-like growth factor-1 receptor kinase inhibitors in hepatocellular carcinoma
WO2012142164A1 (en) 2011-04-12 2012-10-18 The United States Of America, As Represented By The Secretary, Department Of Health & Human Services Human monoclonal antibodies that bind insulin-like growth factor (igf) i and ii
WO2012145183A2 (en) 2011-04-19 2012-10-26 Pfizer Inc. Combinations of anti-4-1bb antibodies and adcc-inducing antibodies for the treatment of cancer
EP3536708A1 (en) 2011-04-19 2019-09-11 Pfizer Inc Combinations of anti-4-1bb antibodies and adcc-inducing antibodies for the treatment of cancer
WO2012149014A1 (en) 2011-04-25 2012-11-01 OSI Pharmaceuticals, LLC Use of emt gene signatures in cancer drug discovery, diagnostics, and treatment
US10875864B2 (en) 2011-07-21 2020-12-29 Sumitomo Dainippon Pharma Oncology, Inc. Substituted imidazo[1,2-B]pyridazines as protein kinase inhibitors
EP3812387A1 (en) 2011-07-21 2021-04-28 Sumitomo Dainippon Pharma Oncology, Inc. Heterocyclic protein kinase inhibitors
EP3409278A1 (en) 2011-07-21 2018-12-05 Tolero Pharmaceuticals, Inc. Heterocyclic protein kinase inhibitors
WO2013042006A1 (en) 2011-09-22 2013-03-28 Pfizer Inc. Pyrrolopyrimidine and purine derivatives
EP3275902A1 (en) 2011-10-04 2018-01-31 IGEM Therapeutics Limited Ige anti-hmw-maa antibody
WO2013050725A1 (en) 2011-10-04 2013-04-11 King's College London Ige anti -hmw-maa antibody
WO2013068902A1 (en) 2011-11-08 2013-05-16 Pfizer Inc. Methods of treating inflammatory disorders using anti-m-csf antibodies
WO2013152252A1 (en) 2012-04-06 2013-10-10 OSI Pharmaceuticals, LLC Combination anti-cancer therapy
US11738017B2 (en) 2012-09-21 2023-08-29 Arog Pharmaceuticals, Inc. Method of inhibiting constitutively active phosphorylated FLT3 kinase
US10780086B2 (en) 2012-09-21 2020-09-22 Arog Pharmaceuticals, Inc. Method of inhibiting constitutively active phosphorylated FLT3 kinase
US9889127B2 (en) 2012-09-26 2018-02-13 Arog Pharmaceuticals, Inc. Method of inhibiting mutant C-KIT
US10835525B2 (en) 2012-09-26 2020-11-17 Arog Pharmaceuticals, Inc. Method of inhibiting mutant C-KIT
US11642340B2 (en) 2012-09-26 2023-05-09 Arog Pharmaceuticals, Inc. Method of inhibiting mutant C-KIT
US9480683B2 (en) 2012-09-26 2016-11-01 Arog Pharmaceuticals, Inc. Method of inhibiting mutant C-kit
US10251877B2 (en) 2012-09-26 2019-04-09 Arog Pharmaceuticals, Inc. Method of inhibiting mutant C-KIT
US10766865B2 (en) 2012-10-16 2020-09-08 Sumitomo Dainippon Pharma Oncology, Inc. PKM2 modulators and methods for their use
WO2014093383A1 (en) 2012-12-14 2014-06-19 Arrien Pharmaceuticals Llc Substituted 1h-pyrrolo [2,3-b] pyridine and 1h-pyrazolo [3, 4-b] pyridine derivatives as salt inducible kinase 2 (sik2) inhibitors
EP2935244B1 (en) * 2012-12-21 2018-06-27 Selvita S.A. Novel benzimidazole derivatives as kinase inhibitors
US10174013B2 (en) 2012-12-21 2019-01-08 Selvita S.A. Benzimidazole derivatives as kinase inhibitors
WO2014107209A2 (en) 2013-01-07 2014-07-10 Arog Pharmaceuticals, Llc Crenolanib for treating flt3 mutated proliferative disorders
US9801870B2 (en) 2013-01-07 2017-10-31 Arog Pharmaceuticals, Inc. Crenolanib for treating FLT3 mutated proliferative disorders
US10213423B2 (en) 2013-01-07 2019-02-26 Arog Pharmaceuticals, Inc. Crenolanib for treating FLT3 mutated proliferative disorders
US9101624B2 (en) 2013-01-07 2015-08-11 Arog Pharmaceuticals, Inc. Crenolanib for treating FLT3 mutated proliferative disorders
US9393240B2 (en) 2013-01-07 2016-07-19 Arog Pharmaceuticals, Inc. Crenolanib for treating FLT3 mutated proliferative disorders
US11007188B2 (en) 2013-01-07 2021-05-18 Arog Pharmaceuticals, Inc. Crenolanib for treating FLT3 mutated proliferative disorders
US11458131B2 (en) 2013-01-07 2022-10-04 Arog Pharmaceuticals, Inc. Crenolanib for treating FLT3 mutated proliferative disorders
US10752594B2 (en) 2013-03-14 2020-08-25 Sumitomo Dainippon Pharma Oncology, Inc. JAK1 and ALK2 inhibitors and methods for their use
EP3590932A1 (en) 2013-03-14 2020-01-08 Tolero Pharmaceuticals, Inc. Jak2 and alk2 inhibitors and methods for their use
EP3545956A1 (en) 2013-04-18 2019-10-02 Arrien Pharmaceuticals LLC 3,5-(un)substituted-1h-pyrrolo[2,3-b]pyridine, 1h-pyrazolo[3,4-b]pyridine and 5h- pyrrolo[2,3-b]pyrazine dual itk and jak3 kinase inhibitors
US10106542B2 (en) 2013-06-04 2018-10-23 Janssen Pharmaceutica Nv Substituted 6,7-dihydropyrazolo[1,5-a]pyrazines as negative allosteric modulators of mGluR2 receptors
US10584129B2 (en) 2013-06-04 2020-03-10 Janssen Pharmaceuticals Nv Substituted 6,7-dihydropyrazolo[1,5-a]pyrazines as negative allosteric modulators of mGluR2 receptors
US9708315B2 (en) 2013-09-06 2017-07-18 Janssen Pharmaceutica Nv 1,2,4-triazolo[4,3-a]pyridine compounds and their use as positive allosteric modulators of MGLUR2 receptors
US10463658B2 (en) 2013-10-25 2019-11-05 Videra Pharmaceuticals, Llc Method of inhibiting FLT3 kinase
WO2015075598A1 (en) 2013-11-21 2015-05-28 Pfizer Inc. 2,6-substituted purine derivatives and their use in the treatment of proliferative disorders
US10537573B2 (en) 2014-01-21 2020-01-21 Janssen Pharmaceutica Nv Combinations comprising positive allosteric modulators or orthosteric agonists of metabotropic glutamatergic receptor subtype 2 and their use
US11369606B2 (en) 2014-01-21 2022-06-28 Janssen Pharmaceutica Nv Combinations comprising positive allosteric modulators or orthosteric agonists of metabotropic glutamatergic receptor subtype 2 and their use
US11103506B2 (en) 2014-01-21 2021-08-31 Janssen Pharmaceutica Nv Combinations comprising positive allosteric modulators or orthosteric agonists of metabotropic glutamatergic receptor subtype 2 and their use
WO2015149721A1 (en) 2014-04-04 2015-10-08 Crown Bioscience, Inc.(Taicang) Methods for determining responsiveness to mek/erk inhibitors
US10501805B2 (en) 2014-04-04 2019-12-10 Crown Bioscience, Inc. (Taicang) Methods for determining responsiveness to MEK/ERK inhibitors
EP4012049A2 (en) 2014-04-04 2022-06-15 Crown Bioscience, Inc. (Taicang) Methods for determining responsiveness to mek/erk inhibitors
WO2015155624A1 (en) 2014-04-10 2015-10-15 Pfizer Inc. Dihydropyrrolopyrimidine derivatives
EP3556757A1 (en) 2014-04-30 2019-10-23 Pfizer Inc Cycloalkyl-linked diheterocycle derivatives
WO2015166373A1 (en) 2014-04-30 2015-11-05 Pfizer Inc. Cycloalkyl-linked diheterocycle derivatives
WO2016001789A1 (en) 2014-06-30 2016-01-07 Pfizer Inc. Pyrimidine derivatives as pi3k inhibitors for use in the treatment of cancer
EP3473271A1 (en) 2014-07-31 2019-04-24 The Government of the United States of America as represented by the Secretary of the Department of Health and Human Services Human monoclonal antibodies against epha4 and their use
WO2016019280A1 (en) 2014-07-31 2016-02-04 The Government Of The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Human monoclonal antibodies against epha4 and their use
WO2016097918A1 (en) 2014-12-18 2016-06-23 Pfizer Inc. Pyrimidine and triazine derivatives and their use as axl inhibitors
US9593097B2 (en) 2014-12-18 2017-03-14 Pfizer Inc. Axl inhibitors
US10624880B2 (en) 2015-04-20 2020-04-21 Tolero Pharmaceuticals, Inc. Predicting response to alvocidib by mitochondrial profiling
WO2016178876A2 (en) 2015-05-01 2016-11-10 Cocrystal Pharma, Inc. Nucleoside analogs for treatment of the flaviviridae family of viruses and cancer
US10562925B2 (en) 2015-05-18 2020-02-18 Tolero Pharmaceuticals, Inc. Alvocidib prodrugs having increased bioavailability
WO2017009751A1 (en) 2015-07-15 2017-01-19 Pfizer Inc. Pyrimidine derivatives
US10568887B2 (en) 2015-08-03 2020-02-25 Tolero Pharmaceuticals, Inc. Combination therapies for treatment of cancer
US10835537B2 (en) 2015-08-03 2020-11-17 Sumitomo Dainippon Pharma Oncology, Inc. Combination therapies for treatment of cancer
US10682356B2 (en) 2015-08-03 2020-06-16 Tolero Pharmaceuticals, Inc. Combination therapies for treatment of cancer
EP3344039A4 (en) * 2015-09-03 2019-08-28 Arizona Board of Regents on behalf of the University of Arizona Small molecule inhibitors of dyrk1a and uses thereof
US10730842B2 (en) 2015-09-03 2020-08-04 Arizona Board Of Regents On Behalf Of The University Of Arizona Small molecule inhibitors of DYRK1A and uses thereof
WO2017096165A1 (en) 2015-12-03 2017-06-08 Agios Pharmaceuticals, Inc. Mat2a inhibitors for treating mtap null cancer
US11078541B2 (en) 2016-11-02 2021-08-03 Arog Pharmaceuticals, Inc. Crenolanib for treating FLT3 mutated proliferative disorders associated mutations
US11279694B2 (en) 2016-11-18 2022-03-22 Sumitomo Dainippon Pharma Oncology, Inc. Alvocidib prodrugs and their use as protein kinase inhibitors
US10422788B2 (en) 2016-12-19 2019-09-24 Tolero Pharmaceuticals, Inc. Profiling peptides and methods for sensitivity profiling
US11497756B2 (en) 2017-09-12 2022-11-15 Sumitomo Pharma Oncology, Inc. Treatment regimen for cancers that are insensitive to BCL-2 inhibitors using the MCL-1 inhibitor alvocidib
WO2019075367A1 (en) 2017-10-13 2019-04-18 Tolero Pharmaceuticals, Inc. Pkm2 activators in combination with reactive oxygen species for treatment of cancer
US11040038B2 (en) 2018-07-26 2021-06-22 Sumitomo Dainippon Pharma Oncology, Inc. Methods for treating diseases associated with abnormal ACVR1 expression and ACVR1 inhibitors for use in the same
US11034710B2 (en) 2018-12-04 2021-06-15 Sumitomo Dainippon Pharma Oncology, Inc. CDK9 inhibitors and polymorphs thereof for use as agents for treatment of cancer
US11530231B2 (en) 2018-12-04 2022-12-20 Sumitomo Pharma Oncology, Inc. CDK9 inhibitors and polymorphs thereof for use as agents for treatment of cancer
US11602533B2 (en) 2018-12-13 2023-03-14 Arog Pharmaceuticals, Inc. Crenolanib combination therapy
US11471456B2 (en) 2019-02-12 2022-10-18 Sumitomo Pharma Oncology, Inc. Formulations comprising heterocyclic protein kinase inhibitors
US11793802B2 (en) 2019-03-20 2023-10-24 Sumitomo Pharma Oncology, Inc. Treatment of acute myeloid leukemia (AML) with venetoclax failure
WO2020198077A1 (en) 2019-03-22 2020-10-01 Sumitomo Dainippon Pharma Oncology, Inc. Compositions comprising pkm2 modulators and methods of treatment using the same
US11712433B2 (en) 2019-03-22 2023-08-01 Sumitomo Pharma Oncology, Inc. Compositions comprising PKM2 modulators and methods of treatment using the same
US11465998B2 (en) 2019-04-25 2022-10-11 Regents Of The University Of Minnesota Therapeutic compounds and methods of use thereof
US11471451B2 (en) 2019-08-19 2022-10-18 Arog Pharmaceuticals, Inc. Uses of crenolanib
WO2021155006A1 (en) 2020-01-31 2021-08-05 Les Laboratoires Servier Sas Inhibitors of cyclin-dependent kinases and uses thereof
US11713310B2 (en) 2020-07-20 2023-08-01 Arog Pharmaceuticals, Inc. Crystal forms of crenolanib and methods of use thereof
US11524006B2 (en) 2020-09-17 2022-12-13 Arog Pharmaceuticals, Inc. Crenolanib for treating TRK kinase associated proliferative disorders
US11857546B2 (en) 2021-10-22 2024-01-02 Arog Pharmaceuticals, Inc. Crenolanib for treating FLT3 mutated proliferative disorders relapsed/refractory to prior treatment

Also Published As

Publication number Publication date
AU744939B2 (en) 2002-03-07
CA2303830A1 (en) 1999-04-08
AU9500398A (en) 1999-04-23
EP1017682A4 (en) 2000-11-08
EP1017682A1 (en) 2000-07-12
JP2001518470A (en) 2001-10-16

Similar Documents

Publication Publication Date Title
AU744939B2 (en) Novel angiogenesis inhibitors
AU760020B2 (en) Novel angiogenesis inhibitors
US6465484B1 (en) Angiogenesis inhibitors
US6162804A (en) Tyrosine kinase inhibitors
US6235741B1 (en) Angiogenesis inhibitors
EP0984692A1 (en) Novel angiogenesis inhibitors
AU2006223070B2 (en) Benzazole derivatives, compositions, and methods of use as B-secretase inhibitors
EP2328871B1 (en) Hedgehog pathway modulators
US6180643B1 (en) Aryl and heteroaryl substituted fused pyrrole antiinflammatory agents
US6265403B1 (en) Angiogenesis inhibitors
CA2336848A1 (en) Novel angiogenesis inhibitors
US7202245B2 (en) Substituted 4-amino[1,2,4]triazolo[4,3-a] quinoxalines
US20050075334A1 (en) Novel compounds
US6380203B1 (en) Angiogenesis inhibitors
US6228871B1 (en) Angiogenesis inhibitors
AU2011244844A1 (en) Benzazole derivatives, compositions, and methods of use as B-secretase inhibitors

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AL AM AU AZ BA BB BG BR BY CA CN CU CZ EE GD GE HR HU ID IL IS JP KG KR KZ LC LK LR LT LV MD MG MK MN MX NO NZ PL RO RU SG SI SK SL TJ TM TR TT UA US UZ VN YU

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): GH GM KE LS MW SD SZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
WWE Wipo information: entry into national phase

Ref document number: 1998948427

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2303830

Country of ref document: CA

Ref country code: CA

Ref document number: 2303830

Kind code of ref document: A

Format of ref document f/p: F

ENP Entry into the national phase

Ref country code: JP

Ref document number: 2000 513841

Kind code of ref document: A

Format of ref document f/p: F

NENP Non-entry into the national phase

Ref country code: KR

WWE Wipo information: entry into national phase

Ref document number: 95003/98

Country of ref document: AU

WWP Wipo information: published in national office

Ref document number: 1998948427

Country of ref document: EP

WWG Wipo information: grant in national office

Ref document number: 95003/98

Country of ref document: AU

WWW Wipo information: withdrawn in national office

Ref document number: 1998948427

Country of ref document: EP