WO2007009773A1 - Pyrazolo[1.5-a]pyrimidin-7-yl amine derivatives as protein kinase inhibitors - Google Patents

Pyrazolo[1.5-a]pyrimidin-7-yl amine derivatives as protein kinase inhibitors Download PDF

Info

Publication number
WO2007009773A1
WO2007009773A1 PCT/EP2006/007109 EP2006007109W WO2007009773A1 WO 2007009773 A1 WO2007009773 A1 WO 2007009773A1 EP 2006007109 W EP2006007109 W EP 2006007109W WO 2007009773 A1 WO2007009773 A1 WO 2007009773A1
Authority
WO
WIPO (PCT)
Prior art keywords
kinase
compounds
formula
compound
acid
Prior art date
Application number
PCT/EP2006/007109
Other languages
French (fr)
Inventor
Patricia Imbach
Pascal Furet
Georg Martiny-Baron
Original Assignee
Novartis Ag
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Novartis Ag filed Critical Novartis Ag
Priority to MX2008000898A priority Critical patent/MX2008000898A/en
Priority to BRPI0613870-5A priority patent/BRPI0613870A2/en
Priority to AU2006271924A priority patent/AU2006271924A1/en
Priority to JP2008521883A priority patent/JP2009501748A/en
Priority to US11/996,337 priority patent/US20080234284A1/en
Priority to EP06762704A priority patent/EP1910368A1/en
Priority to CA002615433A priority patent/CA2615433A1/en
Publication of WO2007009773A1 publication Critical patent/WO2007009773A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/08Drugs for disorders of the urinary system of the prostate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • A61P15/08Drugs for genital or sexual disorders; Contraceptives for gonadal disorders or for enhancing fertility, e.g. inducers of ovulation or of spermatogenesis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • A61P27/06Antiglaucoma agents or miotics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/02Antithrombotic agents; Anticoagulants; Platelet aggregation inhibitors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/08Vasodilators for multiple indications
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis

Definitions

  • the invention relates to pyrazolo[1,5-a]pyrimidin-7-yl amine derivatives, their use in the treatment of protein kinase dependent diseases, their use for the manufacture of pharmaceutical compositions for the treatment of said diseases, methods of use of pyrazolo[1 ,5-a]pyrimidin-7-yl amine derivatives in the treatment of said diseases, pharmaceutical preparations comprising pyrazolo[1 ,5-a]pyrimidin-7-yl amine derivatives for the treatment of said diseases, novel pyrazolo[1 ,5-a]pyrimidin-7-yl amine derivatives, processes for the manufacture of the novel pyrazolo[1 ,5-a]pyrimidin-7-yl amine derivatives and pharmaceutical preparations, the use or methods of use of the pyrazolo[1 ,5-a]pyrimidin- 7-yl amine derivatives as mentioned above, and/or these pyrazolo[1 ,5-a]pyrimidin-7-yl
  • Pyrazolo[1 ,5-a]pyrimidin-7-yl-amine derivatives have been reported in the literature as ligands of benzodiazepine receptors (e.g., S. Selleri et al., Bioorg. Med. Chem 7 (12), 2705- 11 (1999)), antagonists of the corticotropin releasing factor (EP 1097709), angiotensin Il receptor antagonists (e. g., S. Takeshi et al., Japn. Pharm. Bull.
  • pyrazolo[1,5-a]pyrimidin-7-ylamine scaffold can be also be used as a template for the design of potent kinase inhibitors.
  • the invention relates to a compound of the formula (I): wherein:
  • R 2 is benzyl; unsubstituted phenyl or phenyl substituted by one or two substituents chosen from the group consisting of halo, di-lower alkylaminoalkoxy, hydroxy, alkoxy, benzyloxy, cycloalkyl, amino, acetyl amino;
  • R 3 is H and R 4 is hydroxyalkyl or
  • R 3 and R 4 together with the nitrogen atom they are attached to represent morpholinyl, pyrrolidinyl, piperidinyl or piperazinyl, the heterocycles optionally being further substituted by up to four alkyl groups;
  • A is phenyl which is unsubstituted or substituted by one or more of the substituents chosen from the group consisting of mono-, di- or tri-lower alkoxy, di-lower alkylaminyl, di-lower alkylaminoalkoxy, morpholinyl which is optionally di-substituted by alkyl, piperidinyl which is optionally substituted by di-lower alkylaminyl, and piperazinyl which is optionally substituted by lower alkyl, lower alkoxy, lower alkyl piperazinyl, pyrrolidinyl, di-lower or alkylaminyl;
  • a preferred embodiment is a compound of formula I according to the above, wherein:
  • R 1 is H
  • R 2 is phenyl substituted by fluoro or chloro
  • R 3 is H and R 4 is hydroxyethyl or
  • R 3 and R 4 together with the nitrogen atom they are attached to represent piperazinyl
  • A is phenyl which is substituted with one or more of the substituents chosen from the group consisting of; mono-, di- or tri-methoxy, di-methylaminoethoxy and di-ethylamino piperidinyl or a pharmaceutical salt thereof.
  • a further embodiment is a compound of formula I according to the above, wherein:
  • R 2 is phenyl substituted by fluoro or chloro
  • R 3 and R 4 together with the nitrogen atom they are attached to represent piperazinyl
  • A is phenyl which is substituted with one or more of the substituents chosen from the group consisting of; mono-, di- or tri-methoxy, di-methylaminoethoxy and di-ethylamino piperidinyl or a pharmaceutical salt thereof.
  • a further embodiment is a compound of formula I according to the above, wherein:
  • R 1 is H
  • R 2 is phenyl substituted by fluoro or chloro
  • R 3 and R 4 together with the nitrogen atom they are attached to represent piperazinyl
  • A is phenyl which is substituted with one or more of the substituents chosen from the group consisting of; mono-, di- and tri-methoxy or a pharmaceutical salt thereof.
  • Another embodiment is a compound of formula I according to the above, wherein:
  • R 1 is H
  • R 2 is phenyl substituted by fluoro or chloro
  • R 3 is H and R 4 is hydroxyethyl
  • A is phenyl which is substituted with one or more of the substituents chosen from the group consisting of; mono-, di- and tri-methoxy or a pharmaceutical salt thereof.
  • the invention pertains to a compound of formula I for use in the treatment of the human or animal body.
  • Yet another embodiment is the use of a compound of formula I according to the above in the preparation of a pharmaceutical composition.
  • Yet another embodiment is a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of formula I according to the above. - A -
  • the pharmaceutical composition preferably comprises a compound of formula I according to the above and an acceptable pharmaceutical carrier.
  • a protein kinase dependent disease is preferably one that depends on c-Abl, Bcr-Abl, c-Kit, c-Raf, Flt-1 , Flt-3, Her-1 , KDR, PDGFR-kinase, c-Src, RET-receptor kinase, FGF-R1 , FGF- R2, FGF-R3, FGF-R4, Ephrin receptor kinases (e.
  • EphB2 kinase EphB4 kinase and related Eph kinases
  • casein kinases CK-1 , CK-2, G-CK
  • Pak ALK, ZAP70, Jak1 , Jak2, AxI, Cdk1 , cdk4, cdk5, Met, FAK 1 Pyk2, Syk, Insulin receptor kinase, Tie-2 or costitutively activating mutations of kinases (activating kinases) such as of Bcr-Abl, c-Kit, c-Raf, Flt-3, FGF-R3, PDGF-receptors, RET 1 and Met and (especially aberrantly highly expressed or activated) kinase-dependent disease or disease dependent on the activation of the kinase pathways, or a disease dependent on any two or more of the kinases just mentioned.
  • activating kinases such as of Bcr-Abl, c-K
  • a protein kinase dependent disease is more preferably one that depends on c-abl, Flt-3, KDR, c-Src, RET, EphB4, c-kit, cdk1 , FGFR-1 , c-raf, Her-1 , Ins-R or Tek.
  • the disease to be treated is a proliferative disease, preferably a benign or especially malignant tumor, more preferably carcinoma of the brain, kidney, liver, adrenal gland, bladder, breast, stomach (especially gastric tumors), ovaries, colon, rectum, prostate, pancreas, lung, vagina, thyroid, sarcoma, glioblastomas, multiple myeloma or gastrointestinal cancer, especially colon carcinoma or colorectal adenoma, or a tumor of the neck and head, an epidermal hyperproliferation, especially psoriasis, prostate hyperplasia, a neoplasia, especially of epithelial character, preferably mammary carcinoma, or a leukemia.
  • a proliferative disease preferably a benign or especially malignant tumor, more preferably carcinoma of the brain, kidney, liver, adrenal gland, bladder, breast, stomach (especially gastric tumors), ovaries, colon, rectum, prostate, pancreas, lung, vagina, thyroid, s
  • the disease to be treated is a disease which is triggered by persistent angiogenesis, such as psoriasis; Kaposi's sarcoma; restenosis, e.g., stent- induced restenosis; endometriosis; Crohn's disease; Hodgkin's disease; leukemia; arthritis, such as rheumatoid arthritis; hemangioma; angiofibroma; eye diseases, such as diabetic retinopathy and neovascular glaucoma; renal diseases, such as glomerulonephritis; diabetic nephropathy; malignant nephrosclerosis; thrombotic microangiopathic syndromes; transplant rejections and glomerulopathy; fibrotic diseases, such as cirrhosis of the liver; mesangial cell-proliferative diseases; arteriosclerosis; injuries of the nerve tissue.
  • persistent angiogenesis such as psoriasis; Kaposi's sarcoma; restenosis
  • the compounds of the present invention can also be used for inhibiting the re-occlusion of vessels after balloon catheter treatment, for use in vascular prosthetics or after inserting mechanical devices for holding vessels open, such as, e.g., stents, as immunosuppressants, as an aid in scar-free wound healing, and for treating age spots and contact dermatitis.
  • Alkyl includes lower alkyl preferably alkyl with up to 7 carbon atoms, preferably from 1 to and including 5, and is linear or branched; preferably, lower alkyl is pentyl, such as n-pentyl, butyl, such as n-butyl, sec-butyl, isobutyl, tert-butyl, propyl, such as n-propyl or isopropyl, ethyl or methyl.
  • Preferably lower alkyl is methyl, propyl or tert-butyl.
  • Alkyl can be substituted or unsubstituted, and when substituted may be with up to 3 substituents including other alkyl, cycloalkyl, alkenyl, alkynyl, any of the substituents defined above for aryl or any of the functional groups defined below.
  • Halo or halogen is preferably fluoro, chloro, bromo or iodo, most preferably fluoro, chloro or bromo.
  • Salts are especially the pharmaceutically acceptable salts of compounds of formula I.
  • Such salts are formed, for example, as acid addition salts, preferably with organic or inorganic acids, from compounds of formula (I) with a basic nitrogen atom, especially the pharmaceutically acceptable salts.
  • Suitable inorganic acids are, for example, halogen acids, such as hydrochloric acid, sulfuric acid, or phosphoric acid.
  • Suitable organic acids are, for example, carboxylic, phosphonic, sulfonic or sulfamic acids, for example acetic acid, propionic acid, octanoic acid, decanoic acid, dodecanoic acid, glycolic acid, lactic acid, fumaric acid, succinic acid, adipic acid, pimelic acid, suberic acid, azelaic acid, malic acid, tartaric acid, citric acid, amino acids, such as glutamic acid or aspartic acid, maleic acid, hydroxy- maleic acid, methylmaleic acid, cyclohexanecarboxylic acid, adamantanecarboxylic acid, benzoic acid, salicylic acid, 4-aminosalicylic acid, phthalic acid, phenylacetic acid, mandelic acid, cinnamic acid, methane- or ethane-sulfonic acid, 2-hydroxyethanesulfonic acid, ethane- 1 ,
  • salts may also be formed with bases, e.g. metal or ammonium salts, such as alkali metal or alkaline earth metal salts, for example sodium, potassium, magnesium or calcium salts, or ammonium salts with ammonia or suitable organic amines, such as tertiary monoamines, for example triethyl- amine or tri(2-hydroxyethyl)amine, or heterocyclic bases, for example N-ethyl-piperidine or N,N'-dimethylpiperazine.
  • bases e.g. metal or ammonium salts, such as alkali metal or alkaline earth metal salts, for example sodium, potassium, magnesium or calcium salts, or ammonium salts with ammonia or suitable organic amines, such as tertiary monoamines, for example triethyl- amine or tri(2-hydroxyethyl)amine, or heterocyclic bases, for example N-ethyl-piperidine or N,N'-dimethylpiperazine.
  • a compound of formula (I) may also form internal salts.
  • salts for isolation or purification purposes it is also possible to use pharmaceutically unacceptable salts, for example picrates or perchlorates.
  • pharmaceutically acceptable salts or free compounds are employed (where applicable in the form of pharmaceutical preparations), and these are therefore preferred.
  • any reference to the compounds hereinbefore and hereinafter especially the compounds of the formula I is to be understood as referring also to the corresponding tautomers of these compounds, especially of compounds of the formula I, tautomeric mixtures of these compounds, especially of compounds of the formula I, or salts of any of these, as appropriate and expedient and if not mentioned otherwise.
  • Any asymmetric carbon atom may be present in the (R)-, (S)- or (R.S)-configuration, preferably in the (R)- or (S)-configuration.
  • the compounds may thus be present as mixtures of isomers or preferably as pure isomers, preferably as enantiomer-pure diastereomers or pure enantiomers.
  • the present invention also relates to pro-drugs of a compound of formula (I) that convert in vivo to the compound of formula (I) as such. Any reference to a compound of formula (I) is therefore to be understood as referring also to the corresponding pro-drugs of the compound of formula (I), as appropriate and expedient.
  • Salts of compounds of formula (I) having at least one salt-forming group may be prepared in a manner known per se.
  • salts of compounds of formula (I) having acid groups may be formed, for example, by treating the compounds with metal compounds, such as alkali metal salts of suitable organic carboxylic acids, e.g. the sodium salt of 2-ethylhexanoic acid, with organic alkali metal or alkaline earth metal compounds, such as the corresponding hydroxides, carbonates or hydrogen carbonates, such as sodium or potassium hydroxide, carbonate or hydrogen carbonate, with corresponding calcium compounds or with ammonia or a suitable organic amine, stoichiometric amounts or only a small excess of the salt- forming agent preferably being used.
  • metal compounds such as alkali metal salts of suitable organic carboxylic acids, e.g. the sodium salt of 2-ethylhexanoic acid
  • organic alkali metal or alkaline earth metal compounds such as the corresponding hydroxides, carbonates or hydrogen carbonates,
  • Acid addition salts of compounds of formula (I) are obtained in customary manner, e.g. by treating the compounds with an acid or a suitable anion exchange reagent.
  • Internal salts of compounds of formula (I) containing acid and basic salt-forming groups, e.g. a free carboxy group and a free amino group, may be formed, e.g. by the neutralisation of salts, such as acid addition salts, to the isoelectric point, e.g. with weak bases, or by treatment with ion exchangers.
  • Salts can be converted in customary manner into the free compounds; metal and ammonium salts can be converted, for example, by treatment with suitable acids, and acid addition salts, for example, by treatment with a suitable basic agent.
  • diastereoisomers can be separated in a manner known per se into the individual isomers; diastereoisomers can be separated, for example, by partitioning between polyphasic solvent mixtures, recrystallisation and/or chromatographic separation, for example over silica gel or by e.g. medium pressure liquid chromatography over a reversed phase column, and racemates can be separated, for example, by the formation of salts with optically pure salt-forming reagents and separation of the mixture of diastereoisomers so obtainable, for example by means of fractional crystallisation, or by chromatography over optically active column materials. lntermediates and final products can be worked up and/or purified according to standard methods, e.g. using chromatographic methods, distribution methods, (re-) crystallization, and the like.
  • All the above-mentioned process steps can be carried out under reaction conditions that are known p_er se, preferably those mentioned specifically, in the absence or, customarily, in the presence of solvents or diluents, preferably solvents or diluents that are inert towards the reagents used and dissolve them, in the absence or presence of catalysts, condensation or neutralizing agents, for example ion exchangers, such as cation exchangers, e.g.
  • mixtures of isomers that are formed can be separated into the individual isomers, for example diastereoisomers or enantiomers, or into any desired mixtures of isomers, for example racemates or mixtures of diastereoisomers, for example analogously to the methods described under "Additional process steps”.
  • solvents from which those solvents that are suitable for any particular reaction may be selected include those mentioned specifically or, for example, water, esters, such as lower alkyl-lower alkanoates, for example ethyl acetate, ethers, such as aliphatic ethers, for example diethyl ether, or cyclic ethers, for example tetrahydrofurane or dioxane, liquid aromatic hydrocarbons, such as benzene or toluene, alcohols, such as methanol, ethanol or 1- or 2-propanol, nitriles, such as acetonitrile, halogenated hydrocarbons, such as methylene chloride or chloroform, acid amides, such as dimethylformamide or dimethyl acetamide, bases, such as heterocyclic nitrogen bases, for example pyridine or N-methylpyrrolidin-2-one, carboxylic acid anhydrides, such as lower alkanoic acid anhydrides, for example acetic anhydride,
  • the compounds, including their salts, may also be obtained in the form of hydrates, or their crystals may, for example, include the solvent used for crystallization. Different crystalline forms may be present.
  • the invention relates also to those forms of the process in which a compound obtainable as intermediate at any stage of the process is used as starting material and the remaining process steps are carried out, or in which a starting material is formed under the reaction conditions or is used in the form of a derivative, for example in protected form or in the form of a salt, or a compound obtainable by the process according to the invention is produced under the process conditions and processed further jn situ.
  • those starting materials are preferably used which result in new compounds of formula (I) described at the beginning as being especially valuable. Special preference is given to reaction conditions that are identical or analogous to those mentioned in the Examples.
  • the compounds of formula (I) have valuable pharmacological properties. They exhibit their biological activity for instance as inhibitors of different protein kinases, preferably inhibiting c- abl, Flt-3, KDR, c-Src, RET, EphB4, c-kit, cdk1 , FGFR-1 , c-raf, Her-1 , Ins-R or Tek, most preferably as inhibitors of Ephrin B4 receptor (EphB4) kinases. Therefore, the compound of the invention are useful in the treatment of kinase dependent diseases, e.g., as drugs to treat proliferative diseases.
  • treatment of tyrosine protein kinase dependent diseases refers to the prophylactic or preferably therapeutic (including palliative and/or curing) treatment of said diseases, especially of the diseases mentioned below.
  • the inhibition of RET is measured as follows:
  • the baculovirus donor vector pFB-GSTX3 is used to generate a recombinant baculovirus that expresses the amino acid region 658- 1072 (Swiss prot No. Q9BTB0) of the intra-cytoplasmic kinase domain of human RET- Men2A which corresponds to the wild-type kinase domain of RET (wtRET) and RET-Men2B, which differs from the wtRET by the activating mutation in the activation loop M918T.
  • the coding sequences for the cytoplasmic domain of wtRET and RET-Men2B are amplified by PCR from the plasmids pBABEpuro RET-Men2A and pBABEpuro RET-Men2B.
  • the amplified DNA fragments and the pFB-GSTX3 vector are made compatible for ligation by digestion with Sail and Kpnl. Ligation of these DNA fragments results in the baculovirus donor plasmid pFB-GX3-RET-Men2A and pFB-GX3-RET-Men2B, respectively.
  • Transfer vectors containing the kinase domains are transfected into the DhMOBac cell line (GIBCO) and plated on selective agar plates. Colonies without insertion of the fusion sequence into the viral genome (carried by the bacteria) are blue. Single, white colonies are picked and viral DNA (bacmid) are isolated from the bacteria by standard plasmid purification procedures. Sf9 cells or Sf21 (American Type Culture Collection) cells are then transfected in 25 cm 2 flasks with the viral DNA using Cellfectin reagent. Determination of small scale protein expression in Sf9 cells: Virus-containing media is collected from the transfected cell culture and used for infection to increase its titer.
  • Virus- containing media obtained after two rounds of infection is used for large-scale protein expression.
  • 100 cm 2 round tissue culture plates are seeded with 5 x 10 7 cells/plate and infected with 1 mL of virus-containing media (approximately 5 MOIs). After 3 days, the cells are scraped off the plate and centrifuged at 500 rpm for 5 minutes.
  • Cell pellets from 10-20, 100 cm 2 plates are re-suspended in 50 mL of ice-cold lysis buffer (25 mM tris-HCI, pH 7.5, 2 mM EDTA, 1% NP-40, 1 mM DTT, 1 mM P MSF). The cells are stirred on ice for 15 minutes and then centrifuged at 5,000 rpms for 20 minutes.
  • the centrifuged cell lysate is loaded onto a 2 mL glutathione-sepharose column (Pharmacia) and is washed 3 x with 10 mL of 25 mM tris-HCI, pH 7.5, 2 mM EDTA, 1 mM DTT, 200 mM NaCI.
  • the GST-tagged proteins are then eluted by 10 applications (1 mL each) of 25 mM tris-HCI, pH 7.5, 10 mM reduced-glutathione, 100 mM NaCI, 1 mM DTT, 10% glycerol and stored at -70 0 C.
  • Tyrosine protein kinase assays with either purified GST-wtRET or GST-RET-Men2B protein are carried out in a final volume of 30 ⁇ L containing 15 ng of either GST-wtRET or GST-RET-Men2B protein, 20 mM tris-HCI, pH 7.5, 1 mM MnCI2, 10 mM MgCI2, 1 mM DTT, 3 ⁇ g/mL poly(Glu,Tyr) 4:1 , 1% DMSO, 2.0 ⁇ M ATP ( ⁇ -[ 33 P]-ATP 0.1 ⁇ Ci).
  • the activity is assayed in the presence or absence of inhibitors, by measuring the incorporation of 33 P from [ ⁇ 33 P] ATP into poly(Glu.Tyr) 4:1.
  • the assay is carried out in 96- well plates at ambient temperature for 15 minutes under conditions described below and terminated by the addition of 20 ⁇ L of 125 mM EDTA. Subsequently, 40 ⁇ L of the reaction mixture are transferred onto Immobilon-PVDF membrane (Millipore) previously soaked for 5 minutes with methanol, rinsed with water, then soaked for 5 minutes with 0.5% H 3 PO 4 and mounted on vacuum manifold with disconnected vacuum source. After spotting all samples, vacuum is connected and each well-rinsed with 200 ⁇ L 0.5% H 3 PO 4 .
  • Membranes are removed and washed 4 x on a shaker with 1.0% H 3 PO 4 , once with ethanol. Membranes are counted after drying at ambient temperature, mounting in Packard TopCount 96-well frame, and addition of 10 ⁇ L/well of Microscint TM (Packard). IC 50 values are calculated by linear regression analysis of the percentage inhibition of each compound in duplicate, at 4 concentrations (usually 0.01 , 0.1 , 1 and 10 ⁇ M). One unit of protein kinase activity is defined as 1 nmole of 33 P ATP transferred from [ ⁇ 33 P] ATP to the substrate protein/minute/mg of protein at 37°C.
  • IC 50 values are calculated by logarithmic regression analysis of the percentage inhibition of each compound at 4 concentrations (usually 3- or 10-fold dilution series starting at 10 ⁇ M).
  • Normalized IC 50 measured IC 50 average ref. IC 50 / measured ref. IC 50
  • staurosporine or a synthetic staurosporine derivative are used as reference compounds.
  • the compounds of the formula (I) are found to show IC 5O values for RET inhibition in the range from 0.005-100 ⁇ M, preferably in the range from 0.01-2 ⁇ M.
  • a protein of 37 kD (c-Abl kinase) is purified by a two- step procedure over a Cobalt metal chelate column followed by an anion exchange column with a yield of 1-2 mg/L of Sf9 cells (Bhat et al., reference cited).
  • the purity of the c-Abl kinase is >90% as judged by SDS-PAGE after Coomassie blue stai-ning.
  • the assay contains (total volume of 30 ⁇ l_): c-Abl kinase (50 ng), 20 mM Tris HCI, pH 7.5, 10 mM MgCI2, 10 ⁇ M Na3VO4, 1 mM DTT and 0.06 ⁇ Ci/assay [ ⁇ 33 P]-ATP (5 ⁇ M ATP) using 30 ⁇ g/mL poly-Ala,Glu,Lys,Tyr-6:2:5:1 (PoIy-AEKY, Sigma P1152) in the presence of 1 % DMSO.
  • Reactions are terminated by adding 10 ⁇ l_ of 250 mM EDTA and 30 ⁇ L of the reaction mixture is transferred onto Immobilon-PVDF membrane (Millipore, Bedford, MA, USA) previously soaked for 5 min with methanol, rinsed with water, then soaked for 5 min with 0.5 % H3PO4 and mounted on vacuum manifold with disconnected vacuum source. After spotting all samples, vacuum is connected and each well rinsed with 200 ⁇ L 0.5 % H3PO4. Mem-branes are removed and washed on a shaker with 0.5 % H3PO4 (4 times) and once with ethanol.
  • Membranes are counted after drying at ambient temperature, mounting in Packard TopCount 96-well frame, and addition of 10 ⁇ L ⁇ /vell of Microscint TM (Packard). Using this test system, compounds of the formula I show IC 50 values of inhibition for c-Abl inhibition in the range of 0.002 to 100 M, usually between 0.002 and 5 M.
  • VEGF vascular endothelial growth factor
  • the cells are washed twice with ice-cold PBS (phosphate-buffered saline) and immediately lysed in 100 ⁇ l lysis buffer per well.
  • the lysates are then centrifuged to remove the cell nuclei, and the protein concentrations of the supernatants are determined using a commercial protein assay (BIORAD). The lysates can then either be immediately used or, if necessary, stored at -20 0 C.
  • a sandwich ELISA is carried out to measure the VEGF-R2 phosphorylation: a monoclonal antibody to VEGF-R2 (for example Mab 1495.12.14; ProQinase, Freiburg, Germany) is immobilized on black ELISA plates (OptiPlateTM HTRF-96 from Packard). The plates are then washed and the remaining free protein-binding sites are saturated with 3% TopBlock® (Juro, Cat. # TB232010) in phosphate buffered saline with Tween 20® (polyoxyethylen(20)- sorbitane monolaurate, ICI/Uniquema) (PBST).
  • TopBlock® Polyoxyethylen(20)- sorbitane monolaurate, ICI/Uniquema
  • the cell lysates (20 ⁇ g protein per well) are then incubated in these plates overnight at 4°C together with an antiphosphotyrosine antibody coupled with alkaline phosphatase (PY20:AP from Zymed).
  • PY20:AP alkaline phosphatase
  • the (plates are washed again and the) binding of the antiphosphotyrosine antibody to the captured phosphorylated receptor is then demonstrated using a luminescent AP substrate (CDP-Star, ready to use, with Emerald II; Applied Biosystems). The luminescence is measured in a Packard Top Count Microplate Scintillation Counter.
  • VEGF-induced VEGF-R2 phosphorylation 100 %.
  • the activity of the tested substances is calculated as percent inhibition of VEGF-induced VEGF-R2 phosphorylation, wherein the concentration of substance that induces half the maximum inhibition is defined as the IC 50 (inhibitory dose for 50% inhibition).
  • Compounds of the formula I here show an IC 50 in the range of 0.005 to 20 ⁇ M, preferably between 0.005 and 1 ⁇ M for KDR inhibition.
  • Flt3 kinase inhibition is determined as follows:
  • the baculovirus donor vector pFbacGOI (GIBCO) is used to generate a recombinant baculovirus expressing the amino acid region amino acids 563-993 of the cytoplasmic kinase domain of human Flt-3.
  • the coding sequence for the cytoplasmic domain of Flt-3 is amplified by PCR from human c-DNA libraries (Clontech).
  • the amplified DNA fragments and the pFbacGOI vector are made compatible for ligation by digestion with BamH1 and Hindlll. Ligation of these DNA fragments results in the baculovirus donor plasmid Flt-3(1.1).
  • the production of the viruses, the expression of proteins in Sf9 cells and the purification of the GST-fused proteins are performed as follows: Production of virus: Transfer vector (pFbacGOI -Flt-3) containing the Flt-3 kinase domain is transfected into the DHIOBac cell line (GIBCO) and the transfected cells are plated on selective agar plates. Colonies without insertion of the fusion sequence into the viral genome (carried by the bacteria) are blue. Single white colonies are picked and viral DNA (bacmid) is isolated from the bacteria by standard plasmid purification procedures. Sf9 or Sf21 cells (American Type Culture Collection) are then transfected in flasks with the viral DNA using Cellfectin reagent.
  • Transfer vector pFbacGOI -Flt-3 containing the Flt-3 kinase domain is transfected into the DHIOBac cell line (GIBCO) and the transfected cells are plated on selective agar plates. Colonies without insertion of
  • Virus containing media is collected from the transfected cell culture and used for infection to increase its titre. Virus containing media obtained after two rounds of infection is used for large-scale protein expression. For large-scale protein expression 100 cm 2 round tissue culture plates are seeded with 5 x 10 7 cells/plate and infected with 1 mL of virus-containing media (approx. 5 MOIs). After 3 days the cells are scraped off the plate and centrifuged at 500 rpm for 5 min.
  • Cell pellets from 10-20, 100 cm 2 plates, are resuspended in 50 mL of ice-cold lysis buffer (25 mMTris- HCI, pH 7.5, 2mM EDTA, 1% NP-40, 1 mM DTT 1 1 mM PMSF). The cells are stirred on ice for 15 min and then centrifuged at 5000 rpms for 20 min.
  • ice-cold lysis buffer 25 mMTris- HCI, pH 7.5, 2mM EDTA, 1% NP-40, 1 mM DTT 1 1 mM PMSF.
  • the centrifuged cell lysate is loaded onto a 2 mL gluta- thione-sepharose column (Pharmacia) and washed three times with 10 mL of 25 mM Tris- HCI, pH 7.5, 2 mM EDTA, 1 mM DTT, 200 mM NaCI.
  • the GST-tagged protein is then eluted by 10 applications (1 mL each) of 25 mM Tris-HCI, pH 7.5, 10 mM reduced-glutathione, 100 mM NaCI, 1 mM DTT, 10 % Glycerol and stored at -70 0 C.
  • Tyrosine protein kinase assays with purified GST-Flt-3 are carried out in a final volume of 30 ⁇ L containing 200-1800 ng of enzyme protein (depending on the specific activity), 20 mM Tris-HCI, pH 7.6, 3 mM MnCI 2 , 3 mM MgCI 2 , 1 mM DTT, 10 ⁇ M Na 3 VO 4 , 3 ⁇ g/mL poly(Glu.Tyr) 4:1 , 1 % DMSO, 8.0 ⁇ M ATP and 0.1 ⁇ Ci [ ⁇ 33 P] ATP).
  • the activity is assayed in the presence or absence of inhibitors, by measuring the incorporation of 33 P from [ ⁇ 33 P] ATP into the poly(Glu.Tyr) substrate.
  • the assay (30 ⁇ L) is carried out in 96-well plates at ambient temperature for 20 min under conditions described below and terminated by the addition of 20 ⁇ L of 125 mM EDTA. Subsequently, 40 ⁇ L of the reaction mixture is transferred onto Immobilon-PVDF membrane (Millipore, Bedford, MA, USA) previously soaked for 5 min with methanol, rinsed with water, then soaked for 5 min with 0.5 % H 3 PO 4 and mounted on vacuum manifold with disconnected vacuum source.
  • One unit of protein kinase activity is defined as 1 nmole of 33 P ATP transferred from [ ⁇ 33 P] ATP to the substrate protein per minute per mg of protein at 37 0 C.
  • the compounds of the formula I show IC 50 values for Flt-3 inhibition in the range between 0.01 and 100 ⁇ M, preferably between 0.05 and 10 ⁇ M.
  • the compounds of formula I also inhibit other tyrosine protein kinases such as especially the c-Src kinase, c-Kit, VEGF-R and/or FGFR; all of which play a part in growth regulation and transformation in animal, especially mammal cells, including human cells.
  • An appropriate assay is described in Andrejauskas-Buchdunger et al., Cancer Res.
  • compounds of the formula I show IC 50 values for inhibition of c-Src in the range of 0.005 to 100 ⁇ M, usually between 0.005 and 5 ⁇ M.
  • compounds of formula I also show IC 50 values for c-kit inhibition in the range of 0.005 to 10 ⁇ M, usually between 0.005 and 5 ⁇ M; and for inhibition of FGFR-1 , up to 95% inhibition at 10 ⁇ M.
  • the inhibition of IGF-1R and Ins-R can be determined as follows:
  • the baculovirus donor vector pfbgx3IGFIRcd is used to generate a recombinant baculovirus that expresses the amino acid region 950-1337 of the mature peptide cytoplasmic domain of the human IGF-IR.
  • the fragments of the human IGF-IR and Ins-R are cloned, expressed and small-scale purified as a factor Xa-cleavable glutathione-S-transferase (GST)-fusion protein using the Bac-to-BacTM system (GIBCO BRL) of recombinant baculovirus generation.
  • Virus containing media iss collected from the transfected cell culture and used for infection to increase its titer.
  • Virus containing media obtained after two rounds of infection iss used for large-scale protein expression.
  • Cell extracts are prepared and loaded onto a glutathione-Sepharose (Pharmacia) column. After washing, the GST-tagged proteins are then eluted with a glutathione-containing buffer.
  • Tyrosine protein kinase assays with purified GST-IGF-1 R and GST-lns-R are carried in a final volume of 30 ⁇ l containing 20 mM Tris-HCI, pH 7.6, 10 mM MgCI 2 , 0.01 mM Na 3 VO 4 , 1% DMSO, 1 mM DTT, 3 ⁇ g/ml poly(Glu, Tyr) 4:1 and 10 ⁇ M ATP ( ⁇ -[ 33 P]-ATP 0.1 ⁇ Ci).
  • the assay is performed in 96-well plates at ambient temperature for 20 min and terminated by addition of 25 ⁇ l 0.05 M EDTA pH 7.0.
  • the inhibition of Tek can be determined as follows: The procedure of the expression, purification and assay these kinases has been described. Fabbro et al., Pharmacol. Ther. 82(2-3) 293-301 (1999). In brief, the glutathione S-transferase (GST) gene from the pAcG1 vector (Pharmingen) is excised with EcoRV and EcoRI and inserted into the cloning site of the Fast-Bac baculoviral vector (GIBCO) creating a 5530 bp vector with N-terminal cloning sites derived from the pAcG1 fusion vector (FBGO).
  • GST glutathione S-transferase
  • the C-terminal cloning site may be any cloning site (from the Fast-Bac vector) downstream of the N-terminal cloning site used.
  • N- terminally GST-fused (pAcG1 , Pharmingen) KDR 1 Flt-1, Flk-1 , Tek and PDGFR- ⁇ kinase domains are obtained from ProQinase, Freiburg, Germany.
  • Tek is recloned into the FBG1 vector by EcoRI excision and ligation into EcoRI digested FBG1 (FBG1-Tek).
  • the coding sequences for the whole cytoplasmic domain of c-Kit (aa 544-976) and c-Fms (aa 538-972) are amplified by PCR from human uterus and from human bone marrow cDNA libraries (Clontech), respectively.
  • the amplified DNA fragments are fused to GST by cloning them into FBG1 as BamHI-EcoRI insertions, to yield FBG1-c-Kit and FBG1-c-Fms.
  • Tek is recloned into the FBGO transfer vector by EcoRI excision and ligation into EcoRI digested FBGO (FBG-Tie2/Tek).
  • FGFR-1 and c-met kinase domains are obtained by PCR from human A431 cells.
  • N-terminal primers contain an overhanging EcoRI site, while C-terminal primers contain a Xhol site to aid cloning into the transfer vectors.
  • cleavage products are gel-purified and ligated together to form the kinase constructs (FBG-Met, FBG-FGFR-1).
  • Viruses for each of the kinases are made according to the protocol supplied by GIBCO.
  • transfer vectors containing the kinase domains are transfected into the DHIOBac cell line (GIBCO), plated on agar plates containing the recommended concentrations of Blue- Gal, IPTG, Kanamycin, Tetracycline, and Gentamycin. Colonies without insertion of the fusion sequence into the viral genome (carried by the bacteria) are blue. A single white colony is usually picked and viral DNA (bacmid) isolated from the bacteria by standard plasmid mini prep procedures.
  • Sf9 cells or High Five cells are then transfected in 25 cm 2 flasks with the viral DNA using the Cellfectin reagent and protocol supplied with the Bac-to-Bac kit (GIBCO).
  • Virus containing media is collected from the transfected cell culture and used for infection to increase its titer. Virus containing media obtained after two rounds of infection is used for large-scale protein expression. For large-scale protein expression 100 cm 2 round tissue culture plates are seeded with 5x10 7 cells/plate and infected with 1 ml of virus-containing media (about 5 MOIs). After 3 days the cells are scraped off the plate and centrifuged at 500 rpm for 5 min.
  • Cell pellets from 10-20, 100 cm 2 plates, are resuspended in 50 ml of ice-cold lysis buffer (25 mM Tris-HCI, pH 7.5, 2 mM EDTA, 1 % NP-40, 1 mM DTT, 1 mM PMSF).
  • the cells are stirred on ice for 15 min and then centrifuged at 5000 rpms for 20 min.
  • the supernatant is loaded onto a 2 ml glutathione-sepharose column and washed three times with 10 ml of 25 mM Tris-HCI, pH 7.5, 2 mM EDTA 1 1 mM DTT, 200 mM NaCI.
  • the GST-tagged proteins are then eluted by 10 applications (1 ml each) of 25 mM Tris-HCI, pH 7.5, 10 mM reduced- glutathione, 100 mM NaCI, 1 mM DTT, 10% Glycerol and stored at -70 0 C.
  • the assays (30 ⁇ l) contain 200-1800 ng of enzyme protein (depending on the specific activity), 20 mM Tris-HCI, pH 7.6, 3 mM MnCI 2 , 3 mM MgCI 2, 1 mM DTT, 10 ⁇ M Na 3 VO 4 , 3 ⁇ g/ml poly(Glu.Tyr) 4:1 , 8 ⁇ M ATP ( ⁇ -[ 33 P]-ATP 0.1 ⁇ Ci).
  • Cdk1/cycB Cdk1/cycB are obtained from ProQinase, Freiburg, Germany. Starfish oocytes are induced to enter M phase of the cell cycle with 10 ⁇ M 1-methyladenine and frozen in liquid nitrogen and stored at - 80 0 C. When required, the oocytes are homogenized and centrifuged as described (Arion et al., Cell 55: 371-378 (1988) and Rialet et al., Anticancer Res. 11 : 1581-1590 (1991)).
  • Cdk1/cycB kinase is purified on p9 CKShs -sepharose beads and eluted with recombinant human p9 CKShs as described (Azzi et al., Eur. J. Biochem. 203: 353-360. (1992)). Briefly, the supernatant from oocytes is equilibrated for 30 min at 4°C under constant rotation with the p9 CKShs -sepharose beads. The beads are extensively washed and active cdk1/cycB kinase is eluted with purified p ⁇ CKShs (3 mg/ml).
  • Cdk1/cycB The activity of Cdk1/cycB is measured as described (Arion et al., Cell 55: 371-378 (1988), Meijer et al., EMBO J. 1989; 8: 2275-2282 and Meijer et al., EMBO J. 1991 ; 8: 2275-2282).
  • the assay is carried with slight modifications in 96-well plates at ambient temperature for 20 min.
  • the final volume of 30 ⁇ l contains 0.1-0.3U of Cdk1/cycB, 1 mg/ml histone H1 as a substrate, 60 mM ⁇ -glycerophosphate, 30 mM nitrophenylphosphate, 25 mM MOPS, 5 mM EGTA, 15 mM MgCI 2 , 1 mM DTT, 0.1 mM Na 3 VO 4, 15 ⁇ M ATP and 0.1 ⁇ Ci ⁇ - 33 P-ATP (75 ⁇ M, 8800 cpm/pmole).
  • the reaction is terminated by addition of 25 ⁇ l 0.05 M EDTA pH 7.0.
  • the inhibition of c-Raf-1 can be determined as follows: Production of recombinant c-Raf- 1 protein, is obtained by triple infection of Sf21 cells with GST-c-Raf-1 recombinant baculovirus together with v-Src and v-Ras recombinant baculoviruses that are required for active c-Raf-1 kinase production (Williams et al., PNAS 1992; 89: 2922-2926).
  • v- Ras Active Ras
  • c-Raf-1 is required to recruit c-Raf-1 to the cell membrane and v-Src to phosphorylate c-Raf-1 to fully activate it
  • Cells were seeded at 2.5 x 10 7 cells per 150 mm dish and allowed to attach to a 150 mm dish for 1hr at RT.
  • Media SF900II containing 10 % FBS
  • GST-C-Raf-1 , v- Ras and v-Src are added at MOI of 3.0, 2.5 and 2.5 receptively in a total volume of 4-5 mL.
  • Infected cells are incubated for 1 hr at RT and then 15 mL of medium is added. Infected cells are incubated for 48-72 hr at 27 0 C. Infected Sf21 cells are scraped and collected into a 50 mL tube and centrifuged for 10 min at 4 0 C at 1100 g in a Sorvall centrifuge. The cell pellet is washed once with ice cold PBS and lysed with 0.6 mL lysis buffer per 2.5 x 10 7 cells. Complete lysis of cells is achieved after 10 min on ice with occasional pipetting.
  • the cell lysates are centrifuged for 10 min at 4 0 C at 14,500 g in a Sorvall centrifuge with SS-34 rotor and the supernatant is transferred to a fresh tube and stored at -80 0 C.
  • c-Raf-1 is purified from cell lysates using 100 uL of packed Glutathione-Sepharose 4B beads equilibrated in ice cold PBS per 2.5 x 10 7 cells. GST-c-Raf-1 was allowed to bind to the beads at 4 0 C for 1hr with rocking. Bound GST-c-Raf-1 with beads was transferred to a column.
  • the column is washed once with lysis buffer and twice with ice cold Tris buffered saline. Ice cold elution buffer is added and column flow is stopped to allow the free glutathione to disrupt the interaction of GST-c-Raf-1 with glutathione sepharose beads. Fractions (1 mL) are collected into pre-chilled tubes. Each tube contains 10 % glycerol (final concentration) to maintain kinase activity during freeze thaw cycles. Purified fractions of GST-c-Raf-1 kinase protein are stored at -80 0 C.
  • IKB was used as substrate for the c-Raf-1 kinase.
  • IKB is expressed in bacteria as a His- tagged protein (cloned and kindly provided by Dr. Eder; ABM, Novartis, Basel).
  • BL21 LysS bacteria containing the IKB plasmid are grown to an OD 6 oo of 0.6 in LB medium then induced to express the kb with IPTG (final concentration of 1 mM) for 3 hrs at 37° C and then bacteria are lysed by sonication (microtip limit setting for 3 times at 1 min each in sonication buffer [50 mM Tris pH 8.0, 1 mM DTT, 1 mM EDTA] and centrifuged at 10,000 g for 15 min.
  • sonication buffer 50 mM Tris pH 8.0, 1 mM DTT, 1 mM EDTA
  • the supernatant is mixed with ammonium sulfate to give a final concentration of 30 %.
  • This mixture is rocked for 15 min at 4 0 C then spun at 10,000 g for 15 min.
  • the pellet is resuspended in binding buffer (Novagen) containing 10 mM BSA. This solution is applied to Ni-agarose (Novagen) and washed according to the Novagen manual.
  • IKB is eluted from the column using elution buffer (0.4 M imidazole, 0.2 M NaCI, 8 mM Tris pH 7.9). Fractions containing protein are dialysed in 50 mM Tris pH 8, 1 mM DTT.
  • c-Raf-1 protein kinase The activity of c-Raf-1 protein kinase is assayed in the presence or absence of inhibitors, by measuring the incorporation of 33 P from [ ⁇ 33 P] ATP into IB.
  • the assay is carried out in 96- well plates at ambient temperature for 60 min. It contains (total volume of 30 ⁇ l): c-rafl 1 kinase (400 ng), 25 mM Tris HCI, pH 7.5, 5 mM MgCI 2 , 5 mM MnCI 2 , 10 ⁇ M Na 3 VO 4 , 1 mM DTT and 0.3 ⁇ Ci/assay [ ⁇ 33 P]-ATP (10 ⁇ M ATP) using 600 ng IB in the presence of 1 % DMSO.
  • Reactions are terminated by adding 10 ⁇ L of 250 mM EDTA and 30 ⁇ L of the reaction mixture is transferred onto Immobilon-PVDF membrane (Millipore, Bedford, MA, USA) previously soaked for 5 min with methanol, rinsed with water, then soaked for 5 min with 0.5 % H 3 PO 4 and mounted on vacuum manifold with disconnected vacuum source. After spotting all samples, vacuum is connected and each well rinsed with 200 ⁇ L 0.5 % H 3 PO 4 . Membranes are removed and washed 4 x on a shaker with 0.5 % H 3 PO 4 , once with ethanol.
  • Membranes are counted after drying at ambient temperature, mounting in Packard TopCount 96-well frame, and addition of 10 ⁇ L/well of Microscint TM (Packard).
  • Compounds of formula (I) show c-Raf-1 inhibition in the range between 0.1-50 ⁇ M, preferably between 0.1 and 10 ⁇ M.
  • Ephrin B4 receptor (EphB4) kinases can be demonstrated as follows: Generation of Bac-to-BacTM (Invitrogen Life Technologies, Basel, Switzerland) GST-fusion expression vectors: Entire cytoplasmatic coding regions of the EphB-class are amplified by PCR from cDNA libraries derived from human placenta or brain, respectively. Recombinant baculovirus are generated that express the amino acid region 566-987 of the human EphB4 receptor (SwissProt Database, Accession No. P54760).
  • GST sequence is cloned into pFastBad® vector (Invitrogen Life Technologies, Basel, Switzerland) and PCR amplified.
  • cDNAs encoding EphB4- receptor domains, respectively are cloned in frame 3'prime to the GST sequence into this modified FastBad vector to generate pBac-to-BacTM donor vectors.
  • Single colonies arising from the transformation are inoculated to give overnight cultures for small scale plasmid preparation. Restriction enzyme analysis of plasmid DNA reveals several clones to contain inserts of the expected size. By automated sequencing the inserts and approximately 50 bp of the flanking vector sequences are confirmed on both strands.
  • Viruses for each of the kinases are made according to the protocol supplied by GIBCO if not stated otherwise.
  • transfer vectors containing the kinase domains are transfected into the DHIOBac cell line (GIBCO) and plated on selective agar plates. Colonies without insertion of the fusion sequence into the viral genome (carried by the bacteria) are blue. Single white colonies are picked and viral DNA (bacmid) isolated from the bacteria by standard plasmid purification procedures. Sf9 cells or Sf21 cells are then transfected in 25 cm2 flasks with the viral DNA using Cellfectin reagent according to the protocol.
  • GST-tagged kinases The centrifuged cell lysate is loaded onto a 2 mL glutathione-sepharose column (Pharmacia) and washed three times with 10 mL of 25 mM Tris-HCI, pH 7.5, 2mM EDTA, 1 mM DTT 1 200 mM NaCI. The GST-tagged proteins are then eluted by 10 applications (1 mL each) of 25 mM Tris-HCI, pH 7.5, 10 mM reduced- glutathione, 100 mM NaCI, 1 mM DTT, 10 % Glycerol and stored at -70 0 C.
  • Protein kinase assays The activities of protein kinases are assayed in the presence or absence of inhibitors, by measuring the incorporation of 33P from [D33PJATP into a polymer of glutamic acid and tyrosine (poly(Glu.Tyr)) as a substrate.
  • the kinase assays with purified GST-EphB (30ng) are carried out for 15-30 min at ambient temperature in a final volume of 30 HL containing 20 mM Tris-HCI , pH 7.5, 10 mM MgCI2, 3-50 mM MnCI2, 0.01 mM Na3VO4, 1 % DMSO, 1 mM DTT, 3 ⁇ g/mL poly(Glu.Tyr) 4:1 (Sigma; St. Louis, Mo., USA) and 2.0-3.0 ⁇ M ATP ( ⁇ -[33P]-ATP 0.1 ⁇ Ci).
  • the assay is terminated by the addition of 20 ⁇ L of 125 mM EDTA.
  • IC 50 values are calculated by linear regression analysis of the percentage inhibition of each compound in duplicate, at four concentrations (usually 0.01 , 0.1 , 1 and 10 ⁇ M).
  • One unit of protein kinase activity is defined as 1 nmole of 33P ATP transferred from [ ⁇ 33P] ATP to the substrate protein per minute per mg of protein at 37 0 C.
  • Compounds of formula I show EphB4 inhibition down to 1 nM, preferably IC50 values between 0.001-5.0 ⁇ M.
  • Ligand induced autophosphorylation is induced by the addition of 1 microg/ml soluble ephrinB2-Fc (s-ephrinB2- Fc : R&D Biosystems, CatNr 496-EB) and 0.1 microM ortho-vanadate. After a further 20 minutes incubation at 37°C, the cells are washed twice with ice-cold PBS (phosphate-buffered saline) and immediately lysed in 200 ⁇ l lysis buffer per well. The lysates are then centrifuged to remove the cell nuclei, and the protein concentrations of the supernatants are determined using a commercial protein assay (PIERCE). The lysates can then either be immediately used or, if necessary, stored at -20 0 C.
  • PBS phosphate-buffered saline
  • a sandwich ELISA is carried out to measure the EphB4 phosphorylation: To capture phosphorylated EphB4 proteinlOO ng/well of ephrinB2-Fc (s-ephrinB2-Fc : R&D Biosystems, CatNr 496-EB) is immobilized MaxiSorb (Nunc) ELISA plates. The plates are then washed and the remaining free protein-binding sites are saturated with 3% TopBlock® (Juro, Cat. # TB232010) in phosphate buffered saline with Tween 20® (polyoxyethylen(20)sorbitane monolaurate, ICI/Uniquema) (PBST).
  • TopBlock® Polyoxyethylen(20)sorbitane monolaurate, ICI/Uniquema
  • the cell lysates (100 ⁇ g protein per well) are then incubated in these plates for 1 h at room temperature. After washing the wells three times with PBS an antiphosphotyrosine antibody coupled with alkaline phosphatase (PY 20 Alkaline Phosphate conjugated: ZYMED, Cat NrO3-7722) is added and incubated for another hour. The plates are washed again and the binding of the antiphosphotyrosine antibody to the captured phosphorylated receptor is then demonstrated and quantified using 10 mM D- nitrophenylphosphat as subtrate and measuring the OD at 405 nm after 0.5h-1h.
  • PY 20 Alkaline Phosphate conjugated ZYMED, Cat NrO3-7722
  • the activity of the tested substances is calculated as percent inhibition of maximal EphB4 phosphorylation, wherein the concentration of substance that induces half the maximum inhibition is defined as the IC 50 (inhibitory dose for 50% inhibition).
  • mice are killed, and the chambers are removed.
  • the vascularized tissue growing around the chamber is carefully removed and weighed, and the blood content is assessed by measuring the hemoglobin content of the tissue (Drabkins method; Sigma, Deisenhofen, Germany). It has been shown previously that these growth factors induce dose-dependent increases in weight and blood content of this tissue growing (characterized histologically to contain fibroblasts and small blood vessels) around the chambers and that this response is blocked by antibodies that specifically neutralize VEGF (see Wood JM et al., Cancer Res.
  • the invention relates also to pharmaceutical compositions comprising a compound of formula (I), to their use in the therapeutic (in a broader aspect of the invention also prophylactic) treatment or a method of treatment of a kinase dependent disease, especially the preferred diseases mentioned above, to the compounds for said use and to the preparation of pharmaceutical preparations, especially for said uses.
  • the present invention also relates to pro-drugs of a compound of formula (I) that convert in vivo to the compound of formula (I) as such. Any reference to a compound of formula (I) is therefore to be understood as referring also to the corresponding pro-drugs of the compound of formula (I), as appropriate and expedient.
  • the pharmacologically acceptable compounds of the present invention may be used, for example, for the preparation of pharmaceutical compositions that comprise an effective amount of a compound of the formula (I), or a pharmaceutically acceptable salt thereof, as active ingredient together or in admixture with a significant amount of one or more inorganic or organic, solid or liquid, pharmaceutically acceptable carriers.
  • compositions according to the invention are those for enteral, such as nasal, rectal or oral, or parenteral, such as intramuscular or intravenous, administration to warm-blooded animals (especially a human), that comprise an effective dose of the pharmacologically active ingredient, alone or together with a significant amount of a pharmaceutically acceptable carrier.
  • the dose of the active ingredient depends on the species of warm-blooded animal, the body weight, the age and the individual condition, . individual pharmacokinetic data, the disease to be treated and the mode of administration.
  • the invention relates also to a method of treatment for a disease that responds to inhibition of a kinase; which comprises administering an (against the mentioned disease) prophylactically or especially therapeutically effective amount of a compound of formula (l)according to the invention, especially to a warm-blooded animal, for example a human, that, on account of one of the mentioned diseases, requires such treatment.
  • the dose of a compound of the formula (I) or a pharmaceutically acceptable salt thereof to be administered to warm-blooded animals is preferably from approximately 3 mg to approximately 10 g, more preferably from approximately 10 mg to approximately 1.5 g, most preferably from about 100 mg to about 1000 mg /person/day, divided preferably into 1-3 single doses which may, for example, be of the same size. Usually, children receive half of the adult dose.
  • the pharmaceutical compositions comprise from approximately 1% to approximately 95%, preferably from approximately 20% to approximately 90%, active ingredient.
  • Pharmaceutical compositions according to the invention may be, for example, in unit dose form, such as in the form of ampoules, vials, suppositories, dragees, tablets or capsules.
  • compositions of the present invention are prepared in a manner known per se, for example by means of conventional dissolving, lyophilizing, mixing, granulating or confectioning processes.
  • Solutions of the active ingredient, and also suspensions, and especially isotonic aqueous solutions or suspensions are preferably used, it being possible, for example in the case of lyophilized compositions that comprise the active ingredient alone or together with a carrier, for example mannitol, for such solutions or suspensions to be produced prior to use.
  • the pharmaceutical compositions may be sterilized and/or may comprise excipients, for example preservatives, stabilizers, wetting and/or emulsifying agents, solubilizers, salts for regulating the osmotic pressure and/or buffers, and are prepared in a manner known per se, for example by means of conventional dissolving or lyophilizing processes.
  • the said solutions or suspensions may comprise viscosity-increasing substances, such as sodium carboxymethylcellulose, carboxymethylcellulose, dextran, polyvinylpyrrolidone or gelatin.
  • Suspensions in oil comprise as the oil component the vegetable, synthetic or semi-synthetic oils customary for injection purposes.
  • liquid fatty acid esters that contain as the acid component a long-chained fatty acid having from 8- 22, especially from 12-22, carbon atoms, for example lauric acid, tridecylic acid, myristic acid, pentadecylic acid, palmitic acid, margaric acid, stearic acid, arachidic acid, behenic acid or corresponding unsaturated acids, for example oleic acid, elaidic acid, erucic acid, brasidic acid or linoleic acid, if desired with the addition of antioxidants, for example vitamin E, ⁇ -carotene or 3,5-di-tert-butyl-4-hydroxytoluene.
  • the alcohol component of those fatty acid esters has a maximum of 6 carbon atoms and is a mono- or poly-hydroxy, for example a mono-, di- or tri-hydroxy, alcohol, for example methanol, ethanol, propanol, butanol or pentanol or the isomers thereof, but especially glycol and glycerol.
  • fatty acid esters are therefore to be mentioned: ethyl oleate, isopropyl myristate, isopropyl palmitate, "Labrafil M 2375” (polyoxyethylene glycerol trioleate, Gattefosse, Paris), "Miglyol 812” (triglyceride of saturated fatty acids with a chain length of C8 to C12, HuIs AG, Germany), but especially vegetable oils, such as cottonseed oil, almond oil, olive oil, castor oil, sesame oil, soybean oil and more especially groundnut oil.
  • vegetable oils such as cottonseed oil, almond oil, olive oil, castor oil, sesame oil, soybean oil and more especially groundnut oil.
  • compositions for oral administration can be obtained by combining the active ingredient with solid carriers, if desired granulating a resulting mixture, and processing the mixture, if desired or necessary, after the addition of appropriate excipients, into tablets, dragee cores or capsules. It is also possible for them to be incorporated into plastics carriers that allow the active ingredients to diffuse or be released in measured amounts.
  • Suitable carriers are especially fillers, such as sugars, for example lactose, saccharose, mannitol or sorbitol, cellulose preparations and/or calcium phosphates, for example tricalcium phosphate or calcium hydrogen phosphate, and binders, such as starch pastes using for example corn, wheat, rice or potato starch, gelatin, tragacanth, methylcellulose, hydroxypropylmethylcellulose, sodium carboxymethylcellulose and/or polyvinylpyrrolidone, and/or, if desired, disintegrators, such as the above-mentioned starches, and/or carboxymethyl starch, crosslinked polyvinylpyrrolidone, agar, alginic acid or a salt thereof, such as sodium alginate.
  • fillers such as sugars, for example lactose, saccharose, mannitol or sorbitol
  • cellulose preparations and/or calcium phosphates for example tricalcium phosphate or calcium hydrogen phosphate
  • Excipients are especially flow conditioners and lubricants, for example silicic acid, talc, stearic acid or salts thereof, such as magnesium or calcium stearate, and/or polyethylene glycol.
  • Dragee cores are provided with suitable, optionally enteric, coatings, there being used, inter alia, concentrated sugar solutions which may comprise gum arabic, talc, polyvinylpyrrolidone, polyethylene glycol and/or titanium dioxide, or coating solutions in suitable organic solvents, or, for the preparation of enteric coatings, solutions of suitable cellulose preparations, such as ethylcellulose phthalate or hydroxypropylmethylcellulose phthalate.
  • Capsules are dry-filled capsules made of gelatin and soft sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol.
  • the dry-filled capsules may comprise the active ingredient in the form of granules, for example with fillers, such as lactose, binders, such as starches, and/or glidants, such as talc or magnesium stearate, and if desired with stabilizers.
  • the active ingredient is preferably dissolved or suspended in suitable oily excipients, such as fatty oils, paraffin oil or liquid polyethylene glycols, it being possible also for stabilizers and/or antibacterial agents to be added.
  • Dyes or pigments may be added to the tablets or dragee coatings or the capsule casings, for example for identification purposes or to indicate different doses of active ingredient.
  • a compound of the formula (I) may also be used to advantage in combination with other antiproliferative agents.
  • antiproliferative agents include, but are not limited to aro- matase inhibitors; antiestrogens; topoisomerase I inhibitors; topoisomerase Il inhibitors; microtubule active agents; alkylating agents; histone deacetylase inhibitors; compounds which induce cell differentiation processes; cyclooxygenase inhibitors; MMP inhibitors; mTOR inhibitors; antineoplastic antimetabolites; platin compounds; compounds targeting/decreasing a protein or lipid kinase activity and further anti-angiogenic compounds; compounds which target, decrease or inhibit the activity of a protein or lipid phosphatase; gonadorelin agonists; anti-androgens; methionine aminopeptidase inhibitors; bisphosphonates; biological response modifiers; antiproliferative antibodies; heparanase inhibitors; inhibitors of Ras oncogenic isoforms; telomerase inhibitors; proteasome inhibitors; agents used in the treatment of hematologic malign
  • aromatase inhibitor as used herein relates to a compound which inhibits the estrogen production, i.e. the conversion of the substrates androstenedione and testosterone to estrone and estradiol, respectively.
  • the term includes, but is not limited to steroids, especially atamestane, exemestane and formestane and, in particular, non-steroids, especially aminoglutethimide, roglethimide, pyridoglutethimide, trilostane, testolactone, ketokonazole, vorozole, fadrozole, anastrozole and letrozole.
  • Exemestane can be administered, e.g., in the form as it is marketed, e.g.
  • AROMASIN Formestane can be administered, e.g., in the form as it is marketed, e.g. under the trademark LENTARON. Fadrozole can be administered, e.g., in the form as it is marketed, e.g. under the trademark AFEMA. Anastrozole can be administered, e.g., in the form as it is marketed, e.g. under the trademark ARIMIDEX. Letrozole can be administered, e.g., in the form as it is marketed, e.g. under the trademark FEMARA or FEMAR. Aminoglutethimide can be administered, e.g., in the form as it is marketed, e.g. under the trademark ORIMETEN.
  • a combination of the invention comprising a chemotherapeutic agent which is an aromatase inhibitor is particularly useful for the treatment of hormone receptor positive tumors, e.g. breast tumors.
  • antiestrogen as used herein relates to a compound which antagonizes the effect of estrogens at the estrogen receptor level.
  • the term includes, but is not limited to tamoxifen, fulvestrant, raloxifene and raloxifene hydrochloride.
  • Tamoxifen can be admi- nistered, e.g., in the form as it is marketed, e.g. under the trademark NOLVADEX.
  • Raloxifene hydrochloride can be administered, e.g., in the form as it is marketed, e.g. under the trademark EVISTA.
  • Fulvestrant can be formulated as disclosed in US 4,659,516 or it can be administered, e.g., in the form as it is marketed, e.g. under the trademark FASLODEX.
  • a combination of the invention comprising a chemotherapeutic agent which is an antiestrogen is particularly useful for the treatment of estrogen receptor positive tumors, e.g. breast tumors.
  • anti-androgen as used herein relates to any substance which is capable of inhibiting the biological effects of androgenic hormones and includes, but is not limited to, bicalutamide (CASODEX), which can be formulated, e.g. as disclosed in US 4,636,505.
  • CASODEX bicalutamide
  • gonadorelin agonist includes, but is not limited to abarelix, go- serelin and goserelin acetate.
  • Goserelin is disclosed in US 4,100,274 and can be administered, e.g., in the form as it is marketed, e.g. under the trademark ZOLADEX.
  • Abarelix can be formulated, e.g. as disclosed in US 5,843,901.
  • topoisomerase I inhibitor includes, but is not limited to topotecan, gimatecan, irinotecan, camptothecian and its analogues, 9-nitrocamptothecin and the macromolecular camptothecin conjugate PNU-166148 (compound A1 in WO99/ 17804).
  • Irinotecan can be administered, e.g. in the form as it is marketed, e.g. under the trademark CAMPTOSAR.
  • Topotecan can be administered, e.g., in the form as it is marketed, e.g. under the trademark HYCAMTIN.
  • topoisomerase Il inhibitor includes, but is not limited to the an- thracyclines such as doxorubicin (including liposomal formulation, e.g. CAELYX), dauno- rubicin, epirubicin, idarubicin and nemorubicin, the anthraquinones mitoxantrone and lo- soxantrone, and the podophillotoxines etoposide and teniposide.
  • Etoposide can be administered, e.g. in the form as it is marketed, e.g. under the trademark ETOPOPHOS.
  • Teniposide can be administered, e.g. in the form as it is marketed, e.g.
  • Doxorubicin can be administered, e.g. in the form as it is marketed, e.g. under the trademark ADRIBLASTIN or ADRIAMYCIN.
  • Epirubicin can be administered, e.g. in the form as it is marketed, e.g. under the trademark FARMORUBICIN.
  • Idarubicin can be administered, e.g. in the form as it is marketed, e.g. under the trademark ZAVEDOS.
  • Mitoxantrone can be administered, e.g. in the form as it is marketed, e.g. under the trademark NOVANTRON.
  • microtubule active agent relates to microtubule stabilizing, microtubule destabilizing agents and microtublin polymerization inhibitors including, but not limited to taxanes, e.g. paclitaxel and docetaxel, vinca alkaloids, e.g., vinblastine, especially vinblastine sulfate, vincristine especially vincristine sulfate, and vinorelbine, discodermolides, cochicine and epothilones and derivatives thereof, e.g. epothilone B or D or derivatives thereof.
  • Paclitaxel may be administered e.g. in the form as it is marketed, e.g. TAXOL.
  • Docetaxel can be administered, e.g., in the form as it is marketed, e.g. under the trademark TAXOTERE.
  • Vinblastine sulfate can be administered, e.g., in the form as it is marketed, e.g. under the trademark VINBLASTIN R.
  • Vincristine sulfate can be administered, e.g., in the form as it is marketed, e.g. under the trademark FARMISTIN.
  • Discodermolide can be obtained, e.g., as disclosed in US 5,010,099.
  • Epothilone derivatives which are disclosed in WO 98/10121 , US 6,194,181 , WO 98/25929, WO 98/08849, WO 99/43653, WO 98/22461 and WO 00/31247. Especially preferred are Epothilone A and/or B.
  • alkylating agent includes, but is not limited to, cyclophosphamide, ifosfamide, melphalan or nitrosourea (BCNU or Gliadel).
  • Cyclophosphamide can be administered, e.g., in the form as it is marketed, e.g. under the trademark CYCLOSTIN.
  • Ifosfamide can be administered, e.g., in the form as it is marketed, e.g. under the trademark HOLOXAN.
  • histone deacetylase inhibitors or "HDAC inhibitors” relates to compounds which inhibit the histone deacetylase and which possess antiproliferative activity. This includes compounds disclosed in WO 02/22577, especially N-hydroxy-3-[4-[[(2-hydroxyethyl)[2-(1H- indol-3-yl)ethyl]-amino]methyl]phenyl]-2E-2-propenamide, N-hydroxy-3-[4-[[[2-(2-methyl-1H- indol-3-yl)-ethyl]-amino]methyl]phenyl]-2E-2-propenamide and pharmaceutically acceptable salts thereof. It further especially includes Suberoylanilide hydroxamic acid (SAHA).
  • SAHA Suberoylanilide hydroxamic acid
  • antimetabolite includes, but is not limited to, 5-Fluorouracil or 5-FU, capecitabine, gemcitabine, DNA demethylating agents, such as 5-azacytidine and decitabine, methotrexate and edatrexate, and folic acid antagonists such as pemetrexed.
  • Capecitabine can be administered, e.g., in the form as it is marketed, e.g. under the trademark XELODA.
  • Gemcitabine can be administered, e.g., in the form as it is marketed, e.g. under the trademark GEMZAR.
  • trastuzumab which can be administered, e.g., in the form as it is marketed, e.g. under the trademark HERCEPTIN.
  • platinum compound as used herein includes, but is not limited to, carboplatin, cis- platin, cisplatinum and oxaliplatin.
  • Carboplatin can be administered, e.g., in the form as it is marketed, e.g. under the trademark CARBOPLAT.
  • Oxaliplatin can be administered, e.g., in the form as it is marketed, e.g. under the trademark ELOXATIN.
  • compounds targeting/decreasing a protein or lipid kinase activity; or a protein or lipid phosphatase activity; or further anti-angiogenic compounds includes, but is not limited to: protein tyrosine kinase and/or serine and/or threonine kinase inhibitors or lipid kinase inhibitors, e.g.:
  • PDGFR platelet-derived growth factor-receptors
  • compounds which target, decrease or inhibit the activity of PDGFR especially compounds which inhibit the PDGF receptor, e.g. a N-phenyl-2- pyrimidine-amine derivative, e.g. imatinib, SU101 , SU6668, and GFB-111 ;
  • FGFR fibroblast growth factor- receptors
  • IGF-IR insulin-like growth factor receptor
  • compounds targeting, decreasing or inhibiting the activity of the insulin-like growth factor receptor I(IGF-IR) such as compounds which target, decrease or inhibit the activity of IGF- IR, especially compounds which inhibit the IGF-IR receptor, such as those compounds disclosed in WO 02/092599;
  • compounds targeting, decreasing or inhibiting the activity of the C-kit receptor tyrosine kinases - such as compounds which target, decrease or inhibit the activity of the c-Kit receptor tyrosine kinase family, especially compounds which inhibit the c-Kit receptor, e.g imatinib
  • compounds targeting, decreasing or inhibiting the activity of members of the c-Abl family and their gene-fusion products e.g. BCR-AbI kinase
  • compounds which target decrease or inhibit the activity of c-Abl family members and their gene fusion products e.g. a N-phenyl-2-pyrimidine-amine derivative, e.g. imatinib; PD180970; AG957; NSC 680410; or PD173955 from ParkeDavis
  • a N-phenyl-2-pyrimidine-amine derivative e.g. imatinib
  • PD180970 AG957; NSC 680410
  • UCN-01 safingol, BAY 43-9006, Bryostatin 1 , Perifosine; llmofosine; RO 318220 and RO 320432; GO 6976; lsis 3521 ; LY333531/LY379196; isochinoline compounds such as those disclosed in WO 00/09495; FTIs; PD184352 or QAN697( a P13K inhibitor);
  • k) compounds targeting, decreasing or inhibiting the activity of protein-tyrosine kinase inhibitors include imatinib mesylate (GLEEVEC) or tyrphostin.
  • a tyrphostin is preferably a low molecular weight (Mr ⁇ 1500) compound, or a pharmaceutically acceptable salt thereof, especially a compound selected from the benzylidenemalonitrile class or the S- arylbenzenemalonirile or bisubstrate quinoline class of compounds, more especially any compound selected from the group consisting of Tyrphostin A23/RG-50810; AG 99; Tyrphostin AG 213; Tyrphostin AG 1748; Tyrphostin AG 490; Tyrphostin B44; Tyrphostin B44 (+) enantiomer; Tyrphostin AG 555; AG 494; Tyrphostin AG 556, AG957 and adaphostin (4- ⁇ [(2,5-dihydroxyphenyl)methyl]amino ⁇ -benzoic acid adamantyl ester; NSC 680410, adaphostin); and
  • compounds targeting, decreasing or inhibiting the activity of the epidermal growth factor family of receptor tyrosine kinases are especially compounds, proteins or antibodies which inhibit members of the EGF receptor tyrosine kinase family, e.g. EGF receptor, ErbB2, ErbB3 and ErbB4 or bind to EGF or EGF related ligands, and are in particular those compounds, proteins or monoclonal antibodies generically and specifically disclosed in WO 97/02266, e.g. the compound of ex.
  • trastuzumab (HERCEPTIN), cetuximab, Iressa, Tarceva, OSI-774, CM 033, EKB-569, GW- 2016, E1.1 , E2.4, E2.5, E6.2, E6.4, E2.11 , E6.3 or E7.6.3, and 7H-pyrrolo-[2,3-d]pyrimidine derivatives which are disclosed in WO 03/013541.
  • anti-angiogenic compounds include compounds having another mechanism for their activity, e.g. unrelated to protein or lipid kinase inhibition e.g. thalidomide (THALOMID) and TNP-470.
  • TAALOMID thalidomide
  • TNP-470 TNP-470.
  • Compounds which target, decrease or inhibit the activity of a protein or lipid phosphatase are e.g. inhibitors of phosphatase 1 , phosphatase 2A, PTEN or CDC25, e.g. okadaic acid or a derivative thereof.
  • Compounds which induce cell differentiation processes are e.g. retinoic acid, ⁇ - ⁇ - or ⁇ - tocopherol or ⁇ - ⁇ - or ⁇ -tocotrienol.
  • cyclooxygenase inhibitor as used herein includes, but is not limited to, e.g. Cox-2 inhibitors, 5-alkyl substituted 2-arylaminophenylacetic acid and derivatives, such as celecoxib (CELEBREX), rofecoxib (VIOXX), etoricoxib, valdecoxib or a 5-alkyl-2- arylaminophenylacetic acid, e.g. 5-methyl-2-(2'-chloro-6'-fluoroanilino)phenyl acetic acid, lumiracoxib.
  • bisphosphonates as used herein includes, but is not limited to, etridonic, clodronic, tiludronic, pamidronic, alendronic, ibandronic, risedronic and zoledronic acid.
  • Etridonic acid can be administered, e.g., in the form as it is marketed, e.g. under the trademark DIDRONEL.
  • Clodronic acid can be administered, e.g., in the form as it is marketed, e.g. under the trademark BONEFOS.
  • Tiludronic acid can be administered, e.g., in the form as it is marketed, e.g. under the trademark SKELID.
  • “Pamidronic acid” can be administered, e.g. in the form as it is marketed, e.g. under the trademark AREDIATM.
  • “Alendronic acid” can be administered, e.g., in the form as it is marketed, e.g. under the trademark FOSAMAX.
  • “Ibandronic acid” can be administered, e.g., in the form as it is marketed, e.g. under the trademark BONDRANAT.
  • “Risedronic acid” can be administered, e.g., in the form as it is marketed, e.g. under the trademark ACTONEL.
  • "Zoledronic acid” can be administered, e.g. in the form as it is marketed, e.g. under the trademark ZOMETA.
  • mTOR inhibitors relates to compounds which inhibit the mammalian target of rapamycin (mTOR) and which possess antiproliferative activity such as sirolimus (Rapamune®), everolimus (CerticanTM), CCI-779 and ABT578.
  • heparanase inhibitor refers to compounds which target, decrease or inhibit heparin sulphate degradation.
  • the term includes, but is not limited to, PI-88.
  • biological response modifier refers to. a lymphokine or interferons, e.g. interferon ⁇ .
  • inhibitor of Ras oncogenic isoforms e.g. H-Ras, K-Ras, or N-Ras
  • H-Ras, K-Ras, or N-Ras refers to compounds which target, decrease or inhibit the oncogenic activity of Ras e.g. a "farnesyl transferase inhibitor” e.g. L-744832, DK8G557 or P115777 (Zamestra).
  • telomerase inhibitor refers to compounds which target, decrease or inhibit the activity of telomerase.
  • Compounds which target, decrease or inhibit the activity of telomerase are especially compounds which inhibit the telomerase receptor, e.g. telomestatin.
  • methionine aminopeptidase inhibitor refers to compounds which target, decrease or inhibit the activity of methionine aminopeptidase.
  • Compounds which target, decrease or inhibit the activity of methionine aminopeptidase are e.g. bengamide or a derivative thereof.
  • proteasome inhibitor refers to compounds which target, decrease or inhibit the activity of the proteasome.
  • Compounds which target, decrease or inhibit the activity of the proteasome include e.g. PS-341 and MLN 341.
  • matrix metalloproteinase inhibitor or (“MMP inhibitor”) as used herein includes, but is not limited to collagen peptidomimetic and nonpeptidomimetic inhibitors, tetracycline derivatives, e.g. hydroxamate peptidomimetic inhibitor batimastat and its orally bioavailable analogue marimastat (BB-2516), prinomastat (AG3340), metastat (NSC 683551) BMS- 279251 , BAY 12-9566, TAA211 , MMI270B or AAJ996.
  • MMP inhibitor matrix metalloproteinase inhibitor
  • agents used in the treatment of hematologic malignancies includes, but is not limited to FMS-like tyrosine kinase inhibitors e.g. compounds targeting, decreasing or inhibiting the activity of FMS-like tyrosine kinase receptors (Flt-3R); interferon, 1-b-D-arabinofuransylcytosine (ara-c) and bisulfan; and ALK inhibitors e.g. compounds which target, decrease or inhibit anaplastic lymphoma kinase.
  • FMS-like tyrosine kinase inhibitors e.g. compounds targeting, decreasing or inhibiting the activity of FMS-like tyrosine kinase receptors (Flt-3R); interferon, 1-b-D-arabinofuransylcytosine (ara-c) and bisulfan
  • ALK inhibitors e.g. compounds which target, decrease or inhibit anaplastic lymphoma kinase.
  • FMS-like tyrosine kinase receptors are especially compounds, proteins or antibodies which inhibit members of the Flt-3R receptor kinase family, e.g.PKC412, midostaurin, a staurosporine derivative, SU11248 and MLN518.
  • HSP90 inhibitors includes, but is not limited to, compounds targeting, decreasing or inhibiting the intrinsic ATPase activity of HSP90; degrading, targeting, decreasing or inhibiting the HSP90 client proteins via the ubiquitin proteasome pathway.
  • Compounds targeting, decreasing or inhibiting the intrinsic ATPase activity of HSP90 are especially compounds, proteins or antibodies which inhibit the ATPase activity of HSP90 e.g,17-allylamino,17-demethoxygeldanamycin (17AAG), a geldanamycin derivative; other geldanamycin related compounds; radicicol and HDAC inhibitors.
  • antiproliferative antibodies includes, but is not limited to trastuzumab (HerceptinTM), Trastuzumab-DM1 , erlotinib (TarcevaTM), bevacizumab (AvastinTM), rituximab (Rituxan®), PRO64553 (anti-CD40) and 2C4 Antibody.
  • trastuzumab HerceptinTM
  • Trastuzumab-DM1 erlotinib
  • TarcevaTM bevacizumab
  • AvastinTM bevacizumab
  • rituximab Renuxan®
  • PRO64553 anti-CD40
  • compounds of formula (I) can be used in combination with standard leukemia therapies, especially in combination with therapies used for the treatment of AML.
  • compounds of formula (I) can be administered in combination with e.g. farnesyl transferase inhibitors and/or other drugs useful for the treatment of AML, such as Daunorubicin, Adriamycin, Ara-C, VP-16, Teniposide, Mitoxantrone, Idarubicin, Carboplatinum and PKC412.
  • antigenemic compounds includes, for example, Ara-C, a pyrimidine analog, which is the 2 ' -alpha-hydroxy ribose (arabinoside) derivative of deoxycytidine. Also included is the purine analog of hypoxanthine, 6-mercaptopurine (6-MP) and fludarabine phosphate.
  • HDAC histone deacetylase
  • SAHA suberoylanilide hydroxamic acid
  • HDAC inhibitors include MS275, SAHA, FK228 (formerly FR901228), Trichostatin A and compounds disclosed in US 6,552,065, in particular, N-hydroxy-3-[4-[[[2-(2-methyl-1 H-indol-3-yl)-ethyl]- amino]methyl]phenyl]-2E-2-propenamide, or a pharmaceutically acceptable salt thereof and N-hydroxy-3-[4-[(2-hydroxyethyl) ⁇ 2-(1H-indol-3-yl)ethyl]-amino]methyl]phenyl]-2E-2- propenamide, or a pharmaceutically acceptable salt thereof, especially the lactate salt.
  • Compounds which target, decrease or inhibit the activity of serine/theronine mTOR kinase are especially compounds, proteins or antibodies which inhibit members of the mTOR kinase family e.g. RAD, RAD001 , CCI-779, ABT578, SAR543, rapamycin and derivatives thereof; AP23573 from Ariad; everolimus (CERTICAN); and sirolimus.
  • Somatostatin receptor antagonists refers to agents which target, treat or inhibit the somatostatin receptor such as octreoride, and SOM230.
  • Tumor cell damaging approaches refer to approaches such as ionizing radiation.
  • ionizing radiation means ionizing radiation that occurs as either electromagnetic rays (such as X-rays and gamma rays) or particles (such as alpha and beta particles). Ionizing radiation is provided in, but not limited to, radiation therapy and is known in the art. See Hellman, Principles of Radiation Therapy, Cancer, in Principles and Practice of Oncology, Devita et al., Eds., 4th Edition, Vol. 1 , pp. 248-275 (1993).
  • EDG binders refers a class of immunosuppressants that modulates lymphocyte recirculation, such as FTY720.
  • CERTICAN an investigational novel proliferation signal inhibitor that prevents proliferation of T-cells and vascular smooth muscle cells.
  • ribonucleotide reductase inhibitors refers to pyrimidine or puring nucleoside analogs including, but not limited to, fludarabine and/or cytosine arabinoside (ara-C), 6-thioguanine, 5-fluorouracil, cladribine, 6-mercaptopurine (especially in combination with ara-C against ALL) and/or pentostatin.
  • Ribonucleotide reductase inhibitors are especially hydroxyurea or 2-hydroxy-1 H-isoindole-1 ,3-dione derivatives, such as PL-1 , PL-2, PL-3, PL-4, PL-5, PL-6, PL-7 or PL-8 mentioned in Nandy et al., Acta Oncologica, Vol. 33, No. 8, pp. 953-961 (1994).
  • S-adenosylmethionine decarboxylase inhibitors as used herein includes, but is not limited to the compounds disclosed in US 5,461 ,076.
  • VEGF vascular endothelial growth factor
  • WO 98/35958 e.g. 1 -(4-chloroanilino)-4-(4-pyridylmethyl)phthalazine or a pharmaceutically acceptable salt thereof, e.g. the succinate, or in WO 00/09495, WO 00/27820, WO 00/59509, WO 98/11223, WO 00/27819 and EP 0 769 947; those as described by Prewett et al, Cancer Res, Vol. 59, pp. 5209-5218 (1999); Yuan et at., Proc Natl Acad Sci U S A, Vol. 93, pp.
  • Photodynamic therapy refers to therapy which uses certain chemicals known as photosensitizing agents to treat or prevent cancers.
  • Examples of photodynamic therapy includes treatment with agents, such as e.g. VISUDYNE and porfimer sodium.
  • Angiostatic steroids refers to agents which block or inhibit angiogenesis, such as, e.g., anecortave, triamcinolone, hydrocortisone, 11- ⁇ -epihydrocotisol, cortexolone, 17 ⁇ -hydroxyprogesterone, corticosterone, desoxycorticosterone, testosterone, estrone and dexamethasone.
  • Implants containing corticosteroids refers to agents, such as e.g. fluocinolone, dexamethasone.
  • chemotherapeutic agents include, but are not limited to, plant alkaloids, hormonal agents and antagonists; biological response modifiers, preferably lymphokines or interferons; antisense oligonucleotides or oligonucleotide derivatives; or miscellaneous agents or agents with other or unknown mechanism of action.
  • biological response modifiers preferably lymphokines or interferons
  • antisense oligonucleotides or oligonucleotide derivatives or miscellaneous agents or agents with other or unknown mechanism of action.
  • the structure of the active agents identified by code nos., generic or trade names may be taken from the actual edition of the standard compendium "The Merck Index" or from databases, e.g. Patents International (e.g. IMS World Publications).
  • a compound of the formula (I) may also be used to advantage in combination with known therapeutic processes, e.g., the administration of hormones or especially radiation.
  • a compound of formula (I) may in particular be used as a radiosensitizer, especially for the treatment of tumors which exhibit poor sensitivity to radiotherapy.
  • ком ⁇ онент there is meant either a fixed combination in one dosage unit form, or a kit of parts for the combined administration where a compound of the formula (I) and a combination partner may be administered independently at the same time or separately within time intervals that especially allow that the combination partners show a cooperative, e.g. synergistic, effect, or any combination thereof.
  • Ratios of solvents are given in volume by volume (7 V ).
  • Flash chromatography is performed by using silica gel (Merck; 40-63 ⁇ m).
  • silica gel Merck 40-63 ⁇ m
  • pre-coated silica gel Merck 60 F254 plates are used. Detection of the components is made by UV light (254 nm).
  • HPLC is performed on an Agilent HP 1100 using a Nucleosil 100-3 Ci 8 HD 125 x 4.0 mm column [1 mL/min.; 20-100% NeCN / 0.1% TFA in 7 minutes) (Method A); SpectraSystem SP8800/UV2000 using a Nucleosil 100-5 C 18 AB 250 x 4.6 mm column (2 mL/min.; 2-100% MeCN / 0.1% TFA in 10 minutes) (Method B); using a Chromalith Speed ROD RP18 50-4.6 mm column (Merck) (2 mL/min.; 2-100% MeCN / 0.1% TFA in 2 minutes) (Method C); or a C8 2.1-50 mm 3 ⁇ m column (Waters) (2 mL/min.; 5-95% MeCN / 0.1% TFA in 2 minutes) (Method D). Electrospray mass spectra are obtained with a Fisons Instruments VG Platform II. Melting points
  • Stage 1.1 2-(3-Chloro-phenyl)-3-oxo-butyronitrile.
  • Example 3 6-(3-Chloro-phenyl)-3-[4- ⁇ 2-dimethylamino-ethoxy)-phenyl]-5-piperazin- 1-ylmethyl-pyrazolo[1,5-a]pyrimidin-7-ylamine
  • Example 4 2- ⁇ [7-Amino-6-(3-chloro-phenyl)-3-(3,4-dimethoxy-phenyl)-pyrazolo[1,5- a]pyrimidin-5-ylmethyl]-amino ⁇ -ethanol
  • Tablets comprising, as active ingredient, 50 mg of any one of the compounds of formula (I) mentioned in the preceding Examples of the following composition are prepared using routine methods:
  • the active ingredient is combined with part of the wheat starch, the lactose and the colloidal silica and the mixture pressed through a sieve.
  • a further part of the wheat starch is mixed with the 5-fold amount of water on a water bath to form a paste and the mixture made first is kneaded with this paste until a weakly plastic mass is formed.
  • Example 7 Tablets 2 comprising compounds of the formula (I)
  • Tablets comprising, as active ingredient, 100 mg of any one of the compounds of formula (I) mentioned in the preceding Examples are prepared with the following composition, following standard procedures:
  • the active ingredient is mixed with the carrier materials and compressed by means of a tabletting machine (Korsch EKO, Stempel barnmesser 10 mm).
  • Capsules comprising, as active ingredient, 100 mg of any one of the compounds of formula (I) mentioned in the preceding Examples, of the following composition are prepared according to standard procedures:
  • Manufacturing is done by mixing the components and filling them into hard gelatine capsules, size 1.
  • Example 9 Biological data of compounds of Example 1 and 2 as EphB4 and c-Abl protein kinase inhibitors:
  • Table 2 Inhibition of c-Abl protein-tyrosine kinase activity Activity of the compounds of Example 1 and 2 as c-Abl protein tyrosine kinase inhibitors is tested according to the method described in the specification.

Abstract

The invention relates to pyrazolo[1,5a]pyrimidin-7-yl amine derivatives of formula (I) and salts thereof, their use in the treatment of protein kinase dependent diseases.

Description

Pyrazolof 1.5-a1pyrimidin-7-yl amine derivatives as protein kinase inhibitors
The invention relates to pyrazolo[1,5-a]pyrimidin-7-yl amine derivatives, their use in the treatment of protein kinase dependent diseases, their use for the manufacture of pharmaceutical compositions for the treatment of said diseases, methods of use of pyrazolo[1 ,5-a]pyrimidin-7-yl amine derivatives in the treatment of said diseases, pharmaceutical preparations comprising pyrazolo[1 ,5-a]pyrimidin-7-yl amine derivatives for the treatment of said diseases, novel pyrazolo[1 ,5-a]pyrimidin-7-yl amine derivatives, processes for the manufacture of the novel pyrazolo[1 ,5-a]pyrimidin-7-yl amine derivatives and pharmaceutical preparations, the use or methods of use of the pyrazolo[1 ,5-a]pyrimidin- 7-yl amine derivatives as mentioned above, and/or these pyrazolo[1 ,5-a]pyrimidin-7-yl amine derivatives for use in the treatment of the animal or human body. Pyrazolo[1 ,5-a]pyrimidin-7-yl-amine derivatives have been reported in the literature as ligands of benzodiazepine receptors (e.g., S. Selleri et al., Bioorg. Med. Chem 7 (12), 2705- 11 (1999)), antagonists of the corticotropin releasing factor (EP 1097709), angiotensin Il receptor antagonists (e. g., S. Takeshi et al., Japn. Pharm. Bull. 47 (7), 928-38 (1999)), monoxide synthetase inhibitors (JP 10101671), analgesics (WO 9535298), fungicides (EP 071792) or anti-inflammatory reagents (WO 9218504).
We have now found that the pyrazolo[1,5-a]pyrimidin-7-ylamine scaffold can be also be used as a template for the design of potent kinase inhibitors.
In view of the large number of protein kinase inhibitors and the multitude of proliferative and other protein kinase-related diseases, there is an ever-existing need to provide novel classes of compounds that are useful as protein kinase inhibitors and thus in the treatment of related diseases.
What is desirable from the point of view of possible treatments of proliferative diseases is to have a plethora of compound classes each tailored to specific protein kinases or protein kinase classes, thus allowing to come to specific treatments. Therefore, a strong need exists to find new classes of compounds allowing for such specific inhibitory effects.
In one embodiment, the invention relates to a compound of the formula (I):
Figure imgf000003_0001
wherein:
R1 Js H
R2 is benzyl; unsubstituted phenyl or phenyl substituted by one or two substituents chosen from the group consisting of halo, di-lower alkylaminoalkoxy, hydroxy, alkoxy, benzyloxy, cycloalkyl, amino, acetyl amino;
R3 is H and R4 is hydroxyalkyl or
R3 and R4 together with the nitrogen atom they are attached to represent morpholinyl, pyrrolidinyl, piperidinyl or piperazinyl, the heterocycles optionally being further substituted by up to four alkyl groups;
A is phenyl which is unsubstituted or substituted by one or more of the substituents chosen from the group consisting of mono-, di- or tri-lower alkoxy, di-lower alkylaminyl, di-lower alkylaminoalkoxy, morpholinyl which is optionally di-substituted by alkyl, piperidinyl which is optionally substituted by di-lower alkylaminyl, and piperazinyl which is optionally substituted by lower alkyl, lower alkoxy, lower alkyl piperazinyl, pyrrolidinyl, di-lower or alkylaminyl;
or a pharmaceutical salt thereof.
A preferred embodiment is a compound of formula I according to the above, wherein:
R1 is H
R2 is phenyl substituted by fluoro or chloro;
R3 is H and R4 is hydroxyethyl or
R3 and R4 together with the nitrogen atom they are attached to represent piperazinyl;
A is phenyl which is substituted with one or more of the substituents chosen from the group consisting of; mono-, di- or tri-methoxy, di-methylaminoethoxy and di-ethylamino piperidinyl or a pharmaceutical salt thereof. A further embodiment is a compound of formula I according to the above, wherein:
R2 is phenyl substituted by fluoro or chloro;
R3 and R4 together with the nitrogen atom they are attached to represent piperazinyl;
A is phenyl which is substituted with one or more of the substituents chosen from the group consisting of; mono-, di- or tri-methoxy, di-methylaminoethoxy and di-ethylamino piperidinyl or a pharmaceutical salt thereof.
A further embodiment is a compound of formula I according to the above, wherein:
R1 is H;
R2 is phenyl substituted by fluoro or chloro;
R3 and R4 together with the nitrogen atom they are attached to represent piperazinyl;
A is phenyl which is substituted with one or more of the substituents chosen from the group consisting of; mono-, di- and tri-methoxy or a pharmaceutical salt thereof.
Another embodiment is a compound of formula I according to the above, wherein:
R1 is H
R2 is phenyl substituted by fluoro or chloro;
R3 is H and R4 is hydroxyethyl;
A is phenyl which is substituted with one or more of the substituents chosen from the group consisting of; mono-, di- and tri-methoxy or a pharmaceutical salt thereof.
In one embodiment the invention pertains to a compound of formula I for use in the treatment of the human or animal body.
Yet another embodiment is the use of a compound of formula I according to the above in the preparation of a pharmaceutical composition.
Yet another embodiment is a pharmaceutical composition comprising a compound of formula I according to the above. - A -
The pharmaceutical composition preferably comprises a compound of formula I according to the above and an acceptable pharmaceutical carrier.
In another embodiment, there is provided the use of a compound of formula I according to the above in the preparation of a pharmaceutical compositions for use in the treatment of a kinase dependent disease.
A protein kinase dependent disease is preferably one that depends on c-Abl, Bcr-Abl, c-Kit, c-Raf, Flt-1 , Flt-3, Her-1 , KDR, PDGFR-kinase, c-Src, RET-receptor kinase, FGF-R1 , FGF- R2, FGF-R3, FGF-R4, Ephrin receptor kinases (e. g., EphB2 kinase, EphB4 kinase and related Eph kinases), casein kinases (CK-1 , CK-2, G-CK), Pak, ALK, ZAP70, Jak1 , Jak2, AxI, Cdk1 , cdk4, cdk5, Met, FAK1 Pyk2, Syk, Insulin receptor kinase, Tie-2 or costitutively activating mutations of kinases (activating kinases) such as of Bcr-Abl, c-Kit, c-Raf, Flt-3, FGF-R3, PDGF-receptors, RET1 and Met and (especially aberrantly highly expressed or activated) kinase-dependent disease or disease dependent on the activation of the kinase pathways, or a disease dependent on any two or more of the kinases just mentioned.
A protein kinase dependent disease is more preferably one that depends on c-abl, Flt-3, KDR, c-Src, RET, EphB4, c-kit, cdk1 , FGFR-1 , c-raf, Her-1 , Ins-R or Tek.
Most preferably, the disease to be treated is a proliferative disease, preferably a benign or especially malignant tumor, more preferably carcinoma of the brain, kidney, liver, adrenal gland, bladder, breast, stomach (especially gastric tumors), ovaries, colon, rectum, prostate, pancreas, lung, vagina, thyroid, sarcoma, glioblastomas, multiple myeloma or gastrointestinal cancer, especially colon carcinoma or colorectal adenoma, or a tumor of the neck and head, an epidermal hyperproliferation, especially psoriasis, prostate hyperplasia, a neoplasia, especially of epithelial character, preferably mammary carcinoma, or a leukemia.
In a further embodiment, the disease to be treated is a disease which is triggered by persistent angiogenesis, such as psoriasis; Kaposi's sarcoma; restenosis, e.g., stent- induced restenosis; endometriosis; Crohn's disease; Hodgkin's disease; leukemia; arthritis, such as rheumatoid arthritis; hemangioma; angiofibroma; eye diseases, such as diabetic retinopathy and neovascular glaucoma; renal diseases, such as glomerulonephritis; diabetic nephropathy; malignant nephrosclerosis; thrombotic microangiopathic syndromes; transplant rejections and glomerulopathy; fibrotic diseases, such as cirrhosis of the liver; mesangial cell-proliferative diseases; arteriosclerosis; injuries of the nerve tissue. The compounds of the present invention can also be used for inhibiting the re-occlusion of vessels after balloon catheter treatment, for use in vascular prosthetics or after inserting mechanical devices for holding vessels open, such as, e.g., stents, as immunosuppressants, as an aid in scar-free wound healing, and for treating age spots and contact dermatitis.
The general terms used hereinbefore and hereinafter preferably have within the context of this disclosure the following meanings, unless otherwise indicated:
Alkyl includes lower alkyl preferably alkyl with up to 7 carbon atoms, preferably from 1 to and including 5, and is linear or branched; preferably, lower alkyl is pentyl, such as n-pentyl, butyl, such as n-butyl, sec-butyl, isobutyl, tert-butyl, propyl, such as n-propyl or isopropyl, ethyl or methyl. Preferably lower alkyl is methyl, propyl or tert-butyl.
Alkyl can be substituted or unsubstituted, and when substituted may be with up to 3 substituents including other alkyl, cycloalkyl, alkenyl, alkynyl, any of the substituents defined above for aryl or any of the functional groups defined below.
Halo or halogen is preferably fluoro, chloro, bromo or iodo, most preferably fluoro, chloro or bromo.
Where the plural form is used for compounds, salts, pharmaceutical preparations, diseases and the like, this is intended to mean also a single compound, salt, or the like. Salts are especially the pharmaceutically acceptable salts of compounds of formula I. Such salts are formed, for example, as acid addition salts, preferably with organic or inorganic acids, from compounds of formula (I) with a basic nitrogen atom, especially the pharmaceutically acceptable salts. Suitable inorganic acids are, for example, halogen acids, such as hydrochloric acid, sulfuric acid, or phosphoric acid. Suitable organic acids are, for example, carboxylic, phosphonic, sulfonic or sulfamic acids, for example acetic acid, propionic acid, octanoic acid, decanoic acid, dodecanoic acid, glycolic acid, lactic acid, fumaric acid, succinic acid, adipic acid, pimelic acid, suberic acid, azelaic acid, malic acid, tartaric acid, citric acid, amino acids, such as glutamic acid or aspartic acid, maleic acid, hydroxy- maleic acid, methylmaleic acid, cyclohexanecarboxylic acid, adamantanecarboxylic acid, benzoic acid, salicylic acid, 4-aminosalicylic acid, phthalic acid, phenylacetic acid, mandelic acid, cinnamic acid, methane- or ethane-sulfonic acid, 2-hydroxyethanesulfonic acid, ethane- 1 ,2-disulfonic acid, benzenesulfonic acid, 2-naphthalenesulfonic acid, 1 ,5-naphthalene- disulfonic acid, 2-, 3- or 4-methylbenzenesulfonic acid, methylsulfuric acid, ethylsulfuric acid, dodecytsulfuric acid, N-cyclohexylsulfamic acid, N-methyl-, N-ethyl- or N-propyl-sulfamic acid, or other organic protonic acids, such as ascorbic acid.
In the presence of negatively charged radicals, such as carboxy or sulfo, salts may also be formed with bases, e.g. metal or ammonium salts, such as alkali metal or alkaline earth metal salts, for example sodium, potassium, magnesium or calcium salts, or ammonium salts with ammonia or suitable organic amines, such as tertiary monoamines, for example triethyl- amine or tri(2-hydroxyethyl)amine, or heterocyclic bases, for example N-ethyl-piperidine or N,N'-dimethylpiperazine.
When a basic group and an acid group are present in the same molecule, a compound of formula (I) may also form internal salts.
For isolation or purification purposes it is also possible to use pharmaceutically unacceptable salts, for example picrates or perchlorates. For therapeutic use, only pharmaceutically acceptable salts or free compounds are employed (where applicable in the form of pharmaceutical preparations), and these are therefore preferred.
In view of the close relationship between the compounds in free form and those in the form of their salts, including those salts that can be used as intermediates, for example in the purification or identification of the compounds, tautomers or tautomeric mixtures and their salts, any reference to the compounds hereinbefore and hereinafter especially the compounds of the formula I, is to be understood as referring also to the corresponding tautomers of these compounds, especially of compounds of the formula I, tautomeric mixtures of these compounds, especially of compounds of the formula I, or salts of any of these, as appropriate and expedient and if not mentioned otherwise.
Where "a compound ..., a tautomer thereof; or a salt thereof or the like is mentioned, this means "a compound ..., a tautomer thereof, or a salt of the compound or the tautomer".
Any asymmetric carbon atom may be present in the (R)-, (S)- or (R.S)-configuration, preferably in the (R)- or (S)-configuration. Substituents at a ring at atoms with saturated bonds may, if possible, be present in cis- (= Z-) or trans (= E-) form. The compounds may thus be present as mixtures of isomers or preferably as pure isomers, preferably as enantiomer-pure diastereomers or pure enantiomers. The present invention also relates to pro-drugs of a compound of formula (I) that convert in vivo to the compound of formula (I) as such. Any reference to a compound of formula (I) is therefore to be understood as referring also to the corresponding pro-drugs of the compound of formula (I), as appropriate and expedient.
Compounds of formula (I) can be prepared analogously to the procedure described by Alicade, E; De Mendoza, J; Garcia-Marquina, JM; Almera, C; J. Heterocycl. Chem. 11 , 423 (1974) or in EP2005/000602.
Salts of compounds of formula (I) having at least one salt-forming group may be prepared in a manner known per se. For example, salts of compounds of formula (I) having acid groups may be formed, for example, by treating the compounds with metal compounds, such as alkali metal salts of suitable organic carboxylic acids, e.g. the sodium salt of 2-ethylhexanoic acid, with organic alkali metal or alkaline earth metal compounds, such as the corresponding hydroxides, carbonates or hydrogen carbonates, such as sodium or potassium hydroxide, carbonate or hydrogen carbonate, with corresponding calcium compounds or with ammonia or a suitable organic amine, stoichiometric amounts or only a small excess of the salt- forming agent preferably being used. Acid addition salts of compounds of formula (I) are obtained in customary manner, e.g. by treating the compounds with an acid or a suitable anion exchange reagent. Internal salts of compounds of formula (I) containing acid and basic salt-forming groups, e.g. a free carboxy group and a free amino group, may be formed, e.g. by the neutralisation of salts, such as acid addition salts, to the isoelectric point, e.g. with weak bases, or by treatment with ion exchangers.
Salts can be converted in customary manner into the free compounds; metal and ammonium salts can be converted, for example, by treatment with suitable acids, and acid addition salts, for example, by treatment with a suitable basic agent.
Mixtures of isomers obtainable according to the invention can be separated in a manner known per se into the individual isomers; diastereoisomers can be separated, for example, by partitioning between polyphasic solvent mixtures, recrystallisation and/or chromatographic separation, for example over silica gel or by e.g. medium pressure liquid chromatography over a reversed phase column, and racemates can be separated, for example, by the formation of salts with optically pure salt-forming reagents and separation of the mixture of diastereoisomers so obtainable, for example by means of fractional crystallisation, or by chromatography over optically active column materials. lntermediates and final products can be worked up and/or purified according to standard methods, e.g. using chromatographic methods, distribution methods, (re-) crystallization, and the like.
The following applies in general to all processes mentioned hereinbefore and hereinafter, while reaction conditions specifically mentioned above or below are preferred:
All the above-mentioned process steps can be carried out under reaction conditions that are known p_er se, preferably those mentioned specifically, in the absence or, customarily, in the presence of solvents or diluents, preferably solvents or diluents that are inert towards the reagents used and dissolve them, in the absence or presence of catalysts, condensation or neutralizing agents, for example ion exchangers, such as cation exchangers, e.g. in the H+ form, depending on the nature of the reaction and/or of the reactants at reduced, normal or elevated temperature, for example in a temperature range of from about -100 0C to about 19O0C, preferably from approximately -8O0C to approximately 1500C, for example at from -80 to -6O0C, at room temperature, at from -20 to 4O0C or at reflux temperature, under atmospheric pressure or in a closed vessel, where appropriate under pressure, and/or in an inert atmosphere, for example under an argon or nitrogen atmosphere.
At all stages of the reactions, mixtures of isomers that are formed can be separated into the individual isomers, for example diastereoisomers or enantiomers, or into any desired mixtures of isomers, for example racemates or mixtures of diastereoisomers, for example analogously to the methods described under "Additional process steps".
The solvents from which those solvents that are suitable for any particular reaction may be selected include those mentioned specifically or, for example, water, esters, such as lower alkyl-lower alkanoates, for example ethyl acetate, ethers, such as aliphatic ethers, for example diethyl ether, or cyclic ethers, for example tetrahydrofurane or dioxane, liquid aromatic hydrocarbons, such as benzene or toluene, alcohols, such as methanol, ethanol or 1- or 2-propanol, nitriles, such as acetonitrile, halogenated hydrocarbons, such as methylene chloride or chloroform, acid amides, such as dimethylformamide or dimethyl acetamide, bases, such as heterocyclic nitrogen bases, for example pyridine or N-methylpyrrolidin-2-one, carboxylic acid anhydrides, such as lower alkanoic acid anhydrides, for example acetic anhydride, cyclic, linear or branched hydrocarbons, such as cyclohexane, hexane or isopen- tane, or mixtures of those solvents, for example aqueous solutions, unless otherwise indica- ted in the description of the processes. Such solvent mixtures may also be used in working up, for example by chromatography or partitioning.
The compounds, including their salts, may also be obtained in the form of hydrates, or their crystals may, for example, include the solvent used for crystallization. Different crystalline forms may be present.
The invention relates also to those forms of the process in which a compound obtainable as intermediate at any stage of the process is used as starting material and the remaining process steps are carried out, or in which a starting material is formed under the reaction conditions or is used in the form of a derivative, for example in protected form or in the form of a salt, or a compound obtainable by the process according to the invention is produced under the process conditions and processed further jn situ. In the process of the present invention those starting materials are preferably used which result in new compounds of formula (I) described at the beginning as being especially valuable. Special preference is given to reaction conditions that are identical or analogous to those mentioned in the Examples.
The compounds of formula (I) have valuable pharmacological properties. They exhibit their biological activity for instance as inhibitors of different protein kinases, preferably inhibiting c- abl, Flt-3, KDR, c-Src, RET, EphB4, c-kit, cdk1 , FGFR-1 , c-raf, Her-1 , Ins-R or Tek, most preferably as inhibitors of Ephrin B4 receptor (EphB4) kinases. Therefore, the compound of the invention are useful in the treatment of kinase dependent diseases, e.g., as drugs to treat proliferative diseases.
The term "treatment of tyrosine protein kinase dependent diseases" refers to the prophylactic or preferably therapeutic (including palliative and/or curing) treatment of said diseases, especially of the diseases mentioned below.
The inhibition of RET is measured as follows: The baculovirus donor vector pFB-GSTX3 is used to generate a recombinant baculovirus that expresses the amino acid region 658- 1072 (Swiss prot No. Q9BTB0) of the intra-cytoplasmic kinase domain of human RET- Men2A which corresponds to the wild-type kinase domain of RET (wtRET) and RET-Men2B, which differs from the wtRET by the activating mutation in the activation loop M918T. The coding sequences for the cytoplasmic domain of wtRET and RET-Men2B are amplified by PCR from the plasmids pBABEpuro RET-Men2A and pBABEpuro RET-Men2B. The amplified DNA fragments and the pFB-GSTX3 vector are made compatible for ligation by digestion with Sail and Kpnl. Ligation of these DNA fragments results in the baculovirus donor plasmid pFB-GX3-RET-Men2A and pFB-GX3-RET-Men2B, respectively. Production of virus: Transfer vectors containing the kinase domains are transfected into the DhMOBac cell line (GIBCO) and plated on selective agar plates. Colonies without insertion of the fusion sequence into the viral genome (carried by the bacteria) are blue. Single, white colonies are picked and viral DNA (bacmid) are isolated from the bacteria by standard plasmid purification procedures. Sf9 cells or Sf21 (American Type Culture Collection) cells are then transfected in 25 cm2 flasks with the viral DNA using Cellfectin reagent. Determination of small scale protein expression in Sf9 cells: Virus-containing media is collected from the transfected cell culture and used for infection to increase its titer. Virus- containing media obtained after two rounds of infection is used for large-scale protein expression. For large-scale protein expression 100 cm2 round tissue culture plates are seeded with 5 x 107 cells/plate and infected with 1 mL of virus-containing media (approximately 5 MOIs). After 3 days, the cells are scraped off the plate and centrifuged at 500 rpm for 5 minutes. Cell pellets from 10-20, 100 cm2 plates, are re-suspended in 50 mL of ice-cold lysis buffer (25 mM tris-HCI, pH 7.5, 2 mM EDTA, 1% NP-40, 1 mM DTT, 1 mM P MSF). The cells are stirred on ice for 15 minutes and then centrifuged at 5,000 rpms for 20 minutes.
Purification of GST-tagged proteins: The centrifuged cell lysate is loaded onto a 2 mL glutathione-sepharose column (Pharmacia) and is washed 3 x with 10 mL of 25 mM tris-HCI, pH 7.5, 2 mM EDTA, 1 mM DTT, 200 mM NaCI. The GST-tagged proteins are then eluted by 10 applications (1 mL each) of 25 mM tris-HCI, pH 7.5, 10 mM reduced-glutathione, 100 mM NaCI, 1 mM DTT, 10% glycerol and stored at -700C.
Measure of enzyme activity: Tyrosine protein kinase assays with either purified GST-wtRET or GST-RET-Men2B protein are carried out in a final volume of 30 μL containing 15 ng of either GST-wtRET or GST-RET-Men2B protein, 20 mM tris-HCI, pH 7.5, 1 mM MnCI2, 10 mM MgCI2, 1 mM DTT, 3 μg/mL poly(Glu,Tyr) 4:1 , 1% DMSO, 2.0 μM ATP (γ-[33P]-ATP 0.1 μCi). The activity is assayed in the presence or absence of inhibitors, by measuring the incorporation of 33P from [γ33P] ATP into poly(Glu.Tyr) 4:1. The assay is carried out in 96- well plates at ambient temperature for 15 minutes under conditions described below and terminated by the addition of 20 μL of 125 mM EDTA. Subsequently, 40 μL of the reaction mixture are transferred onto Immobilon-PVDF membrane (Millipore) previously soaked for 5 minutes with methanol, rinsed with water, then soaked for 5 minutes with 0.5% H3PO4 and mounted on vacuum manifold with disconnected vacuum source. After spotting all samples, vacuum is connected and each well-rinsed with 200 μL 0.5% H3PO4. Membranes are removed and washed 4 x on a shaker with 1.0% H3PO4, once with ethanol. Membranes are counted after drying at ambient temperature, mounting in Packard TopCount 96-well frame, and addition of 10 μL/well of Microscint TM (Packard). IC50 values are calculated by linear regression analysis of the percentage inhibition of each compound in duplicate, at 4 concentrations (usually 0.01 , 0.1 , 1 and 10 μM). One unit of protein kinase activity is defined as 1 nmole of 33P ATP transferred from [γ33P] ATP to the substrate protein/minute/mg of protein at 37°C.
IC50 calculations input 3 x 4 μL stopped assay on lmmobilon membrane, not washed background (3 wells) assay with H2O instead of enzyme positive control (4 wells) 3% DMSO instead of compound bath control (1 well) no reaction mix
IC50 values are calculated by logarithmic regression analysis of the percentage inhibition of each compound at 4 concentrations (usually 3- or 10-fold dilution series starting at 10 μM).
In each experiment, the actual inhibition by reference compound is used for normalization of
IC50 values to the basis of an average value of the reference inhibitor:
Normalized IC50 = measured IC50 average ref. IC50 / measured ref. IC50
Example: Reference inhibitor in experiment 0.4 μM, average 0.3 μM
Test compound in experiment 1.0 μM, normalization: 0.3/0.4 = 0.75 μM For example, staurosporine or a synthetic staurosporine derivative are used as reference compounds.
Using this protocol, the compounds of the formula (I) are found to show IC5O values for RET inhibition in the range from 0.005-100 μM, preferably in the range from 0.01-2 μM.
The efficacy of the compounds of the invention as inhibitors of c-Abl protein-tyrosine kinase activity can be demonstrated as follows: An in vitro enzyme assay is performed in 96-well plates as a filter binding assay as described by Geissler et al. in Cancer Res. 1992; 52:4492-4498, with the following modifications. The His-tagged kinase domain of c-Abl is cloned and expressed in the baculovirus/Sf9 system as described by Bhat et al. in J.Biol.Chem. 1997; 272:16170-16175. A protein of 37 kD (c-Abl kinase) is purified by a two- step procedure over a Cobalt metal chelate column followed by an anion exchange column with a yield of 1-2 mg/L of Sf9 cells (Bhat et al., reference cited). The purity of the c-Abl kinase is >90% as judged by SDS-PAGE after Coomassie blue stai-ning. The assay contains (total volume of 30 μl_): c-Abl kinase (50 ng), 20 mM Tris HCI, pH 7.5, 10 mM MgCI2, 10 μM Na3VO4, 1 mM DTT and 0.06 μCi/assay [γ33 P]-ATP (5 μM ATP) using 30 μg/mL poly-Ala,Glu,Lys,Tyr-6:2:5:1 (PoIy-AEKY, Sigma P1152) in the presence of 1 % DMSO. Reactions are terminated by adding 10 μl_ of 250 mM EDTA and 30 μL of the reaction mixture is transferred onto Immobilon-PVDF membrane (Millipore, Bedford, MA, USA) previously soaked for 5 min with methanol, rinsed with water, then soaked for 5 min with 0.5 % H3PO4 and mounted on vacuum manifold with disconnected vacuum source. After spotting all samples, vacuum is connected and each well rinsed with 200 μL 0.5 % H3PO4. Mem-branes are removed and washed on a shaker with 0.5 % H3PO4 (4 times) and once with ethanol. Membranes are counted after drying at ambient temperature, mounting in Packard TopCount 96-well frame, and addition of 10 μLΛ/vell of Microscint TM (Packard). Using this test system, compounds of the formula I show IC50 values of inhibition for c-Abl inhibition in the range of 0.002 to 100 M, usually between 0.002 and 5 M.
The efficacy of the compounds of the invention as inhibitors of KDR protein-tyrosine kinase activity can be demonstrated as follows: The inhibition of VEGF-induced receptor autophosphorylation can be confirmed with a further in vitro experiments in cells such as transfected CHO cells, which permanently express human VEGF-R2 receptor (KDR), are seeded in complete culture medium (with 10% fetal calf serum = FCS) in 6-well cell-culture plates and incubated at 37°C under 5% CO2 until they show about 80% confluency. The compounds to be tested are then diluted in culture medium (without FCS, with 0.1% bovine serum albumin) and added to the cells. (Controls comprise medium without test compounds). After two hours' incubation at 37°C, recombinant VEGF is added; the final VEGF concentration is 20 ng/ml). After a further five minutes incubation at 37°C, the cells are washed twice with ice-cold PBS (phosphate-buffered saline) and immediately lysed in 100 μl lysis buffer per well. The lysates are then centrifuged to remove the cell nuclei, and the protein concentrations of the supernatants are determined using a commercial protein assay (BIORAD). The lysates can then either be immediately used or, if necessary, stored at -200C.
A sandwich ELISA is carried out to measure the VEGF-R2 phosphorylation: a monoclonal antibody to VEGF-R2 (for example Mab 1495.12.14; ProQinase, Freiburg, Germany) is immobilized on black ELISA plates (OptiPlateTM HTRF-96 from Packard). The plates are then washed and the remaining free protein-binding sites are saturated with 3% TopBlock® (Juro, Cat. # TB232010) in phosphate buffered saline with Tween 20® (polyoxyethylen(20)- sorbitane monolaurate, ICI/Uniquema) (PBST). The cell lysates (20 μg protein per well) are then incubated in these plates overnight at 4°C together with an antiphosphotyrosine antibody coupled with alkaline phosphatase (PY20:AP from Zymed). The (plates are washed again and the) binding of the antiphosphotyrosine antibody to the captured phosphorylated receptor is then demonstrated using a luminescent AP substrate (CDP-Star, ready to use, with Emerald II; Applied Biosystems). The luminescence is measured in a Packard Top Count Microplate Scintillation Counter. The difference between the signal of the positive control (stimulated with VEGF) and that of the negative control (not stimulated with VEGF) corresponds to VEGF-induced VEGF-R2 phosphorylation (= 100 %). The activity of the tested substances is calculated as percent inhibition of VEGF-induced VEGF-R2 phosphorylation, wherein the concentration of substance that induces half the maximum inhibition is defined as the IC50 (inhibitory dose for 50% inhibition). Compounds of the formula I here show an IC50 in the range of 0.005 to 20 μM, preferably between 0.005 and 1 μM for KDR inhibition.
Flt3 kinase inhibition is determined as follows: The baculovirus donor vector pFbacGOI (GIBCO) is used to generate a recombinant baculovirus expressing the amino acid region amino acids 563-993 of the cytoplasmic kinase domain of human Flt-3. The coding sequence for the cytoplasmic domain of Flt-3 is amplified by PCR from human c-DNA libraries (Clontech). The amplified DNA fragments and the pFbacGOI vector are made compatible for ligation by digestion with BamH1 and Hindlll. Ligation of these DNA fragments results in the baculovirus donor plasmid Flt-3(1.1). The production of the viruses, the expression of proteins in Sf9 cells and the purification of the GST-fused proteins are performed as follows: Production of virus: Transfer vector (pFbacGOI -Flt-3) containing the Flt-3 kinase domain is transfected into the DHIOBac cell line (GIBCO) and the transfected cells are plated on selective agar plates. Colonies without insertion of the fusion sequence into the viral genome (carried by the bacteria) are blue. Single white colonies are picked and viral DNA (bacmid) is isolated from the bacteria by standard plasmid purification procedures. Sf9 or Sf21 cells (American Type Culture Collection) are then transfected in flasks with the viral DNA using Cellfectin reagent.
Determination of small scale protein expression in Sf9 cells: Virus containing media is collected from the transfected cell culture and used for infection to increase its titre. Virus containing media obtained after two rounds of infection is used for large-scale protein expression. For large-scale protein expression 100 cm2 round tissue culture plates are seeded with 5 x 107 cells/plate and infected with 1 mL of virus-containing media (approx. 5 MOIs). After 3 days the cells are scraped off the plate and centrifuged at 500 rpm for 5 min. Cell pellets from 10-20, 100 cm2 plates, are resuspended in 50 mL of ice-cold lysis buffer (25 mMTris- HCI, pH 7.5, 2mM EDTA, 1% NP-40, 1 mM DTT1 1 mM PMSF). The cells are stirred on ice for 15 min and then centrifuged at 5000 rpms for 20 min.
Purification of GST-tagged proteins: The centrifuged cell lysate is loaded onto a 2 mL gluta- thione-sepharose column (Pharmacia) and washed three times with 10 mL of 25 mM Tris- HCI, pH 7.5, 2 mM EDTA, 1 mM DTT, 200 mM NaCI. The GST-tagged protein is then eluted by 10 applications (1 mL each) of 25 mM Tris-HCI, pH 7.5, 10 mM reduced-glutathione, 100 mM NaCI, 1 mM DTT, 10 % Glycerol and stored at -700C.
Measurement of enzyme activity: Tyrosine protein kinase assays with purified GST-Flt-3 are carried out in a final volume of 30 μL containing 200-1800 ng of enzyme protein (depending on the specific activity), 20 mM Tris-HCI, pH 7.6, 3 mM MnCI2, 3 mM MgCI2, 1 mM DTT, 10 μM Na3VO4, 3 μg/mL poly(Glu.Tyr) 4:1 , 1 % DMSO, 8.0 μM ATP and 0.1 μCi [γ33 P] ATP). The activity is assayed in the presence or absence of inhibitors, by measuring the incorporation of 33P from [γ33P] ATP into the poly(Glu.Tyr) substrate. The assay (30 μL) is carried out in 96-well plates at ambient temperature for 20 min under conditions described below and terminated by the addition of 20 μL of 125 mM EDTA. Subsequently, 40 μL of the reaction mixture is transferred onto Immobilon-PVDF membrane (Millipore, Bedford, MA, USA) previously soaked for 5 min with methanol, rinsed with water, then soaked for 5 min with 0.5 % H3PO4 and mounted on vacuum manifold with disconnected vacuum source. After spotting all samples, vacuum is connected and each well rinsed with 200 μL 0.5 % H3PO4. Membranes are removed and washed 4 x on a shaker with 1.0 % H3PO4, once with ethanol. Membranes are counted after drying at ambient temperature, mounting in Packard TopCount 96-well frame, and addition of 10 μL/well of Microscint TM (Packard). IC50 values are calculated by linear regression analysis of the percentage inhibition of each compound in duplicate, at four concentrations (usually 0.01 , 0.1 , 1 and 10 μM). One unit of protein kinase activity is defined as 1 nmole of 33P ATP transferred from [γ33P] ATP to the substrate protein per minute per mg of protein at 37 0C. The compounds of the formula I show IC50 values for Flt-3 inhibition in the range between 0.01 and 100 μM, preferably between 0.05 and 10 μM. The compounds of formula I also inhibit other tyrosine protein kinases such as especially the c-Src kinase, c-Kit, VEGF-R and/or FGFR; all of which play a part in growth regulation and transformation in animal, especially mammal cells, including human cells. An appropriate assay is described in Andrejauskas-Buchdunger et al., Cancer Res. 52, 5353-8 (1992). Using this test system, compounds of the formula I show IC50 values for inhibition of c-Src in the range of 0.005 to 100 μM, usually between 0.005 and 5 μM. Compounds of formula I also show IC50 values for c-kit inhibition in the range of 0.005 to 10 μM, usually between 0.005 and 5 μM; and for inhibition of FGFR-1 , up to 95% inhibition at 10 μM.
The inhibition of IGF-1R and Ins-R can be determined as follows: The baculovirus donor vector pfbgx3IGFIRcd is used to generate a recombinant baculovirus that expresses the amino acid region 950-1337 of the mature peptide cytoplasmic domain of the human IGF-IR. To generate the cDNA fragment encoding the amino acid region 919-1343 of the intra- cytoplasmic kinase domain of the human insulin receptor, pCδhlnsR is used. The fragments of the human IGF-IR and Ins-R are cloned, expressed and small-scale purified as a factor Xa-cleavable glutathione-S-transferase (GST)-fusion protein using the Bac-to-Bac™ system (GIBCO BRL) of recombinant baculovirus generation. Virus containing media iss collected from the transfected cell culture and used for infection to increase its titer. Virus containing media obtained after two rounds of infection iss used for large-scale protein expression. Cell extracts are prepared and loaded onto a glutathione-Sepharose (Pharmacia) column. After washing, the GST-tagged proteins are then eluted with a glutathione-containing buffer. Purified protein is stored at -700C in elution buffer. Tyrosine protein kinase assays with purified GST-IGF-1 R and GST-lns-R are carried in a final volume of 30 μl containing 20 mM Tris-HCI, pH 7.6, 10 mM MgCI2, 0.01 mM Na3VO4, 1% DMSO, 1 mM DTT, 3 μg/ml poly(Glu, Tyr) 4:1 and 10 μM ATP (γ-[33P]-ATP 0.1 μCi). The assay is performed in 96-well plates at ambient temperature for 20 min and terminated by addition of 25 μl 0.05 M EDTA pH 7.0. An aliquot of 40 μl is spotted with a multichannel dispenser on Whatman P81 membranes mounted in a Millipore Microtiter filter manifold connected to a low vacuum source. After elimination of liquid, the membrane is transferred to a sequence of 4 washing baths containing 0.5% H3PO4 and one with EtOH (shaking incubation for 10 min each), dried, mounted onto a Hewlett Packard TopCount manifold added 10 μl Microscint® and counted. Compounds of formula (I) show up to 90% inhibition of Ins-R at 10,00OnM, preferably between 60-90% inhibition.
The inhibition of Tek can be determined as follows: The procedure of the expression, purification and assay these kinases has been described. Fabbro et al., Pharmacol. Ther. 82(2-3) 293-301 (1999). In brief, the glutathione S-transferase (GST) gene from the pAcG1 vector (Pharmingen) is excised with EcoRV and EcoRI and inserted into the cloning site of the Fast-Bac baculoviral vector (GIBCO) creating a 5530 bp vector with N-terminal cloning sites derived from the pAcG1 fusion vector (FBGO). The C-terminal cloning site may be any cloning site (from the Fast-Bac vector) downstream of the N-terminal cloning site used. N- terminally GST-fused (pAcG1 , Pharmingen) KDR1 Flt-1, Flk-1 , Tek and PDGFR-β kinase domains are obtained from ProQinase, Freiburg, Germany. Tek is recloned into the FBG1 vector by EcoRI excision and ligation into EcoRI digested FBG1 (FBG1-Tek). The coding sequences for the whole cytoplasmic domain of c-Kit (aa 544-976) and c-Fms (aa 538-972) are amplified by PCR from human uterus and from human bone marrow cDNA libraries (Clontech), respectively. The amplified DNA fragments are fused to GST by cloning them into FBG1 as BamHI-EcoRI insertions, to yield FBG1-c-Kit and FBG1-c-Fms. Tek is recloned into the FBGO transfer vector by EcoRI excision and ligation into EcoRI digested FBGO (FBG-Tie2/Tek). FGFR-1 and c-met kinase domains are obtained by PCR from human A431 cells. N-terminal primers contain an overhanging EcoRI site, while C-terminal primers contain a Xhol site to aid cloning into the transfer vectors. After digestion of both the PCR fragments and FBGO the cleavage products are gel-purified and ligated together to form the kinase constructs (FBG-Met, FBG-FGFR-1).
Viruses for each of the kinases are made according to the protocol supplied by GIBCO. In brief, transfer vectors containing the kinase domains are transfected into the DHIOBac cell line (GIBCO), plated on agar plates containing the recommended concentrations of Blue- Gal, IPTG, Kanamycin, Tetracycline, and Gentamycin. Colonies without insertion of the fusion sequence into the viral genome (carried by the bacteria) are blue. A single white colony is usually picked and viral DNA (bacmid) isolated from the bacteria by standard plasmid mini prep procedures. Sf9 cells or High Five cells (GIBCO) are then transfected in 25 cm2 flasks with the viral DNA using the Cellfectin reagent and protocol supplied with the Bac-to-Bac kit (GIBCO). Virus containing media is collected from the transfected cell culture and used for infection to increase its titer. Virus containing media obtained after two rounds of infection is used for large-scale protein expression. For large-scale protein expression 100 cm2 round tissue culture plates are seeded with 5x107 cells/plate and infected with 1 ml of virus-containing media (about 5 MOIs). After 3 days the cells are scraped off the plate and centrifuged at 500 rpm for 5 min.
Cell pellets from 10-20, 100 cm2 plates, are resuspended in 50 ml of ice-cold lysis buffer (25 mM Tris-HCI, pH 7.5, 2 mM EDTA, 1 % NP-40, 1 mM DTT, 1 mM PMSF). The cells are stirred on ice for 15 min and then centrifuged at 5000 rpms for 20 min. The supernatant is loaded onto a 2 ml glutathione-sepharose column and washed three times with 10 ml of 25 mM Tris-HCI, pH 7.5, 2 mM EDTA1 1 mM DTT, 200 mM NaCI. The GST-tagged proteins are then eluted by 10 applications (1 ml each) of 25 mM Tris-HCI, pH 7.5, 10 mM reduced- glutathione, 100 mM NaCI, 1 mM DTT, 10% Glycerol and stored at -700C. The assays (30 μl) contain 200-1800 ng of enzyme protein (depending on the specific activity), 20 mM Tris-HCI, pH 7.6, 3 mM MnCI2, 3 mM MgCI2, 1 mM DTT, 10 μM Na3VO4, 3 μg/ml poly(Glu.Tyr) 4:1 , 8 μM ATP (γ-[33P]-ATP 0.1 μCi). Reactions are incubated for 20 min at ambient temperature and then stopped by addition of 25 μl 0.25 M EDTA (pH 7.0). An aliquot of 40 μl is spotted with a multichannel dispenser on lmmobilon P membranes mounted in a Millipore Microtiter filter manifold connected to a low vacuum source. After elimination of liquid, the membrane is transferred to a sequence of 4 washing baths containing 0.5% H3PO4 and one with EtOH (shaking incubation for 10 min each), dried, mounted onto a Hewlett Packard TopCount manifold added 10 μl Microscint® and counted. Compounds of formula (I) show IC50 values, calculated by linear regression analysis, for Tek inhibition of about 0.1-100μM.
The inhibition of Cdk1 can be determined as follows: Cdk1/cycB: Cdk1/cycB are obtained from ProQinase, Freiburg, Germany. Starfish oocytes are induced to enter M phase of the cell cycle with 10 μM 1-methyladenine and frozen in liquid nitrogen and stored at - 800C. When required, the oocytes are homogenized and centrifuged as described (Arion et al., Cell 55: 371-378 (1988) and Rialet et al., Anticancer Res. 11 : 1581-1590 (1991)). Cdk1/cycB kinase is purified on p9CKShs-sepharose beads and eluted with recombinant human p9CKShs as described (Azzi et al., Eur. J. Biochem. 203: 353-360. (1992)). Briefly, the supernatant from oocytes is equilibrated for 30 min at 4°C under constant rotation with the p9CKShs-sepharose beads. The beads are extensively washed and active cdk1/cycB kinase is eluted with purified pθCKShs (3 mg/ml). The activity of Cdk1/cycB is measured as described (Arion et al., Cell 55: 371-378 (1988), Meijer et al., EMBO J. 1989; 8: 2275-2282 and Meijer et al., EMBO J. 1991 ; 8: 2275-2282). The assay is carried with slight modifications in 96-well plates at ambient temperature for 20 min. The final volume of 30 μl contains 0.1-0.3U of Cdk1/cycB, 1 mg/ml histone H1 as a substrate, 60 mM β-glycerophosphate, 30 mM nitrophenylphosphate, 25 mM MOPS, 5 mM EGTA, 15 mM MgCI2, 1 mM DTT, 0.1 mM Na3VO4, 15 μM ATP and 0.1 μCi γ-33P-ATP (75 μM, 8800 cpm/pmole). The reaction is terminated by addition of 25 μl 0.05 M EDTA pH 7.0. An aliquot of 40 μl is spotted with a multichannel dispenser on lmmobilon P membranes mounted in a Millipore Microtiter filter manifold connected to a low vacuum source. After elimination of liquid, the membrane is transferred to a sequence of 4 washing baths containing 0.5% H3PO4 and one with EtOH (shaking incubation for 10 min each), dried, mounted onto a Hewlett Packard TopCount manifold added 10 μl Microscint® and counted. Compounds of formula (I) show up to 100% Cdk1 inhibition at 10,00OnM.
The inhibition of c-Raf-1 can be determined as follows: Production of recombinant c-Raf- 1 protein, is obtained by triple infection of Sf21 cells with GST-c-Raf-1 recombinant baculovirus together with v-Src and v-Ras recombinant baculoviruses that are required for active c-Raf-1 kinase production (Williams et al., PNAS 1992; 89: 2922-2926). Active Ras (v- Ras) is required to recruit c-Raf-1 to the cell membrane and v-Src to phosphorylate c-Raf-1 to fully activate it (Williams et al., PNAS 1992; 89: 2922-2926). Cells were seeded at 2.5 x 107cells per 150 mm dish and allowed to attach to a 150 mm dish for 1hr at RT. Media (SF900II containing 10 % FBS) is aspirated and recombinant baculovirus; GST-C-Raf-1 , v- Ras and v-Src are added at MOI of 3.0, 2.5 and 2.5 receptively in a total volume of 4-5 mL. Cells are incubated for 1 hr at RT and then 15 mL of medium is added. Infected cells are incubated for 48-72 hr at 270C. Infected Sf21 cells are scraped and collected into a 50 mL tube and centrifuged for 10 min at 40C at 1100 g in a Sorvall centrifuge. The cell pellet is washed once with ice cold PBS and lysed with 0.6 mL lysis buffer per 2.5 x 107 cells. Complete lysis of cells is achieved after 10 min on ice with occasional pipetting. The cell lysates are centrifuged for 10 min at 40C at 14,500 g in a Sorvall centrifuge with SS-34 rotor and the supernatant is transferred to a fresh tube and stored at -800C. c-Raf-1 is purified from cell lysates using 100 uL of packed Glutathione-Sepharose 4B beads equilibrated in ice cold PBS per 2.5 x 107 cells. GST-c-Raf-1 was allowed to bind to the beads at 40C for 1hr with rocking. Bound GST-c-Raf-1 with beads was transferred to a column. The column is washed once with lysis buffer and twice with ice cold Tris buffered saline. Ice cold elution buffer is added and column flow is stopped to allow the free glutathione to disrupt the interaction of GST-c-Raf-1 with glutathione sepharose beads. Fractions (1 mL) are collected into pre-chilled tubes. Each tube contains 10 % glycerol (final concentration) to maintain kinase activity during freeze thaw cycles. Purified fractions of GST-c-Raf-1 kinase protein are stored at -800C.
IKB was used as substrate for the c-Raf-1 kinase. IKB is expressed in bacteria as a His- tagged protein (cloned and kindly provided by Dr. Eder; ABM, Novartis, Basel). BL21 LysS bacteria containing the IKB plasmid are grown to an OD6oo of 0.6 in LB medium then induced to express the kb with IPTG (final concentration of 1 mM) for 3 hrs at 37° C and then bacteria are lysed by sonication (microtip limit setting for 3 times at 1 min each in sonication buffer [50 mM Tris pH 8.0, 1 mM DTT, 1 mM EDTA] and centrifuged at 10,000 g for 15 min. The supernatant is mixed with ammonium sulfate to give a final concentration of 30 %. This mixture is rocked for 15 min at 4 0C then spun at 10,000 g for 15 min. The pellet is resuspended in binding buffer (Novagen) containing 10 mM BSA. This solution is applied to Ni-agarose (Novagen) and washed according to the Novagen manual. IKB is eluted from the column using elution buffer (0.4 M imidazole, 0.2 M NaCI, 8 mM Tris pH 7.9). Fractions containing protein are dialysed in 50 mM Tris pH 8, 1 mM DTT.
The activity of c-Raf-1 protein kinase is assayed in the presence or absence of inhibitors, by measuring the incorporation of 33P from [γ33P] ATP into IB. The assay is carried out in 96- well plates at ambient temperature for 60 min. It contains (total volume of 30 μl): c-rafl 1 kinase (400 ng), 25 mM Tris HCI, pH 7.5, 5 mM MgCI2, 5 mM MnCI2, 10 μM Na3VO4, 1 mM DTT and 0.3 μCi/assay [γ33 P]-ATP (10 μM ATP) using 600 ng IB in the presence of 1 % DMSO. Reactions are terminated by adding 10 μL of 250 mM EDTA and 30 μL of the reaction mixture is transferred onto Immobilon-PVDF membrane (Millipore, Bedford, MA, USA) previously soaked for 5 min with methanol, rinsed with water, then soaked for 5 min with 0.5 % H3PO4 and mounted on vacuum manifold with disconnected vacuum source. After spotting all samples, vacuum is connected and each well rinsed with 200 μL 0.5 % H3PO4. Membranes are removed and washed 4 x on a shaker with 0.5 % H3PO4, once with ethanol. Membranes are counted after drying at ambient temperature, mounting in Packard TopCount 96-well frame, and addition of 10 μL/well of Microscint TM (Packard). Compounds of formula (I) show c-Raf-1 inhibition in the range between 0.1-50 μM, preferably between 0.1 and 10 μM.
The efficacy of compounds of the invention as inhibitors of Ephrin B4 receptor (EphB4) kinases can be demonstrated as follows: Generation of Bac-to-BacTM (Invitrogen Life Technologies, Basel, Switzerland) GST-fusion expression vectors: Entire cytoplasmatic coding regions of the EphB-class are amplified by PCR from cDNA libraries derived from human placenta or brain, respectively. Recombinant baculovirus are generated that express the amino acid region 566-987 of the human EphB4 receptor (SwissProt Database, Accession No. P54760). GST sequence is cloned into pFastBad® vector (Invitrogen Life Technologies, Basel, Switzerland) and PCR amplified. cDNAs encoding EphB4- receptor domains, respectively are cloned in frame 3'prime to the GST sequence into this modified FastBad vector to generate pBac-to-Bac™ donor vectors. Single colonies arising from the transformation are inoculated to give overnight cultures for small scale plasmid preparation. Restriction enzyme analysis of plasmid DNA reveals several clones to contain inserts of the expected size. By automated sequencing the inserts and approximately 50 bp of the flanking vector sequences are confirmed on both strands. Production of viruses: Viruses for each of the kinases are made according to the protocol supplied by GIBCO if not stated otherwise. In brief, transfer vectors containing the kinase domains are transfected into the DHIOBac cell line (GIBCO) and plated on selective agar plates. Colonies without insertion of the fusion sequence into the viral genome (carried by the bacteria) are blue. Single white colonies are picked and viral DNA (bacmid) isolated from the bacteria by standard plasmid purification procedures. Sf9 cells or Sf21 cells are then transfected in 25 cm2 flasks with the viral DNA using Cellfectin reagent according to the protocol.
Purification of GST-tagged kinases: The centrifuged cell lysate is loaded onto a 2 mL glutathione-sepharose column (Pharmacia) and washed three times with 10 mL of 25 mM Tris-HCI, pH 7.5, 2mM EDTA, 1 mM DTT1 200 mM NaCI. The GST-tagged proteins are then eluted by 10 applications (1 mL each) of 25 mM Tris-HCI, pH 7.5, 10 mM reduced- glutathione, 100 mM NaCI, 1 mM DTT, 10 % Glycerol and stored at -700C. Protein kinase assays: The activities of protein kinases are assayed in the presence or absence of inhibitors, by measuring the incorporation of 33P from [D33PJATP into a polymer of glutamic acid and tyrosine (poly(Glu.Tyr)) as a substrate. The kinase assays with purified GST-EphB (30ng) are carried out for 15-30 min at ambient temperature in a final volume of 30 HL containing 20 mM Tris-HCI , pH 7.5, 10 mM MgCI2, 3-50 mM MnCI2, 0.01 mM Na3VO4, 1 % DMSO, 1 mM DTT, 3 μg/mL poly(Glu.Tyr) 4:1 (Sigma; St. Louis, Mo., USA) and 2.0-3.0 μM ATP (γ-[33P]-ATP 0.1 μCi). The assay is terminated by the addition of 20 μL of 125 mM EDTA. Subsequently, 40 μl of the reaction mixture are transferred onto Immobilon-PVDF membrane (Millipore, Bedford, MA, USA) previously soaked for 5 min with methanol, rinsed with water, then soaked for 5 min with 0.5 % H3PO4 and mounted on vacuum manifold with disconnected vacuum source. After spotting all samples, vacuum is connected and each well rinsed with 200 μl 0.5 % H3PO4. Membranes are removed and washed 4 x on a shaker with 1.0 % H3PO4, once with ethanol. Membranes are counted after drying at ambient temperature, mounting in Packard TopCount96-well frame, and addition of 10 μL/well of MicroscintTM (Packard). IC50 values are calculated by linear regression analysis of the percentage inhibition of each compound in duplicate, at four concentrations (usually 0.01 , 0.1 , 1 and 10 μM). One unit of protein kinase activity is defined as 1 nmole of 33P ATP transferred from [γ33P] ATP to the substrate protein per minute per mg of protein at 37 0C. Compounds of formula I show EphB4 inhibition down to 1 nM, preferably IC50 values between 0.001-5.0 μM.
Alternatively, EphB4 receptor autophosphorylation can be measured as follows: The inhibition of EphB4 receptor autophosphorylation can be confirmed with an in vitro experiment in cells such as transfected A375 human melanoma cells (ATCC Number: CRL- 1619), which permanently express human EphB4 (SwissProt AccNo P54760), are seeded in complete culture medium (with 10% fetal calf serum = FCS) in 6-well cell-culture plates and incubated at 37°C under 5% CO2 until they show about 90% confluency. The compounds to be tested are then diluted in culture medium (without FCS, with 0.1% bovine serum albumin) and added to the cells. (Controls comprise medium without test compounds). Ligand induced autophosphorylation is induced by the addition of 1 microg/ml soluble ephrinB2-Fc (s-ephrinB2- Fc : R&D Biosystems, CatNr 496-EB) and 0.1 microM ortho-vanadate. After a further 20 minutes incubation at 37°C, the cells are washed twice with ice-cold PBS (phosphate-buffered saline) and immediately lysed in 200 μl lysis buffer per well. The lysates are then centrifuged to remove the cell nuclei, and the protein concentrations of the supernatants are determined using a commercial protein assay (PIERCE). The lysates can then either be immediately used or, if necessary, stored at -200C.
A sandwich ELISA is carried out to measure the EphB4 phosphorylation: To capture phosphorylated EphB4 proteinlOO ng/well of ephrinB2-Fc (s-ephrinB2-Fc : R&D Biosystems, CatNr 496-EB) is immobilized MaxiSorb (Nunc) ELISA plates. The plates are then washed and the remaining free protein-binding sites are saturated with 3% TopBlock® (Juro, Cat. # TB232010) in phosphate buffered saline with Tween 20® (polyoxyethylen(20)sorbitane monolaurate, ICI/Uniquema) (PBST). The cell lysates (100 μg protein per well) are then incubated in these plates for 1 h at room temperature. After washing the wells three times with PBS an antiphosphotyrosine antibody coupled with alkaline phosphatase (PY 20 Alkaline Phosphate conjugated: ZYMED, Cat NrO3-7722) is added and incubated for another hour. The plates are washed again and the binding of the antiphosphotyrosine antibody to the captured phosphorylated receptor is then demonstrated and quantified using 10 mM D- nitrophenylphosphat as subtrate and measuring the OD at 405 nm after 0.5h-1h. The difference between the signal of the positive control (stimulated with vanadate and s- ephrinB2-Fc) and that of the negative control (not stimulated) corresponds to maximal EphB4 phosphorylation (= 100 %). The activity of the tested substances is calculated as percent inhibition of maximal EphB4 phosphorylation, wherein the concentration of substance that induces half the maximum inhibition is defined as the IC50 (inhibitory dose for 50% inhibition).
Experiments to demonstrate the anti-tumor activity of compounds of the formula (I) in vivo: For example, in order to test whether a compound of the formula (I), e.g. that of Example 1 given below, inhibits VEGF-mediated angiogenesis in vivo, its effect on the angiogenic response induced by VEGF in a growth factor imlant model in mice is tested: A porous Teflon chamber (volume 0.5 mL) is filled with 0.8 % w/v agar containing heparin (20 units/ml) with or without growth factor (2 μg/ml human VEGF) is implanted subcutaneously on the dorsal flank of C57/C6 mice. The mice are treated with the test compound (e.g. 25, 50 or 100 mg/kg p.o. once daily) or vehicle starting on the day of implantation of the chamber and continuing for 4 days after. At the end of the treatment, the mice are killed, and the chambers are removed. The vascularized tissue growing around the chamber is carefully removed and weighed, and the blood content is assessed by measuring the hemoglobin content of the tissue (Drabkins method; Sigma, Deisenhofen, Germany). It has been shown previously that these growth factors induce dose-dependent increases in weight and blood content of this tissue growing (characterized histologically to contain fibroblasts and small blood vessels) around the chambers and that this response is blocked by antibodies that specifically neutralize VEGF (see Wood JM et al., Cancer Res. 60(8), 2178-2189, (2000); and Schlaeppi et al., J. Cacner Res. Clin. Oncol. 125, 336-342, (1999)). With this model, inhibition can be shown in the case of compounds of the formula (I).
The invention relates also to pharmaceutical compositions comprising a compound of formula (I), to their use in the therapeutic (in a broader aspect of the invention also prophylactic) treatment or a method of treatment of a kinase dependent disease, especially the preferred diseases mentioned above, to the compounds for said use and to the preparation of pharmaceutical preparations, especially for said uses.
The present invention also relates to pro-drugs of a compound of formula (I) that convert in vivo to the compound of formula (I) as such. Any reference to a compound of formula (I) is therefore to be understood as referring also to the corresponding pro-drugs of the compound of formula (I), as appropriate and expedient. The pharmacologically acceptable compounds of the present invention may be used, for example, for the preparation of pharmaceutical compositions that comprise an effective amount of a compound of the formula (I), or a pharmaceutically acceptable salt thereof, as active ingredient together or in admixture with a significant amount of one or more inorganic or organic, solid or liquid, pharmaceutically acceptable carriers.
The invention relates also to a pharmaceutical composition that is suitable for administration to a warm-blooded animal, especially a human (or to cells or cell lines derived from a warmblooded animal, especially a human, e.g. lymphocytes), for the treatment or, in a broader aspect of the invention, prevention of (= prophylaxis against) a disease that responds to inhibition of kinase activity, comprising an amount of a compound of formula (I) or a pharmaceutically acceptable salt thereof, which is effective for said inhibition, especially the in, together with at least one pharmaceutically acceptable carrier.
The pharmaceutical compositions according to the invention are those for enteral, such as nasal, rectal or oral, or parenteral, such as intramuscular or intravenous, administration to warm-blooded animals (especially a human), that comprise an effective dose of the pharmacologically active ingredient, alone or together with a significant amount of a pharmaceutically acceptable carrier. The dose of the active ingredient depends on the species of warm-blooded animal, the body weight, the age and the individual condition, . individual pharmacokinetic data, the disease to be treated and the mode of administration.
The invention relates also to a method of treatment for a disease that responds to inhibition of a kinase; which comprises administering an (against the mentioned disease) prophylactically or especially therapeutically effective amount of a compound of formula (l)according to the invention, especially to a warm-blooded animal, for example a human, that, on account of one of the mentioned diseases, requires such treatment.
The dose of a compound of the formula (I) or a pharmaceutically acceptable salt thereof to be administered to warm-blooded animals, for example humans of approximately 70 kg body weight, is preferably from approximately 3 mg to approximately 10 g, more preferably from approximately 10 mg to approximately 1.5 g, most preferably from about 100 mg to about 1000 mg /person/day, divided preferably into 1-3 single doses which may, for example, be of the same size. Usually, children receive half of the adult dose. The pharmaceutical compositions comprise from approximately 1% to approximately 95%, preferably from approximately 20% to approximately 90%, active ingredient. Pharmaceutical compositions according to the invention may be, for example, in unit dose form, such as in the form of ampoules, vials, suppositories, dragees, tablets or capsules.
The pharmaceutical compositions of the present invention are prepared in a manner known per se, for example by means of conventional dissolving, lyophilizing, mixing, granulating or confectioning processes.
Solutions of the active ingredient, and also suspensions, and especially isotonic aqueous solutions or suspensions, are preferably used, it being possible, for example in the case of lyophilized compositions that comprise the active ingredient alone or together with a carrier, for example mannitol, for such solutions or suspensions to be produced prior to use. The pharmaceutical compositions may be sterilized and/or may comprise excipients, for example preservatives, stabilizers, wetting and/or emulsifying agents, solubilizers, salts for regulating the osmotic pressure and/or buffers, and are prepared in a manner known per se, for example by means of conventional dissolving or lyophilizing processes. The said solutions or suspensions may comprise viscosity-increasing substances, such as sodium carboxymethylcellulose, carboxymethylcellulose, dextran, polyvinylpyrrolidone or gelatin.
Suspensions in oil comprise as the oil component the vegetable, synthetic or semi-synthetic oils customary for injection purposes. There may be mentioned as such especially liquid fatty acid esters that contain as the acid component a long-chained fatty acid having from 8- 22, especially from 12-22, carbon atoms, for example lauric acid, tridecylic acid, myristic acid, pentadecylic acid, palmitic acid, margaric acid, stearic acid, arachidic acid, behenic acid or corresponding unsaturated acids, for example oleic acid, elaidic acid, erucic acid, brasidic acid or linoleic acid, if desired with the addition of antioxidants, for example vitamin E, β-carotene or 3,5-di-tert-butyl-4-hydroxytoluene. The alcohol component of those fatty acid esters has a maximum of 6 carbon atoms and is a mono- or poly-hydroxy, for example a mono-, di- or tri-hydroxy, alcohol, for example methanol, ethanol, propanol, butanol or pentanol or the isomers thereof, but especially glycol and glycerol. The following examples of fatty acid esters are therefore to be mentioned: ethyl oleate, isopropyl myristate, isopropyl palmitate, "Labrafil M 2375" (polyoxyethylene glycerol trioleate, Gattefosse, Paris), "Miglyol 812" (triglyceride of saturated fatty acids with a chain length of C8 to C12, HuIs AG, Germany), but especially vegetable oils, such as cottonseed oil, almond oil, olive oil, castor oil, sesame oil, soybean oil and more especially groundnut oil.
Pharmaceutical compositions for oral administration can be obtained by combining the active ingredient with solid carriers, if desired granulating a resulting mixture, and processing the mixture, if desired or necessary, after the addition of appropriate excipients, into tablets, dragee cores or capsules. It is also possible for them to be incorporated into plastics carriers that allow the active ingredients to diffuse or be released in measured amounts.
Suitable carriers are especially fillers, such as sugars, for example lactose, saccharose, mannitol or sorbitol, cellulose preparations and/or calcium phosphates, for example tricalcium phosphate or calcium hydrogen phosphate, and binders, such as starch pastes using for example corn, wheat, rice or potato starch, gelatin, tragacanth, methylcellulose, hydroxypropylmethylcellulose, sodium carboxymethylcellulose and/or polyvinylpyrrolidone, and/or, if desired, disintegrators, such as the above-mentioned starches, and/or carboxymethyl starch, crosslinked polyvinylpyrrolidone, agar, alginic acid or a salt thereof, such as sodium alginate. Excipients are especially flow conditioners and lubricants, for example silicic acid, talc, stearic acid or salts thereof, such as magnesium or calcium stearate, and/or polyethylene glycol. Dragee cores are provided with suitable, optionally enteric, coatings, there being used, inter alia, concentrated sugar solutions which may comprise gum arabic, talc, polyvinylpyrrolidone, polyethylene glycol and/or titanium dioxide, or coating solutions in suitable organic solvents, or, for the preparation of enteric coatings, solutions of suitable cellulose preparations, such as ethylcellulose phthalate or hydroxypropylmethylcellulose phthalate. Capsules are dry-filled capsules made of gelatin and soft sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol. The dry-filled capsules may comprise the active ingredient in the form of granules, for example with fillers, such as lactose, binders, such as starches, and/or glidants, such as talc or magnesium stearate, and if desired with stabilizers. In soft capsules the active ingredient is preferably dissolved or suspended in suitable oily excipients, such as fatty oils, paraffin oil or liquid polyethylene glycols, it being possible also for stabilizers and/or antibacterial agents to be added. Dyes or pigments may be added to the tablets or dragee coatings or the capsule casings, for example for identification purposes or to indicate different doses of active ingredient. A compound of the formula (I) may also be used to advantage in combination with other antiproliferative agents. Such antiproliferative agents include, but are not limited to aro- matase inhibitors; antiestrogens; topoisomerase I inhibitors; topoisomerase Il inhibitors; microtubule active agents; alkylating agents; histone deacetylase inhibitors; compounds which induce cell differentiation processes; cyclooxygenase inhibitors; MMP inhibitors; mTOR inhibitors; antineoplastic antimetabolites; platin compounds; compounds targeting/decreasing a protein or lipid kinase activity and further anti-angiogenic compounds; compounds which target, decrease or inhibit the activity of a protein or lipid phosphatase; gonadorelin agonists; anti-androgens; methionine aminopeptidase inhibitors; bisphosphonates; biological response modifiers; antiproliferative antibodies; heparanase inhibitors; inhibitors of Ras oncogenic isoforms; telomerase inhibitors; proteasome inhibitors; agents used in the treatment of hematologic malignancies; compounds which target, decrease or inhibit the activity of Flt-3; Hsp90 inhibitors; temozolomide (TEMODAL®); and leucovorin.
The term "aromatase inhibitor" as used herein relates to a compound which inhibits the estrogen production, i.e. the conversion of the substrates androstenedione and testosterone to estrone and estradiol, respectively. The term includes, but is not limited to steroids, especially atamestane, exemestane and formestane and, in particular, non-steroids, especially aminoglutethimide, roglethimide, pyridoglutethimide, trilostane, testolactone, ketokonazole, vorozole, fadrozole, anastrozole and letrozole. Exemestane can be administered, e.g., in the form as it is marketed, e.g. under the trademark AROMASIN. Formestane can be administered, e.g., in the form as it is marketed, e.g. under the trademark LENTARON. Fadrozole can be administered, e.g., in the form as it is marketed, e.g. under the trademark AFEMA. Anastrozole can be administered, e.g., in the form as it is marketed, e.g. under the trademark ARIMIDEX. Letrozole can be administered, e.g., in the form as it is marketed, e.g. under the trademark FEMARA or FEMAR. Aminoglutethimide can be administered, e.g., in the form as it is marketed, e.g. under the trademark ORIMETEN. A combination of the invention comprising a chemotherapeutic agent which is an aromatase inhibitor is particularly useful for the treatment of hormone receptor positive tumors, e.g. breast tumors.
The term "antiestrogen" as used herein relates to a compound which antagonizes the effect of estrogens at the estrogen receptor level. The term includes, but is not limited to tamoxifen, fulvestrant, raloxifene and raloxifene hydrochloride. Tamoxifen can be admi- nistered, e.g., in the form as it is marketed, e.g. under the trademark NOLVADEX. Raloxifene hydrochloride can be administered, e.g., in the form as it is marketed, e.g. under the trademark EVISTA. Fulvestrant can be formulated as disclosed in US 4,659,516 or it can be administered, e.g., in the form as it is marketed, e.g. under the trademark FASLODEX. A combination of the invention comprising a chemotherapeutic agent which is an antiestrogen is particularly useful for the treatment of estrogen receptor positive tumors, e.g. breast tumors.
The term "anti-androgen" as used herein relates to any substance which is capable of inhibiting the biological effects of androgenic hormones and includes, but is not limited to, bicalutamide (CASODEX), which can be formulated, e.g. as disclosed in US 4,636,505.
The term "gonadorelin agonist" as used herein includes, but is not limited to abarelix, go- serelin and goserelin acetate. Goserelin is disclosed in US 4,100,274 and can be administered, e.g., in the form as it is marketed, e.g. under the trademark ZOLADEX. Abarelix can be formulated, e.g. as disclosed in US 5,843,901.
The term "topoisomerase I inhibitor" as used herein includes, but is not limited to topotecan, gimatecan, irinotecan, camptothecian and its analogues, 9-nitrocamptothecin and the macromolecular camptothecin conjugate PNU-166148 (compound A1 in WO99/ 17804). Irinotecan can be administered, e.g. in the form as it is marketed, e.g. under the trademark CAMPTOSAR. Topotecan can be administered, e.g., in the form as it is marketed, e.g. under the trademark HYCAMTIN.
The term "topoisomerase Il inhibitor" as used herein includes, but is not limited to the an- thracyclines such as doxorubicin (including liposomal formulation, e.g. CAELYX), dauno- rubicin, epirubicin, idarubicin and nemorubicin, the anthraquinones mitoxantrone and lo- soxantrone, and the podophillotoxines etoposide and teniposide. Etoposide can be administered, e.g. in the form as it is marketed, e.g. under the trademark ETOPOPHOS. Teniposide can be administered, e.g. in the form as it is marketed, e.g. under the trademark VM 26-BRISTOL. Doxorubicin can be administered, e.g. in the form as it is marketed, e.g. under the trademark ADRIBLASTIN or ADRIAMYCIN. Epirubicin can be administered, e.g. in the form as it is marketed, e.g. under the trademark FARMORUBICIN. Idarubicin can be administered, e.g. in the form as it is marketed, e.g. under the trademark ZAVEDOS. Mitoxantrone can be administered, e.g. in the form as it is marketed, e.g. under the trademark NOVANTRON. The term "microtubule active agent" relates to microtubule stabilizing, microtubule destabilizing agents and microtublin polymerization inhibitors including, but not limited to taxanes, e.g. paclitaxel and docetaxel, vinca alkaloids, e.g., vinblastine, especially vinblastine sulfate, vincristine especially vincristine sulfate, and vinorelbine, discodermolides, cochicine and epothilones and derivatives thereof, e.g. epothilone B or D or derivatives thereof. Paclitaxel may be administered e.g. in the form as it is marketed, e.g. TAXOL. Docetaxel can be administered, e.g., in the form as it is marketed, e.g. under the trademark TAXOTERE. Vinblastine sulfate can be administered, e.g., in the form as it is marketed, e.g. under the trademark VINBLASTIN R. P. Vincristine sulfate can be administered, e.g., in the form as it is marketed, e.g. under the trademark FARMISTIN. Discodermolide can be obtained, e.g., as disclosed in US 5,010,099. Also included are Epothilone derivatives which are disclosed in WO 98/10121 , US 6,194,181 , WO 98/25929, WO 98/08849, WO 99/43653, WO 98/22461 and WO 00/31247. Especially preferred are Epothilone A and/or B.
The term "alkylating agent" as used herein includes, but is not limited to, cyclophosphamide, ifosfamide, melphalan or nitrosourea (BCNU or Gliadel). Cyclophosphamide can be administered, e.g., in the form as it is marketed, e.g. under the trademark CYCLOSTIN. Ifosfamide can be administered, e.g., in the form as it is marketed, e.g. under the trademark HOLOXAN.
The term "histone deacetylase inhibitors" or "HDAC inhibitors" relates to compounds which inhibit the histone deacetylase and which possess antiproliferative activity. This includes compounds disclosed in WO 02/22577, especially N-hydroxy-3-[4-[[(2-hydroxyethyl)[2-(1H- indol-3-yl)ethyl]-amino]methyl]phenyl]-2E-2-propenamide, N-hydroxy-3-[4-[[[2-(2-methyl-1H- indol-3-yl)-ethyl]-amino]methyl]phenyl]-2E-2-propenamide and pharmaceutically acceptable salts thereof. It further especially includes Suberoylanilide hydroxamic acid (SAHA).
The term "antineoplastic antimetabolite" includes, but is not limited to, 5-Fluorouracil or 5-FU, capecitabine, gemcitabine, DNA demethylating agents, such as 5-azacytidine and decitabine, methotrexate and edatrexate, and folic acid antagonists such as pemetrexed. Capecitabine can be administered, e.g., in the form as it is marketed, e.g. under the trademark XELODA. Gemcitabine can be administered, e.g., in the form as it is marketed, e.g. under the trademark GEMZAR. Also included is the monoclonal antibody trastuzumab which can be administered, e.g., in the form as it is marketed, e.g. under the trademark HERCEPTIN. The term "platin compound" as used herein includes, but is not limited to, carboplatin, cis- platin, cisplatinum and oxaliplatin. Carboplatin can be administered, e.g., in the form as it is marketed, e.g. under the trademark CARBOPLAT. Oxaliplatin can be administered, e.g., in the form as it is marketed, e.g. under the trademark ELOXATIN.
The term "compounds targeting/decreasing a protein or lipid kinase activity; or a protein or lipid phosphatase activity; or further anti-angiogenic compounds" as used herein includes, but is not limited to: protein tyrosine kinase and/or serine and/or threonine kinase inhibitors or lipid kinase inhibitors, e.g.:
a) compounds targeting, decreasing or inhibiting the activity of the platelet-derived growth factor-receptors (PDGFR), such as compounds which target, decrease or inhibit the activity of PDGFR, especially compounds which inhibit the PDGF receptor, e.g. a N-phenyl-2- pyrimidine-amine derivative, e.g. imatinib, SU101 , SU6668, and GFB-111 ;
b) compounds targeting, decreasing or inhibiting the activity of the fibroblast growth factor- receptors (FGFR);
c) compounds targeting, decreasing or inhibiting the activity of the insulin-like growth factor receptor I(IGF-IR), such as compounds which target, decrease or inhibit the activity of IGF- IR, especially compounds which inhibit the IGF-IR receptor, such as those compounds disclosed in WO 02/092599;
d) compounds targeting, decreasing or inhibiting the activity of the Trk receptor tyrosine kinase family;
e) compounds targeting, decreasing or inhibiting the activity of the AxI receptor tyrosine kinase family;
f) compounds targeting, decreasing or inhibiting the activity of the c-Met receptor;
g) compounds targeting, decreasing or inhibiting the activity of the Kit/SCFR receptor tyrosine kinase;
h) compounds targeting, decreasing or inhibiting the activity of the C-kit receptor tyrosine kinases - (part of the PDGFR family), such as compounds which target, decrease or inhibit the activity of the c-Kit receptor tyrosine kinase family, especially compounds which inhibit the c-Kit receptor, e.g imatinib; i) compounds targeting, decreasing or inhibiting the activity of members of the c-Abl family and their gene-fusion products (e.g. BCR-AbI kinase), such as compounds which target decrease or inhibit the activity of c-Abl family members and their gene fusion products, e.g. a N-phenyl-2-pyrimidine-amine derivative, e.g. imatinib; PD180970; AG957; NSC 680410; or PD173955 from ParkeDavis;
j) compounds targeting, decreasing or inhibiting the activity of members of the protein kinase C (PKC) and Raf family of serine/threonine kinases, members of the MEK, SRC, JAK, FAK, PDK and Ras/MAPK family members, or Pl(3) kinase family, or of the Pl(3)- kinase-related kinase family, and/or members of the cyclin-dependent kinase family (CDK) and are especially those staurosporine derivatives disclosed in US 5,093,330, e.g. midostaurin; examples of further compounds include e.g. UCN-01 , safingol, BAY 43-9006, Bryostatin 1 , Perifosine; llmofosine; RO 318220 and RO 320432; GO 6976; lsis 3521 ; LY333531/LY379196; isochinoline compounds such as those disclosed in WO 00/09495; FTIs; PD184352 or QAN697( a P13K inhibitor);
k) compounds targeting, decreasing or inhibiting the activity of protein-tyrosine kinase inhibitors, such as compounds which target, decrease or inhibit the activity of protein- tyrosine kinase inhibitors include imatinib mesylate (GLEEVEC) or tyrphostin. A tyrphostin is preferably a low molecular weight (Mr < 1500) compound, or a pharmaceutically acceptable salt thereof, especially a compound selected from the benzylidenemalonitrile class or the S- arylbenzenemalonirile or bisubstrate quinoline class of compounds, more especially any compound selected from the group consisting of Tyrphostin A23/RG-50810; AG 99; Tyrphostin AG 213; Tyrphostin AG 1748; Tyrphostin AG 490; Tyrphostin B44; Tyrphostin B44 (+) enantiomer; Tyrphostin AG 555; AG 494; Tyrphostin AG 556, AG957 and adaphostin (4-{[(2,5-dihydroxyphenyl)methyl]amino}-benzoic acid adamantyl ester; NSC 680410, adaphostin); and
I) compounds targeting, decreasing or inhibiting the activity of the epidermal growth factor family of receptor tyrosine kinases (EGFR, ErbB2, ErbB3, ErbB4 as homo- or heterodimers), such as compounds which target, decrease or inhibit the activity of the epidermal growth factor receptor family are especially compounds, proteins or antibodies which inhibit members of the EGF receptor tyrosine kinase family, e.g. EGF receptor, ErbB2, ErbB3 and ErbB4 or bind to EGF or EGF related ligands, and are in particular those compounds, proteins or monoclonal antibodies generically and specifically disclosed in WO 97/02266, e.g. the compound of ex. 39, or in EP 0 564 409, WO 99/03854, EP 0520722, EP 0 566 226, EP 0 787 722, EP 0 837 063, US 5,747,498, WO 98/10767, WO 97/30034, WO 97/49688, WO 97/38983 and, especially, WO 96/30347 (e.g. compound known as CP 358774), WO 96/33980 (e.g. compound ZD 1839) and WO 95/03283 (e.g. compound ZM105180); e.g. trastuzumab (HERCEPTIN), cetuximab, Iressa, Tarceva, OSI-774, CM 033, EKB-569, GW- 2016, E1.1 , E2.4, E2.5, E6.2, E6.4, E2.11 , E6.3 or E7.6.3, and 7H-pyrrolo-[2,3-d]pyrimidine derivatives which are disclosed in WO 03/013541.
Further anti-angiogenic compounds include compounds having another mechanism for their activity, e.g. unrelated to protein or lipid kinase inhibition e.g. thalidomide (THALOMID) and TNP-470.
Compounds which target, decrease or inhibit the activity of a protein or lipid phosphatase are e.g. inhibitors of phosphatase 1 , phosphatase 2A, PTEN or CDC25, e.g. okadaic acid or a derivative thereof.
Compounds which induce cell differentiation processes are e.g. retinoic acid, α- γ- or δ- tocopherol or α- γ- or δ-tocotrienol.
The term cyclooxygenase inhibitor as used herein includes, but is not limited to, e.g. Cox-2 inhibitors, 5-alkyl substituted 2-arylaminophenylacetic acid and derivatives, such as celecoxib (CELEBREX), rofecoxib (VIOXX), etoricoxib, valdecoxib or a 5-alkyl-2- arylaminophenylacetic acid, e.g. 5-methyl-2-(2'-chloro-6'-fluoroanilino)phenyl acetic acid, lumiracoxib.
The term "bisphosphonates" as used herein includes, but is not limited to, etridonic, clodronic, tiludronic, pamidronic, alendronic, ibandronic, risedronic and zoledronic acid. "Etridonic acid" can be administered, e.g., in the form as it is marketed, e.g. under the trademark DIDRONEL. "Clodronic acid" can be administered, e.g., in the form as it is marketed, e.g. under the trademark BONEFOS. Tiludronic acid" can be administered, e.g., in the form as it is marketed, e.g. under the trademark SKELID. "Pamidronic acid" can be administered, e.g. in the form as it is marketed, e.g. under the trademark AREDIA™. "Alendronic acid" can be administered, e.g., in the form as it is marketed, e.g. under the trademark FOSAMAX. "Ibandronic acid" can be administered, e.g., in the form as it is marketed, e.g. under the trademark BONDRANAT. "Risedronic acid" can be administered, e.g., in the form as it is marketed, e.g. under the trademark ACTONEL. "Zoledronic acid" can be administered, e.g. in the form as it is marketed, e.g. under the trademark ZOMETA.
The term "mTOR inhibitors" relates to compounds which inhibit the mammalian target of rapamycin (mTOR) and which possess antiproliferative activity such as sirolimus (Rapamune®), everolimus (Certican™), CCI-779 and ABT578.
The term "heparanase inhibitor" as used herein refers to compounds which target, decrease or inhibit heparin sulphate degradation. The term includes, but is not limited to, PI-88.
The term " biological response modifier" as used herein refers to. a lymphokine or interferons, e.g. interferon γ.
The term "inhibitor of Ras oncogenic isoforms", e.g. H-Ras, K-Ras, or N-Ras, as used herein refers to compounds which target, decrease or inhibit the oncogenic activity of Ras e.g. a "farnesyl transferase inhibitor" e.g. L-744832, DK8G557 or P115777 (Zamestra).
The term "telomerase inhibitor" as used herein refers to compounds which target, decrease or inhibit the activity of telomerase. Compounds which target, decrease or inhibit the activity of telomerase are especially compounds which inhibit the telomerase receptor, e.g. telomestatin.
The term "methionine aminopeptidase inhibitor" as used herein refers to compounds which target, decrease or inhibit the activity of methionine aminopeptidase. Compounds which target, decrease or inhibit the activity of methionine aminopeptidase are e.g. bengamide or a derivative thereof.
The term "proteasome inhibitor" as used herein refers to compounds which target, decrease or inhibit the activity of the proteasome. Compounds which target, decrease or inhibit the activity of the proteasome include e.g. PS-341 and MLN 341.
The term "matrix metalloproteinase inhibitor" or ("MMP inhibitor") as used herein includes, but is not limited to collagen peptidomimetic and nonpeptidomimetic inhibitors, tetracycline derivatives, e.g. hydroxamate peptidomimetic inhibitor batimastat and its orally bioavailable analogue marimastat (BB-2516), prinomastat (AG3340), metastat (NSC 683551) BMS- 279251 , BAY 12-9566, TAA211 , MMI270B or AAJ996. The term "agents used in the treatment of hematologic malignancies" as used herein includes, but is not limited to FMS-like tyrosine kinase inhibitors e.g. compounds targeting, decreasing or inhibiting the activity of FMS-like tyrosine kinase receptors (Flt-3R); interferon, 1-b-D-arabinofuransylcytosine (ara-c) and bisulfan; and ALK inhibitors e.g. compounds which target, decrease or inhibit anaplastic lymphoma kinase.
Compounds which target, decrease or inhibit the activity of FMS-like tyrosine kinase receptors (Flt-3R) are especially compounds, proteins or antibodies which inhibit members of the Flt-3R receptor kinase family, e.g.PKC412, midostaurin, a staurosporine derivative, SU11248 and MLN518.
The term "HSP90 inhibitors" as used herein includes, but is not limited to, compounds targeting, decreasing or inhibiting the intrinsic ATPase activity of HSP90; degrading, targeting, decreasing or inhibiting the HSP90 client proteins via the ubiquitin proteasome pathway. Compounds targeting, decreasing or inhibiting the intrinsic ATPase activity of HSP90 are especially compounds, proteins or antibodies which inhibit the ATPase activity of HSP90 e.g,17-allylamino,17-demethoxygeldanamycin (17AAG), a geldanamycin derivative; other geldanamycin related compounds; radicicol and HDAC inhibitors.
The term "antiproliferative antibodies" as used herein includes, but is not limited to trastuzumab (Herceptin™), Trastuzumab-DM1 , erlotinib (Tarceva™), bevacizumab (Avastin™), rituximab (Rituxan®), PRO64553 (anti-CD40) and 2C4 Antibody. By antibodies is meant e.g. intact monoclonal antibodies, polyclonal antibodies, multispecific antibodies formed from at least 2 intact antibodies, and antibodies fragments so long as they exhibit the desired biological activity.
For the treatment of acute myeloid leukemia (AML), compounds of formula (I) can be used in combination with standard leukemia therapies, especially in combination with therapies used for the treatment of AML. In particular, compounds of formula (I) can be administered in combination with e.g. farnesyl transferase inhibitors and/or other drugs useful for the treatment of AML, such as Daunorubicin, Adriamycin, Ara-C, VP-16, Teniposide, Mitoxantrone, Idarubicin, Carboplatinum and PKC412.
The term "antileukemic compounds" includes, for example, Ara-C, a pyrimidine analog, which is the 2'-alpha-hydroxy ribose (arabinoside) derivative of deoxycytidine. Also included is the purine analog of hypoxanthine, 6-mercaptopurine (6-MP) and fludarabine phosphate. Compounds which target, decrease or inhibit activity of histone deacetylase (HDAC) inhibitors such as sodium butyrate and suberoylanilide hydroxamic acid (SAHA) inhibit the activity of the enzymes known as histone deacetylases. Specific HDAC inhibitors include MS275, SAHA, FK228 (formerly FR901228), Trichostatin A and compounds disclosed in US 6,552,065, in particular, N-hydroxy-3-[4-[[[2-(2-methyl-1 H-indol-3-yl)-ethyl]- amino]methyl]phenyl]-2E-2-propenamide, or a pharmaceutically acceptable salt thereof and N-hydroxy-3-[4-[(2-hydroxyethyl){2-(1H-indol-3-yl)ethyl]-amino]methyl]phenyl]-2E-2- propenamide, or a pharmaceutically acceptable salt thereof, especially the lactate salt.
Compounds which target, decrease or inhibit the activity of serine/theronine mTOR kinase are especially compounds, proteins or antibodies which inhibit members of the mTOR kinase family e.g. RAD, RAD001 , CCI-779, ABT578, SAR543, rapamycin and derivatives thereof; AP23573 from Ariad; everolimus (CERTICAN); and sirolimus.
Somatostatin receptor antagonists as used herein refers to agents which target, treat or inhibit the somatostatin receptor such as octreoride, and SOM230.
Tumor cell damaging approaches refer to approaches such as ionizing radiation. The term "ionizing radiation" referred to above and hereinafter means ionizing radiation that occurs as either electromagnetic rays (such as X-rays and gamma rays) or particles (such as alpha and beta particles). Ionizing radiation is provided in, but not limited to, radiation therapy and is known in the art. See Hellman, Principles of Radiation Therapy, Cancer, in Principles and Practice of Oncology, Devita et al., Eds., 4th Edition, Vol. 1 , pp. 248-275 (1993).
The term EDG binders as used herein refers a class of immunosuppressants that modulates lymphocyte recirculation, such as FTY720.
CERTICAN (everolimus, RAD) an investigational novel proliferation signal inhibitor that prevents proliferation of T-cells and vascular smooth muscle cells.
The term ribonucleotide reductase inhibitors refers to pyrimidine or puring nucleoside analogs including, but not limited to, fludarabine and/or cytosine arabinoside (ara-C), 6-thioguanine, 5-fluorouracil, cladribine, 6-mercaptopurine (especially in combination with ara-C against ALL) and/or pentostatin. Ribonucleotide reductase inhibitors are especially hydroxyurea or 2-hydroxy-1 H-isoindole-1 ,3-dione derivatives, such as PL-1 , PL-2, PL-3, PL-4, PL-5, PL-6, PL-7 or PL-8 mentioned in Nandy et al., Acta Oncologica, Vol. 33, No. 8, pp. 953-961 (1994). The term "S-adenosylmethionine decarboxylase inhibitors" as used herein includes, but is not limited to the compounds disclosed in US 5,461 ,076.
Also included are in particular those compounds, proteins or monoclonal antibodies of VEGF disclosed in WO 98/35958, e.g. 1 -(4-chloroanilino)-4-(4-pyridylmethyl)phthalazine or a pharmaceutically acceptable salt thereof, e.g. the succinate, or in WO 00/09495, WO 00/27820, WO 00/59509, WO 98/11223, WO 00/27819 and EP 0 769 947; those as described by Prewett et al, Cancer Res, Vol. 59, pp. 5209-5218 (1999); Yuan et at., Proc Natl Acad Sci U S A, Vol. 93, pp. 14765-14770 (1996); Zhu et al., Cancer Res, Vol. 58, pp. 3209-3214 (1998); and Mordenti et al., Toxicol Pathol, Vol. 27, No. 1 , pp. 14-21 (1999); in WO 00/37502 and WO 94/10202; ANGIOSTATIN, described by O'Reilly et al., Cell, Vol. 79, pp. 315-328 (1994); ENDOSTATIN, described by O'Reilly et al., Cell, Vol. 88, pp. 277-285 (1997); anthranilic acid amides; ZD4190; ZD6474; SU5416; SU6668; bevacizumab; or anti-VEGF antibodies or anti-VEGF receptor antibodies, e.g. rhuMAb and RHUFab, VEGF aptamer e.g. Macugon; FLT-4 inhibitors, FLT-3 inhibitors, VEGFR-2 IgGI antibody, Angiozyme (RPI 4610) and Avastan.
Photodynamic therapy as used herein refers to therapy which uses certain chemicals known as photosensitizing agents to treat or prevent cancers. Examples of photodynamic therapy includes treatment with agents, such as e.g. VISUDYNE and porfimer sodium.
Angiostatic steroids as used herein refers to agents which block or inhibit angiogenesis, such as, e.g., anecortave, triamcinolone, hydrocortisone, 11-α-epihydrocotisol, cortexolone, 17α-hydroxyprogesterone, corticosterone, desoxycorticosterone, testosterone, estrone and dexamethasone.
Implants containing corticosteroids refers to agents, such as e.g. fluocinolone, dexamethasone.
Other chemotherapeutic agents include, but are not limited to, plant alkaloids, hormonal agents and antagonists; biological response modifiers, preferably lymphokines or interferons; antisense oligonucleotides or oligonucleotide derivatives; or miscellaneous agents or agents with other or unknown mechanism of action. The structure of the active agents identified by code nos., generic or trade names may be taken from the actual edition of the standard compendium "The Merck Index" or from databases, e.g. Patents International (e.g. IMS World Publications).
The above-mentioned compounds, which can be used in combination with a compound of the formula (I), can be prepared and administered as described in the art such as in the documents cited above.
A compound of the formula (I) may also be used to advantage in combination with known therapeutic processes, e.g., the administration of hormones or especially radiation.
A compound of formula (I) may in particular be used as a radiosensitizer, especially for the treatment of tumors which exhibit poor sensitivity to radiotherapy.
By "combination", there is meant either a fixed combination in one dosage unit form, or a kit of parts for the combined administration where a compound of the formula (I) and a combination partner may be administered independently at the same time or separately within time intervals that especially allow that the combination partners show a cooperative, e.g. synergistic, effect, or any combination thereof.
EXAMPLES
The following examples serve to illustrate the invention without limiting the scope thereof:
Abbreviations
DMSO dimethylsulfoxide
MS(ESI) mass spectrometry using electrospray ionization
EtOAc Ethyl Acetate
HPLC high-pressure liquid chromatography mL mililiter(s)
RT room temperature tREτ HPLC retention time in minutes
TFA trifluoroacetic acid
THF tetrahydrofuran
TLC thin layer chromatography
TMSCI Trimethylsilyl chloride Where no temperatures are given, the reaction takes place at ambient (room) temperature.
Ratios of solvents, e.g., in eluents or solvent mixtures, are given in volume by volume (7V).
Syntheses
Flash chromatography is performed by using silica gel (Merck; 40-63 μm). For thin layer chromatograhy, pre-coated silica gel (Merck 60 F254) plates are used. Detection of the components is made by UV light (254 nm). HPLC is performed on an Agilent HP 1100 using a Nucleosil 100-3 Ci8 HD 125 x 4.0 mm column [1 mL/min.; 20-100% NeCN / 0.1% TFA in 7 minutes) (Method A); SpectraSystem SP8800/UV2000 using a Nucleosil 100-5 C18 AB 250 x 4.6 mm column (2 mL/min.; 2-100% MeCN / 0.1% TFA in 10 minutes) (Method B); using a Chromalith Speed ROD RP18 50-4.6 mm column (Merck) (2 mL/min.; 2-100% MeCN / 0.1% TFA in 2 minutes) (Method C); or a C8 2.1-50 mm 3 μm column (Waters) (2 mL/min.; 5-95% MeCN / 0.1% TFA in 2 minutes) (Method D). Electrospray mass spectra are obtained with a Fisons Instruments VG Platform II. Melting points are measured with a Bϋchi 510 melting point apparatus. Commercially-available solvents and chemicals are used for syntheses.
Analytical HPLC conditions:
System 1
Linear gradient 20-100% CH3CN (0.1%TFA) and H2O (0.1% TFA) in 7min + 2min 100%
CH3CN (0.1%TFA); detection at 215 nm, flow rate 1 mL/min at 300C. Column: Nucleosil 100-
3 C18HD (125 x 4mm).
System 2
Linear gradient 2-100% CH3CN (0.1%TFA) and H2O (0.1% TFA) in 7min + 2min 100%
CH3CN (0.1%TFA); detection at 215 nm, flow rate 1 mL/min at 300C. Column: Nucleosil 100-
3 C18HD (125 x 4mm).
Example 1 : 6-(3-Chloro-phenyl)-3-(3,4-dimethoxy-phenyl)-5-piperazin-1-ylmethyl- pyrazolo[1 ,5-a]pyrimidin-7-ylamine
5-Chloromethyl-6-(3-chloro-phenyl)-3-(3,4-dimethoxy-phenyl)-pyrazolo[1 ,5-a]pyrimidin-7- ylamine (example 1 ; stage 1.3) (1.5 g; 3.49 mMol) is dissolved in N,N'-dimethylacetamide (30 mL) at rt, followed by addition of anhydrous piperazine (3.01 g; 34.9 mMol). The yellow solution is heated at 8O0C for 90 min. After cooling the mixture is concentrated under reduced pressure. The residue is taken into diethyl ether (20 mL) and stirred for 15 min, followed by filtering off the remaining crude crystalline product. Further purification was performed by repeated chromatography (silica gel, 12Og RediSep, ISCO Sg-100, eluting with CH2CI2 / MeOH 4:1).
Stage 1.1 : 2-(3-Chloro-phenyl)-3-oxo-butyronitrile.
355 ml of ethanol is heated to 55°C under N2. To this solution is added sodium (3.91 g; 0.17 mol) within 30 min. and stirred for 1.5 h until all metal is dissolved. 3-Chlorobenzyl cyanide (15.31 g; 0.1 mol) and ethyl acetate (28.53 mL; 0.29 mol) are added to the colorless solution, followed by stirring under reflux for 5 h. After completion of the reaction, the yellow mixture is cooled to rt. and evaporated under reduced pressure. The crude material is taken up into water (200 mL) and neutralized by addition of 25 g of citric acid. The aqueous layer is extracted with CH2CI2 (2x 250 mL). The combined organic phases are washed with H2O (2 x 150 mL,), dried (Na2SO4), concentrated under reduced pressure and chromatographed (silica gel, 1 kg, Merck 60 (0.040 -0.063), eluting with EtOAc/Hexanes 1:1) to obtain the title compound as yellowish crystals; mp. 92-97 0C; MS(ESI+):m/z= 302.9 (M+H)+; HPLC: A tRet = 5.67 minutes (Systemi).
Stage 1.2 : 4-Bromo-2-(3-chloro-phenyl)-3-oxo-butyronitrile
2-(3-Chloro-phenyl)-3-oxo-butyronitrile (12 g; 62 mmol) is dissolved in acetic acid (400 mL) at rt, followed by addition of brom (3.84 mL; 74.4 mMol), the condenser covered with a CaCI2 tube. The yellow solution is stirred for 90 min at 900C. After cooling to rt the solvent is removed under reduced pressure. The oily residue is taken up into toluene (50 mL) and freed from solvent under reduced pressure; this procedure being repeated four times. After these purification steps, the title compound is isolated as fine, bright yellow crystals; mp.
124-132°C; MS(ESI-):m/z= 271.9 (M-H) ' ; HPLC: ,Re, = 6.31 minutes (System 2).
Stage 1.3 : 5-Chloromethyl-6-(3-chloro-phenyl)-3-(3,4-dimethoxy-phenyl)-pyrazolo[1 ,5- a]pyrimidin-7-ylamine
4-Bromo-2-(3-chloro-phenyl)-3-oxo-butyronitrile (example 1 ; stage 1.2); (9.43g; 34.6 mmol) is dissolved in ethanol (200 mL) followed by addition of 4-(3,4-dimethoxy-phenyl)-2H-pyrazol- 3-ylamine (15.2 g; 69.2 mMol) (see example 93; stage 93.1 EP 2005/000602) and HCI in ethanol (111 mL of 1.25 M solution; 138 mmol) at rt. The lightly yellowish suspension was heated to reflux for a total of 163 h; the condenser covered with a CaCI2 tube. After cooling to rt the crystalline product was filtered off and wshed with ethanol. Further purification was performed by flash chromatography (silica gel, eluting with CH2CI2 / MeOH 98:2) to obtain the title compound as yellow crystals; mp. 209-2110C; MS(ESI+):m/z= 429 (M+H)+; HPLC: tRet = 7.06 minutes (System 2).
Example 2: 3-(3,4-Dimethoxy-phenyl)-6-(4-fluoro-phenyl)-5-piperazin-1-ylmethyl- pyrazolo[1,5-a]pyrimidin-7-ylamine
The title compound is prepared as described in example 1 ; using 5-Chloromethyl-3-(3,4- dimethoxy-phenyl)-6-(4-fluoro-phenyl)-pyrazolo[1 ,5-a]pyrimidin-7-ylamine instead.
Stape 2.1 : 5-Chloromethyl-3-(3,4-dimethoxy-phenyl)-6-(4-fluoro-phenyl)-pyrazolo[1 ,5- a]pyrimidin-7-ylamine
The title compound is prepared as described in Example 1 ; using 2-(4-Fluoro-phenyl)-3-oxo- butyronitrile (example 79; stage 79.1 EP 2005/000602) instead. Titel compound: yellowish crystals mp. 256-259°C; MS(ESI+):m/z= 413 (M+H)+; HPLC: ,Ret = 6.65 minutes (System 2).
Physico-chemical Properties of Examples land 2
Example HPLC tRet = MS [ESI] Nr. MP [min] (M+H)+ from to
1 202 213.25 3.70 /S1 479,1 2 201 205 4.49 /S2 463,1
The following Examples 3 to 5 can be prepared analogously
Example 3: 6-(3-Chloro-phenyl)-3-[4-{2-dimethylamino-ethoxy)-phenyl]-5-piperazin- 1-ylmethyl-pyrazolo[1,5-a]pyrimidin-7-ylamine Example 4 2-{[7-Amino-6-(3-chloro-phenyl)-3-(3,4-dimethoxy-phenyl)-pyrazolo[1,5- a]pyrimidin-5-ylmethyl]-amino}-ethanol
Example 5 2-{[7-Amino-3-(3,4-dimethoxy-phenyl)-6-(4-fluoro-phenyl)-pyrazolo[1,5- a]pyrimidin-5-ylmethyl]-amino}-ethanol
Example 6 Tablets 1 comprising compounds of the formula (I)
Tablets, comprising, as active ingredient, 50 mg of any one of the compounds of formula (I) mentioned in the preceding Examples of the following composition are prepared using routine methods:
Figure imgf000041_0001
Manufacture: The active ingredient is combined with part of the wheat starch, the lactose and the colloidal silica and the mixture pressed through a sieve. A further part of the wheat starch is mixed with the 5-fold amount of water on a water bath to form a paste and the mixture made first is kneaded with this paste until a weakly plastic mass is formed.
The dry granules are pressed through a sieve having a mesh size of 3 mm, mixed with a pre-sieved mixture (1 mm sieve) of the remaining corn starch, magnesium stearate and talcum and compressed to form slightly biconvex tablets. Example 7 Tablets 2 comprising compounds of the formula (I)
Tablets, comprising, as active ingredient, 100 mg of any one of the compounds of formula (I) mentioned in the preceding Examples are prepared with the following composition, following standard procedures:
Figure imgf000042_0001
Manufacture: The active ingredient is mixed with the carrier materials and compressed by means of a tabletting machine (Korsch EKO, Stempeldurchmesser 10 mm).
Example 8 Capsules
Capsules, comprising, as active ingredient, 100 mg of any one of the compounds of formula (I) mentioned in the preceding Examples, of the following composition are prepared according to standard procedures:
Figure imgf000042_0002
Figure imgf000043_0001
Manufacturing is done by mixing the components and filling them into hard gelatine capsules, size 1.
Example 9 Biological data of compounds of Example 1 and 2 as EphB4 and c-Abl protein kinase inhibitors:
. EphB4
Figure imgf000043_0002
Table 1 : Inhibition of Ephrin B4 receptor kinase:
Column I : Activity of the compounds of Example 1 and 2 as Ephrin B4 receptor kinase inhibitors is tested on recombinant Ephrin B4 receptor domains according to the method described in the specification. Column Il : Activity of the compounds of Examples 1 and 2 as inhibitors of the EphB4 receptor auto-phosphorylation in vitro on cells is measured as described in the specification.
. c-Abl
Figure imgf000043_0003
Table 2: Inhibition of c-Abl protein-tyrosine kinase activity Activity of the compounds of Example 1 and 2 as c-Abl protein tyrosine kinase inhibitors is tested according to the method described in the specification.

Claims

What is claimed is;
1. A compound of formula (I)
Figure imgf000044_0001
wherein: R1 is H
R2 is benzyl; unsubstituted phenyl or phenyl substituted by one or two substituents chosen from the group consisting of halo, di-lower alkylaminoalkoxy, hydroxy, alkoxy, benzyloxy, cycloalkyl, amino, acetyl amino; R3 is H and R4 is hydroxyalkyl or
R3 and R4 together with the nitrogen atom they are attached to represent morpholinyl, pyrrolidinyl, piperidinyl or piperazinyl, the heterocycles optionally being further substituted by up to four alkyl groups;
A is phenyl which is unsubstituted or substituted by one or more of the substituents chosen from the group consisting of mono-, di- or tri-lower alkoxy, di-lower alkylaminyl, di-lower alkylaminoalkoxy, morpholinyl which is optionally di-substituted by alkyl, piperidinyl which is optionally substituted by di-lower alkylaminyl, and piperazinyl which is optionally substituted by lower alkyl, lower alkoxy, lower alkyl piperazinyl, pyrrolidinyl, di-lower or alkylaminyl; or a pharmaceutical salt thereof.
2. A compound of formula (I) according to claim 1 , wherein:
R1 is H
R2 is phenyl substituted by fluoro or chloro;
R3 is H and R4 is hydroxyethyl or
R3 and R4 together with the nitrogen atom they are attached to represent piperazinyl;
A is phenyl which is substituted with one or more of the substituents chosen from the group consisting of; mono-, di- or tri-methoxy, di-methylaminoethoxy and di-ethylamino piperidinyl or a pharmaceutical salt thereof.
3. Compound of formula (I) according to claim 1 or 2 for use in the treatment of the human or animal body.
4. A pharmaceutical composition comprising a compound of formula (I) according to claim 1 or 2.
5. A pharmaceutical composition comprising a compound of formula (I) according to claim 1 or 2 and an acceptable pharmaceutical carrier.
6. Use of a compound of formula (I) according to claim 1 or 2 in the preparation of a pharmaceutical compositions for use in the treatment of a kinase dependent disease.
7. Use of a compound of formula I according to claim 6 or pharmaceutically acceptable salts thereof for treating a protein kinase dependent disease, wherein the kinase dependent disease is one depending on c-Abl, Bcr-Abl, c-Kit, c-Raf, Flt-1 , Flt-3, Her-1 , KDR1 PDGFR- kinase, c-Src, RET-receptor kinase, FGF-R1 , FGF-R2, FGF-R3, FGF-R4, Ephrin receptor kinases (e. g., EphB2 kinase, EphB4 kinase and related Eph kinases), casein kinases (CK-1 , CK-2, G-CK), Pak, ALK, ZAP70, Jak1 , Jak2, AxI, Cdk1 , cdk4, cdk5, Met, FAK1 Pyk2, Syk, Insulin receptor kinase, Tie-2 or costitutively activating mutations of kinases (activating kinases) such as of Bcr-Abl, c-Kit, c-Raf, Flt-3, FGF-R3, PDGF-receptors, RET, and Met and (especially aberrantly highly expressed or activated) kinase-dependent disease or disease dependent on the activation of the kinase pathways, or a disease dependent on any two or more of the kinases just mentioned.
8. A use according to claim 6 or 7 wherein the kinase dependent disease is one depending on c-abl, Flt-3, KDR1 c-Src, RET, EphB4, c-kit, cdk1 , FGFR-1 , c-raf, Her-1 , Ins-R or Tek.
9. A use according to any one of claims 6-8, wherein the disease to be treated is a proliferative disease, preferably a benign or especially malignant tumor, more preferably carcinoma of the brain, kidney, liver, adrenal gland, bladder, breast, stomach (especially gastric tumors), ovaries, colon, rectum, prostate, pancreas, lung, vagina, thyroid, sarcoma, glioblastomas, multiple myeloma or gastrointestinal cancer, especially colon carcinoma or colorectal adenoma, or a tumor of the neck and head, an epidermal hyperproliferation, especially psoriasis, prostate hyperplasia, a neoplasia, especially of epithelial character, preferably mammary carcinoma, or a leukemia.
10. A use according to any one of claims 6 to 9, wherein the disease to be treated is triggered by persistent angiogenesis, such as psoriasis; Kaposi's sarcoma; restenosis, e.g., stent-induced restenosis; endometriosis; Crohn's disease; Hodgkin's disease; leukemia; arthritis, such as rheumatoid arthritis; hemangioma; angiofibroma; eye diseases, such as diabetic retinopathy and neovascular glaucoma; renal diseases, such as glomerulonephritis; diabetic nephropathy; malignant nephrosclerosis; thrombotic microangiopathic syndromes; transplant rejections and glomerulopathy; fibrotic diseases, such as cirrhosis of the liver; mesangial cell-proliferative diseases; arteriosclerosis; injuries of the nerve tissue; and for inhibiting the re-occlusion of vessels after balloon catheter treatment, for use in vascular prosthetics or after inserting mechanical devices for holding vessels open, such as, e.g., stents, as immunosuppressants, as an aid in scar-free wound healing, and for treating age spots and contact dermatitis.
PCT/EP2006/007109 2005-07-21 2006-07-19 Pyrazolo[1.5-a]pyrimidin-7-yl amine derivatives as protein kinase inhibitors WO2007009773A1 (en)

Priority Applications (7)

Application Number Priority Date Filing Date Title
MX2008000898A MX2008000898A (en) 2005-07-21 2006-07-19 Pyrazolo[1.5-a]pyrimidin-7-yl amine derivatives as protein kinase inhibitors.
BRPI0613870-5A BRPI0613870A2 (en) 2005-07-21 2006-07-19 pyrazol [1,5-a] pyrimidinyl-7-ylamine derivatives as protein kinase inhibitors
AU2006271924A AU2006271924A1 (en) 2005-07-21 2006-07-19 Pyrazolo[1.5-a]pyrimidin-7-yl amine derivatives as protein kinase inhibitors
JP2008521883A JP2009501748A (en) 2005-07-21 2006-07-19 Pyrazolo [1,5-a] pyrimidin-7-ylamine derivatives as protein kinase inhibitors
US11/996,337 US20080234284A1 (en) 2005-07-21 2006-07-19 Pyrazolo[1,5-a]Pyrimidin-7-Yl Amine Derivatives as Protein Kinase Inhibitors
EP06762704A EP1910368A1 (en) 2005-07-21 2006-07-19 Pyrazolo[1.5-a]pyrimidin-7-yl amine derivatives as protein kinase inhibitors
CA002615433A CA2615433A1 (en) 2005-07-21 2006-07-19 Pyrazolo[1.5-a]pyrimidin-7-yl amine derivatives as protein kinase inhibitors

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
GBGB0515026.3A GB0515026D0 (en) 2005-07-21 2005-07-21 Organic compounds
GB0515026.3 2005-07-21

Publications (1)

Publication Number Publication Date
WO2007009773A1 true WO2007009773A1 (en) 2007-01-25

Family

ID=34976344

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2006/007109 WO2007009773A1 (en) 2005-07-21 2006-07-19 Pyrazolo[1.5-a]pyrimidin-7-yl amine derivatives as protein kinase inhibitors

Country Status (12)

Country Link
US (1) US20080234284A1 (en)
EP (1) EP1910368A1 (en)
JP (1) JP2009501748A (en)
KR (1) KR20080036997A (en)
CN (1) CN101228167A (en)
AU (1) AU2006271924A1 (en)
BR (1) BRPI0613870A2 (en)
CA (1) CA2615433A1 (en)
GB (1) GB0515026D0 (en)
MX (1) MX2008000898A (en)
RU (1) RU2008106056A (en)
WO (1) WO2007009773A1 (en)

Cited By (45)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007103432A2 (en) * 2006-03-08 2007-09-13 Novartis Ag Use of pyrazolo[1,5a]pyrimidin-7-yl amine derivatives in the treatment of neurological disorders
WO2007113000A1 (en) * 2006-04-04 2007-10-11 Novartis Ag 3-SUBSTITUTED N-(ARYL- OR HETEROARYL)-PYRAZO[1,5-a]PYRIMIDINES AS KINASE INHIBITORS
WO2008037459A1 (en) * 2006-09-28 2008-04-03 Novartis Ag Pyrazolo [1, 5-a] pyrimidine derivatives and their therapeutic use
FR2918986A1 (en) * 2007-07-19 2009-01-23 Sanofi Aventis Sa New 6-cycloamino-3-(pyridazin-4-yl)imidazo(1,2-b)-pyridazine compounds are casein kinase 1 delta inhibitors useful to treat or prevent e.g. primary insomnia, bipolar disorders, anxiety, depression, Alzheimer's disease and lung cancer
JP2009511491A (en) * 2005-10-06 2009-03-19 シェーリング コーポレイション Pyrazolopyrimidines as protein kinase inhibitors
WO2010010186A1 (en) * 2008-07-25 2010-01-28 Galapagos Nv Novel compounds useful for the treatment of degenerative and inflammatory diseases
WO2010010191A1 (en) 2008-07-25 2010-01-28 Galapagos Nv Novel compounds useful for the treatment of degenerative and inflammatory diseases.
WO2010010188A1 (en) * 2008-07-25 2010-01-28 Galapagos Nv Novel compounds useful for the treatment of degenerative and inflammatory diseases.
WO2010070237A1 (en) * 2008-12-19 2010-06-24 Sanofi-Aventis Derivatives of 6-cycloamino-2-thienyl-3-(pyridin-4-yl)imidazo[1,2-b]-pyridazine and 6-cycloamino-2-furanyl-3-(pyridin-4-yl)imidazo[1,2-b]-pyridazine, preparation and therapeutic application thereof
WO2010070238A1 (en) * 2008-12-19 2010-06-24 Sanofi-Aventis Derivatives of 6-cycloamino-2,3-di-pyridinyl-imidazo[1,2-b]-pyridazine, preparation and therapeutic application thereof
WO2010074947A1 (en) * 2008-12-16 2010-07-01 Eli Lilly And Company Amino pyrazole compound
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
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
WO2010099139A2 (en) 2009-02-25 2010-09-02 Osi Pharmaceuticals, Inc. Combination anti-cancer therapy
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
WO2011002887A1 (en) * 2009-07-02 2011-01-06 Schering Corporation FUSED TRICYCLIC COMPOUNDS AS NOVEL mTOR INHIBITORS
WO2011041152A1 (en) 2009-09-30 2011-04-07 Schering Corporation Novel compounds that are erk inhibitors
WO2012036997A1 (en) 2010-09-16 2012-03-22 Schering Corporation Fused pyrazole derivatives as novel erk inhibitors
WO2012149014A1 (en) 2011-04-25 2012-11-01 OSI Pharmaceuticals, LLC Use of emt gene signatures in cancer drug discovery, diagnostics, and treatment
WO2013152252A1 (en) 2012-04-06 2013-10-10 OSI Pharmaceuticals, LLC Combination anti-cancer therapy
US8563545B2 (en) 2009-06-26 2013-10-22 Galapagos Nv Compound useful for the treatment of degenerative and inflammatory diseases
US8591943B2 (en) 2009-04-09 2013-11-26 Merck Sharp & Dohme Corp. Pyrazolo[1,5-a]pyrimidine derivatives as mTOR inhibitors
US8796457B2 (en) 2009-06-26 2014-08-05 Galapagos Nv Compound useful for the treatment of degenerative and inflammatory diseases
US8987456B2 (en) 2011-10-05 2015-03-24 Merck Sharp & Dohme Corp. 3-pyridyl carboxamide-containing spleen tyrosine kinase (SYK) inhibitors
US9006444B2 (en) 2011-10-05 2015-04-14 Merck Sharp & Dohme Corp. Phenyl carboxamide-containing spleen tyrosine kinase (SYK) inhibitors
US9120785B2 (en) 2011-05-10 2015-09-01 Merck Sharp & Dohme Corp. Pyridyl aminopyridines as Syk inhibitors
US9145391B2 (en) 2011-05-10 2015-09-29 Merck Sharp & Dohme Corp. Bipyridylaminopyridines as Syk inhibitors
US9216173B2 (en) 2011-10-05 2015-12-22 Merck Sharp & Dohme Corp. 2-Pyridyl carboxamide-containing spleen tyrosine kinase (SYK) inhibitors
US9242984B2 (en) 2012-06-20 2016-01-26 Merck Sharp & Dohme Corp. Pyrazolyl derivatives as Syk inhibitors
US9290490B2 (en) 2011-05-10 2016-03-22 Merck Sharp & Dohme Corp. Aminopyrimidines as Syk inhibitors
US9353066B2 (en) 2012-08-20 2016-05-31 Merck Sharp & Dohme Corp. Substituted phenyl-Spleen Tyrosine Kinase (Syk) inhibitors
US9376418B2 (en) 2012-06-22 2016-06-28 Merck Sharp & Dohme Corp. Substituted pyridine spleen tyrosine kinase (SYK) inhibitors
US9416111B2 (en) 2012-06-22 2016-08-16 Merck Sharp & Dohme Corp. Substituted diazine and triazine spleen tyrosine kinease (Syk) inhibitors
US9487504B2 (en) 2012-06-20 2016-11-08 Merck Sharp & Dohme Corp. Imidazolyl analogs as syk inhibitors
US9499534B2 (en) 2013-04-26 2016-11-22 Merck Sharp & Dohme Corp. Thiazole-substituted aminopyrimidines as spleen tyrosine kinase inhibitors
US9586931B2 (en) 2012-09-28 2017-03-07 Merck Sharp & Dohme Corp. Triazolyl derivatives as Syk inhibitors
US9598405B2 (en) 2012-12-21 2017-03-21 Merck Sharp & Dohme Corp. Thiazole-substituted aminopyridines as spleen tyrosine kinase inhibitors
US9624210B2 (en) 2012-12-12 2017-04-18 Merck Sharp & Dohme Corp. Amino-pyrimidine-containing spleen tyrosine kinase (Syk) inhibitors
US9670196B2 (en) 2013-12-20 2017-06-06 Merck Sharp & Dohme Corp. Thiazole-substituted aminoheteroaryls as Spleen Tyrosine Kinase inhibitors
US9745295B2 (en) 2013-04-26 2017-08-29 Merck Sharp & Dohme Corp. Thiazole-substituted aminoheteroaryls as spleen tyrosine kinase inhibitors
US9775839B2 (en) 2014-03-13 2017-10-03 Merck Sharp & Dohme Corp. 2-pyrazine carboxamides as spleen tyrosine kinase inhibitors
US9783531B2 (en) 2013-12-20 2017-10-10 Merck Sharp & Dohme Corp. Thiazole-substituted aminoheteroaryls as spleen tyrosine kinase inhibitors
US9822107B2 (en) 2013-12-20 2017-11-21 Merck Sharp & Dohme Corp. Thiazole-substituted aminoheteroaryls as spleen tyrosine kinase inhibitors
US10493158B2 (en) 2014-02-07 2019-12-03 Galapagos Nv Pharmaceutical compositions for the treatment of inflammatory disorders
US10708263B2 (en) 2014-02-07 2020-07-07 Galapagos Nv Salts and pharmaceutical compositions thereof for the treatment of inflammatory disorders

Families Citing this family (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2483407A2 (en) 2009-09-30 2012-08-08 President and Fellows of Harvard College Methods for modulation of autophagy through the modulation of autophagy-enhancing gene products
CR20170098A (en) 2010-05-20 2017-07-17 Array Biopharma Inc MACROCICLICAL COMPOUNDS AS QUINASA TRK INHIBITORS
US8975264B2 (en) * 2010-08-31 2015-03-10 Amorepacific Corporation Compound acting as a cannabinoid receptor-1 inhibitor
CN105906630B (en) * 2015-04-06 2018-10-23 四川百利药业有限责任公司 Two substitutional amine-group compounds of N- (1H- pyrazoles -5- bases) pyrimido pyrazoles -4,6- as FGFR inhibitor
MX2018000577A (en) 2015-07-16 2018-09-05 Array Biopharma Inc Substituted pyrazolo[1,5-a]pyridine compounds as ret kinase inhibitors.
TWI704148B (en) 2016-10-10 2020-09-11 美商亞雷生物製藥股份有限公司 Substituted pyrazolo[1,5-a]pyridine compounds as ret kinase inhibitors
JOP20190077A1 (en) 2016-10-10 2019-04-09 Array Biopharma Inc Substituted pyrazolo[1,5-a]pyridine compounds as ret kinase inhibitors
WO2018136663A1 (en) 2017-01-18 2018-07-26 Array Biopharma, Inc. Ret inhibitors
EP3571203B1 (en) 2017-01-18 2023-06-07 Array BioPharma Inc. Substituted pyrazolo[1,5-a]pyrazine compounds as ret kinase inhibitors
JOP20190213A1 (en) 2017-03-16 2019-09-16 Array Biopharma Inc Macrocyclic compounds as ros1 kinase inhibitors
TWI791053B (en) 2017-10-10 2023-02-01 美商亞雷生物製藥股份有限公司 Crystalline forms of 6-(2-hydroxy-2-methylpropoxy)-4-(6-(6-((6-methoxypyridin-3-yl)methyl)-3,6-diazabicyclo[3.1.1]heptan-3-yl)pyridin-3-yl)pyrazolo[1,5-a]pyridine-3-carbonitrile and pharmaceutical composition thereof
TWI812649B (en) 2017-10-10 2023-08-21 美商絡速藥業公司 Formulations of 6-(2-hydroxy-2-methylpropoxy)-4-(6-(6-((6-methoxypyridin-3-yl)methyl)-3,6-diazabicyclo[3.1.1]heptan-3-yl)pyridin-3-yl)pyrazolo[1,5-a]pyridine-3-carbonitrile
US11472802B2 (en) 2018-01-18 2022-10-18 Array Biopharma Inc. Substituted pyrazolyl[4,3-c]pyridine compounds as RET kinase inhibitors
JP7061195B2 (en) 2018-01-18 2022-04-27 アレイ バイオファーマ インコーポレイテッド Substituted pyrazolo [3,4-d] pyrimidine compound as a RET kinase inhibitor
CN111971286B (en) 2018-01-18 2023-04-14 阿雷生物药品公司 Substituted pyrrolo [2,3-d ] pyrimidine compounds as RET kinase inhibitors
WO2020055672A1 (en) 2018-09-10 2020-03-19 Array Biopharma Inc. Fused heterocyclic compounds as ret kinase inhibitors

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6380203B1 (en) * 1998-01-14 2002-04-30 Merck & Co., Inc. Angiogenesis inhibitors
WO2004022561A1 (en) * 2002-09-04 2004-03-18 Schering Corporation Pyrazolopyrimidines as cyclin-dependent kinase inhibitors
WO2005070431A1 (en) * 2004-01-22 2005-08-04 Novartis Ag Pyrazolo[1,5-a]pyrimidin-7-yl-amine derivatives for use in the treatment of protein kinase dependent diseases

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6380203B1 (en) * 1998-01-14 2002-04-30 Merck & Co., Inc. Angiogenesis inhibitors
WO2004022561A1 (en) * 2002-09-04 2004-03-18 Schering Corporation Pyrazolopyrimidines as cyclin-dependent kinase inhibitors
WO2005070431A1 (en) * 2004-01-22 2005-08-04 Novartis Ag Pyrazolo[1,5-a]pyrimidin-7-yl-amine derivatives for use in the treatment of protein kinase dependent diseases

Cited By (75)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2009511491A (en) * 2005-10-06 2009-03-19 シェーリング コーポレイション Pyrazolopyrimidines as protein kinase inhibitors
WO2007103432A3 (en) * 2006-03-08 2007-11-22 Novartis Ag Use of pyrazolo[1,5a]pyrimidin-7-yl amine derivatives in the treatment of neurological disorders
WO2007103432A2 (en) * 2006-03-08 2007-09-13 Novartis Ag Use of pyrazolo[1,5a]pyrimidin-7-yl amine derivatives in the treatment of neurological disorders
WO2007113000A1 (en) * 2006-04-04 2007-10-11 Novartis Ag 3-SUBSTITUTED N-(ARYL- OR HETEROARYL)-PYRAZO[1,5-a]PYRIMIDINES AS KINASE INHIBITORS
WO2008037459A1 (en) * 2006-09-28 2008-04-03 Novartis Ag Pyrazolo [1, 5-a] pyrimidine derivatives and their therapeutic use
EA016376B1 (en) * 2007-07-19 2012-04-30 Санофи-Авентис 6-cycloamino-s-(pyridazin-4-yl)imidazo[1,2-b]-pyridazine and derivatives thereof preparation and therapeutic application thereof
FR2918986A1 (en) * 2007-07-19 2009-01-23 Sanofi Aventis Sa New 6-cycloamino-3-(pyridazin-4-yl)imidazo(1,2-b)-pyridazine compounds are casein kinase 1 delta inhibitors useful to treat or prevent e.g. primary insomnia, bipolar disorders, anxiety, depression, Alzheimer's disease and lung cancer
WO2009037394A2 (en) * 2007-07-19 2009-03-26 Sanofi-Aventis 6-cycloamino-s-(pyridazin-4-yl)imidazo[1,2-b]-pyridazine and derivatives thereof preparation and therapeutic application thereof
WO2009037394A3 (en) * 2007-07-19 2009-06-18 Sanofi Aventis 6-cycloamino-s-(pyridazin-4-yl)imidazo[1,2-b]-pyridazine and derivatives thereof preparation and therapeutic application thereof
US8455491B2 (en) 2007-07-19 2013-06-04 Sanofi 6-cycloamino-3-(pyridazin-4-yl)imidazo[1,2-b]pyridazine and derivatives and pharmaceutical compositions comprising the same
US9108971B2 (en) 2007-07-19 2015-08-18 Sanofi 6-cycloamino-3-(pyridazin-4-yl)imidazo[1,2-b]-pyridazine and derivatives thereof preparation and therapeutic application thereof
WO2010010190A1 (en) * 2008-07-25 2010-01-28 Galapagos Nv Novel compounds useful for the treatment of degenerative and inflammatory diseases
WO2010010191A1 (en) 2008-07-25 2010-01-28 Galapagos Nv Novel compounds useful for the treatment of degenerative and inflammatory diseases.
US8853240B2 (en) 2008-07-25 2014-10-07 Galapagos Nv Compounds useful for the treatment of degenerative and inflammatory diseases
WO2010010186A1 (en) * 2008-07-25 2010-01-28 Galapagos Nv Novel compounds useful for the treatment of degenerative and inflammatory diseases
US20150150856A1 (en) * 2008-07-25 2015-06-04 Christel Jeanne Marie Menet Novel compounds useful for the treatment of degenerative and inflammatory diseases
EA018587B1 (en) * 2008-07-25 2013-09-30 Галапагос Нв 2-CYCLOPROPYLCARBONYLANINO-5-PHENYL[1,2,4]TRIAZOLO[1,5-a]PYRIDINE DERIVATIVES FOR THE TREATMENT OF DEGENERATIVE AND INFLAMMATORY DISEASES
EA018080B1 (en) * 2008-07-25 2013-05-30 Галапагос Нв Compounds useful for the treatment of degenerative and inflammatory diseases
CN102105472B (en) * 2008-07-25 2013-04-17 加拉帕戈斯股份有限公司 Novel compounds useful for the treatment of degenerative and inflammatory diseases
US8242274B2 (en) 2008-07-25 2012-08-14 Galapagos Nv Triazolopyridine kinase inhibitors useful for the treatment of degenerative and inflammatory diseases
WO2010010188A1 (en) * 2008-07-25 2010-01-28 Galapagos Nv Novel compounds useful for the treatment of degenerative and inflammatory diseases.
US10206907B2 (en) 2008-07-25 2019-02-19 Galapagos Nv Compounds useful for the treatment of degenerative and inflammatory diseases
US9415037B2 (en) 2008-07-25 2016-08-16 Galapagos Nv Compounds useful for the treatment of degenerative and inflammatory diseases
KR101676391B1 (en) 2008-07-25 2016-11-15 갈라파고스 엔.브이. Novel compounds useful for the treatment of degenerative and inflammatory diseases
KR20110053435A (en) * 2008-07-25 2011-05-23 갈라파고스 엔.브이. Novel compounds useful for the treatment of degenerative and inflammatory diseases
KR101300458B1 (en) * 2008-12-16 2013-08-30 일라이 릴리 앤드 캄파니 Amino pyrazole compound
EA019554B1 (en) * 2008-12-16 2014-04-30 Эли Лилли Энд Компани Amino pyrazole compound
WO2010074947A1 (en) * 2008-12-16 2010-07-01 Eli Lilly And Company Amino pyrazole compound
AU2009330503B2 (en) * 2008-12-16 2012-06-07 Eli Lilly And Company Amino pyrazole compound
US7897600B2 (en) 2008-12-16 2011-03-01 Eli Lilly And Company Amino pyrazole compound
WO2010070237A1 (en) * 2008-12-19 2010-06-24 Sanofi-Aventis Derivatives of 6-cycloamino-2-thienyl-3-(pyridin-4-yl)imidazo[1,2-b]-pyridazine and 6-cycloamino-2-furanyl-3-(pyridin-4-yl)imidazo[1,2-b]-pyridazine, preparation and therapeutic application thereof
CN102325773A (en) * 2008-12-19 2012-01-18 赛诺菲 6-ring amino-2,3-bipyridyl imidazo [1,2-b]-pyridazine derivatives and preparation thereof and treatment are used
FR2940284A1 (en) * 2008-12-19 2010-06-25 Sanofi Aventis 6-CYCLOAMINO-2,3-DI-PYRIDINYL-IMIDAZO-1,2-B-PYRIDAZINE DERIVATIVES, THEIR PREPARATION AND THEIR THERAPEUTIC USE
FR2940285A1 (en) * 2008-12-19 2010-06-25 Sanofi Aventis 6-CYCLOAMINO-2-THIENYL-3- (PYRIDIN-4-YL) IMIDAZO-1,2-B! -PYRIDAZINE AND 6-CYCLOAMINO-2-FURANYL-3- (PYRIDIN-4-YL) IMIDAZO-1 DERIVATIVES , 2-B! -PYRIDAZINE, THEIR PREPARATION AND THEIR THERAPEUTIC APPLICATION
WO2010070238A1 (en) * 2008-12-19 2010-06-24 Sanofi-Aventis Derivatives of 6-cycloamino-2,3-di-pyridinyl-imidazo[1,2-b]-pyridazine, preparation and therapeutic application thereof
WO2010099139A2 (en) 2009-02-25 2010-09-02 Osi Pharmaceuticals, Inc. Combination anti-cancer therapy
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
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
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
US8591943B2 (en) 2009-04-09 2013-11-26 Merck Sharp & Dohme Corp. Pyrazolo[1,5-a]pyrimidine derivatives as mTOR inhibitors
US8563545B2 (en) 2009-06-26 2013-10-22 Galapagos Nv Compound useful for the treatment of degenerative and inflammatory diseases
US11000528B2 (en) 2009-06-26 2021-05-11 Galapagos Nv Compound useful for the treatment of degenerative and inflammatory diseases
US10328081B2 (en) 2009-06-26 2019-06-25 Galapagos Nv Compound useful for the treatment of degenerative and inflammatory diseases
US8796457B2 (en) 2009-06-26 2014-08-05 Galapagos Nv Compound useful for the treatment of degenerative and inflammatory diseases
US9505754B2 (en) 2009-06-26 2016-11-29 Galapagos Nv Compound useful for the treatment of degenerative and inflammatory diseases
US8609675B2 (en) 2009-07-02 2013-12-17 Merck Sharp & Dohme Corp. Fused Tricyclic Compounds as novel mTOR inhibitors
WO2011002887A1 (en) * 2009-07-02 2011-01-06 Schering Corporation FUSED TRICYCLIC COMPOUNDS AS NOVEL mTOR INHIBITORS
WO2011041152A1 (en) 2009-09-30 2011-04-07 Schering Corporation Novel compounds that are erk inhibitors
WO2012036997A1 (en) 2010-09-16 2012-03-22 Schering Corporation Fused pyrazole derivatives as novel erk inhibitors
WO2012149014A1 (en) 2011-04-25 2012-11-01 OSI Pharmaceuticals, LLC Use of emt gene signatures in cancer drug discovery, diagnostics, and treatment
US9120785B2 (en) 2011-05-10 2015-09-01 Merck Sharp & Dohme Corp. Pyridyl aminopyridines as Syk inhibitors
US9145391B2 (en) 2011-05-10 2015-09-29 Merck Sharp & Dohme Corp. Bipyridylaminopyridines as Syk inhibitors
US9290490B2 (en) 2011-05-10 2016-03-22 Merck Sharp & Dohme Corp. Aminopyrimidines as Syk inhibitors
US9216173B2 (en) 2011-10-05 2015-12-22 Merck Sharp & Dohme Corp. 2-Pyridyl carboxamide-containing spleen tyrosine kinase (SYK) inhibitors
US8987456B2 (en) 2011-10-05 2015-03-24 Merck Sharp & Dohme Corp. 3-pyridyl carboxamide-containing spleen tyrosine kinase (SYK) inhibitors
US9006444B2 (en) 2011-10-05 2015-04-14 Merck Sharp & Dohme Corp. Phenyl carboxamide-containing spleen tyrosine kinase (SYK) inhibitors
WO2013152252A1 (en) 2012-04-06 2013-10-10 OSI Pharmaceuticals, LLC Combination anti-cancer therapy
US9242984B2 (en) 2012-06-20 2016-01-26 Merck Sharp & Dohme Corp. Pyrazolyl derivatives as Syk inhibitors
US9487504B2 (en) 2012-06-20 2016-11-08 Merck Sharp & Dohme Corp. Imidazolyl analogs as syk inhibitors
US9376418B2 (en) 2012-06-22 2016-06-28 Merck Sharp & Dohme Corp. Substituted pyridine spleen tyrosine kinase (SYK) inhibitors
US9416111B2 (en) 2012-06-22 2016-08-16 Merck Sharp & Dohme Corp. Substituted diazine and triazine spleen tyrosine kinease (Syk) inhibitors
US9353066B2 (en) 2012-08-20 2016-05-31 Merck Sharp & Dohme Corp. Substituted phenyl-Spleen Tyrosine Kinase (Syk) inhibitors
US9586931B2 (en) 2012-09-28 2017-03-07 Merck Sharp & Dohme Corp. Triazolyl derivatives as Syk inhibitors
US9624210B2 (en) 2012-12-12 2017-04-18 Merck Sharp & Dohme Corp. Amino-pyrimidine-containing spleen tyrosine kinase (Syk) inhibitors
US9598405B2 (en) 2012-12-21 2017-03-21 Merck Sharp & Dohme Corp. Thiazole-substituted aminopyridines as spleen tyrosine kinase inhibitors
US9745295B2 (en) 2013-04-26 2017-08-29 Merck Sharp & Dohme Corp. Thiazole-substituted aminoheteroaryls as spleen tyrosine kinase inhibitors
US9499534B2 (en) 2013-04-26 2016-11-22 Merck Sharp & Dohme Corp. Thiazole-substituted aminopyrimidines as spleen tyrosine kinase inhibitors
US9783531B2 (en) 2013-12-20 2017-10-10 Merck Sharp & Dohme Corp. Thiazole-substituted aminoheteroaryls as spleen tyrosine kinase inhibitors
US9822107B2 (en) 2013-12-20 2017-11-21 Merck Sharp & Dohme Corp. Thiazole-substituted aminoheteroaryls as spleen tyrosine kinase inhibitors
US9670196B2 (en) 2013-12-20 2017-06-06 Merck Sharp & Dohme Corp. Thiazole-substituted aminoheteroaryls as Spleen Tyrosine Kinase inhibitors
US10493158B2 (en) 2014-02-07 2019-12-03 Galapagos Nv Pharmaceutical compositions for the treatment of inflammatory disorders
US10708263B2 (en) 2014-02-07 2020-07-07 Galapagos Nv Salts and pharmaceutical compositions thereof for the treatment of inflammatory disorders
US10919890B2 (en) 2014-02-07 2021-02-16 Galapagos Nv Salts and pharmaceutical compositions thereof for the treatment of inflammatory disorders
US11667633B2 (en) 2014-02-07 2023-06-06 Galapagos Nv Salts and pharmaceutical compositions thereof for the treatment of inflammatory disorders
US9775839B2 (en) 2014-03-13 2017-10-03 Merck Sharp & Dohme Corp. 2-pyrazine carboxamides as spleen tyrosine kinase inhibitors

Also Published As

Publication number Publication date
CA2615433A1 (en) 2007-01-25
US20080234284A1 (en) 2008-09-25
MX2008000898A (en) 2008-03-18
KR20080036997A (en) 2008-04-29
CN101228167A (en) 2008-07-23
GB0515026D0 (en) 2005-08-31
AU2006271924A1 (en) 2007-01-25
JP2009501748A (en) 2009-01-22
BRPI0613870A2 (en) 2011-02-15
EP1910368A1 (en) 2008-04-16
RU2008106056A (en) 2009-10-27

Similar Documents

Publication Publication Date Title
US20080234284A1 (en) Pyrazolo[1,5-a]Pyrimidin-7-Yl Amine Derivatives as Protein Kinase Inhibitors
AU2005205915B2 (en) Pyrazolo[1,5-a]pyrimidin-7-yl-amine derivatives for use in the treatment of protein kinase dependent diseases
US20050222171A1 (en) Organic compounds
EP1687305B1 (en) 1h-imidazoquinoline derivatives as protein kinase inhibitors
US20100069395A1 (en) Pyrazolo[1,5-a]pyrimidine-3-carboxylic acid compounds as protein kinase inhibitors
US20080096868A1 (en) 1,4 Substituted Pyrazolopyrimidines as Kinase Inhibitors
US20060035897A1 (en) Trifluoromethyl substituted benzamides as kinase inhibitors
US20070213355A1 (en) 1H-Imidazo[4,5-C]Quinoline Derivatives in the Treatment of Protein Kinase Dependent Diseases
KR20090074791A (en) 3-amino-pyrazole-4-carboxamide derivatives useful as inhibitors of protein kinases
MXPA06008303A (en) Pyrazolo[1,5-a]pyrimidin-7-yl-amine derivatives for use in the treatment of protein kinase dependent diseases

Legal Events

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

Ref document number: 2006762704

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 9828/DELNP/2007

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: 2615433

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: MX/a/2008/000898

Country of ref document: MX

WWE Wipo information: entry into national phase

Ref document number: 2008521883

Country of ref document: JP

Ref document number: 200680026732.1

Country of ref document: CN

Ref document number: 11996337

Country of ref document: US

Ref document number: 2006271924

Country of ref document: AU

Ref document number: 1020087001656

Country of ref document: KR

NENP Non-entry into the national phase

Ref country code: DE

WWW Wipo information: withdrawn in national office

Ref document number: DE

ENP Entry into the national phase

Ref document number: 2006271924

Country of ref document: AU

Date of ref document: 20060719

Kind code of ref document: A

WWP Wipo information: published in national office

Ref document number: 2006271924

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2008106056

Country of ref document: RU

WWP Wipo information: published in national office

Ref document number: 2006762704

Country of ref document: EP

ENP Entry into the national phase

Ref document number: PI0613870

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20080121