WO2005070431A1 - Derives de pyrazolo[1,5-a]pyrimidin-7-yl-amine destines a etre utilises dans le traitement de maladies dependantes de la proteine kinase - Google Patents

Derives de pyrazolo[1,5-a]pyrimidin-7-yl-amine destines a etre utilises dans le traitement de maladies dependantes de la proteine kinase Download PDF

Info

Publication number
WO2005070431A1
WO2005070431A1 PCT/EP2005/000602 EP2005000602W WO2005070431A1 WO 2005070431 A1 WO2005070431 A1 WO 2005070431A1 EP 2005000602 W EP2005000602 W EP 2005000602W WO 2005070431 A1 WO2005070431 A1 WO 2005070431A1
Authority
WO
WIPO (PCT)
Prior art keywords
phenyl
pyrazolo
pyrimidin
methyl
ylamine
Prior art date
Application number
PCT/EP2005/000602
Other languages
English (en)
Inventor
Guido Bold
Andreas Floersheimer
Pascal Furet
Patricia Imbach
Keiichi Masuya
Joseph Schoepfer
Georg Martiny-Baron
Original Assignee
Novartis Ag
Novartis Pharma Gmbh
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
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=34807167&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=WO2005070431(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Novartis Ag, Novartis Pharma Gmbh filed Critical Novartis Ag
Priority to CA002552885A priority Critical patent/CA2552885A1/fr
Priority to RU2006130003/15A priority patent/RU2006130003A/ru
Priority to BRPI0507071-6A priority patent/BRPI0507071A/pt
Priority to EP05706961A priority patent/EP1708710A1/fr
Priority to AU2005205915A priority patent/AU2005205915B2/en
Priority to JP2006550056A priority patent/JP2007519662A/ja
Publication of WO2005070431A1 publication Critical patent/WO2005070431A1/fr
Priority to IL176737A priority patent/IL176737A0/en
Priority to TNP2006000226A priority patent/TNSN06226A1/en
Priority to NO20063758A priority patent/NO20063758L/no

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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • 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
    • 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
    • A61P17/02Drugs for dermatological disorders for treating wounds, ulcers, burns, scars, keloids, or the like
    • 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
    • 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
    • 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
    • A61P9/00Drugs for disorders of the cardiovascular system
    • 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 the use of pyrazolo[1 ,5a]pyrimidin-7-yl amine derivatives in the treatment of protein kinase dependent diseases, or for the manufacture of pharmaceutical compositions for use in the treatment of said diseases, methods of use of pyrazolo[1,5a]pyrimidin-7-yl amine derivatives in the treatment of said diseases, pharmaceutical preparations comprising pyrazolo[1 ,5a]pyrimidin-7-yl amine derivatives for the treatment of said diseases, novel pyrazo!o[1,5a]pyrimidin-7-yl amine derivatives, processes for the manufacture of the novel pyrazolo[1,5a]pyrimidin-7-yl amine derivatives and pharmaceutical preparations, the use or methods of use of the pyrazolo[1 ,5a]pyrimidin- 7-yl amine derivatives as mentioned above, and/or these pyrazolo[1,5a]pyrimidin-7-yl amine derivatives for use in the treatment of the animal or
  • the class of pyrazolo[1,5a]pyrimidin-7-yl amine compounds described herein, especially novel compounds falling under this class, has surprisingly been found to have pharmaceutically advantageous properties, allowing for the inhibition of specific types or classes or groups of kinases, especially c-Abl, Bcr-Abl, c-Kit, c-Raf, FIt-1 , Flt-3, KDR, Her-1 , 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, Axl, Cdk1 , cdk4, cdk5, Met, FAK, Pyk2, Syk, Insulin receptor kinase, Tie-2 or constitutively activating mutations of kinases (activating kinases) such as of Bcr-Abl, c-Kit, c-Raf, Flt-3, FGF-R3, PDGF-receptors, RET, and Met.
  • activating kinases such as of Bcr-Abl, c-Kit, c-Raf, Flt-3, FGF-R3, PDGF-receptors, RET, and Met.
  • the class of pyrazolo[1 ,5a]pyrimidin-7-yl amine compounds described herein further inhibit mutants of said kinases.
  • the pyrazolo[1 ,5a]pyrimidin-7-yl amine derivatives have the advantage in that their backbone allows for a plethora of substitution patterns that offer a broad possibility to achieve a fine tuning for specific interaction with the binding site of the targeted kinase or kinases, thus opening a new perspective and providing kinase inhibitors of various degrees of specificity.
  • the compounds can be used for the treatment of diseases related to especially aberrant or excessive activity of such types of kinases, especially those mentioned.
  • the invention relates to the use of a compound of the formula (I):
  • R 2 is H; substituted or unsubstituted aryl; substituted or unsubstituted heteroaryl; substituted or unsubstituted aliphatic residue; a functional group; or a substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl or substituted or unsubstituted aliphatic residue which is connected by one connecting group or atom to the pyrazolo[1 ,5a]pyrimidinyl ring;
  • R 3 can be H, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, substituted or unsubstituted aliphatic residue, a functional group, or a substituted or unsubstituted aliphatic residue which may be connected by a connecting group or atom to the pyrazolo[1 ,5a]pyrimidinyl ring, at least one of R 2 or R 3 is substituted or unsubstituted aryl;
  • A is H, halogen (such as bromo), an aliphatic moiety, a functional group, substituted or unsubstituted aryl or substituted or unsubstituted heteroaryl; and
  • R ⁇ is H, halogen or lower alkyl, or pharmaceutically acceptable salts thereof for treating a protein kinase dependent disease.
  • a preferred embodiment is the use of a compound according to the above, wherein:
  • R 2 is H; lower alkyl; cycloalkyl; benzyl; benzo thienyl, indyl substituted by lower alkyl, pyridyl or thiazolyl optionally substituted by lower alkyl; unsubstituted phenyl or phenyl substituted by one or two substituents chosen from the group consisting of; halo, hydroxy, alkoxy, benzyloxy, cycloalkyl, amino, acetyl amino, lower alkyl sulfonamide and benzene sulfonamide substituted by one or two halo;
  • R3 is H; lower alkyl optionally substituted by halo; phenyl, pyridyl, or oxazolyl; A is
  • phenyl which is unsubstituted or substituted with one or more of the substituents chosen from the group consisting of; mono-, di- or tri-lower alkoxy, di-lower alkylaminyl, morpholinyl which is optionally di-substituted by alkyl, piperazinyl which is substituted with one or more of the substituents chosen from the group consisting of lower alkyl, lower alkoxy, lower alkyl piperazinyl, pyrrolidinyl, dialkyl aminyl and lower alkanol; and
  • 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, Axl, Cdk1 , cdk4, cdk ⁇ , Met, FAK, 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.
  • activating kinases such as of Bcr-Abl, c-Kit,
  • a protein kinase dependent disease is more preferably one that depends on c-abl, Flt-3, KDR, c-Src, RET, EphB4, c-kit, cdkl , 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.
  • the invention relates to a compound of formula (I):
  • R 2 is H; substituted or unsubstituted aryl; substituted or unsubstituted heteroaryl; an aliphatic residue; a functional group; or a substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl or aliphatic residue which is connected by one connecting group or atom to the pyrazolo[1 ,5a]pyrimidinyl ring;
  • R 3 can be H, substituted or unsubstituted aryl, heteroaryl, an aliphatic residue, a functional group, or an aliphatic residue which may be connected by a connecting group or atom to the pyrazolo[1 ,5a]pyrimidinyl ring, at least one of R 2 or R 3 is substituted or unsubstituted aryl; substituted or unsubstituted heteroaryl; or a substituted or unsubstituted heteroaryl or substituted or unsubstituted aryl residue which is connected by one connecting group or atom to the pyrazolo[1 ,5a]pyrimidinyl ring, and provided that both R 2 and A cannot both be unsubstituted phenyl;
  • A is H, halogen (such as bromo), an aliphatic moiety, a functional group, substituted or unsubstituted aryl or heteroaryl; and
  • Ri is H, halogen or lower alkyl, or a pharmaceutically acceptable salt thereof.
  • a preferred embodiment is a compound according to the above, wherein; R 2 is H; lower alkyl; cycloalkyl; benzyl; benzo thienyl, indyl substituted by lower alkyl, pyridyl or thiazolyl optionally substituted by lower alkyl; unsubstituted phenyl or phenyl substituted by one or two substituents chosen from the group consisting of; halo, hydroxy, alkoxy, benzyloxy, cycloalkyl, amino, acetyl amino, lower alkyl sulfonamide and benzene sulfonamide substituted by one or two halo;
  • R3 is H; lower alkyl optionally substituted by halo; phenyl, pyridyl, or oxazolyl; A is
  • phenyl which is unsubstituted or substituted with one or more of the substituents chosen from the group consisting of; mono-, di- or tri-lower alkoxy, di-lower alkylaminyl, morpholinyl which is optionally di-substituted by alkyl, piperazinyl which is substituted with one or more of the substituents chosen from the group consisting of lower alkyl, lower alkoxy, lower alkyl piperazinyl, pyrrolidinyl, dialkyl aminyl and lower alkanol; and
  • R- is H; and provided that both R 2 and A cannot both be unsubstituted phenyl.
  • the compound is selected from the group consisting of: 3- ⁇ 7-Amino-3-[4-(4-methyl-piperazin-1-yI)-phenyl]-pyrazolo[1 ,5-a]pyrimidin-6-yl ⁇ -phenol;
  • Yet another embodiment is the use of a compound according to the above in the preparation of a pharmaceutical composition.
  • Yet another embodiment is a pharmaceutical composition comprising a compound according to the above.
  • the pharmaceutical composition preferably comprises a compound according to the above and an acceptable pharmaceutical carrier.
  • step (b) condensing the substituted 3-oxo-propionitriles of step (a) with hydrazine monohydrate in an organic solvent to form a 2H-pyrazol-3-ylamine of formula (III):
  • the invention in particular relates to pyrazolo[1,5a]pyrimidin-7-yl amine compounds of the formula (I):
  • R 2 is H; substituted or unsubstituted aryl; substituted or unsubstituted heteroaryl; substituted or unsubstituted aliphatic residue; a functional group; or a substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl or substituted or unsubstituted aliphatic residue which is connected by one connecting group or atom to the pyrazolo[1 ,5a]pyrimidinyl ring;
  • R 3 is H, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, substituted or unsubstituted aliphatic residue, a functional group, or an aliphatic residue which may be connected by a connecting group or atom to the pyrazolo[1 ,5a]pyrimidinyl ring, at least one of R 2 or R 3 is substituted or unsubstituted aryl; substituted or unsubstituted heteroaryl; or a substituted or unsubstituted heteroaryl or substituted or unsubstituted aryl residue which is connected by one connecting group or atom to the pyrazolo[1,5a]pyrimidinyl ring;
  • A is H, halogen (such as bromo), an aliphatic moiety, a functional group, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl; and
  • Ri is H, halogen or lower alkyl, or pharmaceutically acceptable salts thereof, in the treatment of protein kinase (especially tyrosine protein kinase) dependent diseases or for the manufacture of pharmaceutical compositions for use in the treatment of said diseases, methods of use of compounds of formula (I) in the treatment of said diseases, or pharmaceutical preparations comprising compounds of formula (I) for the treatment of said diseases.
  • protein kinase especially tyrosine protein kinase
  • Ri is H, halogen or lower alkyl, or pharmaceutically acceptable salts thereof, in the treatment of protein kinase (especially tyrosine protein kinase) dependent diseases or for the manufacture of pharmaceutical compositions for use in the treatment of said diseases, methods of use of compounds of formula (I) in the treatment of said diseases, or pharmaceutical preparations comprising compounds of formula (I) for the treatment of said diseases.
  • the present invention is especially related to a compound of formula (I) wherein R 2 is H; substituted or unsubstituted aryl; substituted or unsubstituted heteroaryl; substituted or unsubstituted aliphatic residue; a functional group; or a substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl or substituted or unsubstituted aliphatic residue which is connected by one connecting group or atom to the pyrazolo[1 ,5a]pyrimidinyl ring; R 3 is H, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, substituted or unsubstituted aliphatic residue, a functional group, or an aliphatic residue which may be connected by a connecting group or atom to the pyrazolo[1 ,5a]pyrimidinyl ring, at least one of R 2 or R is substituted or unsubstituted
  • the present invention also relates to a method of treating kinase dependent diseases comprising administering pyrazolo[1 ,5a]pyrimidin-7-yl amine compounds ofthe formula (I) to a warm-blooded animal, especially a human.
  • the present invention also relates to pharmaceutical preparations comprising an pyrazolo[1,5a]pyrimidin-7-yl amine compound of the formula (I), especially for the treatment of a kinase dependent disease, novel pyrazolo[1,5a]pyrimidin-7-yl amine compounds of the formula (I), a process for the manufacture of the pyrazolo[1,5a]pyrimidin-7-yl amine compounds of the formula (I), and novel starting materials and intermediates for their manufacture.
  • the present invention also relates to use of a compound of formula 1 in the manufacture of a pharmaceutical preparation for the treatment of a kinase dependent disease.
  • Aryl is an aromatic radical having 6 to 14 carbon atoms, especially phenyl, naphthyl, indenyl, azulenyl, or anthryl, and is unsubstituted or substituted by one or more, preferably one or two substituents, wherein the substituents are selected from any of the functional groups defined below, and including: lower halo, alkyl, substituted alkyl, halo lower alkyl e.g.
  • P and R 5 together with the N atom form a 3- to 8-membered heterocyclic ring containing 1-4 nitrogen, oxygen or sulfur atoms (e.g. piperazinyl, lower alkyl-piperazinyl, azetidinyl, pyrrolidinyl, piperidino, morpholinyl, imidazolinyl).
  • 1-4 nitrogen, oxygen or sulfur atoms e.g. piperazinyl, lower alkyl-piperazinyl, azetidinyl, pyrrolidinyl, piperidino, morpholinyl, imidazolinyl.
  • Aryl is more preferably phenyl which is either unsubstituted or independently substituted by one or two substituents selected from a solubilizing group selected from the group consisting of: halo (such as Cl, Br or F); hydroxy; lower alkyl (such as C C 3 lower alkyl or methyl); aryl (such as phenyl or benzyl); amino; amino lower alkyl (such as dimethylamino); acetyl amino; amino lower alkoxy (such as ethoxyamine); substituted lower alkyl (such as fluror ethyl); alkoxy (such as methoxy or benzyloxy where the benzyl ring may be substituted or unsubstituted, such as 3, 4 -dichlorobenzyloxy); sulfoamino; substituted or unsubstituted sulfonamide (such as benzo sulfonamide, chlorobenzene sulfonamide or 2,3-dich
  • R 5 together with the N atom form a 3- to 8-membered heterocyclic ring containing 1-4 nitrogen, oxygen or sulfur atoms (e.g. piperazinyl, lower alkyl-piperazinyl, pyridyl, indolyl, thiophenyl, thiazolyl, morpholinyl n-methyl piperazinyl, benzothiophenyl, azetidinyl, pyrrolidinyl, piperidino or imidazolinyl) where when R 4 and R 5 together with the N form an heterocyclic ring, said ring may be substituted with 1 , 2 or more of any of the substituents described herein, preferably piperazinyl, pyrrolidinyl, alkyl such as methyl, or hydroxy alkyl such as ethanyl.
  • substituents described herein preferably piperazinyl, pyrrolidinyl, alkyl such as methyl, or hydroxy al
  • heteroring formed by R 4 and R 5 together with the N examples include morpholinyl, which can be unsubstituted or substituted with methyl or dimethyl; piperazinyl which can be unsubstituted or substituted with 1 , 2 or 3 substituents prefereably methyl, oxy or ethanol; or piperadinyl which can be unsubstituted or substituted with 1 , 2 or 3 substituents prefereably pyrrolidinyl, amine, alkyl amine, methyl amine, dialkyl amine, dimethylamine or diethylamine;
  • a heteroaryl group is preferably monocyclic, but may be bi- or tri-cyclic, and comprises 3-24, preferably 4-16 ring atoms, wherein at least one or more, preferably one to four ring carbons are replaced by a heteroatom selected from O, N or S.
  • the heteroaryl group is selected from pyridyl, indolyl, pyrimidyl, pyrazolyl, oxazolyl, thiophenyl, benzothiophenyl, 2H- pyrrolyl, pyrrolyl, imidazolyl, benzimidazolyl, pyrazolyl, indazolyl, purinyl, pyrazinyl, pyridazinyl, 4H-quinolizinyl, isoquinolyl, quinolyl, phthalazinyl, naphthyridinyl, quinoxalyl, quinazolinyl, quinnolinyl, indolizinyl, 3H-indoIyl, isoindolyl, isoxazolyl, thiazolyl, isothiazolyl, triazolyl, tetrazolyl, furazanyl and benzo[d]pyrazol.
  • heteroaryl group is selected from the group consisting of pyridyl, indolyl, pyrimidyl, pyrazolyl, oxazolyl, thiophenyl or benzothiophenyl.
  • the heteroaryl group may be unsubstituted or substituted by one or more substituents selected from the group defined above as substituents for aryl, most preferably by hydroxy, halogen, lower alkyl, such as methyl or lower alkoxy, such as methoxy or ethoxy.
  • Aliphatic as used herein refers to any non-aromatic carbon based residue. Examples of aliphatic residues include substituted or unsubstituted alkyl, cycloalkyl, alkenyl and alkynyl.
  • 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.
  • a cycloalkyl group is preferably cyclopentyl, cyclohexyl or cycloheptyl, and may be unsubstituted or substituted by one or more, especially one or two, substituents selected from the group defined above as substituents for aryl, most preferably by lower alkyl such as methyl, lower alkoxy such as methoxy or ethoxy, or hydroxy.
  • Alkenyl and alkynyl preferably have up to 7 carbon atoms, preferably from 1 to and including 5, and can be linear or branched.
  • Alkyl, cycloalkyl, alkenyl and alkynyl 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.
  • connecting atom or group includes alkyl, (such as -CH 2 -); oxy -O- ; keto -CO-; thio -S-; sulfonyl -SO 2 -; sulfoxides -SO-; amines -NH- or-NR-; carboxylic acid; alcohol; esters (-COO-); amides (-.
  • sulfonamides (, -SO 2 NH-, -SO 2 NR'- ); sulfones (-SO 2 -); sulfoxides (-SO-); amino-group; ureas ( -NH-CO-NH-, -NR-CO-NH-, -NH- CO-NR-, -NR-CO-NR-); ethers (-O-); carbamates (-NH-CO-O-, -NR-CO-O-); or inverse amides sulfonamides and esters (-NH-CO-, -NR-CO-, -NH-SO 2 -, -NR-SO 2 -, -OOC-).
  • the term "functional group" as used herein includes: carboxylic acid; hydroxyl; halogen; cyano (-CN); ethers (-OR); ketones (-CO-R); esters (-COOR); amides (-CONH2, -CONHR, - CONRR'); thioethers (-SR); sulfonamides (-SO 2 NH 2 , -SO 2 NHR, -SO 2 NRR'); sulfones (-SO 2 - R); sulfoxides (-SO-R); amines (-NHR, NR'R); ureas (-NH-CO-NH 2 , -NH-CO-NHR); ethers (- O-R); halogens; carbamates (-NH-CO-OR); aldehyde-function (-CHO); then also inverse amides; sulfonamides and esters (-NH-CO-R, -NH-SO 2 -R, -OOC-R); R
  • 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, pro- pionic 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.
  • 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.
  • the compounds of formula (I) have valuable pharmacological properties and 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 term "USE” includes any one or more of the following embodiments of the invention, respectively: the use in the treatment of (especially tyrosine) protein kinase dependent diseases, the use for the manufacture of pharmaceutical compositions for use in the treatment of said diseases, methods of use of pyrazolo[1,5a]pyrimidin-7-yI amine derivatives in the treatment of said diseases, pharmaceutical preparations comprising pyrazolo[1,5a]pyrimidin-7-yl amine derivatives for the treatment of said diseases, and pyrazolo[1,5a]pyrimidin-7-yI amine derivatives for use in the treatment of said diseases, as appropriate and expedient, if not stated otherwise.
  • diseases to be treated and are thus preferred for USE of a compound of formula (I) are selected from (especially tyrosine) protein kinase dependent ("dependent” meaning also “supported”, not only “solely dependent”) diseases mentioned below, especially corresponding proliferative diseases, more especially diseases that depend on c-Abl, Bcr- Abl, c-Kit, c-Raf, Flt-1 , Flt-3, KDR, Her-1 , 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, Axl, Cdk1 , cdk4, cdk ⁇ , Met, FAK, Pyk2, Syk, Insulin receptor kinase, Tie-2 or constitutively activating mutations of kinases (activating kinases) such as of Bcr-Abl, c-Kit, c- Raf, Flt-3, FGF-R3, PDGF-receptors, RET, and Met, (hereinafter "said kinases”) can therefore be used in the treatment of kinase dependent diseases, especially diseases depending on said kinases and (especially aberrantly highly-expressed or constitutively activated) said kinase-dependent disease or disease dependent on the activation of the said kinase
  • a compound of formula (I) for treating diseases dependant upon c- abl, Flt-3, KDR, c-Src, RET, EphB4, c-kit, cdkl , FGFR-1 , c-raf, Her-1 , Ins-R and Tek, and use of a compound of formula (I) as an inhibitor of c-abl, Flt-3, KDR, c-Src, RET, EphB4, c- kit, FGFR-1 , c-raf, cdkl , Her-1 , Ins-R and Tek.
  • the compounds of formula (I) have valuable pharmacological properties and are useful in the treatment of protein kinase dependent diseases, e.g., as drugs to treat proliferative diseases.
  • 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-
  • 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
  • Colonies without insertion of the fusion sequence into the viral genome 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.
  • 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°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 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
  • Example: Reference inhibitor in experiment 0.4 ⁇ M, average 0.3 ⁇ M Test compound in experiment 1.0 ⁇ M, normalization: 0.3/0.4 0.75 ⁇ M
  • staurosporine or a synthetic staurosporine derivative are used as reference compounds.
  • the compounds of the formula (I) are found to show IC 50 values for RET inhibition in the range from 0.005-100 ⁇ M, preferably in the range from 0.01-2 ⁇ M.
  • 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.
  • c-Abl kinase 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 staining.
  • the assay contains (total volume of 30 ⁇ L): c-Abl kinase (50 ng), 20 mM Tris-HCI, pH 7.5, 10 mM MgCI 2 , 10 ⁇ M Na 3 VO 4 , 1 mM DTT and 0.06 ⁇ Ci/assay [ ⁇ 33 PJ-ATP (5 ⁇ M ATP) using 30 ⁇ g/mL poly- Ala,Glu,Lys,Tyr-6:2:5:1 (Poly-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 % 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 on a shaker with 0.5 % H 3 PO 4 (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 ⁇ lJwell 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°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 alkaiine phosphatase (PY20:AP from Zymed).
  • PY20:AP alkaiine 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 ofthe 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 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 mM PMSF.
  • 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, 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°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 °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-1R 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 (FBG0).
  • 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, 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 mM DTT, 200 mM NaCI.
  • the GST-tagged proteins are then eluted by 10 applications.
  • 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°C. When required, the oocytes are homogenized and centrifuged as described (Arion et al., Cell 55: 371-378 (1988) and Riaiet 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 cdkl/cycB kinase is eluted with purified p9CKShs (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).
  • Active 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 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°C. Infected Sf21 cells are scraped and collected into a 50 mL tube and centrifuged for 10 min at 4°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.
  • 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°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°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 600 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 °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.
  • 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.
  • c-rafl1 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
  • 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 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.
  • 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.
  • step (b) condensing the substituted 3-oxo-propionitriles of step (a) with hydrazine monohydrate in an organic solvent to form a 2H-pyrazol-3-ylamine of formula (III):
  • compounds of formula (I) are prepared by condensing 3-oxo- propionitriles (II) and the corresponding 2H-pyrazol-3-ylamines (III) in the presence of ethanolic HCl ( Figure 2).
  • the 2H-pyrazol-3-ylamines (III) are prepared by condensing hydrazine monohydrate with the corresponding 3-oxo-propionitriles dissolved in an organic solvent, such as EtOH, dioxane or AcOH and heated at elevated temperatures (preferably at 100 °C) for several hours.
  • the preferred procedure for preparing the pyrazolo moiety of the title compounds was stirring the hydrazine monohydrate with the corresponding 3-oxo-propionitriles in acetic acid at 100 °C for 2-3 h followed by addition of aqueous HCl and further refluxing the reaction mixture for further 20 min.
  • R1 is not H
  • the corresponding substituted hydrazines are used.
  • the 3-oxo-propionitriles (I) and (II) are synthesized from the corresponding nitriles by classical formylation reaction using freshly prepared sodium ethanolate and formic acid ethyl ester (refluxing for 1 h in EtOH).
  • the corresponding 3,3- dialkoxy-propionitiles in analogy to the procedure described by Seneci, P, Nicola, M, Inglesi, M, Vanotti, E, Resnati, G. Synth. Commun. 29 (2), 311-341 (1999)
  • 3- dimethylamino-acrylonitriles can be used.
  • compounds of formula (I) can be prepared by first synthesizing the pyrazolo[1 ,5-a]pyrimidin-7-ylamine core scaffold carrying a corresponding functional groups (X, see Figure 3) where residues A, R 2 , or R 3 , respectively, can be introduced by known reactions as indicated in Figure 3.
  • X functional group for further chemical modifications
  • R ⁇ R 2 , R 3, and X are as defined for compounds of the formula I,
  • 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.
  • Mixtures of isomers obtainable according to the invention can be separated in a manner known perse 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 dia- stereoisomers so obtainable, for example by means of fractional crystallisation, or by chromatography over optically active column materials.
  • Intermediates 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 ⁇ 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 in situ.
  • 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 in 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.
  • tyrosine protein kinase dependent disease to be treated is a proliferative disease depending on any one or more of of the following tyrosine protein kinases: especially c-Abl, Bcr-Abl, c-Kit, c-Raf, Flt-1, Flt-3, KDR, Her-1, 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, Axl, Cdkl , cdk4, cdk ⁇ , Met, FAK, Pyk2, Syk, Insulin receptor kinase, Tie-2 or constitutively activating mutations of kinases (activating kinases) such as of Bcr-Abl, c-Kit, cdkl, c-Raf, Flt-3, FGF-R3, PDGF-receptors, RET, and Met.
  • activating kinases such as of Bcr-Abl, c-Kit, cdkl, c-Raf, Flt-3, FGF-R3, PDGF-receptors, RET, and Met.
  • compounds of formula (I) may be used to treat a proliferative disease depending on the following kinases: c-abl, Flt-3, KDR, c-Src, RET, EphB4, c-kit, cdkl, FGFR-1 , c-raf, Her-1, Ins-R and Tek.
  • the invention relates especially to a compound of the formula (I),
  • R 2 is H; substituted or unsubstituted aryl; substituted or unsubstituted heteroaryl; substituted or unsubstituted aliphatic residue; a functional group; or a substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl or substituted or unsubstituted aliphatic residue which is connected by one connecting group or atom to the pyrazolo[1 ,5a]pyrimidinyl ring;
  • R 3 can be H, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, substituted or unsubstituted aliphatic residue, a functional group, or an aliphatic residue which may be connected by a connecting group or atom to the pyrazolo[1,5a]pyrimidinyl ring,
  • R 2 or R 3 is substituted or unsubstituted aryl; substituted or unsubstituted heteroaryl; or a substituted or unsubstituted heteroaryl or substituted or unsubstituted aryl residue which is connected by one connecting group or atom to the pyrazolo[1,5a]pyrimidinyI ring;
  • A is H, halogen (such as bromo), an aliphatic moiety, a functional group, substituted or unsubstituted aryl or substituted or unsubstituted heteroaryl; and
  • R-i is H, halogen or lower alkyl, or pharmaceutically acceptable salts thereof, and use of compounds of formula (I) in the treatment of kinase dependent diseases or for the manufacture of pharmaceutical preparations for the treatment of kinase dependent diseases.
  • the invention further relates to a compound of the formula (I),
  • R 2 is H; substituted or unsubstituted aryl; substituted or unsubstituted heteroaryl; substituted or unsubstituted aliphatic residue; a functional group; or a substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl or substituted or unsubstituted aliphatic residue which is connected by one connecting group or atom to the pyrazolo[1 ,5a]pyrimidinyl ring;
  • R 3 can be H, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, substituted or unsubstituted aliphatic residue, a functional group, or a substituted or unsubstituted aliphatic residue which may be connected by a connecting group or atom to the pyrazolo[1,5a]pyrimidinyl ring,
  • R 2 or R 3 is substituted or unsubstituted aryl; substituted or unsubstituted heteroaryl; or a substituted or unsubstituted heteroaryl or substituted or unsubstituted aryl residue which is connected by one connecting group or atom to the pyrazolo[1 ,5a]pyrimidinyl ring;
  • A is H, halogen (such as bromo), an aliphatic moiety, a functional group, substituted or unsubstituted aryl or heteroaryl; and
  • Ri is H, halogen or lower alkyl, or pharmaceutically acceptable salts thereof, and use of compounds of formula (I) in the treatment of kinase dependent diseases or for the manufacture of pharmaceutical preparations for the treatment of kinase dependent diseases.
  • the connecting atom or group is selected from the group consisting of: alkyl, (such as -CH 2 - ); oxy -O-; keto -CO-; thio -S-; sulfonyl -SO 2 -; sulfoxides -SO-; amines -NH- or -NR-; carboxylic acid; alcohol; esters (-COO-); amides (- -CONR-, -CONHR'-); sulfonamides ( - SO 2 NH-, -SO 2 NR'-); (-SO 3 -); sulfoxides (-SO-); amino-group; ureas ( -NH-CO-NH-, -NR-CO- NH-, -NH-CO-NR-, -NR-CO-NR-); ethers (-O-); carbamates (-NH-CO-O-, -NR-CO-O-); or inverse amides sulfonamides and esters
  • the functional group is selected from the group consisting of: carboxylic acid; hydroxyl; halogens; cyano (-CN); ethers (-OR); ketones (-CO-R); esters (-COOR); amides (-CONH 2 , - CONHR, -CONRR'); thioethers (-SR); sulfonamides (-SO 2 NH 2 , -SO 2 NHR, -SO 2 NRR'); sulfones (-SO 2 -R); sulfoxides (-SO-R); amines (-NHR, NR'R); ureas (-NH-CO-NH 2 , -NH-CO- NHR); ethers (-O-R); halogens; carbamates (-NH-CO-OR); aldehyde-function (-CHO); then also inverse amides; sulfonamides and esters (-NH-CO-R, -NH-SO 2 -R, -OOC-
  • A is H; a halo (such as Br); or aryl (such as phenyl or benzyl) or heterocyclyl (such as pyridinyl, indolyl or benzothiophenyl),
  • aryl or heterocyclyl may be substituted or unsubstituted with up to 4, preferably up to 2 substituents, wherein the substituents are the same or different and are independently selected from halo (such as Cl or Br); hydroxy; amino; amino lower alkyl (such as dimethylamino); amino lower alkoxy (such as ethoxyamine); lower alkyl (such as methyl); lower alkoxy (such as methoxy); substituted or unsubstituted sulfonamide (such as benzo sulfonamide, chlorobenzene sulfonamide or dichloro benzene sulfonamide); carbamates; R 4 R 5 , wherein R and R 5 can be the same or different and are independently H; lower alkyl (e.g.
  • R 4 and R 5 together with the N atom form a 3- to 8-membered heterocyclic ring containing 1-4 nitrogen, oxygen or sulfur atoms (e.g. piperazinyl or lower alkyl piperazinyl) where when R 4 and R 5 together with the N form an heterocyclic ring, said ring may be substituted with 1 , 2 or more of any of the substituents described herein, preferably piperazinyl, pyrrolidinyl, alkyl such as methyl, or hydroxy alkyl such as ethanyl.
  • heteroring formed by R 4 and R 5 together with the N examples include morpholinyl, which can be unsubstituted or substituted with methyl or dimethyl; piperazinyl which can be unsubstituted or substituted with 1 , 2 or 3 substituents prefereably methyl, oxy or ethanol; or piperadinyl which can be unsubstituted or substituted with 1 , 2 or 3 substituents prefereably pyrrolidinyl, amine, alkyl amine, methyl amine, dialkyl amine, dimethylamine or diethylamine;
  • R 2 is H, C C 3 lower alkyl (such as methyl) or aryl (such as phenyl or benzyl) or heterocyclyl (such as pyridyl, indolyl, thiophenyl, thiazolyl or benzothiophenyl), wherein the aryl or heterocyclyl may be substituted or unsubstituted with up to 4, preferably up to 2 substituents, wherein the substituents are the same or different and are independently selected from halo (such as Cl, F or Br); hydroxy; amino; amino lower alkyl; C C 3 lower alkyl; alkoxy (such as methoxy and benzyloxy where the benzyl ring may be substituted or unsubstituted, such as 3, 4 -dichlorobenzyloxy); sulfoamino; substituted or unsubstituted benzosulfonamide (such as 2, 3-dichlorobenzene sulfonamide); substitute
  • R 3 is H; C C 3 alkyl, methyl; phenyl; pyridinyl or oxaz-5-yl;
  • a compound of formula (I), or a pharmaceutically acceptable salt thereof in the manufacture of a pharmaceutical preparation for the treatment of a kinase dependent disease.
  • a compound of the formula (I), or a pharmaceutically acceptable salt thereof as shown above for use in the treatment of a kinase dependent disease, especially one depending on said kinases and (especially aberrantly highly expressed or constitutively activated) said kinases-dependent disease or disease dependent on the activation of the said kinases pathways or disease dependent on any two or more of said kinases.
  • a kinase dependant disease may be a proliferative disease including a hyperproliferative condition, such as leukemias, hyperplasias, fibrosis (especially pulmonary, but also other types of fibrosis, such as renal fibrosis), angiogenesis, psoriasis, atherosclerosis and smooth muscle proliferation in the blood vessels, such as stenosis or restenosis following angioplasty.
  • a hyperproliferative condition such as leukemias, hyperplasias, fibrosis (especially pulmonary, but also other types of fibrosis, such as renal fibrosis), angiogenesis, psoriasis, atherosclerosis and smooth muscle proliferation in the blood vessels, such as stenosis or restenosis following angioplasty.
  • a method of treating a kinase dependent disease comprising administering a compound of formula (I), where 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 (especially SCLC), 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. Also for the treatment of atherosclerosis, thrombosis, psoriasis, scleroderma and fibrosis.
  • Compounds of formula (I) are able to bring about the regression of tumors and to prevent the formation of tumor metastases and the growth of (also micro)metastases.
  • they can be used in epidermal hyperproliferation (e.g. psoriasis), in prostate hyperplasia, and in the treatment of neoplasias, especially of epithelial character, for example mammary carcinoma.
  • the compounds of formula (I) in the treatment of diseases of the immune system insofar as several or, especially, individual tyrosine protein kinases are involved; furthermore, the compounds of formula (I) can be used also in the treatment of diseases of the central or peripheral nervous system where signal transmission by at least one tyrosine protein kinase, especially selected from those mentioned specifically, is involved.
  • KDR inhibitors are thus especially appropriate for the therapy of diseases related to VEGF receptor tyrosine kinase overexpression.
  • diseases especially retinopathies, age-related macula degeneration, psoriasis, haemangioblastoma, haemangioma, arteriosclerosis, inflammatory diseases, such as rheumatoid or rheumatic inflammatory diseases, especially arthritis, such as rheumatoid arthritis, or other chronic inflammatory disorders, such as chronic asthma, arterial or post-transplantational atherosclerosis, endometriosis, and especially neoplastic diseases, for example so-called solid tumors (especially cancers of the gastrointestinal tract, the pancreas, breast, stomach, cervix, bladder, kidney, prostate, ovaries, endometrium, lung, brain, melanoma, Kaposi's sarcoma, squamous cell carcinoma of head and neck, malignant pleural mesotherioma,
  • Flt3 (FMD-like tyrosine kinase) is especially expressed in hematopoietic progenitor cells and in progenitors of the lymphoid and myeloid series.
  • Aberrant expression of the Flt3 gene has been documented in both adult and childhood leukemias including AML (acute myelogenous leukemia), AML with trilineage myelodysplasia (AML/TMDS), ALL (acute lymphoblastic leukemia), CML (chronic myelogenous leukemia) and myelodysplastic syndrome (MDS), which are therefore the preferred diseases to be treated with compounds of the formula I.
  • AML acute myelogenous leukemia
  • AML/TMDS trilineage myelodysplasia
  • ALL acute lymphoblastic leukemia
  • CML chronic myelogenous leukemia
  • MDS myelodysplastic syndrome
  • CML chronic myelogeous leukemia
  • HSCs hematopoietic stem cells
  • the BCR-ABL fusion gene encodes as constitutively activated kinase, which transforms HSCs to produce a phenotype exhibiting deregulated clonal proliferation, reduced ca- pacity to adhere to the bone marrow stroma and a reduces apoptotic response to mutagenic stimuli, which enable it to accumulate progressively more malignant transformations.
  • the resulting granulocytes fail to develop into mature lymphocytes and are released into the circulation, leading to a deficiency in the mature cells and increased susceptibility to infection.
  • ATP-competitive inhibitors of Bcr-Abl have been described which prevent the kinase from activating mitogenic and anti-apoptotic pathways (e.g. P-3 kinase and STAT5), leading to the death of the BCR-ABL phenotype cells and thereby providing an effective therapy against CML.
  • the pyrazolo[1 ,5a]pyrimidin-7-yl amine derivatives useful according to the present invention, especially the compounds of formula I, as Bcr-Abl inhibitors are thus especially appropriate for the therapy of diseases related to its overexpression, especially leukemias, such as leukemias, e.g. CML or ALL.
  • the compounds of formula (I) which inhibit the tyrosine kinase activity of the EGF-R or of the other protein tyrosine kinases mentioned are therefore useful, for example, in the treatment of benign or malignant tumors.
  • the compounds of formula (I) are e.g. able to simultaneously inhibit the growth of tumors with deregulated EGF-R and/or ErbB-2 activity as well as to inhibit the vascularisation of solid tumors triggered by VEGF. This combined activity leads to an improved antitumor effect (see also WO 02/41882).
  • the use of a dual inhibitor reduces the risk of drug-drug interactions and further reduces the total drug load as compared to a combination therapy.
  • the compounds of formula (I) are capable of slowing down tumor growth or effecting tumor regression and of preventing the formation of tumor metastases and the growth of micrometastases. They can be used especially in the case of epidermal hyperproliferation (psoriasis), in the treatment of solid cancers like for example non-small cell lung cancer, squameous carcinoma (head and neck), breast, gastric, ovarian, colon and prostate cancers as well as gliomas and in the treatment of leukemias, such as especially acute myeloid leukemia (AML) and chronic myeloid leukemia (CML).
  • AML acute myeloid leukemia
  • CML chronic myeloid leukemia
  • the compounds of formula (I) can be used in the treatment of those disorders of the immune system in which several or, especially, individual protein tyrosine kinases and/or (furthermore) serine/threonine protein kinases are involved; the compounds of formula (I) can also be used in the treatment of those disorders of the central or peripheral nervous system in which signal transmission by several or, especially, a single protein tyrosine kinase(s) and/or (furthermore) serine/threonine protein kinase(s) is/are involved.
  • Angiogenesis is regarded as an absolute prerequisite for those tumors which grow beyond a maximum diameter of about 1-2 mm; up to this limit, oxygen and nutrients may be supplied to the tumor cells by diffusion. Every tumor, regardless of its origin and its cause, is thus dependent on angiogenesis for its growth after it has reached a certain size.
  • the present invention can also be used to prevent or treat diseases that are 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
  • 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.
  • 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, H ⁇ ls 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.
  • the injection compositions are prepared in customary manner under sterile conditions; the same applies also to introducing the compositions into ampoules or vials and sealing the containers.
  • 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.
  • 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 dose
  • 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 II 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 isoform
  • 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 administered, 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 II 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.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.
  • 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-(1 H- indol-3-yl)ethyl]-amino]methyl]phenyl]-2E-2-propenamide, N-hydroxy-3-[4-[[[2-(2-methyl-1 H- 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
  • anti-plastic antimetabolite includes, but is not limited to, 5-FIuorouracil 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.
  • the monoclonal antibody 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 l
  • compounds targeting, decreasing or inhibiting the activity of the insulin-like growth factor receptor l(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 members of the c-Abl family and their gene-fusion products e.g. BCR-Abl 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;
  • UCN-01 safingol, BAY 43-9006, Bryostatin 1, Perifosine; llmofosine; RO 318220 and RO 320432; GO 6976; Isis 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, CI-1033, 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 ⁇ -toc ⁇ trienol.
  • 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-methyI-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.
  • titaniumudronic 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 (Zarnestra).
  • 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.
  • 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.
  • 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. famesyl 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.
  • famesyl 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-(1 H-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 everolimus, RAD
  • 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-1H-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 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-pyridyImethyl)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 al, Proc Natl Acad Sci U S A, Vol. 93, pp.
  • 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 lgG1 antibody, Angiozyme (RPI 4610) and Avastan.
  • 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.
  • 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.
  • 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 e.g, in eluents or solvent mixtures, are given in volume by volume
  • 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 C 18 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).
  • 1 H-NMR measurements are performed on a Varian Gemini 400 or a Bruker DRX 500 spectrometer using tetraethylsilane as internal standard. Chemical shifts are expressed in ppm downfield from tetraethylsilane and coupling constants (J) are expressed in Hertz (Hz). Electrospray mass spectra are obtained with a Fisons Instruments VG Platform II. Melting points are measured with a B ⁇ chi 510 melting point apparatus. Commercially-available solvents and chemicals are used for syntheses.
  • Examples 5-69 The following Examples enlisted on Table 1 are synthesized analogously to the preparation of Example 1. The syntheses of intermediates for the preparation of compounds of Examples 5-69 which are not commercially available are described in the text below Table 1. In cases where the title compounds carry a free amino group (Examples 52 - 54), the final products are generated from their corresponding nitro-function carrying precursors by hydrogenation in the presence of Pd/C (10 %) in THF/MeOH during several hours.
  • 2-Methyl-3-oxo-3-phenyl-propionitrile is prepared analogously to the procedure of Yoo et al., Tetrahedron Lett, Vol. 43, No. 27, pp. 4813-4815 (2002).
  • 2-Bromo-propionitrile (0.965 mL, 11.05 mmol) and In-powder (975 mg, 8.5 mmol) are stirred under Ar in THF (15 mL) for 1 hour.
  • benzoylnitrile (735 mg, 5.6 mmol) the reaction mixture is stirred at 60°C in a microwave ofen (Emrys optimizer, personal chemistry, Sweden) for 30 minutes.
  • Example 10 The compound of Example 10 is synthesized analogously to the preparation of compound of Stage 1.1 by condensing 2,3-dichloro- ⁇ /-[4-(cyano-formyl-methyl)-phenyl]- benzenesulfonamide (Stage 10.1) and 4-(4-dimethylamino-phenyl)-2H-pyrazol-3-ylamine (Stage 10.3).
  • 4-(4-Dimethylamino-phenyl)-2f/-pyrazol-3-ylamine is prepared from 2-(4-dimethylamino- phenyl)-3-oxo-propionitrile (Stage 10.4) and hydrazine hydrate as described in U.S. Patent No. 2,989,539 (20.6.61; Anderson and Reiff; Example 18).
  • 2-(4-Dimethylamino-phenyl)-3-oxo-propionitrile is prepared from (4-dimethylamino-phenyl)- acetonitrile, ethyl formate and sodium as described in U.S. Patent No. 2,989,539 (Example 18).
  • Example 11 is prepared analogously to the synthesis of the compound of Example 10 using 4-(4-dimethylamino-phenyl)-2H-pyrazol-3-ylamine (Stage 10.3) and 4- chloro-benzenesulfonic acid 4-(cyano-formyl-methyl)-phenyl ester (Stage 11.1).
  • 4-Chloro-benzenesulfonic acid 4-(cyano-formyl-methyI)-phenyl ester is prepared as described in Example 10 (Stage 10.1), using commercially-available 4-(cyanomethyl)phenyl- 4-chlorobenzene-1 -sulfonate instead.
  • Compounds 61 , 62, 64, 67, and 68 carrying sulfonamide and acetylamide functions are prepared by reacting the amino precursor with the corresponding sulfonic acid chloride or acetic acid anhydride in the presence of pyridine.
  • Example 72 The title compound is prepared as described in example 72; using 4-[4-(4-Methyl-piperazin- 1-yl)-phenyl]-2H-pyrazol-3-ylamine (Example 1 ; Stage 1.2) and 2-(3-Chloro-phenyl)-3-oxo- butyronitrile (Example 73, Stage 73.1) instead.
  • Example 72 The title compound is prepared as described in example 72; using 4-[2-Methoxy-5-(4-methyl- piperazin-1 -yl)-phenyl]-2H-pyrazol-3-ylamine and 2-(3-Chloro-phenyl)-3-oxo-butyronitrile (Example 72, Stage 72.1) instead .
  • Stage 74.1 4-f2-Methoxv-5-(4-methyl-piperazin-1 -yl)-phenvll-2H-pyrazol-3-vlamine
  • Example 72 The title compound is prepared as described in example 72; using 4-[2-Methoxy-4-(4-methyl- piperazin-1 -yl)-phenyl]-2H-pyrazol-3-ylamine and 2-(3-Chloro-phenyl)-3-oxo-butyronitrile (Example 72, Stage 72.1) instead .
  • Example 76 3- ⁇ 7-Amino-3-[2-methoxy-4-(4-methyl-piperazin-1-yl)-phenyl]- pyrazolo[1,5-a]pyrimidin-6-yl ⁇ -phenol
  • the title compound is prepared by dissolving 6-(3-Benzyloxy-phenyl)-3-[2-methoxy-4-(4- methyl-piperazin-1-yl)-phenyl]-pyrazolo[1 ,5-a]pyrimidin-7-ylamine in methanol and subjecting it to catalytic hydrogenation in the presence of Pd/C as described in example 1.
  • Stage 77.1 (Z)-2-(2-Chloro-phenyl)-3-dimethvlamino-acrylonitrile.
  • N,N-Dimethylformamide-dimethylacetal (9.06 mL; 64.3 mMol) and 2-chlorobenzylcyanide (1.95 g; 12.86 mMol) is heated under stirring to 100°C under an atmosphere of Argon.
  • Example 78 6-(2-Chloro-phenyl)-3-[3-(4-methyl-piperazin-1-yl)-phenyl]-pyrazolo[1, 5-a]pyrimidin-7-ylamine
  • Stage 79.1 2-(4-Fluoro-phenvl)-3-oxo-butyronitrile.
  • the title compound is prepared as described for example 72, Stage 72.1 using (4-Fluoro- phenyl)-acetonitrile instead.
  • Stage 82.1 2-(3-Chloro-4-fluoro-phenvl)-3-oxo-butyronitrile
  • the title compound is prepared as described for example 72, Stage 72.1 using (3-ChIoro-4- fluoro-phenyl)-acetonitrile instead.
  • White crystals; mp. 133-134°C; MS(ESl " ):m/z 209.9 (M-
  • Example 84 6-(3-Bromo-phenyl)-5-methyI-3-[3-(4-methyl-piperazin-1-yl)-phenyI]- pyrazolo[1,5-a]pyrimidin-7-ylamine
  • the title compound is prepared as described for example 72, Stage 72.1 using 2-(3-Bromo- phenyl)-3-oxo-butyronitrile instead.
  • Stage 84.1 2-(3-Bromo-phenvl)-3-oxo-butyronitrile.
  • the title compound is prepared as described for example 72, Stage 72.1 using (3-Bromo- phenyl)-acetonitrile instead.
  • White crystals; mp. 96-100°C; MS(ESl “ ):m/z 235.9 (M-H) " ;
  • Stage 85.1 3-(3-Bromo-phenvl)-2-formvl-propionitrile.
  • 3-(3-Bromophenyl)propionitriIe (0.703 mL; 4.66 mMol) and ethyl formate (1.499 mL; 18.64 mMol) are dissolved in THF anhydrous (12.5 mL) followed by the addition of NaH (60% in mineral oil; 670 mg) at rt. After 17 h at rt, additional NaH (448 mg) and ethyl formate (0.765 mL) is added. Since this results in a strong exothermic reaction, additional solvent is added (15 mL of THF).
  • reaction mixture After completion (3 days), the reaction mixture is cooled to 0°C, treated with a few little ice cubes, followed by addition of 6N HCl (3 mL) to acidify the mixture..After addition of water (50 mL), the mixture is extracted with EtOAc (3x 100 mL).
  • Example 86 6-(3-Bromo-phenyl)-3-[3-(4-methyl-piperazin-1-yl)-phenyl]- pyrazolo[1,5-a]pyrimidin-7-ylamine
  • the title compound is prepared as described in example 72; using (Z)-2-(3-Bromo-phenyl)-3- dimethylamino-acrylonitrile instead.
  • Stage 90.1 2- ⁇ 4-[3-(5-Amino-1 H-pyrazol-4-yl)-phenyl]-piperazin-1 -yl ⁇ -ethanol
  • Example 92 6-(3-Chloro-phenyl)-3-(3,4-dimethoxy-phenyl)-5-fluoromethyl- pyrazolo[1,5-a]pyrimidin-7-ylamine
  • Stage 92.1 2-(3-Chloro-phenyl)-4-fluoro-3-oxo-butyronitrile
  • the title compound is prepared as described for example 72, Stage 72.1 using fluoro-acetic acid ethyl ester instead.
  • Beige crystals; mp. 90-96°C; MS(ESf):m/z 209.9 (M-H) " ; HPLC:
  • Stage 102.1 4-[3-(4-Pyrrolidin-1 -yl-piperidin-1 -yl)-phenyl]-1 H-pyrazol-3-ylamine
  • the title compound is prepared as described in example 72; using 4-[3-(4-Pyrrolidin-1 -yl- piperidin-1 -yl)-phenyl]-1 H-pyrazol-3-ylamine (Example 102; Stage 102.1) and 2-(4-Fluoro- phenyl)-3-oxo-butyronitrile (Example 79; Stage 79.1) instead.
  • Example 104 6-(3-Chloro-phenyl)-3-[3-(4-diethylamino-piperidin-1-yl)-phenyl]-5-methyl-pyrazolo[1,5- a]pyrimidin-7-ylamine
  • the title compound is prepared as described in example 72; using ⁇ 1-[3-(3-Amino-1 H- pyrazol-4-yl)-phenyl]-piperidin-4-yl ⁇ -diethyl-amine instead.
  • Stage 104.1 ⁇ 1 -[3-(3-Amino-1 H-pyrazoI-4-yl)-phenyl]-piperidin-4-yI ⁇ -diethyl-amine
  • the title compound is prepared as described in example 1 , (Stage 1.2 and 1.4 and 1.5); using (3-bromo-phenyl)-acetonitrile and diethyl-piperidin-4-yl-amine instead.
  • Example 104 The title compound is prepared as described in example 72; using ⁇ 1-[3-(3-Amino-1H- pyrazol-4-yl)-phenyl]-piperidin-4-yl ⁇ -diethyl-amine (Example 104, Stage 104.1) and 2-(4- Fluoro-phenyl)-3-oxo-butyronitrile (Example 79; Stage 79.1) instead.
  • the title compound is prepared as described in example 1 , (Stage 1.2 and 1.4 and 1.5); using (3-bromo-phenyl)-acetonitrile and dimethyI-piperidin-4-yl-amine instead.
  • Example 108 The title compound is prepared as described in example 72; using ⁇ 1-[3-(5-Amino-1H- pyrazol-4-yl)-phenyl]-piperidin-4-yl ⁇ -dimethyl-amine (Example 108; Stage 108.1) and 2-(3,4- difluoro-phenyl)-3-oxo-butyronitrile (Example 98; Stage 98.1) instead.
  • Stage 110.1 4-(3,4,5-Trimethoxy-phenyl)-2H-pyrazol-3-ylamine
  • the title compound is prepared as described in example 1, (Stage 1.4 and 1.2); using (3,4,5-trimethoxy-phenyl)-acetonitrile instead.
  • Example 110 The title compound is prepared as described in example 72; using 4-(3,4,5-trimethoxy- phenyl)-2H-pyrazol-3-ylamine (Example 110; Stage 110.1) and 2-(3,4-difluoro-phenyl)-3-oxo- butyronitrile (Example 98; Stage 98.1) instead.
  • Stage 112.1 4-(3-Methoxy-phenyl)-2H-pyrazol-3-ylamine
  • the title compound is prepared as described in example 1 , (Stage 1.4 and 1.2); using (3- methoxy-phenyl)-acetonitrile instead.
  • 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 1-115 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.
  • 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.
  • Tablets 2 comprising compounds of the formula (I)
  • Tablets comprising, as active ingredient, 100 mg of any one of the compounds of formula (I) of Examples 1-115 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) given in Examples 1-115, 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.
  • Activity as used herein is defined as having IC 50 values for kinase inhibition of 10 ⁇ M or less than:

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Immunology (AREA)
  • Ophthalmology & Optometry (AREA)
  • Dermatology (AREA)
  • Urology & Nephrology (AREA)
  • Epidemiology (AREA)
  • Cardiology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Rheumatology (AREA)
  • Transplantation (AREA)
  • Pain & Pain Management (AREA)
  • Vascular Medicine (AREA)
  • Oncology (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Endocrinology (AREA)
  • Reproductive Health (AREA)
  • Hematology (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

La présente invention a trait à l'utilisation des composés de pyrazolo[1,5a]pyrimidin-7-yl amine et de leurs sels dans le traitement de maladies dépendantes de la protéine kinase et pour la fabrication de préparations pharmaceutiques pour le traitement desdites maladies, à de nouveaux composés de pyrazolo[1,5a]pyrimidin-7-yl amine, et à un procédé pour la préparation de composés de pyrazolo[1,5a]pyrimidin-7-yl amine
PCT/EP2005/000602 2004-01-22 2005-01-21 Derives de pyrazolo[1,5-a]pyrimidin-7-yl-amine destines a etre utilises dans le traitement de maladies dependantes de la proteine kinase WO2005070431A1 (fr)

Priority Applications (9)

Application Number Priority Date Filing Date Title
CA002552885A CA2552885A1 (fr) 2004-01-22 2005-01-21 Derives de pyrazolo[1,5-a]pyrimidin-7-yl-amine destines a etre utilises dans le traitement de maladies dependantes de la proteine kinase
RU2006130003/15A RU2006130003A (ru) 2004-01-22 2005-01-21 Применение производных пиразоло[1, 5-а]пиримидин-7-иламина для лечения заболеваний, зависимых от протеинкиназ
BRPI0507071-6A BRPI0507071A (pt) 2004-01-22 2005-01-21 derivados de pirazol[1,5-a]pirimidin-7-il-amina para emprego no tratamento de doenças dependentes de proteìna cinase
EP05706961A EP1708710A1 (fr) 2004-01-22 2005-01-21 Derives de pyrazolo [1,5-a]pyrimidin-7-yl-amine destines a etre utilises dans le traitement de maladies dependantes de la proteine kinase
AU2005205915A AU2005205915B2 (en) 2004-01-22 2005-01-21 Pyrazolo[1,5-a]pyrimidin-7-yl-amine derivatives for use in the treatment of protein kinase dependent diseases
JP2006550056A JP2007519662A (ja) 2004-01-22 2005-01-21 タンパク質キナーゼ依存性疾患の処置に使用するためのピラゾロ[1,5−a]ピリミジン−7−イル−アミン誘導体
IL176737A IL176737A0 (en) 2004-01-22 2006-07-06 Pyrazolo[1,5-a] pyrimidin-7-yl-amine derivatives for use in the treatment of protein kinase dependent diseases
TNP2006000226A TNSN06226A1 (en) 2004-01-22 2006-07-21 Pyrazolo[1,5-a]pyrimidin-7-yl-amine derivatives for use in the treatment of protein kinase dependent diseases
NO20063758A NO20063758L (no) 2004-01-22 2006-08-22 Pyrazolo[1,5-A]pyrimidin-7-yl-amin derivater for anvendelse i behandling av proteinkinase avhengige sykdommer

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US53822004P 2004-01-22 2004-01-22
US60/538,220 2004-01-22

Publications (1)

Publication Number Publication Date
WO2005070431A1 true WO2005070431A1 (fr) 2005-08-04

Family

ID=34807167

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2005/000602 WO2005070431A1 (fr) 2004-01-22 2005-01-21 Derives de pyrazolo[1,5-a]pyrimidin-7-yl-amine destines a etre utilises dans le traitement de maladies dependantes de la proteine kinase

Country Status (17)

Country Link
EP (1) EP1708710A1 (fr)
JP (1) JP2007519662A (fr)
KR (1) KR20070009546A (fr)
CN (1) CN1909908A (fr)
AR (1) AR049769A1 (fr)
AU (1) AU2005205915B2 (fr)
BR (1) BRPI0507071A (fr)
CA (1) CA2552885A1 (fr)
EC (1) ECSP066718A (fr)
IL (1) IL176737A0 (fr)
MA (1) MA28400B1 (fr)
NO (1) NO20063758L (fr)
PE (1) PE20051089A1 (fr)
RU (1) RU2006130003A (fr)
TN (1) TNSN06226A1 (fr)
TW (1) TW200528103A (fr)
WO (1) WO2005070431A1 (fr)

Cited By (37)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007009773A1 (fr) * 2005-07-21 2007-01-25 Novartis Ag Derives de pyrazolo[1,5a]pyrimidin-7-yl-amine utilises en tant qu'inhibiteurs des proteines kinases
US7196078B2 (en) 2002-09-04 2007-03-27 Schering Corpoartion Trisubstituted and tetrasubstituted pyrazolopyrimidines as cyclin dependent kinase inhibitors
WO2007041712A1 (fr) 2005-10-06 2007-04-12 Schering Corporation Pyrazolopyrimidines en tant qu’inhibiteurs de protéines kinases
WO2007044441A2 (fr) * 2005-10-06 2007-04-19 Schering Corporation Methodes destinees a inhiber des proteines kinases
WO2007103432A2 (fr) * 2006-03-08 2007-09-13 Novartis Ag Utilisation de derives de pyrazolo[1,5a]pyrimidin-7-yl amine dans le traitement de troubles neurologiques
WO2007109183A2 (fr) * 2006-03-20 2007-09-27 Novartis Ag Mutations et polymorphismes de la tyrosine kinase 1 liée au fms
WO2007112998A1 (fr) * 2006-04-04 2007-10-11 Novartis Ag N-(aryl- ou hétéroaryl)-pyrazolo-[1,5-a]pyrimidines 3-non substituées utilisées comme inhibiteurs de kinase
WO2007113000A1 (fr) * 2006-04-04 2007-10-11 Novartis Ag N-(ARYL- OU HÉTÉROARYL)-PYRAZOLO[1,5-a]PYRIMIDINES SUBSTITUÉES EN POSITION 3 UTILISÉS EN TANT QU'INHIBITEURS DE KINASES
WO2008037459A1 (fr) * 2006-09-28 2008-04-03 Novartis Ag Dérivés pyrazolo[1,5-a]pyrimidine et leur utilisation thérapeutique
WO2008037477A1 (fr) 2006-09-29 2008-04-03 Novartis Ag PYRAZOLOPYRIMIDINES UTILISÉES COMME INHIBITEURS DES LIPIDES KINASES Pl3K
EP1918291A1 (fr) * 2006-10-30 2008-05-07 Novartis AG Dérives de pyrazole fusionnes substitués de 3-aminocarbonyle comme modulateurs de protéine kinase
JP2010505838A (ja) * 2006-10-04 2010-02-25 シェーリング コーポレイション サイクリン依存性キナーゼインヒビターとしてのピラゾロピリミジン
JP2010505839A (ja) * 2006-10-04 2010-02-25 シェーリング コーポレイション サイクリン依存性キナーゼインヒビターのピラゾロピリミジン
JP2010529123A (ja) * 2007-06-07 2010-08-26 シェーリング コーポレイション 置換−3−アミノピラゾールの合成
WO2011041152A1 (fr) 2009-09-30 2011-04-07 Schering Corporation Nouveaux composés inhibiteurs d'erk
WO2012024586A1 (fr) 2010-08-19 2012-02-23 E. I. Du Pont De Nemours And Company Pyrazoles fongicides
WO2012036997A1 (fr) 2010-09-16 2012-03-22 Schering Corporation Dérivés condensés de pyrazole utilisés comme nouveaux inhibiteurs erk
US8591943B2 (en) 2009-04-09 2013-11-26 Merck Sharp & Dohme Corp. Pyrazolo[1,5-a]pyrimidine derivatives as mTOR inhibitors
US10023570B2 (en) 2015-07-16 2018-07-17 Array Biopharma Inc. Substituted pyrazolo[1,5-A]pyridine compounds as RET kinase inhibitors
WO2018136661A1 (fr) 2017-01-18 2018-07-26 Andrews Steven W Composés de pyrazolo[1,5-a]pyrazine substitués utilisés en tant qu'inhibiteurs de la kinase ret
WO2018136663A1 (fr) 2017-01-18 2018-07-26 Array Biopharma, Inc. Inhibiteurs de ret
US10112942B2 (en) 2016-10-10 2018-10-30 Array Biopharma Inc. Substituted pyrazolo[1,5-A]pyridine compounds as RET kinase inhibitors
US10144734B2 (en) 2016-10-10 2018-12-04 Array Biopharma Inc. Substituted pyrazolo[1,5-A]pyridine compounds as RET kinase inhibitors
EP3417912A1 (fr) 2005-12-23 2018-12-26 ARIAD Pharmaceuticals, Inc. Composants hétéroaromatiques bicycliques
WO2019075108A1 (fr) 2017-10-10 2019-04-18 Metcalf Andrew T Formes cristallines
WO2019075114A1 (fr) 2017-10-10 2019-04-18 Mark Reynolds Formulations comprenant du 6-(2-hydroxy-2-méthylpropoxy)-4-(6-(6-((6-méthoxypyridin-3-yl)méthyl)-3,6-diazabicyclo[3.1.1]heptan-3-yl)pyridin-3-yl)pyrazolo[1,5-a]pyridine-3-carbonitrile
WO2019143977A1 (fr) 2018-01-18 2019-07-25 Array Biopharma Inc. Composés de pyrrolo[2,3-d]pyrimidines substitués utilisés en tant qu'inhibiteurs de la kinase ret
WO2019143994A1 (fr) 2018-01-18 2019-07-25 Array Biopharma Inc. Composés de pyrazolyl[4,3-c]pyridine substitués utilisés en tant qu'inhibiteurs de la kinase ret
WO2020055672A1 (fr) 2018-09-10 2020-03-19 Array Biopharma Inc. Composés hétérocycliques condensés comme inhibiteurs de kinases ret
US10647730B2 (en) 2010-05-20 2020-05-12 Array Biopharma Inc. Macrocyclic compounds as TRK kinase inhibitors
US10966985B2 (en) 2017-03-16 2021-04-06 Array Biopharma Inc. Macrocyclic compounds as ROS1 kinase inhibitors
EP3746075A4 (fr) * 2018-01-29 2021-09-08 Merck Patent GmbH Inhibiteurs de gcn2 et leurs utilisations
US11512097B2 (en) 2019-11-25 2022-11-29 Amgen Inc. Heterocyclic compounds as Delta-5 desaturase inhibitors and methods of use
US11524963B2 (en) 2018-01-18 2022-12-13 Array Biopharma Inc. Substituted pyrazolo[3,4-d]pyrimidines as RET kinase inhibitors
EP3996736A4 (fr) * 2019-07-10 2023-05-24 Musc Foundation for Research Development Peptides d'endostatine pour le traitement de tumeurs, d'une fibrose et d'une lésion pulmonaire aiguë
WO2023154282A1 (fr) * 2022-02-08 2023-08-17 Theras, Inc. Composés ayant une structure t formée par au moins quatre cycles destinés à être utilisés dans le traitement du cancer et d'autres indications
RU2811408C2 (ru) * 2018-01-29 2024-01-11 Мерк Патент Гмбх Ингибиторы gcn2 и их применение

Families Citing this family (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
UY33227A (es) * 2010-02-19 2011-09-30 Novartis Ag Compuestos de pirrolopirimidina como inhibidores de la cdk4/6
US8710231B2 (en) * 2010-08-09 2014-04-29 Pharmadesign, Inc. Inhibitor of casein kinase 1delta and casein kinase 1E
US20150038500A1 (en) * 2011-11-25 2015-02-05 Catholic University Industry-Academic Cooperation Foundation Pharmaceutical composition for preventing and treating ophthalmic disorders
CN105906630B (zh) * 2015-04-06 2018-10-23 四川百利药业有限责任公司 用作fgfr抑制剂的n-(1h-吡唑-5-基)嘧啶并吡唑-4,6-二取代胺类化合物
CN106588893A (zh) * 2016-12-01 2017-04-26 北京万全德众医药生物技术有限公司 维拉佐酮双氧化物的制备
CN109836428A (zh) * 2019-02-27 2019-06-04 华东师范大学 具有免疫抑制活性的吡唑[4,3-d]嘧啶衍生物及用途
CN114751910B (zh) * 2022-05-17 2023-02-24 重庆文理学院 一种可诱导细胞巨泡式死亡的化合物及其制备方法和应用

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0591528A1 (fr) * 1991-04-22 1994-04-13 Otsuka Pharmaceutical Factory, Inc. DERIVE DE PYRAZOLO 1,5-a]PYRIMIDINE ET AGENT ANTI-INFLAMMATOIRECONTENANT CE DERIVE
EP0714898A1 (fr) * 1994-06-21 1996-06-05 Otsuka Pharmaceutical Factory, Inc. DERIVE DE PYRAZOLO [1,5-a]PYRIMIDINE
WO1998010590A1 (fr) * 1996-09-02 1998-03-12 Sony Corporation Dispositif et procede de transmission d'un signal video

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE3130633A1 (de) * 1981-08-01 1983-02-17 Basf Ag, 6700 Ludwigshafen 7-amino-azolo(1,5-a)pyrimidine und diese enthaltende fungizide
JP3163413B2 (ja) * 1994-06-21 2001-05-08 株式会社大塚製薬工場 鎮痛剤
ATE376548T1 (de) * 2002-09-04 2007-11-15 Schering Corp Pyrazolopyrimidine als hemmstoffe cyclin- abhängiger kinasen
JP2006500391A (ja) * 2002-09-04 2006-01-05 シェーリング コーポレイション サイクリン依存性キナーゼインヒビターとしてのピラゾロピリミジン

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0591528A1 (fr) * 1991-04-22 1994-04-13 Otsuka Pharmaceutical Factory, Inc. DERIVE DE PYRAZOLO 1,5-a]PYRIMIDINE ET AGENT ANTI-INFLAMMATOIRECONTENANT CE DERIVE
EP0714898A1 (fr) * 1994-06-21 1996-06-05 Otsuka Pharmaceutical Factory, Inc. DERIVE DE PYRAZOLO [1,5-a]PYRIMIDINE
WO1998010590A1 (fr) * 1996-09-02 1998-03-12 Sony Corporation Dispositif et procede de transmission d'un signal video

Cited By (70)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8586576B2 (en) 2002-09-04 2013-11-19 Merck Sharp & Dohme Corp. Substituted pyrazolo[1,5-a]pyrimidines as cyclin dependent kinase inhibitors
US7196078B2 (en) 2002-09-04 2007-03-27 Schering Corpoartion Trisubstituted and tetrasubstituted pyrazolopyrimidines as cyclin dependent kinase inhibitors
WO2007009773A1 (fr) * 2005-07-21 2007-01-25 Novartis Ag Derives de pyrazolo[1,5a]pyrimidin-7-yl-amine utilises en tant qu'inhibiteurs des proteines kinases
WO2007044441A2 (fr) * 2005-10-06 2007-04-19 Schering Corporation Methodes destinees a inhiber des proteines kinases
WO2007044441A3 (fr) * 2005-10-06 2007-07-26 Schering Corp Methodes destinees a inhiber des proteines kinases
US7776865B2 (en) 2005-10-06 2010-08-17 Schering Corporation Substituted pyrazolo[1,5-a]pyrimidines as protein kinase inhibitors
CN101360499A (zh) * 2005-10-06 2009-02-04 先灵公司 吡唑并[1,5-a]嘧啶衍生物在抑制蛋白激酶的方法中抑制蛋白激酶的用途
TWI421078B (zh) * 2005-10-06 2014-01-01 Merck Sharp & Dohme 關卡激酶抑制劑及其用途
JP2009511487A (ja) * 2005-10-06 2009-03-19 シェーリング コーポレイション サイクリン依存性キナーゼインヒビターとしての新規ピラゾロピリミジン
JP2009511486A (ja) * 2005-10-06 2009-03-19 シェーリング コーポレイション プロテインキナーゼを阻害するためのピラゾロ[1,5−a]ピリミジン誘導体の使用およびプロテインキナーゼを阻害する方法
JP2009511491A (ja) * 2005-10-06 2009-03-19 シェーリング コーポレイション プロテインキナーゼインヒビターとしてのピラゾロピリミジン
WO2007041712A1 (fr) 2005-10-06 2007-04-12 Schering Corporation Pyrazolopyrimidines en tant qu’inhibiteurs de protéines kinases
JP2012082234A (ja) * 2005-10-06 2012-04-26 Schering Corp プロテインキナーゼを阻害するためのピラゾロ[1,5−a]ピリミジン誘導体の使用およびプロテインキナーゼを阻害する方法
CN101360499B (zh) * 2005-10-06 2015-10-07 默沙东公司 吡唑并[1,5-a]嘧啶衍生物在制备抑制蛋白激酶的药物中的用途
EP3747441A1 (fr) 2005-12-23 2020-12-09 ARIAD Pharmaceuticals, Inc. Composants hétéroaryles bicycliques
EP3417912A1 (fr) 2005-12-23 2018-12-26 ARIAD Pharmaceuticals, Inc. Composants hétéroaromatiques bicycliques
WO2007103432A3 (fr) * 2006-03-08 2007-11-22 Novartis Ag Utilisation de derives de pyrazolo[1,5a]pyrimidin-7-yl amine dans le traitement de troubles neurologiques
WO2007103432A2 (fr) * 2006-03-08 2007-09-13 Novartis Ag Utilisation de derives de pyrazolo[1,5a]pyrimidin-7-yl amine dans le traitement de troubles neurologiques
WO2007109183A3 (fr) * 2006-03-20 2008-09-18 Novartis Ag Mutations et polymorphismes de la tyrosine kinase 1 liée au fms
WO2007109183A2 (fr) * 2006-03-20 2007-09-27 Novartis Ag Mutations et polymorphismes de la tyrosine kinase 1 liée au fms
WO2007113000A1 (fr) * 2006-04-04 2007-10-11 Novartis Ag N-(ARYL- OU HÉTÉROARYL)-PYRAZOLO[1,5-a]PYRIMIDINES SUBSTITUÉES EN POSITION 3 UTILISÉS EN TANT QU'INHIBITEURS DE KINASES
JP2009532400A (ja) * 2006-04-04 2009-09-10 ノバルティス アクチエンゲゼルシャフト キナーゼ阻害剤としての3−非置換N−(アリール−もしくはヘテロアリール)−ピラゾロ[1,5−a]ピリミジン
JP2009532401A (ja) * 2006-04-04 2009-09-10 ノバルティス アクチエンゲゼルシャフト キナーゼ阻害剤としての3−置換N−(アリール−またはヘテロアリール)−ピラゾロ[1,5−a]ピリミジン
WO2007112998A1 (fr) * 2006-04-04 2007-10-11 Novartis Ag N-(aryl- ou hétéroaryl)-pyrazolo-[1,5-a]pyrimidines 3-non substituées utilisées comme inhibiteurs de kinase
WO2008037459A1 (fr) * 2006-09-28 2008-04-03 Novartis Ag Dérivés pyrazolo[1,5-a]pyrimidine et leur utilisation thérapeutique
WO2008037477A1 (fr) 2006-09-29 2008-04-03 Novartis Ag PYRAZOLOPYRIMIDINES UTILISÉES COMME INHIBITEURS DES LIPIDES KINASES Pl3K
JP2010505839A (ja) * 2006-10-04 2010-02-25 シェーリング コーポレイション サイクリン依存性キナーゼインヒビターのピラゾロピリミジン
JP2010505838A (ja) * 2006-10-04 2010-02-25 シェーリング コーポレイション サイクリン依存性キナーゼインヒビターとしてのピラゾロピリミジン
EP1918291A1 (fr) * 2006-10-30 2008-05-07 Novartis AG Dérives de pyrazole fusionnes substitués de 3-aminocarbonyle comme modulateurs de protéine kinase
WO2008052964A1 (fr) * 2006-10-30 2008-05-08 Novartis Ag Dérivés pyrazolo fusionnés substitués par 3-aminocarbonyle comme modulateurs des protéines kinases
JP2010529123A (ja) * 2007-06-07 2010-08-26 シェーリング コーポレイション 置換−3−アミノピラゾールの合成
US8591943B2 (en) 2009-04-09 2013-11-26 Merck Sharp & Dohme Corp. Pyrazolo[1,5-a]pyrimidine derivatives as mTOR inhibitors
WO2011041152A1 (fr) 2009-09-30 2011-04-07 Schering Corporation Nouveaux composés inhibiteurs d'erk
US10647730B2 (en) 2010-05-20 2020-05-12 Array Biopharma Inc. Macrocyclic compounds as TRK kinase inhibitors
WO2012024586A1 (fr) 2010-08-19 2012-02-23 E. I. Du Pont De Nemours And Company Pyrazoles fongicides
WO2012036997A1 (fr) 2010-09-16 2012-03-22 Schering Corporation Dérivés condensés de pyrazole utilisés comme nouveaux inhibiteurs erk
US10023570B2 (en) 2015-07-16 2018-07-17 Array Biopharma Inc. Substituted pyrazolo[1,5-A]pyridine compounds as RET kinase inhibitors
US10174027B2 (en) 2015-07-16 2019-01-08 Array Biopharma Inc. Substituted pyrazolo[1,5-a]pyridine compounds as RET kinase inhibitors
US10138243B2 (en) 2015-07-16 2018-11-27 Array Biopharma Inc. Substituted pyrazolo[1,5-a]pyridine compounds as RET kinase inhibitors
US10174028B2 (en) 2015-07-16 2019-01-08 Array Biopharma Inc. Substituted pyrazolo[1,5-A]pyridine compounds as RET kinase inhibitors
US10555944B2 (en) 2016-10-10 2020-02-11 Eli Lilly And Company Substituted pyrazolo[1,5-A]pyridine compounds as RET kinase inhibitors
US10112942B2 (en) 2016-10-10 2018-10-30 Array Biopharma Inc. Substituted pyrazolo[1,5-A]pyridine compounds as RET kinase inhibitors
US10172851B2 (en) 2016-10-10 2019-01-08 Array Biopharma Inc. Substituted pyrazolo[1,5-A]pyridine compounds as RET kinase inhibitors
US10172845B2 (en) 2016-10-10 2019-01-08 Array Biopharma Inc. Substituted pyrazolo[1,5-A]pyridine compounds as RET kinase inhibitors
US10137124B2 (en) 2016-10-10 2018-11-27 Array Biopharma Inc. Substituted pyrazolo[1,5-a]pyridine compounds as RET kinase inhibitors
US10953005B1 (en) 2016-10-10 2021-03-23 Array Biopharma Inc. Substituted pyrazolo[1,5-a]pyridine compounds as RET kinase inhibitors
US10881652B2 (en) 2016-10-10 2021-01-05 Array Biopharma Inc. Substituted pyrazolo[1,5-A]pyridine compounds as RET kinase inhibitors
US11648243B2 (en) 2016-10-10 2023-05-16 Array Biopharma Inc. Substituted pyrazolo[1,5-A]pyridine compounds as RET kinase inhibitors
US10144734B2 (en) 2016-10-10 2018-12-04 Array Biopharma Inc. Substituted pyrazolo[1,5-A]pyridine compounds as RET kinase inhibitors
US10441581B2 (en) 2016-10-10 2019-10-15 Array Biopharma Inc. Substituted pyrazolo[1,5-A]pyridine compounds as RET kinase inhibitors
WO2018136661A1 (fr) 2017-01-18 2018-07-26 Andrews Steven W Composés de pyrazolo[1,5-a]pyrazine substitués utilisés en tant qu'inhibiteurs de la kinase ret
US11851434B2 (en) 2017-01-18 2023-12-26 Array Biopharma Inc. Substituted pyrazolo[1,5-A]pyrazine compounds as ret kinase inhibitors
WO2018136663A1 (fr) 2017-01-18 2018-07-26 Array Biopharma, Inc. Inhibiteurs de ret
US11168090B2 (en) 2017-01-18 2021-11-09 Array Biopharma Inc. Substituted pyrazolo[1,5-a]pyrazines as RET kinase inhibitors
US10966985B2 (en) 2017-03-16 2021-04-06 Array Biopharma Inc. Macrocyclic compounds as ROS1 kinase inhibitors
WO2019075114A1 (fr) 2017-10-10 2019-04-18 Mark Reynolds Formulations comprenant du 6-(2-hydroxy-2-méthylpropoxy)-4-(6-(6-((6-méthoxypyridin-3-yl)méthyl)-3,6-diazabicyclo[3.1.1]heptan-3-yl)pyridin-3-yl)pyrazolo[1,5-a]pyridine-3-carbonitrile
WO2019075108A1 (fr) 2017-10-10 2019-04-18 Metcalf Andrew T Formes cristallines
WO2019143994A1 (fr) 2018-01-18 2019-07-25 Array Biopharma Inc. Composés de pyrazolyl[4,3-c]pyridine substitués utilisés en tant qu'inhibiteurs de la kinase ret
US11472802B2 (en) 2018-01-18 2022-10-18 Array Biopharma Inc. Substituted pyrazolyl[4,3-c]pyridine compounds as RET kinase inhibitors
US11524963B2 (en) 2018-01-18 2022-12-13 Array Biopharma Inc. Substituted pyrazolo[3,4-d]pyrimidines as RET kinase inhibitors
US11603374B2 (en) 2018-01-18 2023-03-14 Array Biopharma Inc. Substituted pyrrolo[2,3-d]pyrimidines compounds as ret kinase inhibitors
WO2019143977A1 (fr) 2018-01-18 2019-07-25 Array Biopharma Inc. Composés de pyrrolo[2,3-d]pyrimidines substitués utilisés en tant qu'inhibiteurs de la kinase ret
EP3746075A4 (fr) * 2018-01-29 2021-09-08 Merck Patent GmbH Inhibiteurs de gcn2 et leurs utilisations
RU2811408C2 (ru) * 2018-01-29 2024-01-11 Мерк Патент Гмбх Ингибиторы gcn2 и их применение
WO2020055672A1 (fr) 2018-09-10 2020-03-19 Array Biopharma Inc. Composés hétérocycliques condensés comme inhibiteurs de kinases ret
US11964988B2 (en) 2018-09-10 2024-04-23 Array Biopharma Inc. Fused heterocyclic compounds as RET kinase inhibitors
EP3996736A4 (fr) * 2019-07-10 2023-05-24 Musc Foundation for Research Development Peptides d'endostatine pour le traitement de tumeurs, d'une fibrose et d'une lésion pulmonaire aiguë
US11512097B2 (en) 2019-11-25 2022-11-29 Amgen Inc. Heterocyclic compounds as Delta-5 desaturase inhibitors and methods of use
WO2023154282A1 (fr) * 2022-02-08 2023-08-17 Theras, Inc. Composés ayant une structure t formée par au moins quatre cycles destinés à être utilisés dans le traitement du cancer et d'autres indications
US11998545B2 (en) 2023-06-27 2024-06-04 Array Biopharma Inc. Substituted pyrazolo[1,5-a]pyridine compounds as RET kinase inhibitors

Also Published As

Publication number Publication date
IL176737A0 (en) 2006-10-31
JP2007519662A (ja) 2007-07-19
CN1909908A (zh) 2007-02-07
PE20051089A1 (es) 2006-01-25
BRPI0507071A (pt) 2007-06-19
KR20070009546A (ko) 2007-01-18
ECSP066718A (es) 2006-10-31
TW200528103A (en) 2005-09-01
TNSN06226A1 (en) 2007-12-03
CA2552885A1 (fr) 2005-08-04
AU2005205915B2 (en) 2009-05-21
AU2005205915A1 (en) 2005-08-04
RU2006130003A (ru) 2008-04-10
EP1708710A1 (fr) 2006-10-11
AR049769A1 (es) 2006-09-06
NO20063758L (no) 2006-10-23
MA28400B1 (fr) 2007-01-02

Similar Documents

Publication Publication Date Title
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
US20080234284A1 (en) Pyrazolo[1,5-a]Pyrimidin-7-Yl Amine 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
US20080275054A1 (en) 3-(Substituted Amino)-Pyrazolo[3, 4-D]Pyrimidines as Ephb and Vegfr2 Kinase Inhibitors
EP2004653B1 (fr) N-(aryl- ou hétéroaryl)-pyrazolo[1,5-a]pyrimidines substituées en position 3 utilisés en tant qu'inhibiteurs de kinases
CA2575316A1 (fr) Benzamides substitues trifluoromethyle comme inhibiteurs de kinase
US20090118277A1 (en) 3 Unsubstituted N-(aryl- or heteroaryl)-pyrazolo[1,5-a]pyrimidines as Kinase Inhibitors
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
AK Designated states

Kind code of ref document: A1

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

AL Designated countries for regional patents

Kind code of ref document: A1

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

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

Ref document number: 2005205915

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 548320

Country of ref document: NZ

WWE Wipo information: entry into national phase

Ref document number: 176737

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: 2552885

Country of ref document: CA

Ref document number: 12006501322

Country of ref document: PH

WWE Wipo information: entry into national phase

Ref document number: 2006/05735

Country of ref document: ZA

Ref document number: 200605735

Country of ref document: ZA

WWE Wipo information: entry into national phase

Ref document number: 06071058

Country of ref document: CO

ENP Entry into the national phase

Ref document number: 2005205915

Country of ref document: AU

Date of ref document: 20050121

Kind code of ref document: A

WWP Wipo information: published in national office

Ref document number: 2005205915

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: PA/a/2006/008303

Country of ref document: MX

Ref document number: 2006550056

Country of ref document: JP

Ref document number: 1020067014683

Country of ref document: KR

Ref document number: 2681/CHENP/2006

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: 200580003037.9

Country of ref document: CN

WWE Wipo information: entry into national phase

Ref document number: 2005706961

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2006130003

Country of ref document: RU

WWP Wipo information: published in national office

Ref document number: 2005706961

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 1020067014683

Country of ref document: KR

ENP Entry into the national phase

Ref document number: PI0507071

Country of ref document: BR