WO2008031594A1 - Benzoxazoles and oxazolopyridines being useful as janus kinases inhibitors - Google Patents

Benzoxazoles and oxazolopyridines being useful as janus kinases inhibitors Download PDF

Info

Publication number
WO2008031594A1
WO2008031594A1 PCT/EP2007/007983 EP2007007983W WO2008031594A1 WO 2008031594 A1 WO2008031594 A1 WO 2008031594A1 EP 2007007983 W EP2007007983 W EP 2007007983W WO 2008031594 A1 WO2008031594 A1 WO 2008031594A1
Authority
WO
WIPO (PCT)
Prior art keywords
phenyl
alkyl
benzooxazol
methyl
piperazin
Prior art date
Application number
PCT/EP2007/007983
Other languages
French (fr)
Inventor
Marc Gerspacher
Pascal Furet
Eric Vangrevelinghe
Original Assignee
Novartis Ag
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=37717673&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=WO2008031594(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Priority to BRPI0716841-1A2A priority Critical patent/BRPI0716841A2/en
Priority to US12/440,298 priority patent/US8629168B2/en
Priority to PL07802300T priority patent/PL2066647T3/en
Priority to ES07802300T priority patent/ES2377148T3/en
Priority to CN2007800339907A priority patent/CN101516860B/en
Priority to CA002660987A priority patent/CA2660987A1/en
Priority to AU2007296916A priority patent/AU2007296916B2/en
Application filed by Novartis Ag filed Critical Novartis Ag
Priority to JP2009527741A priority patent/JP5325105B2/en
Priority to MX2009002812A priority patent/MX2009002812A/en
Priority to EA200900388A priority patent/EA200900388A1/en
Priority to EP07802300A priority patent/EP2066647B1/en
Priority to AT07802300T priority patent/ATE532774T1/en
Publication of WO2008031594A1 publication Critical patent/WO2008031594A1/en
Priority to IL196860A priority patent/IL196860A0/en
Priority to TN2009000070A priority patent/TN2009000070A1/en
Priority to SM200900023T priority patent/SMP200900023B/en
Priority to NO20091469A priority patent/NO20091469L/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D263/00Heterocyclic compounds containing 1,3-oxazole or hydrogenated 1,3-oxazole rings
    • C07D263/52Heterocyclic compounds containing 1,3-oxazole or hydrogenated 1,3-oxazole rings condensed with carbocyclic rings or ring systems
    • C07D263/54Benzoxazoles; Hydrogenated benzoxazoles
    • C07D263/58Benzoxazoles; Hydrogenated benzoxazoles with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached in position 2
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D498/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D498/02Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D498/04Ortho-condensed systems
    • 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
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/04Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D498/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D498/02Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms in which the condensed system contains two hetero rings

Definitions

  • the invention relates to 2,7-disubstituted benzoxazole and 2,4-disubstituted-oxazolo[5,4- c]py ⁇ dine compounds of the formula I given below, as well as salts thereof, processes for the preparation thereof, the application thereof in a process for the treatment of the human or animal body, these compounds for use in the treatment (including prophylaxis) of the animal, especially human, body (especially with regard to a proliferative disease), the use thereof - alone or in combination with one or more other pharmaceutically active compounds - for the treatment especially of a protein tyrosine kinase mediated disease (such as a tumor disease) or for the manufacture of a pharmaceutical preparation for use in the treatment of such a disease, a method for the treatment of such a disease and a pharmaceutical preparation for the treatment of a disease as mentioned.
  • a protein tyrosine kinase mediated disease such as a tumor disease
  • JAKs Janus kinases
  • JAK2 activation by chromosome re-arrangements and/or loss of negative JAK/STAT (STAT signal transducing and activating factor(s)) pathway regulators has been observed in hematological malignancies as well as in certain solid tumors.
  • the compounds of formula I are suitable, for example, to be used in the treatment of diseases depending on the tyrosine kinase activity of JAK2 (and/or JAK3) kinase, especially proliferative diseases such as tumor diseases, leukaemias, polycythemia vera, essential thrombocythemia, and myelofibrosis with myeloid metaplasia.
  • compounds of the invention also have utility as immunosuppressive agents, for example for the treatment of diseases such as organ transplant rejection, lupus erythematodes, multiple sclerosis, rheumatoid arthritis, psoriasis, dermatitis, Crohn's disease, type-1 diabetes and complications from type-1 diabetes.
  • diseases such as organ transplant rejection, lupus erythematodes, multiple sclerosis, rheumatoid arthritis, psoriasis, dermatitis, Crohn's disease, type-1 diabetes and complications from type-1 diabetes.
  • the invention relates to compounds of the formula I,
  • X is N or CR 5 , wherein R 5 is halo, cyano, hydroxyl, d-Cy-alkoxy, C 1 -C 7 -alkyl, amino, N- mono- or N.N-di-d-Cy-alkyl or preferably hydrogen;
  • R 1 is unsubstituted or substituted aryl or is unsubstituted or substituted heterocyclyl; and R 2 is unsubstituted or substituted aryl or is unsubstituted or substituted heterocyclyl which is bound via a ring carbon atom (to the carbon at position 7 of the benzoxazole ring in formula I or (if X is N) to the carbon at position 4 in the oxazolopyridine ring of formula I); R 3 is cyano, hydroxyl, C 1 -C 7 -SIkVi, amino, N-mono- or N.N-di-d-C T' -alkyl or preferably hydrogen; and
  • R 4 is hydroxyl, amino or preferably hydrogen; or salts thereof.
  • the invention relates to a compound of the formula I wherein X is CR 5 or N, wherein R 5 is halo, cyano, hydroxyl, C 1 -C 7 ⁇ IkOXy, Ci-C 7 -alkyl, amino, N- mono- or N,N-di-CrC 7 -alkyl or preferably hydrogen;
  • R 1 is unsubstituted or substituted aryl or is unsubstituted or substituted heterocyclyl, especially R 1 is phenyl, naphthyl, indanyl, pyridyl, oxo-1H-pyridyl, indolyl, dihydroindolyl or oxo-dihydroindolyl, each of which is bound via a ring carbon atom and is unsubstituted or substituted by one to three moieties independently selected from the group consisting of C 1 - C 7 -alkyl, amino-d-d-alkyl, halo-d-C 7 -alkyl, N-d-d-alkanoylamino-d-d-alkyl, N-C 1 -C 7 - alkanesulfonyl-amino-d-C 7 -alkyl, pyrrolidino-d-C 7 -alkyl, oxo-
  • R 3 is cyano, hydroxyl, d-C 7 -alkyl, amino, N-mono- or N.N-di-d-C ⁇ alkyl or preferably hydrogen;
  • R 4 is hydroxyl, amino or preferably hydrogen
  • the invention relates to a compound of the formula I wherein X is CR 5 or N, wherein R 5 is halo, cyano, hydroxyl, C 1 -C ⁇ aIkOXy, C 1 -C 7 -alkyl, amino, N- mono- or N.N-di-d-Cr-alkyl or preferably hydrogen;
  • R 1 is unsubstituted or substituted aryl or is unsubstituted or substituted heterocyclyl
  • R 2 is phenyl, naphthyl, pyridyl, pyrazolyl, thiophenyl, quinolinyl, isoquinolinyl or fH-benzo- imidazolyl, each of which is bound via a ring carbon atom and is unsubstituted or substituted by one to three moieties independently selected from the group consisting of d-C 7 -alkyl, amino-d-C 7 -alkyl, N-d-C T -alkanoylamino-d-d-alkyl, N-d-C 7 -alkanesulfonyl-amino-d-C 7 - alkyl, carbamoyl-d-C 7 -alkyl, [N-mono- or N,N-di-(C 1 -C 7 -alkyl)
  • R 4 is hydroxyl, amino or preferably hydrogen
  • the invention relates to a compound of the formula I wherein
  • X is CR 5 or N, wherein R 5 is halo, cyano, hydroxyl, d-C 7 -alkoxy, d-C 7 -alkyl, amino, N- mono- or N,N-di-d-C 7 -alkyl or preferably hydrogen;
  • R 1 is unsubstituted or substituted aryl or is unsubstituted or substituted heterocyclyl
  • R 2 is phenyl, naphthyl, pyridyl, pyrazolyl, thiophenyl, quinolinyl, isoquinolinyl or 7H-benzo- imidazolyl, each of which is bound via a ring carbon atom and is unsubstituted or substituted by one to three moieties independently selected from the group consisting of d-C ⁇ alkyl, amino-d-d-alkyl, N-C 1 -C 7 -alkanoylamino-C 1 -C 7 -alkyl, N-d-d-alkanesulfonyl-amino-d-d- alkyl, carbamoyl-d-C ⁇ alkyl, [N-mono- or N.N-dHd-d-alkyO-carbamoyrj-d-d-alkyl, C 1 -C 7 - alkanesulfinyl-C,-C 7 -alky
  • R 3 is cyano, hydroxyl, d-C ⁇ alkyl, amino, N-mono- or N,N-di-d-C 7 -alkyl or preferably hydrogen;
  • R 4 is hydroxyl, amino or preferably hydrogen
  • the invention relates very especially to a compound of the formula I, wherein X is CH or N;
  • R 1 is phenyl, (especially 3,4,5-)trimethoxyphenyr, (especially 3,4- or 3,5-)dimethoxyphenyl * , (especially 4-)morpholinophenyl, (especially 4-) N-(2-methoxyethyl)-carbamoylphenyr, or (especially 4-)N,N-(2-dimethylamino-ethyl)-carbamoylphenyr, (especially 4-)dimethylamino- carbonyl-(especially 3-)methyl-phenyl*, (especially 4-)-(preferably 4-)-(2-methoxy-ethyl- piperazin-(especially 1 -)yl-(especially 3-)-methyl-phenyl, (especially 4-)pyrrolidin-1 -carbonyl- (especially 3-)methyl-phenyl * , (especially 3-)methyl-(especially 4-)-4-methylpiperazin-1- carbonyl-phenyl, (especially 3- or 4-)4-methyl-piperazin-1-yl
  • R 2 is phenyl, (especially 4-)methylphenyl, (especially 3-)methylphenyl, (especially 2-)me- thylphenyl, (especially 4-)-hydroxymethyl-phenyl, (especially 4-)aminomethyl-phenyl, (especially 3-)aminomethyl-phenyl, (especially 4-)acetylaminomethyl-phenyl * , (especially 4- )methanesulfonylaminomethyl-phenyl, (especially 3-)acetylaminomethyl-phenyl, (especially 3-)methanesulfonylaminomethyl-phenyl*, (especially 4-)methanesulfonylaminomethyl-phenyl, (especially 4-)(N-methylcarbamoyl)-methylphenyr, (especially 4-)methanesulfinylmethyl- phenyl, (especially 4-)methanesulfonylmethylphenyl, (especially 3-)chlorophenyl, (especially 3-)hydroxypheny
  • each of R 3 and R 4 is hydrogen; or a pharmaceutically acceptable salt thereof.
  • the invention relates especially also to a compound of the formula I, wherein X is CH or N;
  • R 1 is (especially 3,4,5- )trimethoxyphenyl, (especially 3,4- or 3,5-)dimethoxyphenyl, (especially 4-)morpholinophenyl, (especially 4-) N-(2-methoxyethyl)-carbamoylphenyl, or (especially 4-)N,N-(2-dimethylamino-ethyl)-carbamoylphenyl,
  • R 2 is phenyl, (especially 4-)methylphenyl, (especially 3-)methylphenyl, (especially 2-)me- thylphenyl, (especially 4-)aminomethyl-phenyl, (especially 3-)aminomethyl-phenyl, (especially 4-)acetylaminomethyl-phenyl * , (especially 4-)methanesulfonylaminomethyl- phenyl, (especially 3-)acetylaminomethyl-phenyl, (especially 3-)methanesulfonylamino- methyl-phenyl, (especially 4-)methanesulfonylaminomethyl-phenyl, (especially 4-)(N- methylcarbamoyl)-methylphenyl, (especially 4-)methanesulfinylmethylphenyl, (especially 4- )methanesulfonylmethylphenyl, (especially 3-)chlorophenyl, (especially 3-)hydroxyphenyl, (especially 4-)methoxypheny
  • the invention relates especially to the compounds of the formula I given in the Examples, as well as the methods of manufacture described therein.
  • Any asymmetric carbon atoms 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 as pure isomers, preferably as enantiomer-pure diastereomers.
  • the invention also relates to tautomers where tautomeric forms are possible.
  • CrCy-alkyl is preferably alkyl with from and including 1 up to and including 7, preferably from and including 1 to and including 4, and is linear or branched; preferably, lower alkyl is butyl, such as n-butyl, sec-butyl, isobutyl, tert-butyl, propyl, such as n-propyl or isopropyl, ethyl or preferably methyl.
  • Halogen is especially fluorine, chlorine, bromine, or iodine, especially fluorine, chlorine, or bromine.
  • That a heterocyclyl is bound via a ring carbon atom means that it is not bound via a nitrogen atom to the rest of the molecule in formula I (that is, to the 7-position of the central benzoxazole ring or if X is N to the 4-position of the central oxazolopyridine ring).
  • aryl is preferably an aromatic moiety with 6 to 14 ring carbon atoms, more preferably with 6 to 10 ring carbon atoms, such as phenyl or naphthyl, which is unsubstituted or substituted by one or more, preferably up to three, more preferably up to two substituents independently selected from the group consisting of unsubstituted or substituted heterocyclyl as described below, especially pyrrolidinyl, such as pyrrolidino, oxopyrrolidinyl, such as oxopyrrolidino, C 1 -C 7 -alkyl-pyrrolidinyl, 2,5-di-(C 1 -C 7 alkyl)pyrrolidinyl, such as 2,5-di-(C 1 -C 7 alkyl)-pyrrolidino, tetrahydrofuranyl, thiophenyl, C 1 -C 7 -alkyl
  • halo especially fluoro, chloro, bromo or iodo, halo- lower alkyl, such as trifluoromethyl, hydroxy, lower alkoxy, azido, amino, N-mono- or N,N-di- (lower alkyl and/or d-d-alkanoy ⁇ -amino-, nitro, carboxy, lower-alkoxycarbonyl, carbamoyl, cyano and/or sulfamoyl.
  • substituents independently selected from halo, especially fluoro, chloro, bromo or iodo, halo- lower alkyl, such as trifluoromethyl, hydroxy, lower alkoxy, azido, amino, N-mono- or N,N-di- (lower alkyl and/or d-d-alkanoy ⁇ -amino-, nitro, carboxy, lower-alkoxycarbonyl, carbamoyl, cyano and/or s
  • the substituents of substituted aryl are up to three substituents independently selected from the group consisting of d-C 7 -alkyl, hydroxyl-d-C 7 -alkyl, C 1 -C 7 - alkoxy-d-C 7 -alkyl, amino-d-C 7 -alkyl, halo-d-C 7 -alkyl, N-C 1 -C 7 -alkanoylamino-C 1 -C 7 -alkyl, N-C 1 -C 7 -alkanesulfonyl-amino-C 1 -C 7 -alkyl, pyrrolidino-d-C 7 -alkyl, oxo- pyrrcHidino-d-Cr- alkyl, piperidino-CrC 7 -alkyl, piperazin-1-yl-d-C ⁇ -alkyl, 4-(C 1 -C 7 -alkyl,
  • unsubstituted or substituted aryl is preferably phenyl, naphthyl or indanyl, each of which is unsubstituted or substituted by one to three moieties independently selected from the group consisting of d-C ⁇ alky!, ammo-d-d-alky!, halo-C r C 7 -alkyl, N-C 1 -C 7 -alkanoylamino-C 1 -C 7 -alkyl, N-C 1 -C 7 -alkanesulfonyl-amino-C 1 -C 7 - alkyl, pyrrolidino-C 1 -C 7 -alkyl, oxo- pyrrolidino-d-C ⁇ alkyl, piperidino-d-C ⁇ alkyl, piperazin-1- yl-d-C 7 -alkyl, 4-(C 1 -C 7 -alkyl,
  • R 2 is preferably phenyl which is unsubstituted or substituted by one to three moieties independently selected from the group consisting of d-d-alkyl, hydroxyl-C 1 -C 7 -alkyl, amino-d-C 7 -alkyl, halo-d-C 7 -alkyl, N-C 1 -C 7 - alkanoylamino-d-d-alkyl, N-d-d-alkanesulfonyl-amino-d-d-alkyl, d-C 7 -alkanesulfonyl- d-C ⁇ alkyl, d-d-alkenesulfinyl-d-C T -alkyl, pyrrolidino-C ⁇ C T -alkyl, OXO-Py ⁇ oIJdJnO-C 1 -C 7 - alkyl,
  • unsubstituted or substituted heterocyclyl is preferably pyrrolyl, oxo-pyrrolyl, 2,3-dihydroindolyl, 2-oxo-2,3-dihydroindolyl or 1H-pyridin-2-onyl, each of which is unsubstituted or substituted by one to three substituents independently selected from those mentioned above for unsubstituted or substituted aryl R 1 .
  • unsubstituted or substituted heterocyclyl is preferably pyridyl, pyrazolyl, thiophenyl, quinolinyl, isoquinolinyl or fH-benzoimidazofyl, each of which is unsubstituted or substituted by one to three moieties independently selected from those mentioned above as substituents for aryl R 2 , or especially from the group consisting of d-C ⁇ -alkyl, amino-d-d- alkyl, N-d-Cr-alkanoylamino-d-Cr-alkyl, N-d-C T -alkanesulfonyl-amino-d-C T -alkyl, carba- moyl-d-Cy-alkyl, [N-mono- or N,N-di-(C 1 -C 7 -alkyl)-carbamoyl]-C 1 -C 7 -alkyl
  • X is preferably CH (especially for JAK2 inhibitors of the formula I) or preferably N (especially for JAK3 inhibitors of the formula I).
  • R 2 preferably not more than one substituent (if a substituent is present at all) is present in ortho-position and in meta position. That is, the substituent or substituents is or are present preferably in para-position and not more than one is present in ortho- and meta-position.
  • Treatment includes both prophylactic and therapeutic treatment.
  • Protein tyrosine kinase (especially JAK2 and/or JAK3 kinase) mediated diseases are especially such disorders that respond in a beneficial way (e.g. amelioration of one or more symptoms, delay of the onset of a disease, up to temporary or complete cure from a disease) to the inhibition of a protein tyrosine kinase, especially inhibition of a JAK (preferably JAK2 and/or JAK3) kinase or TYK2, more especially inhibition of JAK2 kinase (where among the diseases to be treated, especially proliferative diseases such as tumor diseases, leukaemias, polycythemia vera, essential thrombocythemia, and myelofibrosis with myeloid metaplasia may be mentioned) and/or of JAK3 kinase (where preferably the treatment (e.g.
  • Salts are preferably pharmaceutically acceptable salts.
  • 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, e.g., carboxylic acids or sulfonic acids, such as fumaric acid or methansulfonic acid.
  • salts for isolation or purification purposes it is also possible to use pharmaceutically unacceptable salts, for example picrates or perchlorates.
  • pharmaceutically acceptable salts or free compounds are employed (where applicable in the form of pharmaceutical preparations), and these are therefore preferred.
  • any reference to the free compounds hereinbefore and hereinafter is to be understood as referring also to the corresponding salts, as appropriate and expedient.
  • the compounds of formula I thereof have valuable pharmacological properties, as described hereinbefore and hereinafter.
  • Baculovirus including the amino acid domain ASP751-VAL1129 of the JAK2 protein is obtainable by ProQinase, Freiburg, Germany.
  • the virus is scaled up as following: 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 5 x 10 7 ceHs/plate and infected with 1 m!_ 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 re-suspended in 50 mL of ice-cold lysis buffer (25mMTris-HCI, pH7.5, 2mMEDTA, 1%NP-40, 1mM DTT, 1mMP MSF).
  • the cells are stirred on ice for 15 min and then centrifuged at 5000 rpms for 20 min.
  • the protein is purified by loading the centrifuged ceH lysate 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 (1 mL each) of 25 mM Tris-HCI, pH 7.5, 10 mM reduced-glutathione, 100 mM NaCI, 1 mM DTT, 10 % Glycerol and stored at -70 0 C.
  • JAK2 The activity of JAK2 is assayed in the presence or absence of inhibitor measuring the incorporation of 33P from [ ⁇ 33P]ATP into appropriate substrates [Garcia-Echeverria C, Pearson MA, Marti A, et al (2004) In vivo antitumor activity of NVP-AEW541 - A novel, potent, and selective inhibitor of the IGF-IR kinase. Cancer Cell; 5: 231 -2391.
  • the test compound is dissolved in DMSO (10 mM) and stored at -20°C. Serial dilutions are freshly made in DMSO and are 1000 times concentrated than test solutions ("pre-dilution plates"). They are further diluted with pure water to yield "master plates” containing 3 times concentrated test solutions in 3% DMSO.
  • the final volume of the assay is 30 ⁇ L containing 10 ⁇ l_ of test solution (1% DMSO), 10 ⁇ l_ assay mix including the assay components descry- bed by Garcia-Echeverria (2004) and in the following section as well as 10 ⁇ L enzyme.
  • the pipetting steps can be programmed to be performed either on the MultiPROBE lix, MultiPROBE MLx or HamiltonSTAR robots in the 96 well format.
  • the protein kinase assays are carried as described in details by Garcia-Echeverria (see above).
  • the assay for JAK2 is carried out in 96-well plates at ambient temperature for 10 min (filter-biding method) or 30 min (flash plates) in a finial volume of 30 ⁇ l_ including the following components: 300 ng of GST-JAK2, 20 mM Tris-HCI, pH 7.5, 1.0 mM MnCI 2 , 10 mM MgCI 2 , 1 mM DTT, 3 ⁇ g/mL poiy(Glu,Tyr) 4:1 , 1 % DMSO and 1.0 ⁇ M ATP ( ⁇ -[ 33 PJ-ATP 0.1 ⁇ Ci); The assays are terminated by the addition of 20 ⁇ l of 125 mM EDTA.
  • the capturing of the phosphorylated- peptides by the filter-binding method is performed as following: 40 ⁇ l_ of the reaction mixture are transferred onto Immobilon-PVDF membranes 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 . Free membranes are removed and washed 4 x on a shaker with 1.0 % H3PO4, once with ethanol. Membranes are counted after drying at ambient temperature, mounting in Packard TopCount 96-well frame, and addition of 10 ⁇ l/well of Microscint. The plates are eventually sealed and counted in a microplate scintillation counter (TopCount NXT, TopCount NXT HTS, PerkinElmer, Brussels, Belgium).
  • the assays for the flash plate method is carried out in a total volume of 30 ⁇ l_ at RT in conventional 96-well flash plates. The reaction is stopped after 30 min by the addition of 20 ⁇ L of 125 mM EDTA The assay plates are then washed three times with PBS and dried at room temperature. The plates are sealed and counted in a microplate scintillation counter (TopCount NXT, TopCount NXT HTS). IC50 values are calculated by linear regression analysis of the percentage inhibition of the compound either in duplicate, at four concentrations (usually 0.01 , 0.1 , 1 and 10 ⁇ M) or as 8 single point IC 50 starting at 10 ⁇ M following by 1 :3 dilutions. With compounds according to the invention, IC 50 values in the range from 5 nM to 5 ⁇ M can be found with compounds of the formula I.
  • JAK-2 or JAK-3 enzymatic activity is determined using a time-resolved fluorescence energy transfer technology.
  • the phosphorylation of a synthetic biotinylated peptide substrate (GGEEEEYFELVKKKK, SEQ ID NO: 3)) by either JAK-2 or JAK-3 in the presence of ATP is quantified using Europium labeled anti phosphotyrosine antibody and Streptavidin- Allophycocyanin
  • Both JAK-2 and JAK-3 enzymes used in these assays contain the kinase domain (JH-1 domain) of the full length proteins and are used as glutathione S-transferase (GST) fusion proteins.
  • Inhibitors are dissolved in dimethylsulfoxide (DMSO). Dilutions are prepared in 90% DMSO followed by additional dilutions steps as required to perform a 8-point concentration- response.
  • DMSO dimethylsulfoxide
  • the reaction mix consists of 5 ⁇ L of diluted compound, 10 ⁇ l_ of assay buffer and 5 ⁇ l_ of enzyme dilution. After incubation for 60 minutes at room temperature the reaction is stopped by the addition of EDTA. For detection of the product anti-phosphotyrosine antibody and Streptavidin-APC are added and after 60 minutes the samples are measured in an EnVision 2102 Multilabel Reader (Perkin Elmer, Inc., Wellesley, MA, USA, in the following mentioned as "PerkinElmer”) with excitation wavelength of 320nm and emission at 665nm.
  • the kinase assays are performed as described in details by Garcia-Echeverria et al [(2004), Cancer Cell; 5: 231-239] in 96-well plates at ambient temperature for 10 min (filter-biding method) or 30 min (flash plates) in a final volume of 30 ⁇ L including the following components: GST-JAK-2 or GST-JAK-3, 20 mM Tris-HCI, pH 7.5, 0-1.0 mM MnCI 2 , 1-10 mM MgCI 2 , 1 mM dithiothreitol (DTT), 3 ⁇ g/mL poly(Glu.Tyr) 4:1, 1 % DMSO and 1.0 ⁇ M ATP ( ⁇ -[ 33 P]-ATP 0.1 ⁇ Ci); The assays are terminated by the addition of 20 ⁇ l of 125 mM ethylendiamine tetraacetate (EDTA).
  • EDTA ethylendiamine tetraacetate
  • the capturing of the phosphorylated peptides by the filter-binding method is performed as following: 40 ⁇ L of the reaction mixture are transferred onto Immobilon-PVDF membranes 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 . Free 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 (now PerkinElmer), and addition of 10 ⁇ l/well of Microscint (PerkinElmer). The plates are eventually sealed and counted in a microplate scintillation counter (TopCount NXT, TopCount NXT HTS, PerkinElmer, Brussels, Belgium).
  • the compounds of the invention have IC 50 values of from ca. 0.1 -1000 nM.
  • JAK-2 and JAK-3 assays filter binding/flash plate kinase assays
  • Enzyme activities are measured by mixing 10 ⁇ L of a 3-fold concentrated compound solution with 10 ⁇ L of the corresponding substrate mixture (peptidic substrate,
  • FB in a finial volume of 30 ⁇ L including the following components:
  • JAK-2 200 ng GST-JAK-2, 20 mM Tris-HCI, pH 7.5, 10 mM MgCI 2 , 1.0 mM MnCI 2 , 1 mM
  • JAK-3 15 ng GST-JAK-3, 20 mM Tris-HCI, pH 7.5, 10 mM MgCI 2 , 1 mM DTT, 1% DMSO,
  • Filter binding method The capturing of the phosphorylated peptides by the FB method is performed as following: 40 ⁇ L of the stopped reaction mixture were transferred onto Immobilon-PVDF (Millipore, Eschbom, Germany) membranes previously soaked for 5 min with methanol, rinsed with water, soaked for 5 min with 0.5% H 3 PO 4 and mounted on vacuum manifold with disconnected vacuum source. After spotting, vacuum is connected and each well rinsed with 200 ⁇ L 0.5% H 3 PO 4 . Free membranes are removed and washed 4 times on a shaker with 1% H 3 PO 4 and once with ethanol.
  • Immobilon-PVDF Micropore, Eschbom, Germany
  • Membranes are counted after drying, mounting in Packard TopCount 96-well frame, and addition of 10 ⁇ L/well of MicroscintTM. The plates are eventually sealed and counted in a microplate scintillation counter (TopCount NXT, TopCount NXT HTS).
  • Flash plate method For the capturing of the phosphorylated substrates (60 min, RT), 96-well standard FPs (i.e. polystyrene microplates in which the interior of each well is permanently coated with a thin layer of polystyrene-based scintillant) are used. The assay plates are then washed three times with PBS and dried at room temperature. The plates are sealed and counted in a microplate scintillation counter (TopCount NXT, TopCount NXT HTS).
  • a microplate scintillation counter TopCount NXT, TopCount NXT HTS
  • IC 50 1 S A 4 Parameter logistic equation is used to calculate IC 50 values (IDBS XLfit) of the percentage inhibition of each compound at 8 concentrations (usually 10, 3.0, 1.0, 0.3, 0.1 , 0.030,0.010 and 0.003 ⁇ M).
  • test system as described in Example 186 is used.
  • IC 50 values in the range from 0.1 nM to 10 ⁇ M, e.g. from less than 3 nM to 5 ⁇ M, most preferably from 0.1 nM to 1000 nM can be found in the above-mentioned test systems.
  • the assay can be performed as described by G. Wernig, T. Mercher, R. Okabe, R.L. Levine, B. H. Lee, D.G. Gilliland-, Blood First Edition paper, published online February 14, 2006; DOI 10, 1182/blood-2005-12-4824.
  • a compound of formula I according to the invention shows therapeutic efficacy especially against disorders dependent on protein kinase, especially proliferative diseases mediated JAK2 kinase activity.
  • protein kinases can be inhibited by compounds of the present invention, such as Tyk 2, c-src, Flt-3, KDR and others, for each of which test systems are known in the art.
  • the dosage of the active ingredient to be applied to a warm-blooded animal depends upon a variety of factors including type, species, age, weight, sex and medical condition of the patient; the severity of the condition to be treated; the route of administration; the renal and hepatic function of the patient; and the particular compound employed.
  • a physician, clinician or veterinarian of ordinary skill can readily determine and prescribe the effective amount of the drug required to prevent, counter or arrest the progress of the condition.
  • Optimal precision in achieving concentration of drug within the range that yields efficacy without toxicity requires a regimen based on the kinetics of the drug's availability to target sites. This involves a consideration of the distribution, equilibrium, and elimination of a drug.
  • the dose of a compound of the formula I or a pharmaceutically acceptable salt thereof to be administered to warmblooded animals, for example humans of approximately 70 kg body weight, is preferably from approximately 3 mg to approximately 5 g, more preferably from approximately 10 mg to approximately 1.5 g per person per day, divided preferably into 1 to 3 single doses which may, for example, be of the same size. Usually, children receive half of the adult dose.
  • a compound of formula I can be administered alone or in combination with one or more other therapeutic agents, possible combination therapy taking the form of fixed combinations or the administration ⁇ f a compound of the invention and one or more other therapeutic agents being staggered or given independently of one another, or the combined administration of fixed combinations and one or more other therapeutic agents.
  • a compound of formula I can besides or in addition be administered especially for tumor therapy in combination with chemotherapy, radiotherapy, immunotherapy, surgical intervention, or a combination of these. Long-term therapy is equally possible as is adjuvant therapy in the context of other treatment strategies, as described above. Other possible treatments are therapy to maintain the patient's status after tumor regression, or even chemopreventive therapy, for example in patients at risk.
  • a compound of the formula I may be used to advantage in combination with other antiproliferative compounds.
  • antiproliferative compounds include, but are not limited to aromatase inhibitors; antiestrogens; topoisomerase I inhibitors; topoisomerase Il inhibitors; microtubule active compounds; alkylating compounds; histone deacetylase inhibitors; compounds which induce cell differentiation processes; cyclooxygenase inhibitors; MMP inhibit- tors; 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; bisphospho- nates; biological response modifiers; antiproliferative antibodies; heparanase inhibitors; inhibitors of Ras oncogenic isoform
  • tumor treatment approaches including surgery, ionizing radiation, photo- dynamic therapy, implants, e.g. with corticosteroids, hormones, or they may be used as radiosensitizers.
  • implants e.g. with corticosteroids, hormones, or they may be used as radiosensitizers.
  • anti-inflammatory and/or antiproliferative treatment combination with anti-inflammatory drugs is included. Combination is also possible with antihistamine drug substances, bronchodilatatory drugs, NSAID or antagonists of chemokine receptors.
  • 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 atame- stane, exemestane and formestane and, in particular, non-steroids, especially aminogluteth- imide, roglethimide, pyridoglutethimide, trilostane, testolactone, ketokonazole, vorozole, fadro- zole, 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, ful- vestrant, 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.
  • bicalutamide CASODEX
  • gonadorelin agonist as used herein includes, but is not limited to abarelix, goserelin and goserelin acetate. Goserelin is disclosed in US 4,100,274 and can be administered, e.g., in the form as it is marketed, e.g. under the trademark ZOLADEX.
  • Abarelix can be formulated, e.g. as disclosed in US 5,843,901.
  • topoisomerase I inhibitor includes, but is not limited- to topotecan, gimatecan, irinotecan, camptothecian and its analogues, 9-nitrocamptothecin and the macromolecular camptothecin conjugate PNU-166148 (compound A1 in WO99/ 17804).
  • Irinotecan can be administered, e.g. in the form as it is marketed, e.g. under the trademark CAMPTOSAR.
  • Topotecan can be administered, e.g., in the form as it is marketed, e.g. under the trademark HYCAMTIN.
  • topoisomerase Il inhibitor includes, but is not limited to the an- thracyclines such as doxorubicin (including liposomal formulation, e.g. CAELYX), daunorubicin, epirubicin, idarubicin and nemorubicin, the anthraquinones mitoxantrone and losoxantrone, 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 compound relates to microtubule stabilizing, microtubule destabilizing compounds 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 compound includes, but is not limited to, cyclophosphamide, ifosfamide, melphalan or nitrosourea (BCNU or Gliadel).
  • Cyclophosphamide can be administered, e.g., in the form as it is marketed, e.g. under the trademark CYCLOSTIN.
  • Ifosfamide can be administered, e.g., in the form as it is marketed, e.g. under the trademark HOLOXAN.
  • histone deacetylase inhibitors or "HDAC inhibitors” relates to compounds which inhibit the histone deacetylase and which possess antiproliferative activity. This includes compounds disclosed in WO 02/22577, especially N-hydroxy-3-[4-[[(2-hydroxyethyl)[2-(1H-indol- 3-yl)ethyl]-amino]methyl]phenyl]-2E-2-propenamide, N-hydroxy-3-[4-[[[2-(2-methyl-1H-indol-3- yl)-ethyl]-amino]methyl]phenyl]-2£-2-propenamide and pharmaceutically acceptable salts thereof.
  • SAHA Suberoylanilide hydroxamic acid
  • antimetabolite includes, but is not limited to, 5-Fluorouracil or 5-FU, capecitabine, gemcitabine, DNA demethylating compounds, 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..
  • 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 includes, but is not limited to, protein tyrosine kinase and/or serine and/or threonine kinase inhibitors or lipid kinase inhibitors, e.g., a) compounds targeting, decreasing or inhibiting the activity of the platelet-derived growth factor-receptors (PDGFR), such as compounds which target, decrease or inhibit the activity of PDGFR, especially compounds which inhibit the PDGF receptor.
  • PDGFR platelet-derived growth factor-receptors
  • a N-phenyl-2-pyrimidine-amine derivative e.g. imatinib, SU101 , SU6668 and GFB-111 ; b) compounds targeting, decreasing or inhibiting the activity of the fibroblast growth factor-receptors (FGFR); c) compounds targeting, decreasing or inhibiting the activity of the insulin-like growth factor receptor I (IGF-IR), such as compounds which target, decrease or inhibit the activity of IGF-IR, especially compounds which inhibit the kinase activity of IGF-I receptor, such as those compounds disclosed in WO 02/092599, or antibodies that target the extracellular domain of IGF-I receptor or its growth factors; d) compounds targeting, decreasing or inhibiting the activity of the Trk receptor tyrosine kinase family, or ephrin B4 inhibitors; e) compounds targeting, decreasing or inhibiting the activity of the AxI receptor tyrosine kinase family; f) compounds targeting, decreasing or inhibiting the activity of the Ret receptor
  • imatinib compounds targeting, decreasing or inhibiting the activity of the C-kit receptor tyrosine kinases - (part of the PDGFR family), such as compounds which target, decrease or inhibit the activity of the c-Kit receptor tyrosine kinase family, especially compounds which inhibit the c-Kit receptor, e.g. imatinib; i) compounds targeting, decreasing or inhibiting the activity of members of the c-Abl family, their gene-fusion products (e.g. BCR-AbI kinase) and mutants, such as compounds which target decrease or inhibit the activity of c-Abl family members and their gene fusion products, e.g.
  • N-phenyl-2-pyrimidine-amine derivative e.g. imatinib or nilotinib (AMN107); PD180970; AG957; NSC 680410; PD173955 from ParkeDavis; or dasatinib (BMS-354825) j) compounds targeting, decreasing or inhibiting the activity of members of the protein kinase C (PKC) and Raf family of serine/threonine kinases, members of the MEK.
  • PDC protein kinase C
  • Raf family of serine/threonine kinases members of the MEK.
  • UCN-01 safingol, BAY 43-9006, Bryostatin 1 , Perifosine; llmofosine; RO 318220 and RO 320432; GO 6976; lsis 3521 ; LY333531/LY379196; isochinoline compounds such as those disclosed in WO 00/09495; FTIs; PD184352 or QAN697 (a P13K inhibitor) or AT7519 (CDK inhibitor); k) compounds targeting, decreasing or inhibiting the activity of protein-tyrosine kinase inhibitors, such as compounds which target, decrease or inhibit the activity of protein-tyrosine kinase inhibitors include imatinib mesylate (GLEEVEC) or tyrphostin.
  • GLEEVEC imatinib mesylate
  • tyrphostin 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; Tyrphostirv 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);
  • compounds targeting, decreasing or inhibiting the activity of the epidermal growth factor family of receptor tyrosine kinases (EGFR, ErbB2, ErbB3, ErbB4 as homo- or heterodimers) and their mutants, such as compounds which target, decrease or inhibit the activity of the epidermal growth factor receptor family are especially compounds, proteins or antibodies which inhibit members of the EGF receptor tyrosine kinase family, e.g. EGF receptor, ErbB2, ErbB3 and ErbB4 or bind to EGF or EGF related ligands, and are in particular those compounds, proteins or monoclonal antibodies generically and specifically disclosed in WO 97/02266, e.g. the compound of ex.
  • trastuzumab HerceptinTM
  • cetuximab ErbituxTM
  • Iressa Tarceva
  • OSI-774 Cl- 1033
  • EKB-569 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 ; and m) compounds targeting, decreasing or inhibiting the activity of the c-Met receptor, such as compounds which target, decrease or inhibit the activity of c-Met, especially compounds which inhibit the kinase activity of c-Met receptor, or antibodies that target the extracellular domain of c-Met or bind to HGF.
  • compounds targeting, decreasing or inhibiting the activity of the c-Met receptor such as compounds which target, decrease or inhibit the activity of c-Met, especially compounds which inhibit the kinase activity of c-Met receptor, or antibodies that target the extracellular domain of c-Met or bind to HGF
  • 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, or CDC25, e.g. okadaic acid or a derivative thereof.
  • Compounds which induce cell differentiation processes are e.g. retinoic acid, ⁇ - ⁇ - or ⁇ - tocopherol or ⁇ - ⁇ - or ⁇ -tocotrienol.
  • cyclooxygenase inhibitor as used herein includes, but is not limited to, e.g. Cox-2 inhibitors, 5-alkyl substituted 2-arylaminophenylacetic acid and derivatives, such as celecoxib (CELEBREX), rofecoxib (VIOXX), etoricoxib, valdec ⁇ xib or a 5-alkyl-2- arylaminophenylacetic acid, e.g. 5-methyl-2-(2'-chloro-6'-fluoroanilino)phenyl acetic acid, lumiracoxib.
  • Cox-2 inhibitors 5-alkyl substituted 2-arylaminophenylacetic acid and derivatives, such as celecoxib (CELEBREX), rofecoxib (VIOXX), etoricoxib, valdec ⁇ xib or a 5-alkyl-2- arylaminophenylacetic acid, e.g. 5-methyl-2-(2'-chlor
  • 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.
  • mTOR inhibitors relates to compounds which inhibit the mammalian target of rapamycin (mTOR) and which possess antiproliferative activity such as siroh ' mus (RapamuneCD), everolimus (CerticanTM), CCI-779 and ABT578.
  • heparanase inhibitor refers to compounds which target, decrease or inhibit heparin sulfate 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, as used herein refers to compounds which target, decrease or inhibit the oncogenic activity of Ras e.g. a
  • farnesyl transferase inhibitor e.g. L-744832, DK8G557 or R115777 (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. Bortezomid (VelcadeTM)and MLN 341.
  • matrix metalloproteinase inhibitor or (“MMP” inhibitory 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-
  • FMS-like tyrosine kinase inhibitors e.g. compounds targeting, decreasing or inhibiting the activity of FMS-like tyrosine kinase receptors (Flt-3R); interferon,
  • 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,
  • 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 proteosome pathway. Compounds targeting, decreasing or inhibiting the intrinsic ATPase activity of HSP90; degrading, targeting, decreasing or inhibiting the HSP90 client proteins via the ubiquitin proteosome 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-DMI.erbitux, 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., farnesyf 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, ⁇ /-hydroxy-3-[4-[[[2-(2-methyl-1H-indol-3-yl)-ethyl]-amino]me- thyl]phenyl]-2E-2-propenamide, or a pharmaceutically acceptable salt thereof and ⁇ /-hydro- xy-3-[4-[(2-hydroxyethyl) ⁇ 2-(1/-/-indol-3-yl)ethyl]-amino]methyl]phenyl]-2E-2-propenamide, or a pharmaceutically acceptable salt thereof, especially the lactate salt.
  • Somatostatin receptor antagonists refers to compounds which target, treat or inhibit the somatostatin receptor such as octreotide, 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., 4 th Edition, Vol. 1 , pp. 248-275 (1993).
  • EDG binders refers a class of immunosuppressants that modulates lymphocyte recirculation, such as FTY720.
  • ribonucleotide reductase inhibitors refers to pyrimidine or purine nucleoside analogs including, but not limited to, fludarabine and/or cytosine arabinoside (ara-C),
  • Ribonucleotide reductase inhibitors are especially hydroxyurea or 2-hydroxy-1/-/-isoindole-1 ,3-dione derivatives, such as PL-1 , PL-2, PL-3,
  • 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-pyridylmethyl)phthalazine or a pharmaceutically acceptable salt thereof, e.g. the succinate, or in WO 00/09495,
  • VEGF aptamer e.g. Macugon
  • FLT-4 inhibitors FLT-3 inhibitors
  • VEGFR-2 IgGI antibody Angiozyme (RPI 4610) and Bevacizumab (AvastinTM).
  • Photodynamic therapy refers to therapy which uses certain chemicals known as photosensitizing compounds to treat or prevent cancers.
  • Examples of photodynamic therapy includes treatment with compounds, such as e.g. VISUDYNE and porfimer sodium.
  • Angiostatic steroids as used herein refers to compounds which block or inhibit angiogenesis, such as, e.g., anecortave, triamcinolone, hydrocortisone, 11- ⁇ -epihydrocotisol, cortexolone, 17 ⁇ -hydroxyprogesterone, corticosterone, desoxycorticosterone, testosterone, estrone and dexamethasone.
  • Implants containing corticosteroids refers to compounds, such as e.g. fluocinolone, dexamethasone.
  • “Other chemotherapeutic compounds” include, but are not limited to, plant alkaloids, hormonal compounds and antagonists; biological response modifiers, preferably lymphokines or interferons; antisense oligonucleotides or oligonucleotide derivatives; shRNA or siRNA; or miscellaneous compounds or compounds with other or unknown mechanism of action.
  • the compounds of the invention are also useful as co-therapeutic compounds for use in combination with other drug substances such as anti-inflammatory, bronchodilatory or antihistamine drug substances, particularly in the treatment of inflammatory diseases such as those mentioned hereinbefore, for example as potentiators of therapeutic activity of such drugs or as a means of reducing required dosaging or potential side effects of such drugs.
  • a compound of the invention may be mixed with the other drug substance in a fixed pharmaceutical composition or it may be administered separately, before, simultaneously with or after the other drug substance.
  • the invention includes a combination of a compound of the invention as hereinbefore described with an anti-inflammator or antihistamine drug substance, said compound of the invention and said drug substance being in the same or different pharmaceutical composition.
  • Suitable anti-inflammatory drugs include steroids, in particular glucocorticosteroids such as budesonide, beclamethasone dipropionate, fluticasone propionate, ciclesonide or mometasone furcate, or steroids described in WO 02/88167, WO 02/12266, WO 02/100879, WO 02/00679 (especially those of Examples 3, 11 , 14, 17, 19, 26, 34, 37, 39, 51 , 60, 67, 72, 73, 90, 99 and 101), WO 03/035668, WO 03/048181 , WO 03/062259, WO 03/064445, WO 03/072592, non-steroidal glucocorticoid receptor agonists such as those described in WO 00/00531 , WO 02/10143, WO 03/082280, WO 03/082787, WO 03/104195, WO 04/005229; LTB4 antagonists such LY293111 , CGS
  • Suitable bronchodilatory drugs include anticholinergic or antimuscarinic compounds, in particular ipratropium bromide, oxitropium bromide, tiotropium salts and CHF 4226 (Chiesi), and glycopyrrolate, but also those described in WO 01/04118, WO 02/51841 , WO 02/53564, WO 03/00840, WO 03/87094, WO 04/05285, WO 02/00652, WO 03/53966, EP 424021 , US 5171744, US 3714357, WO 03/33495 and WO 04/018422.
  • Suitable antihistamine drug substances include cetirizine hydrochloride, acetaminophen, clemastine fumarate, promethazine, loratidine, desloratidine, diphenhydramine and fexofenadine hydrochloride, activastine, astemizole, azelastine, ebastine, epinastine, mizolastine and tefenadine as well as those disclosed in WO 03/099807, WO 04/026841 and JP 2004107299.
  • chemokine receptors e.g. CCR-1 , CCR-2, CCR-3, CCR-4, CCR-5, CCR-6, CCR-7, CCR-8, CCR-9 and CCR10, CXCR1 , CXCR2, CXCR3, CXCR4, CXCR5, particularly CCR-5 antagonists such as Schering-Plough antagonists SC-351125, SCH- 55700 and SCH-D, Takeda antagonists such as N-[[4-[[[[6,7-dihydro-2-(4-methylphenyl)-5H- benzo-cyclohepten-8-yl]carbonyl]amino]phenyl]-methyl]tetrahydro-N,N-dimethyl-2H-pyran-4- amin-ium chloride (TAK-770), and CCR-5 antagonists described in US 6166037 (particularly claims 18 and 19), WO 00/66558 (particularly claim 8), and CCR-5 antagonists described in US 6166037 (particularly claims 18 and 19
  • Therapeutic agents for possible combination are especially one or more antiproliferative, cytostatic or cytotoxic compounds, for example one or several agents selected from the group which includes, but is not limited to, an inhibitor of polyamine biosynthesis, an inhibitor of a protein kinase, especially of a serine/threonine protein kinase, such as protein kinase C, or of a tyrosine protein kinase, such as the EGF receptor tyrosine kinase, e.g. Iressa®, the VEGF receptor tyrosine kinase, e.g. PTK787 or Avastin®, or the PDGF receptor tyrosine kinase, e.g.
  • an inhibitor of polyamine biosynthesis an inhibitor of a protein kinase, especially of a serine/threonine protein kinase, such as protein kinase C, or of a tyrosine protein kinase,
  • STI571 (Glivec®), a cytokine, a negative growth regulator, such as TGF- ⁇ or IFN- ⁇ , an aromatase inhibitor, e.g. letrozole (Femara®) or anastrozole, an inhibitor of the interaction of an SH2 domain with a phosphorylated protein, antiestrogens, topoisomerase I inhibitors, such as irinotecan, topoisomerase Il inhibitors, microtubule active agents, e.g.
  • paclitaxel or an epothilone alkylating agents, antiproliferative antimetabolites, such as gemcitabine or capecitabine, platin compounds, such as carboplatin or cis-platin, bisphosphonates, e.g. AREDIA®or ZOMETA®, and monoclonal antibodies, e.g. against HER2, such as trastuzumab.
  • alkylating agents such as gemcitabine or capecitabine
  • antiproliferative antimetabolites such as gemcitabine or capecitabine
  • platin compounds such as carboplatin or cis-platin
  • bisphosphonates e.g. AREDIA®or ZOMETA®
  • monoclonal antibodies e.g. against HER2, such as trastuzumab.
  • 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, especially where these time intervals allow that the combination partners show a cooperative, e.g. synergistic effect.
  • the invention also provides a pharmaceutical preparation, comprising a compound of formula I as defined herein, or a pharmaceutically acceptable salt of such a compound, or a hydrate or solvate thereof, and at least one pharmaceutically acceptable carrier.
  • the compounds of the invention may be administered by any conventional route, in particular parenterally, for example in the form of injectable solutions or suspensions, enterally, e.g. orally, for example in the form of tablets or capsules, topically, e.g. in the form of lotions, gels, ointments or creams, or in a nasal or a suppository form.
  • Topical administration is e.g. to the skin.
  • a further form of topical administration is to the eye.
  • Pharmaceutical compositions comprising a compound of the invention in association with at least one pharmaceutical acceptable carrier or diluent may be manufactured in conventional manner by mixing with a pharmaceutically acceptable carrier or diluent.
  • pharmaceutically acceptable carriers that are suitable for topical, enteral, for example oral or rectal, or parenteral administration and that may be inorganic or organic, solid or liquid.
  • diluents for example lactose, dextrose, mannitol, and/or glycerol, and/or lubricants and/or polyethylene glycol.
  • Tablets may also comprise binders, for example magnesium aluminum silicate, starches, such as corn, wheat or rice starch, gelatin, methylcellulose, sodium carboxymethylcellulose and/or polyvinylpyrrolidone, and, if desired, disintegrators, for example starches, agar, alginic acid or a salt thereof, such as sodium alginate, and/or effervescent mixtures, or adsorbents, dyes, flavorings and sweeteners. It is also possible to use the pharmacologically active compounds of the present invention in the form of parenterally administrable compositions or in the form of infusion solutions.
  • binders for example magnesium aluminum silicate, starches, such as corn, wheat or rice starch, gelatin, methylcellulose, sodium carboxymethylcellulose and/or polyvinylpyrrolidone
  • disintegrators for example starches, agar, alginic acid or a salt thereof, such as sodium alginate, and/or effervescent mixtures, or
  • the pharmaceutical compositions may be sterilized and/or may comprise excipients, for example preservatives, stabilisers, wetting compounds and/or emulsifiers, so- lubilisers, salts for regulating the osmotic pressure and/or buffers.
  • excipients for example preservatives, stabilisers, wetting compounds and/or emulsifiers, so- lubilisers, salts for regulating the osmotic pressure and/or buffers.
  • the present pharmaceutical compositions which may, if desired, comprise other pharmacologically active substances are prepared in a manner known per se, for example by means of conventional mixing, granulating, confectionning, dissolving or lyophilising processes, and comprise approximately from 1% to 99%, especially from approximately 1% to approximately 20%, active ingredients).
  • the present invention provides a compound of formula I or an N-oxide or a tau- tomer thereof, or a pharmaceutically acceptable salt of such a compound, for use in a method for the treatment of the human or animal body, especially for the treatment of a disease mentioned herein, most especially in a patient requiring such treatment.
  • the invention relates to a method for the treatment of a disease which responds to an inhibition of JAK-2 and/or Jak-3 kinase, which comprises administering a compound of formula I or a pharmaceutically acceptable salt thereof, wherein the radicals and symbols have the meanings as defined above, especially in a quantity effective against said disease, to a warm-blooded animal requiring such treatment.
  • a pharmaceutically acceptable carrier e.g. of solid or liquid tumours in warm-blooded animals, including humans
  • a compound of the formula I may be prepared by processes that, though not applied hitherto for the new compounds of the present invention where they thus form new processes, are known per se: preferably, a process for the manufacture of a compound of the formula I comprises either
  • R 2 is as defined for a compound of the formula I, or a reactive derivative thereof, or b) reacting a compound of the formula II,
  • R 2 is as defined for a compound of the formula I and alk is alkyl, preferably C 1 -C 7 alkyl,
  • reaction a) preferably takes place under Suzuki(-Miyaura) conditions, that is, by palladium-catalyzed crosscoupling of organoboranes, by reacting the halo- carrying compound of the formula Il with the boronic acid of the formula III, or a reactive derivative thereof.
  • a reactive derivative of a boronic acid of the formula II! is preferably one wherein instead of the hydroxyl groups at the boron atom an aryl, alkenyl or especially alkyl moiety is present, or wherein the OH groups are present in bridged form, e.g. , together with the boron atom, forming a group of the formula (A)
  • the reaction preferably takes place in a mixture of a polar aprotic solvent, such as dimethyl- formamide (DMF) or tetrahydrofurane, and water in the presence of a catalyst for the cross- coupling, especially a noble metal catalyst, preferably a palladium catalyst, such as palla- dium(ll) complex, for example bis(triphenylphosphine)palladium (II) dichloride, in the presence of a base, such as potassium carbonate, sodium hydroxide or sodium carbonate, at a preferred temperature in the range from 60 0 C to 130 0 C, e.g. at about 80 0 C; or according to a another preferred method in an ether solvent, e.g.
  • a polar aprotic solvent such as dimethyl- formamide (DMF) or tetrahydrofurane
  • a catalyst for the cross- coupling especially a noble metal catalyst, preferably a palladium catalyst, such as palla- dium(ll) complex
  • a catalyst for the cross coupling especially a noble metal catalyst, preferably a palladium (0) complex, for example tris(dibenzylideneacetone)- dipalladium(O) or tetrakis (triphenylphosphin)palladium(O), in the presence of a base, such as sodium hydroxide, potassium carbonate of sodium carbonate, if desired in the presence of an appropriate ligand, such as 2-dicyclohexylphosphino-2',6'-dimethoxybiphenyl (SPhos), at a preferred temperature in the range from 60 to 150 °C, preferably from 70 to 110 0 C; if required conducting the reaction in a sealed vessel (e.g. a seal reactor) if the boiling point of the reaction mixture is exceeded and especially if the heating is effected by microwave excitation.
  • oxygen is excluded, e.g. by the
  • alk is alkyl, preferably d-Cz-alkyl, more preferably methyl
  • alk is alkyl, preferably d-Cz-alkyl, more preferably methyl
  • an appropriate polar solvent such as N,N-dimethylacetamide or N,N-dimethylformamide
  • an ether such as tetrahydrofurane or dimethoxy-ethane, and/or a mixture of two or more such solvents
  • a a palladium catalyst especially a palladium (0) complex, for example tetrakistriphenylpalladium, e.g.
  • a sealed vessel e.g. a seal reactor or a microwave vessel
  • the heating is effected by microwave excitation.
  • one or more other functional groups for example carboxy, hydroxy, amino, sulfhydryl or the like are or need to be protected in a starting material of the formula Il or III or any other precursor, because they should not take part in the reaction or disturb the reaction, these are such groups as are usually used in the synthesis of peptide compounds, and also of cephalosporins and penicillins, as well as nucleic acid derivatives and sugars.
  • Protecting groups are such groups that are no longer present in the final compounds once they are removed, while groups that remain as substituents are not protecting groups in the sense used here which is groups that are added at a starting material or intermediate stage and removed to obtain a final compound. Also in the case of conversions of a compound of the formula I into a different compound of the formula I, protecting groups may be introduced and removed, if useful or required.
  • the protecting groups may already be present in precursors and should protect the functional groups concerned against unwanted secondary reactions, such as acylations, etheri- fications, esterifications, oxidations, solvolysis, and similar reactions. It is a characteristic of protecting groups that they lend themselves readily, i.e. without undesired secondary reactions, to removal, typically by acetolysis, protonolysis, solvolysis, reduction, photolysis or also by enzyme activity, for example under conditions analogous to physiological conditions, and that they are not present in the end-products.
  • the specialist knows, or can easily establish, which protecting groups are suitable with the reactions mentioned above and below.
  • a compound of the formula I may be converted into a different compounds of the formula I.
  • the amino can be converted into acylamino, e.g. C 1 -C 7 - alkanoylamino or Ci-Cy-alkanesulfonylamino, by reaction with a corresponding C 1 -C 7 ⁇ l- kanoylhalogenide or Ci-Cr-alkanesulfonylhalogenide, e.g.
  • a corresponding chloride in the presence of a tertiary nitrogen base, such as triethylamine or pyridine, in the absence or presence of an appropriate solvent, such a methylene chloride, for example at temperatures in the range from -20 to 50 0 C, e.g. at about room temperature.
  • a tertiary nitrogen base such as triethylamine or pyridine
  • an appropriate solvent such as a methylene chloride
  • the cyano may be converted to an aminomethyl group, e.g. by hydrogenation in the presence of an appropriate metal catalyst, such as Raney Nickel or Raney Cobalt, in an appropriate solvent, e.g. a lower alkanol, such as methanol and/or ethanol, for example at temperatures in the range from -20 to 50 0 C, e.g. at about room temperature.
  • an appropriate metal catalyst such as Raney Nickel or Raney Cobalt
  • an appropriate solvent e.g. a lower alkanol, such as methanol and/or ethanol, for example at temperatures in the range from -20 to 50 0 C, e.g. at about room temperature.
  • R 1 or especially R 2 carries a carboxyl (COOH) substituent
  • the latter can be converted into an amide group, e.g. an N-C ⁇ Cr-alkyl-carba- moyl group, by reaction with the corresponding amine, e.g.
  • a coupling agent that forms a preferred reactive derivative of the carboxyl group in situ
  • a coupling agent that forms a preferred reactive derivative of the carboxyl group in situ
  • DCC/ HOBT dicyclohexylcarbodiimide/i-hydroxybenzotriazole
  • BOPCI bis(2-oxo-3-oxaz- olidinyl)phosphinic chloride
  • TPTU O-(1 ,2-dihydro-2-oxo-1-pyridyl)- ⁇ /, ⁇ /, ⁇ /',/V-tetramethyl- uronium tetrafluoroborate
  • TBTU O-benzotriazol-1-yl)-N,N,N',N'-tetramethyluronium tetrafluoroborate
  • PyBOP (benzotriazol-1 -yloxy)-tripyrrolidinophosphonium-hexafluoro- phosphate
  • PyBOP O-(1
  • the reaction mixture is preferably stirred at a temperature. of between approximately -20 and 50 °C, especially between 0 0 C and 30 0 C, e.g. at room temperature.
  • the reaction preferably takes place by first reacting the compound of the formula I carrying the two vicinal amino groups with formic acid, e.g.
  • a coupling agent such as EDC hydrochloride
  • a base such as N,N-dimethylaminopyridine (DMAP)
  • an appropriate solvent such as methylene chloride
  • the intermediate with the formylamino group obtainable by the first reaction in the preceding paragraph is also a compound of the formula I, so that this first reaction also is a conversion reaction according to the invention.
  • Salts of a compound of formula I with a salt-forming group may be prepared in a manner known per se. Acid addition salts of compounds of formula I may thus be obtained by treatment with an acid or with a suitable anion exchange reagent.
  • a salt with two acid molecules (for example a dihalogenide of a compound of formula I) may also be converted into a salt with one acid molecule per compound (for example a monohalogenide); this may be done by heating to a melt, or for example by heating as a solid under a high vacuum at elevated temperature, for example from 130 to 170 0 C, one molecule of the acid being expelled per molecule of a compound of formula I.
  • Salts can usually be converted to free compounds, e.g. by treating with suitable basic compounds, for example with alkali metal carbonates, alkali metal hydrogencarbonates, or alkali metal hydroxides, typically potassium carbonate or sodium hydroxide.
  • Stereoisomeric mixtures e.g. mixtures of diastereomers
  • Dia- stereomeric mixtures for example may be separated into their individual diastereomers by means of fractionated crystallization, chromatography, solvent distribution, and similar procedures. This separation may take place either at the level of a starting compound or in a compound of formula I itself.
  • Enantiomers may be separated through the formation of dia- stereomeric salts, for example by salt formation with an enant ⁇ omer-pure chiral acid, or by means of chromatography, for example by HPLC, using chromatographic substrates with chiral ligands.
  • the starting materials of the formulae Il and III, as well as other starting materials mentioned herein, e.g. below, can be prepared according to or in analogy to methods that are known in the art, are known in the art and/or are commercially available. Novel starting- materials, as " well as processes for the preparation thereof, are likewise an embodiment of the present invention. In the preferred embodiments, such starting materials are used and the reaction chosen are selected so as to enable the preferred compounds to be obtained.
  • R 1 , R 2 , R 3 , R 4 and X are preferably as defined for a compound of the formula I.
  • Hal is halogen, especially chloro or bromo.
  • a compound of the formula Il can, for example, be obtained by reacting a thiourea compound of the formula IV, under cyclization in the presence of an organic sulfonyl chloride, such as toluene-4-sulfonyl chloride, in an appropriate solvent, e.g. a cyclic ether, such as tetrahydrofurane, in the presence of water and a base, such as sodium hydroxide, at temperatures in the range from -20 to 50 0 C, e.g. at about room temperature.
  • an organic sulfonyl chloride such as toluene-4-sulfonyl chloride
  • an appropriate solvent e.g. a cyclic ether, such as tetrahydrofurane
  • a base such as sodium hydroxide
  • a thiourea compound of the formula IV can, for example, be prepared from an amino phenol of the formula V 1
  • an appropriate solvent e.g. a cyclic ether, such as tetrahydrofurane
  • temperatures e.g. in the range from -20 to 50 0 C, e.g. at about room temperature.
  • a compound of the formula V may, for example, be prepared by reducing a nitro compound of the formula VII,
  • VI e.g. with hydrogen in the presence of a catalyst, such as Raney-Nickel (Ra-Ni) or Raney Cobalt or the like, in an appropriate solvent, such as an alcohol, e.g. methanol, and/or a cyclic ether, such as tetrahydrofurane, at temperatures e.g. in the range from -20 to 50 0 C, e.g. at about room temperature.
  • a catalyst such as Raney-Nickel (Ra-Ni) or Raney Cobalt or the like
  • an appropriate solvent such as an alcohol, e.g. methanol, and/or a cyclic ether, such as tetrahydrofurane
  • a compound of the formula Il can be prepared by reacting a methyl sulfanyl compound of .the formula VIII,
  • This reaction can preferably be conducted, in the presence of an agent capable of oxidising the methanesulfanyl at the oxazole ring to the methanesulfinyl, e.g. in the presence of a peroxide, such as m-chloroperbenzoic acid, and appropriate solvent, such as dichloromethane, preferably at temperatures in the range from 0 to 50 0 C, e.g. at about room temperature.
  • a peroxide such as m-chloroperbenzoic acid
  • solvent such as dichloromethane
  • a compound of the formula VIII can, for example, be prepared by reacting a thiol compound of the formula X,
  • X in an appropriate solvent, e.g. a dialkyl carboxylic acid amide, such as dimethylformamide, in the presence of a base, e.g. an alkali metal (such as potassium) carbonate, at temperatures e.g. in the range from -20 to 50 0 C, e.g. at about room temperature, with a methyl halogenide, e.g. methyl iodide.
  • a solvent e.g. a dialkyl carboxylic acid amide, such as dimethylformamide
  • a base e.g. an alkali metal (such as potassium) carbonate
  • a compound of the formula X can, for example, be prepared from a compound of the formula V described (and obtainable as) above by reacting it with an alkali metal ethyl xanthoge- nate, such as potassium ethyl xanthogenate, in an appropriate solvent, such as an alcohol, e.g. ethanol, preferably at elevated temperatures, e.g. in the range from 50 0 C to the reflux temperature of the reaction mixture, e.g. under reflux.
  • an alkali metal ethyl xanthoge- nate such as potassium ethyl xanthogenate
  • an appropriate solvent such as an alcohol, e.g. ethanol
  • elevated temperatures e.g. in the range from 50 0 C to the reflux temperature of the reaction mixture, e.g. under reflux.
  • a compound of the formula III * can, for example, be prepared from a compound of the formula Xl,
  • Hal is hal ⁇ , especially bromo, by reaction with a hexa-alkyl tin, especially hexa-C,- C 7 -alkyl-tin, e.g. hexamethyltin, in an appropriate solvent, such as toluene, and a customary noble metal catalyst, such as tetrakis(triphenylphosphine)palladium, preferably at elevated temperatures e.g. in the range from 50 to 150 0 C.
  • a hexa-alkyl tin especially hexa-C,- C 7 -alkyl-tin, e.g. hexamethyltin
  • an appropriate solvent such as toluene
  • a customary noble metal catalyst such as tetrakis(triphenylphosphine)palladium
  • Example 1 (7-m-Tolyl-benzooxazol-2-ylH3,4,5-trimethoxy-phenyl)-amine.
  • the starting materials can be prepared as follows:
  • reaction of (7-bromo- benzooxazol-2-yl)-(3,4,5-trimethoxy-phenyl)-amine and the appropriate boronic acid derivative leads to the following examples:
  • Example 2 (7-Phenyl-benzooxazol-2-yl)-(3,4,5-trimethoxy-phenyl)-amine.
  • Example 3 (7-Pyridin-3-yl-benzooxazol-2-ylH3.4,5-trimethoxy-phenyl)-amine.
  • Example 4 f7-(3-Methoxy-phenyl)-benzooxazol-2-yll-(3,4,5-trimethoxy-phenyl)-amine.
  • Example 5 f7-(2-Methoxy-phenyl)-benzooxazol-2-yll-(3,4.5-trimethoxy-phenyl)-amine.
  • Example 6 f7-(3-Hvdroxy-phenyl)-benzooxazol-2-yll-(3,4,5-trimethoxy-phenyl)-amine.
  • Example 7 [7-(4-Methoxy-phenyl)-benzooxazol-2-yll-(3,4,5-trimethoxy-phenyl)-amine.
  • R, 2.44 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH 3 CN in H 2 O in 2min + 0.05% TFA, flow rate 1.Oml/min); MS: 407 (M+ 1 ) + ; m.p. 179-182°C.
  • Example 8 (7-lsoquinolin-4- ⁇ l-benzooxazol-2-yl)-(3.4.5-trimethoxy-phenyl)-amine.
  • Example 9 f7-(3-Chloro-phenyl)-benzooxazol-2-vn-(3 1 4.5-trimethoxy-phenyl)-amine.
  • R 1 2.60 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH 3 CN in H 2 O in 2min + 0.05% TFA, flow rate 1. Oml/min); MS: 411 (M+1 , 35 CI) + ; m.p. 199-201 0 C.
  • Example 10 f7-(3-Amino-phenyl)-benzooxazol-2-yll-(3,4,5-trimethoxy-phenyl)-amine.
  • Example 11 f7-(4-Amino-phenyl)-benzooxazol-2-yll-(3,4,5-trimethoxy-phenyl)-amine.
  • Example 12 [7-(6-Methoxy-pyridin-3-yl)-benzooxazol-2-yll-(3,4.5-trimethoxy-phenyl)-amine.
  • Example 13 f7-(3-Amino-4-methyl-phenyl)-benzooxazol-2-vn-(3,4,5-trimethoxy-phenyl)- amine.
  • Example 15 (7-Quinolin-6-yl-benzooxazol-2-yl)-(3.4,5-trimetr ⁇ oxy-phenyl)-amine.
  • Example 16 4-f2-(3,4,5-Trimethoxy-phenylamino)-benzooxazol-7-v ⁇ -benzamide.
  • Example 17 f7-(4-Methanesulfonyl-phenyl)-benzooxazol-2-ylH3,4,5-trimethoxy-phenyl)- amine.
  • Example 18 4-f2-(3,4,5-Trimethoxy-phenylamino)-benzooxazol-7-yll-benzenesulfonamide.
  • Example 19 4-f2-(3,4,5-Trimethoxy-phenylamino)-benzooxazol-7-vn-benzonitrile.
  • Example 20 f7-(2H-Pvrazol-3-vl)-benzooxazol-2-vn-(3.4.5-trimethoxv-phenyl)-amine.
  • R, 2.00 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH 3 CN in H 2 O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 367 (M+1 ) + ; m.p. 185-189°C.
  • Example 21 N-Methyl-4-[2-(3,4,5-trimethoxy-phenylamino)-benzooxazol-7-yll- benzenesulfonamide.
  • Example 22 N,N-Dimethyl-4-[2-(3,4,5-trimethoxy-phenylamino)-benzooxazol-7-vn- benzenesulfonamide.
  • Example 23 ⁇ 7-f4-(Morpholine-4-sulfonyl)-phenyl1-benzooxazol-2-ylH3,4,5-trimethoxy- phenyQ-amine.
  • Example 24 N-Methyl-4-f2-(3,4,5-trimethoxy-phenylamino)-benzooxazol-7-yl1-benzamide.
  • Example 25 N,N-Dimethyl-4-f2-(3,4.5-trimethoxy-phenylamino)-benzooxazol-7-yl1-benz- amide.
  • Example 26 N- ⁇ 4-f2-(3,4 I 5-Trimethoxy-phenylamino)-benzooxazol-7-vn-phenyl>- methanesulfonamide.
  • Example 27 N- ⁇ 4-f2-(3,4,5-Trimethoxy-phenylamino)-benzooxazol-7-vn-phenyl)-acetamide.
  • Example 28 N- ⁇ 3-f2-(3,4.5-Trimethoxy-phenylamino)-benzooxazol-7-v ⁇ -phenyl)-acetamide.
  • Example 29 r7-(4-Aminomethvl-phenvl)-benzooxazol-2-vlM3,4,5-trimethoxv-phenvl)-amine.
  • 5ml THF and 30mg Ra-Ni (B113W EtOH, Degussa) are added. Then this mixture is hydrogenated under normal pressure for 2Oh at room temperature.
  • Example 30 N- ⁇ 4-[2-(3,4,5-Trimethoxy-phenylaminoVbenzooxazol-7-yll-benzyl)-acetamide.
  • Example 31 N- ⁇ 4-[2-(3,4,5-Trimethoxy-phenylamino)-benzooxazol-7-v ⁇ -benzyl)- methanesulfonamide.
  • Example 32 r7-(3-Aminomethyl-phenyl)-benzooxazol-2-ylH3,4,5-trimethoxy-phenyl)-amine.
  • Example 34 4-f2-(3,4,5-Trimethoxy-phenylaminoV-benzooxazol-7-v ⁇ -thiophene-2-carboxylic acid methylamide.
  • the starting material 4-[2-(3,4,5-trimethoxy-phenylamino)-benzooxazol-7-yl]-thiophene-2- carboxylic acid is prepared from 2-carboxythiophene-4-boronic acid pinacol ester as described in Example 1.
  • Example 35 f7-(1 H-Benzoimidazol-5-yl)-benzooxazol-2-yn-(3,4,5-trimethoxy-phenyl)-amine.
  • a solution of 0.182g (0.243mmol) N- ⁇ 2-amino-4-[2-(3,4,5-trimethoxy-phenylamino)- benzooxazol-7-yl]-phenyl ⁇ -formamide in 3ml acetic acid is stirred at 100 0 C for 1h. Then EtOAc is added to the reaction mixture and the organic layer is washed with 4N NaOH solution (2x) and with water (2x), dried over MgSO 4 , filtered and the filtrate is concentrated in vacuo.
  • 4-(4,4,5,5-tetramethyl-[1 ,3,2]dioxaborolan-2-yl)-benzene-1 ,2-diamine is prepared from the commercially available 2-nitro-4-(4,4,5,5-tetramethyl-[1 ,3,2]dioxaborolan-2-yl)-phenylamine using the Ra-Ni catalyzed nitro reduction method described in Example 1 (step c).
  • Example 36 f4-(4-Amino-phenyl)-oxazolo[5,4-clpyridin-2-yll-(3,4,5-trimethoxy-phenyl)- amine.
  • the starting materials can be prepared as follows:
  • Example 37 f4-(3-Amino-phenyl)-oxazolof5,4-c1pyridin-2-yll-(3,4,5-trimethoxy-phenyl)- amine.
  • Example 38 4-f2-(3,4.5-Trimethoxy-phenylamino)-oxazolof5,4-clpyridin-4-yll- benzenesulfonamide.
  • Example 39 [7-(3-Methoxy-phenyl)-benzooxazol-2-yll-(4-morpholin-4-yl-phenyl)-amine. (from 4-morpholinophenyl-isothiocyanate)
  • Example 40 r7-(4-Amino-phenyl)-benzooxazol-2-yll-(4-morpholin-4-yl-prienyl)-amine. (from 4-morpholinophenyl-isothiocyanate)
  • Example 41 N- ⁇ 4-[2-(4-Morpholin-4-yl-phenylamino)-benzooxazol-7-vn-phenyl ⁇ -methane- sulfonamide. (from 4-morpholinophenyl-isothiocyanate)
  • Example 42 f7-(3-Amino-phenyl)-benzooxazol-2-vn-(6-morpholin-4-yl-pyridin-3-yl)-amine. (from 4-(5-lsothiocyanato-pyridin-2-yl)-morpholine)
  • Example 64 4-f7-(3-Fluoro-4-morpholin-4-ylmethyl-phenyl)-benzooxazol-2-ylamino1-2,N,N- trimethyl-benzamide:
  • Example 65 4-(7-f4-(1 ,1-Dioxo-thiomorpholin-4-ylmethyl)-3,5-difluoro-phenv ⁇ -benzooxazol- 2-ylamino ⁇ -2,N,N-trimethyl-benzamide:
  • Tetrakis (triphenylphosphine) palladium (0.0095g, 0.0082mmol) is added and the reaction mixture is stirred at 100 0 C for 8h. After that the reaction mixture is poured on water and extracted 3x with EtOAc. The combined organic layers are washed with water and saturated NaCI solution, dried over MgSO 4 , filtered and the filtrate is concentrated in vacuo. The residue is purified by chromatography (silicagel, EtOAc), followed by recrystallisation diethylether/methanol to afford the title compound as white crystals.
  • a solution (degassed with argon) of 1.02g (3.0mmol) 4-(4-bromo-2,6-difluoro-benzyl)- thiomorpholine 1 ,1 -dioxide in 4ml dimethylacetamide is added to a solution (degassed with argon) of 0.855g (3.3mmol) bis-(pinacolato)-diboron and 0.594g ( ⁇ .Ommol) dried KOAc in 4ml dimethylacetamide. After that 0.076g (0.092mmol) Pd(dppf)CI 2 -CH 2 CI 2 is added. The reaction mixture is heated to 80 0 C and stirred at this temperature for 4h.
  • Example 66 r4-(2-(4-f4-(2-Methoxy-ethyl)-piperazin-1-vn-3-methyl-phenylamino ⁇ - benzooxazol-7-yl)-2-methyl-phenv ⁇ -morpholin-4-yl-methanone:
  • the title compound is prepared from (7-bromo-benzooxazol-2-yl)- ⁇ 4-[4-(2-methoxy-ethyl)- piperazin-1 -yl]-3-methyl-phenyl ⁇ -amine and [2-methyl-4-(4,4,5,5-tetramethyl-[1 ,3,2Jdioxa- borolan-2-yl)-phenyl]-morpholin-4-yl-methanone using methodology described in the preparation of example 65.
  • the title compound is obtained as an off-white foam.
  • the title compound is prepared from 4-[4-(2-methoxy-ethyl)-piperazin-1-yl]-3-methyl- phenylamine and 7-bromo-2-methylsulfanyl-benzooxazole using methodology described in the preparation of example 64.
  • Example 68 4-f7-(3,5-Difluoro-4-morpholin-4-ylmethvt-phenyl)-benzooxazol-2-ylaminol- 2,N,N-trimethyl-benzamide:
  • Example 70 2 1 N,N-Trimethyl-4- ⁇ 7-f4-(2-mo ⁇ holin-4-yl-2-oxo-ethyl)-phenylVbenzooxazol-2- ylamino)-benzamide:
  • Example 71 4-[7-(4-Methanesulfonylmethyl-phenyl)-benzooxazol-2-ylaminoT-N,N-dimetr ⁇ yl- benzamide:
  • Example 72 2-(4- ⁇ 2-f3-Methyl-4-(4-methyl-piperazine-1-carbonyl)-phenylamino1- benzooxazol-7-yl ⁇ -phen ⁇ l)-1-morpholin-4-yl-ethanone:
  • Example 76 4-(7-f2-Fluoro-4-(morpholine-4-carbonyl)-phenvn-benzooxazol-2-ylaminoV- 2,N,N-trimethyl-benzamide:
  • Example 77 4-(7- ⁇ 4-f2-(1.1-Dioxo-1lambda * 6 * -thiomorpholin-4-yl)-2-oxo-ethyll-phenyl>- benzooxazol-2-ylamino)-2, N , N-trimethyl-benzamide :
  • Example 78 4- ⁇ 7-[3-Fluoro-4-(morpholine-4-carbonyl)-phenyn-benzooxazol-2-ylamino)- 2, N . N-trimethyl-benzamide :
  • Example 79 4-(7-f4-(1,1-Dioxo-1lambda * 6 * -thiomorpholin-4-ylmethyl)-phenyll-benzooxazol- 2-ylamino ⁇ -2,N 1 N-trimethyl-benzamide:
  • Example 80 H J-Dioxo-1lambda * 6Mhiomorpholin-4-ylH2-fluoro-4- ⁇ 2-r4-(4-methyl- piperazin-1-yl)-phenylamino ⁇ -benzooxazol-7-yl)-phenyl)-methanone:
  • Example 82 (2-Methyl-4- ⁇ 2-f4-(4-methyl-piperazin-1-yl)-phenylaminol-benzooxazol-7-yl ⁇ - phenyl)-morpholin-4-yl-methanone:
  • Example 83 4-(7-(4-f2-(1 ,1-Dioxo-1 lambda * 6 * -thiomorpholin-4-yl)-2-oxo-ethvH-3-fluoro- phenyl)-benzooxazol-2-ylarnino)-2,N,N-trimethyl-benzamide:
  • Example 84 4- ⁇ 7-f4-(1 ,1-Dioxo-1lambda * 6Mhiomorpholin-4-ylmethyl)-3-fluoro-phenv ⁇ - benzooxazol-2-ylamino>-2.N,N-trimetriyl-benzamide:
  • Example 88 (7-[4-(1,1-Dioxo-1 lambda * 6 * -thiomorpholin-4-ylmethyl)-3-fluoro-phenyll- benzooxazol-2-ylH4-(4-ethyl-p ⁇ perazin-1-yl)-3-methyl-pheny ⁇ -amine:
  • Example 90 5-(7-[3-Fluoro-4-(morpholine-4-caronyl)-phenyll-benzooxazol-2- ⁇ lamino ⁇ -2-(4- methyl-piperazin-1-yl)-benzonitrile:
  • Example 95 (7-[4-(1 ,1-Dioxo-1lambda*6*-thiomorpholin-4-ylmethyl)-3,5-difluoro-phenyll- benzooxazol-2-ylH4-(4-methyl-piperazin-1-yl)-phenyll-amine:
  • Example 96 (4- ⁇ 7-f4-(1 ,1-Dioxo-1lambda * 6 * -thiomorpholin-4-ylmethyl)-3,5-difluoro-phenyll- benzooxazol-2-ylamino)-2-methyl-phenyl)-pyrrolidin-1-yl-methanone:
  • Example 101 ⁇ 7-f4-(1 ,1-Dioxo-1lambda * 6 * -thiomorpholin-4-ylmethyl)-3,5-clifluoro-phenyll- benzooxazol-2-ylH4-(4-isoprop ⁇ l-piperazin-1-yl)-3-methyl-phenv ⁇ -amine:
  • Example 102 [4-(2- ⁇ 4-r4-(2-Dimethylamino-ethyl)-piperazin-1-vn-3-methyl-phenylamino>- benzooxazol-7-yl)-2-methyl-phenyn-morpholin-4-yl-methanone:
  • Example 103 4-[7-(3,5-Difluoro-4-morpholin-4-ylmeth ⁇ l-phenyl>-benzooxazol-2-ylamino1- N , N-diethyl-2-meth yl-benzamide:
  • Example 104 ⁇ 4-r7-(3,5-Difluoro-4-morpholin-4-ylmethyl-phenyl)-benzooxazol-2-ylarnino1-2- methyl-phenyl)-(4-ethyl-piperazin-1-yl)-methanone:
  • Example 105 f4-(4-lsopropyl-piperazin-1 -yl)-3-methyl-phenylH7-(4-methanesulfonyl- phenyl)-benzooxazol-2-yri-amine:
  • Example 106 (4- ⁇ 2-f4-(4-lsopropyl-piperazin-1 -yl)-3-methyl-phenylaminol-benzooxazol-7-yl>- 2-methyl-phenyl)-morpholin-4-yl-methanone:
  • Example 107 (4-f4-(2-Dimethylamino-ethyl)-piperazin-1-v ⁇ -3-methyl-phenylH7-f4-(1.1- dioxo-1lambda * 6 * -thiomorpholin-4-ylmethyl)-3,5-difluoro-phenv ⁇ -benzooxazol-2-yl>-amine:
  • Example 108 (7-f4-(1 ,1-Dioxo-1lambda * 6Mhiomorpholin-4-ylmethyl)-3.5-difluoro-phenylI- benzooxazol-2-ylH4-f4-(2-methoxy-ethyl)-piperazin-1-v ⁇ -3-methyl-phenyl>-amine:
  • Example 109 (4- ⁇ 2-f4-(4-Ethyl-piperazin-1 -yl)-3-methyl-phenylamino1-benzooxazol-7-yl)-2,6- difluoro-phenyl)-morpholin-4-yl-methanone:
  • Example 110 ⁇ 2-Methyl-4-f7-(3-methyl-4-morpholin-4-ylmethyl-phenyl)-benzooxazol-2- ylamino1-phenyl ⁇ -pyrrolidin-1-yl-methanone:
  • Example 111 2-(4- ⁇ 2-[4-(4-Ethyl-piperazin-1-yl)-3-methyl-phenylaminol-benzooxazol-7-yl>- phenyl)-2-methyl-1-morpholin-4-yl-propan-1-one:
  • Example 1 12 4-(2-f4-(4-Ethyl-piperazin-1-yl)-3-methyl-phenylamino1-benzooxazol-7-yl>- benzenesulfonamide:
  • Example 113 f4-(4-Cyclopropyl-piperazin-1-yl)-phenylH7-(4-methanesulfonyl-phenyl)- benzooxazol-2-vH-amine:
  • Example 114 r4-(4-Cyclopropyl-piperazin-1-ylH>henylH7-f4-(1 ,1-dioxo-1lambda * 6 * - thiomorpholin-4-ylmethyl)-3,5-difluoro-phenyll-benzooxazol-2-yl ⁇ -amine:
  • Example 116 (1 ,1-Dioxo-1lambda*6*-thiomorpholin-4-yl)-(4- ⁇ 2-f4-(4-ethyl-piperazin-1-yl)-3- methyl-phenylamino
  • Example 117 4-(7-f4-(4-Acetyl-pip ⁇ razin-1-ylmethyl)-3,5-difluoro-phenvn-benzooxazol-2- ylamino)-N,N-diethyl-2-methyl-benzamide:
  • Example 118 4-f7-(3,5-Difluoro-4-piperazin-1- ⁇ lmethyl-phenyl)-benzooxazol-2-ylaminol-N,N- diethyl-2-methyl-benzamicle:
  • Example 118 is prepared from 4- ⁇ 4-[2-(4-diethylcarbamoyl-3-methyl-phenylamino)- benzooxazol-7-yl]-2,6-difluoro-benzyl ⁇ -piperazine-1-carboxylic acid tert-butyl ester as follows:
  • Example 119 The 1-ethyl-4-(2-methyl-4-nitro-benzyl)-piperazine used for the preparation of Example 119 is prepared as follows:
  • Example 120 4-f7-(3,5-Difluoro-4-morpholin-4-ylmethyl-phenyl)-benzooxazol-2-ylaminol- 2,N-dimethyl-benzamide:
  • Example 121 f7-(3,5-Difluoro-4-morpholin-4-ylmethyl-phenyl)-benzooxazol-2-ylH3-methyl-4- r4-(3,3,3-trifluoro-propyl)-piperazin-1-v ⁇ -phenyl ⁇ -amine:
  • Example 122 4-[7-(3,5-Difluoro-4-morpholin-4-ylmethyl-phenyl)-benzooxazol-2-ylamino]-N- (2-dimethylamino-ethyl)-2-methyl-benzamide:
  • Example 123 4-(7-(4-f2-(1 J-Dioxo-1lambda*6Mhiomorpholin-4-yl)-2-oxo-ethyl ⁇ -pr)enyl)- benzooxazol-2-ylamino).-N,N-diethyl-2-methoxy-benzarnide:
  • Example 125 1-(4-(7-f4-(1 ,1-Dioxo-1lambda * 6 * -thiomorpholin-4-ylmethyl)-3,5-difluoro- phenvn-benzooxazol-2-ylamino>-phenyl)-pyrrolidin-2-one:
  • Example 126 1- ⁇ 4-[7-(3,5-Difluoro-4-morpholin-4-ylmethyl-phenyl)-benzooxazol-2-ylamino1- phenyl)-pyrrolidin-2-one:
  • Example 127 (4-(2-f4-Methoxy-3-(4-methyl-piperazin-1 -yl)-phenylamino1-benzooxazol-7-yl)- 2-methyl-phenyl)-morpholin-4-yl-methanone:
  • Example 128 (7-f4-(1 ,1-Dioxo-1lambda * 6 * -thiomorpholin-4-ylmethyl)-3,5-difluoro-phenylV benzooxazol-2-yl ⁇ -[3-(4-methyl-piperazin-1-yl)-phenv ⁇ -amine:
  • Example 130 (7-f4-(1 ,1-Dioxo-1lambda * 6 * -thiomorpholin-4-ylmethyl)-3,5-difluoro-phenyn- benzooxazol-2-ylM4-methoxy-3-(4-methyl-piperazin-1-yl)-phenyll-amine:
  • the 4-(2-methyl-4-nitro-benzyl)-morpholine used in the preparation of example 131 is prepared as described in example 119 by using morpholine instead of 1-ethyl-piperazine.
  • Example 132 N- ⁇ 5-f7-(3-Fluoro-4-morpholin-4-ylmethyl-phenyl)-benzooxazol-2-ylaminol- indan-2-yl)-acetamide:
  • Example 133 N-(5-(7-f4-(1.1-Dioxo-1lambda * 6 * -thiomorpholin-4-ylmethyl)-phenyll- benzooxazol-2-ylamino)-indan-2-yl)-acetamide:
  • Example 135 2- ⁇ 4-f2-(4-Methanesulfin ⁇ l-phenylamino)-benzooxazol-7-yl]-phenyl)-1 - morpholin-4-yl-ethanone:
  • Example 135 4-Methanesulfinyl-phenylamine that is needed for the preparation of example 135 can be prepared as described by C. Almansa et al. in Journal of Medicinal Chemistry (2003), 46(16), 3463-3475.
  • Example 136 r7-(4-lmidazol-1-ylmethyl-phenyl)-benzooxazol-2-ylH3A5-trimethoxy-phenyl)- amine:
  • Example 137 (7-[4-(4-Methyl-piperazin-1 -ylmethyl)-phenv ⁇ -benzooxazol-2-ylH3,4,5- trimethoxy-phenyl famine :
  • Example 138 [7-(4-Morpholin-4-ylmethyl-phenyl)-benzooxazol-2-yll-(3,4,5-trimethoxy- phenyQ-amine:
  • Example 140 1 -Morpholin-4-yl-2-(4-f2-(3,4.5-trimethoxy-phenylamino>-benzooxazol-7-vn- phenyD-ethanone:
  • Example 141 Morpholin-4-yl-(4-f2-(3,4,5-trimethoxy-phenylamino)-benzooxazol-7-yll- phenyll-methanone:
  • Example 142 (4-Methyl-piperazin-1-ylH4-f2-(3A5-trimethoxy-phenylamino)-benzooxazol-7- v ⁇ -phenyl)-methanone:
  • Example 144 4-f2-(3-Methoxy-4-methyl-phenylamino)-benzooxazol-7-v ⁇ -N-methyl- benzenesulfonamide:
  • Example 150 2-Methoxy-N,N-dimethyl-4-(7-[4-(morpholine-4-carbonyl)-phenyll- benzooxazol-2-ylamino)-benzamide:
  • Example 151 2-Methyl-2-(4- ⁇ 2-r4-(4-methyl-piperazin-1-yl)-phenylamino1-benzooxazol-7-yl)- phenvD-1 -morpholin-4- ⁇ l-propan-1 -one:
  • Example 153 (4-f4-(1 l 1-Dioxo-Hambda * 6 * -thiomorpholin-4-ylmethylV3,5-difluoro-phenyll- oxazolof5,4-c1pyridin-2-yl)-f4-(4-ethyl-piperazin-1-yl)-3-methyl-prieny ⁇ -amine:
  • Example 154 f4-(4-Cyclopropyl-piperazin-1-yl)-3-methyl-phenylH7-[4-(1 ,1-dioxo- 1 lambda * 6 * -thiomorpholin-4-ylmethyl)-3,5-difluoro-phenvn-benzooxazol-2-yl ⁇ -amine:
  • Example 156 (4- ⁇ 2-f4-(4-Cyclopropyl-piperazin-1 -yl)-3-methyl-phenylamino1-benzooxazol-7- Vl>-2-fluoro-phenyl)-morpholin-4-yl-methanone:
  • Example 157 2-(4- ⁇ 2-f4-(4-Cyclopropyl-piperazin-1 -yl)-3-methyl-phenylamino1-benzooxazol- 7-yl ⁇ -phenyl)-2-methyl-1 -morpholin-4-yl-propan-1 -one:
  • Example 158 4-f7-(3.5-Difluoro-4-morpholin-4-ylmethyl-phenyl)-benzooxazol-2-ylaminol-N- (2-dimethylamino-ethyl)-2,N-dimethyl-benzamide:
  • Example 159 N-(2-Dimethylamino-ethyl)-4- ⁇ 7-f4-( 1 , 1 -dioxo- 1 lambda * 6 * -thiomorpholin-4- ylmethyl)-3,5-difluoro-phenv ⁇ -benzooxazol-2-ylamino ⁇ -2,N-dimethyl-benzamide:
  • Example 160 f4-(4-Cvclopropyl-piperazin-1 -yl)-3-methyl-phenyl1-f7-(3.5-difluoro-4- morpholin-4-ylmethyl-phenyl)-benzooxazol-2-vn-amine:
  • Example 161 5-
  • the 5-amino-1 ,3-dimethyl-1 H-pyridin-2-one needed for the preparation of the title compound is as follows:
  • Example 162 (4- ⁇ 2-f4-(4-Cvclopropyl-piperazin-1 -yl)-3-methyl-phenylamino1-benzooxazol-7- yl)-2-methyl-phenylH1.1-dioxo-1lambda * 6 * -thiomorpholin-4-yl)-methanone:
  • Example 163 f7-(3,5-Difluoro-4-morpholin-4-ylmethyl-phenyl)-benzooxazol-2-vn-(3-methyl-4- morpholin-4-yl-phenyl)-amine:
  • Example 164 (7-f4-(1 ,1-Dioxo-1lambda*6 * -thiomorpholin-4- ⁇ lmethyl)-3,5-difluoro-phenyll- benzooxazol-2-ylH3-meth ⁇ l-4-morpholin-4-yl-phenyl)-amine:
  • Example 168 4- ⁇ 4-f4-(1 ,1-Dioxo-1 lambda * 6Mhiomorpholin-4-ylmethyl)-3,5-difluoro-phenyl1- oxazolof5,4-clpyridin-2-ylamino ⁇ -2.N,N-trimethyl-benzamide:
  • Example 169 4-(7-r3,5-Difluoro-4-(3-oxo-piperazin-1 -ylmethyl)-phenv ⁇ -benzooxazol-2- ylamino)-2,N,N-trirnethyl-benzamide:
  • Example 170 4- ⁇ 2,6-Difluoro-4-f2-(3-methyl-4-morpholin-4-yl-phenylamino)-benzooxazol-7- ylT-benzyl ⁇ -piperazin-2-one:
  • Example 171 4-(4-(2-f4-(4-Cyclopropyl-piperazin-1-yl)-3-methyl-phenylaminol-benzooxazol- 7-yl)-2,6-difluoro-benzyl)-piperazin-2-one:
  • Example 172 4-(7-r3,5-Difluoro-4-(4-methyl-3-oxo-piperazin-1 -ylmethyQ-phenyH- benzooxazol-2-ylamino>-2,N,N-trinnethyl-benzamide:
  • Example 173 4- ⁇ 2,6-Difluoro-4-f2-(3-methyl-4-morpholin-4-yl-phenylamino)-benzooxazol-7- yl]-benzyl)-1-methyl-piperazin-2-one:
  • Example 174 4-(4- ⁇ 2-f4-(4-Cvclopropyl-piperazin-1-yl)-3-methyl-phenylaminol-benzooxazol- 7-yl ⁇ -2,6-difluoro-benzyl)-1-methyl-piperazin-2-one:
  • Example 175 4-f7-(4-Methanesulfinylmethyl-3-methyl-phenyl)-benzooxazol-2-ylaminol- 2,N.N-trimethyl-benzamide:
  • Example 176 f4-(4-Cvclopropyl-piperazin-1-yl)-3-methyl-phenv ⁇ -f7-(4- methanesulfonylmethyl-3-methyl-phenyl)-benzooxazol-2-vH-amine:
  • Example 178 (4-[7-(3,5-Difluoro-4-morpholin-4-ylmethyl-phenyl)-benzooxazol-2-ylamino1-2- methyl-phenyl)-morpholin-4-yl-methanone:
  • Example 179 4-[7-(3,5-Difluoro-4-morpholin-4-ylmeth ⁇ l-phenyl)-benzooxazol-2-ylaminol-N- (2-methoxy-ethyl)-2-meth ⁇ l-benzamide:
  • Example 180 4-f7-(3 1 5-Difluoro-4-morpholin-4-ylmethyl-phenyl)-benzooxazol-2-ylaminol-N- (3-dimethylamino-propyl)-2-methyl-benzamide:
  • Example 181 f7-(3,5-Difluoro-4-morpholin-4-ylmethyl-phenyl)-benzooxazol-2-v ⁇ -(6-methoxy- 5-methyl-pyridin-3-yl)-amine:
  • Example 182 4-f7-(3,5-Difluoro-4-morpholin-4-ylmethyl-phenyl)-benzooxazol-2-ylamino
  • Example 184 4-f7-(3,5-Difluoro-4-morpholin-4-ylmethyl-phenyl)-benzooxazol-2-ylamino1-N- (2-methoxy-ethyl)-2,N-dimethyl-benzamide:
  • Example 185 4-f7-(3,5-Difluoro-4-morpholin-4-ylnnethyl-phenyl)-benzooxazol-2-ylaminol-2- morpholin-4-yl-benzonitrile:
  • Example 186 Soft capsules
  • Preparation process The pulverized active ingredient is suspended in Lauroglykol® (propylene glycol laurate, Gattefosse S.A., Saint Priest, France) and ground in a wet pulverizer to produce a particle size of about 1 to 3 ⁇ m. 0.419 g portions of the mixture are then introduced into soft gelatin capsules using a capsule-filling machine.
  • Lauroglykol® propylene glycol laurate, Gattefosse S.A., Saint Priest, France
  • All four kinases of the JAK/TYK-kinase family are used as purified recombinant GST-fusion proteins, containing the active kinase domains.
  • GST-JAK1 (866- 1154), GST-JAK3(811- 1124), and GST-TYK2(888-1187) are expressed and purified by affinity chromatography.
  • GST-JAK2(808-1132) is purchased from Invitrogen (Carlsbad, USA, #4288).
  • the kinase assays are based on the Caliper mobility shift assay using the LabChip 3000 systems. This technology is similar to capillary electrophoresis and uses charge driven separation of substrate and product in a microfluidic chip.
  • All kinase reactions are performed in 384 well microtiter plates in a total reaction volume of 18 ⁇ l.
  • the assay plates are prepared with 0.1 ⁇ l per well of test compound in the appropriate test concentration, as described under the section "preparation of compound dilutions".
  • the reactions are started by combining 9 ⁇ l of substrate mix (consisting of peptide and ATP) with 9 ⁇ l of kinase dilution.
  • the reactions are incubated for 60 minutes at 30 0 C and stopped by adding 70 ⁇ l of stop buffer (100 mM Hepes, 5% DMSO, 0.1% Coating reagent, 10 mM EDTA, O. ⁇ i5% Brij 35).
  • Fluorescently labeled synthetic peptides are used as substrates in all reactions.
  • a peptide derived from the sequence of IRS-1 IRS-1 peptide, FITC-AhX-KKSRGDYMTMQIG-NH 2 (SEQ ID NO: 1 ); see J. Biol. Chem. 268(33), 25146-51 (1993)
  • JAK1 and TYK2 JAK1 and TYK2
  • JAK3tide FITC-GGEEEEYFELVKKKK-NH 2 (SEQ ID NO: 2); Upstate (Millipore), Temecula, California, USA)
  • Specific assay conditions are described in Table 1 : GGEEEYFELVKKKK
  • the terminated reactions are transferred to a Caliper LabChip 3000 reader (Caliper Life Siciences, Mountain View, California, USA) and the turnover of each reaction is measured by determining the substrate/product ratio.
  • Test compounds are dissolved in DMSO (10 mM) and transferred into 1.4mL flat bottom or V-shaped Matrix tubes carrying a unique 2D matrix chip by individual compound hubs. The numbers of these chips are distinctively linked to the individual compound identification numbers.
  • the stock solutions are stored at -20 0 C if not used immediately.
  • the vials are defrosted and identified by a scanner whereby a working sheet is generated that guides the subsequent working steps.
  • Compound dilutions are made in 96 well plates. This format enabled the assay of maximally 40 individual test compounds at 8 concentrations (single points) including 4 reference compounds.
  • the dilution protocol includes the production of pre-dilution plates, master plates and assay plates:
  • Pre-dilution plates 96 polypropylene well plates are used as pre-dilution plates. A total of 4 pre-difution plates are prepared including 10 test compounds each on the plate positions A1- A10, one standard compound at A11 and one DMSO control at A12. All dilution steps are done on a Hamilton STAR robot (Hamilton, Co.., Reno, NV, USA).
  • Master plates 100 ⁇ l_ of individual compound dilutions including standard compound and controls of the 4 "pre-dilution plates” are transferred into a 384 "master plate” including the following concentrations 1'820, 564, 182, 54.6, 18.2, 5.46, 1.82 and 0.546 ⁇ M, respectively in 90 % of DMSO.
  • Assay plates Identical assay plates are then prepared by pipetting 100 nl_ each of compound dilutions of the master plates into 384-well "assay plates". In the following the compounds are mixed with 9 ⁇ l_ of assays components plus 9 ⁇ L enzyme corresponding to a 1 :181 dilution steps enabling the final concentration of 10, 3.0, 1.0, 0.3, 0.1 , 0.03, 0.01 and 0.003 ⁇ M, respectively.
  • the preparation of the master plates are handled by the Matrix PlateMate Plus robot (Thermo Fisher Scientific, Handforth, Cheshire, United Kingdom) and replication of assay plates by the HummingBird robot (Genomic Solutions, Inc., Ann Arbor, Michigan, USA).
  • a compound of the invention shows therapeutic efficacy especially against disorders dependent on protein kinase, especially proliferative diseases mediated by JAK/TYK kinase activity.
  • Exemplified compounds show JAK2 enzyme inhibitory activities with IC50-values of 0.1 to 1000 nM as shown in the following table:

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Diabetes (AREA)
  • Immunology (AREA)
  • Hematology (AREA)
  • Dermatology (AREA)
  • Endocrinology (AREA)
  • Neurology (AREA)
  • Obesity (AREA)
  • Pain & Pain Management (AREA)
  • Rheumatology (AREA)
  • Oncology (AREA)
  • Biomedical Technology (AREA)
  • Transplantation (AREA)
  • Neurosurgery (AREA)
  • Emergency Medicine (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Heterocyclic Carbon Compounds Containing A Hetero Ring Having Nitrogen And Oxygen As The Only Ring Hetero Atoms (AREA)
  • Nitrogen And Oxygen Or Sulfur-Condensed Heterocyclic Ring Systems (AREA)
  • Plural Heterocyclic Compounds (AREA)

Abstract

The invention relates to 2,7-disubstituted benzoxazole and 2,4-disubstituted oxazolo[5,4- c]pyridine compounds of the formula I given below, as well as salts thereof, processes for the preparation thereof, the application thereof in a process for the treatment of the human or animal body, these compounds for use in the treatment (including prophylaxis) of the animal, especially human, body (especially with regard to a proliferative disease), the use thereof - alone or in combination with one or more other pharmaceutically active compounds - for the treatment especially of a protein tyrosine kinase mediated disease (such as a tumor disease) or for the manufacture of a pharmaceutical preparation for use in the treatment of such a disease, a method for the treatment of such a disease and a pharmaceutical preparation for the treatment of a disease as mentioned. The compounds are of the formula I, wherein the symbols are as defined in the description. The compounds inhibit, for example, JAK2 and JAK3.

Description

BENZOXAZOLES AND OXAZOLOPYRIDINES BEING USEFUL AS JANUS KINASES INHIBITORS
The invention relates to 2,7-disubstituted benzoxazole and 2,4-disubstituted-oxazolo[5,4- c]pyιϊdine compounds of the formula I given below, as well as salts thereof, processes for the preparation thereof, the application thereof in a process for the treatment of the human or animal body, these compounds for use in the treatment (including prophylaxis) of the animal, especially human, body (especially with regard to a proliferative disease), the use thereof - alone or in combination with one or more other pharmaceutically active compounds - for the treatment especially of a protein tyrosine kinase mediated disease (such as a tumor disease) or for the manufacture of a pharmaceutical preparation for use in the treatment of such a disease, a method for the treatment of such a disease and a pharmaceutical preparation for the treatment of a disease as mentioned.
Janus kinases (JAKs) form a family of intracellular protein tyrosine kinases with four members, JAK1 , JAK2, JAK3 and TYK2. These kinases are important in the mediation of cytokine receptor signaling which induces various biological responses including cell proliferation, differentiation and apoptosis. Knock-out experiments in mice have shown that JAKs are inter alia important in hematopoiesis. In addition, JAK2 was shown to be implicated in myeloproliferative diseases and cancers. JAK2 activation by chromosome re-arrangements and/or loss of negative JAK/STAT (STAT = signal transducing and activating factor(s)) pathway regulators has been observed in hematological malignancies as well as in certain solid tumors.
It has now been found that the 2,7-substituted benzoxazole and 2,4-disubstituted-oxazo- lo[5,4-c]pyridine compounds of the formula I, described below, have advantageous pharmacological properties and inhibit, for example, the tyrosine kinase activity of Janus kinases, such as JAK2 kinase and/or JAK3- (but also JAK-1-) kinase. Hence, the compounds of formula I are suitable, for example, to be used in the treatment of diseases depending on the tyrosine kinase activity of JAK2 (and/or JAK3) kinase, especially proliferative diseases such as tumor diseases, leukaemias, polycythemia vera, essential thrombocythemia, and myelofibrosis with myeloid metaplasia. Through the inhibition of JAK-3 kinase, compounds of the invention also have utility as immunosuppressive agents, for example for the treatment of diseases such as organ transplant rejection, lupus erythematodes, multiple sclerosis, rheumatoid arthritis, psoriasis, dermatitis, Crohn's disease, type-1 diabetes and complications from type-1 diabetes.
The invention relates to compounds of the formula I,
Figure imgf000003_0001
wherein
X is N or CR5, wherein R5 is halo, cyano, hydroxyl, d-Cy-alkoxy, C1-C7-alkyl, amino, N- mono- or N.N-di-d-Cy-alkyl or preferably hydrogen;
R1 is unsubstituted or substituted aryl or is unsubstituted or substituted heterocyclyl; and R2 is unsubstituted or substituted aryl or is unsubstituted or substituted heterocyclyl which is bound via a ring carbon atom (to the carbon at position 7 of the benzoxazole ring in formula I or (if X is N) to the carbon at position 4 in the oxazolopyridine ring of formula I); R3 is cyano, hydroxyl, C1-C7-SIkVi, amino, N-mono- or N.N-di-d-CT'-alkyl or preferably hydrogen; and
R4 is hydroxyl, amino or preferably hydrogen; or salts thereof.
Preferably, the invention relates to a compound of the formula I wherein X is CR5 or N, wherein R5 is halo, cyano, hydroxyl, C1-C7^IkOXy, Ci-C7-alkyl, amino, N- mono- or N,N-di-CrC7-alkyl or preferably hydrogen;
R1 is unsubstituted or substituted aryl or is unsubstituted or substituted heterocyclyl, especially R1 is phenyl, naphthyl, indanyl, pyridyl, oxo-1H-pyridyl, indolyl, dihydroindolyl or oxo-dihydroindolyl, each of which is bound via a ring carbon atom and is unsubstituted or substituted by one to three moieties independently selected from the group consisting of C1- C7-alkyl, amino-d-d-alkyl, halo-d-C7-alkyl, N-d-d-alkanoylamino-d-d-alkyl, N-C1-C7- alkanesulfonyl-amino-d-C7-alkyl, pyrrolidino-d-C7-alkyl, oxo- pyrrolidino-C1-C7-alkyl, piperidino-C1-C7-alkyl, piperazin-1-yl-d-d-alkyl, 4-(C1-C7-alkyl, C1-C7-alkoxy-C1-C7-alkyl, halo-d-C7-alkyl or Cs-do-cycloalkyO-piperazin-i-yl-d-d-alkyl, 4-(amino-C1-C7-alkyl)- piperazin-1-yl-CrC^alkyl, 4-[N-mono- or N,N-di-(C1-C7-alkylamino)-C1-C7-alkyl]-piperazin-1- yl-d-C7-alkyl, morpholino-C1-C7-alkyl, thiomorpholino-C1-C7-alkyl, S-mono- or S,S-dioxo- thiomorpholino-C1-C7-alkyl, carbamoyl-d-d-alkyl, [N-mono- or N,N-di-(CrC7-alkyl)- carbamoylJ-CVCValkyl, C1-C7-alkanesulfinyl-C1-C7-alkyl, d-C^alkanesulfonyl-d-CT-alkyl, halo, hydroxyl, d-C7-alkoxy, amino, N-mono- or N.N-dKd-CT-alkyO-amino, C1-C7- alkanoylamino, pyrrolidino, oxo-pyrrolidino, piperidino, piperazin-1-yl, 4-(d-C7-alkyl, C1-C7- alkoxy-d-C7-alkyl, halo-d-C7-alkyl or C3-C10-cycloalkyl)-piperazin-1-yl, 4-(amino-d-C7- alkyl)-piperazin-1-yl, 4-[N-mono- or N.N-dHd-d-alkylaminoJ-d-d-alkyrj-piperazin-i-yl, morpholino, thiomorpholino, S-oxo- or S,S-dioxothiomorpholino, d-C7-alkanesulfonylamino, carbamoyl, N-mono- or N,N-di-(d-C7-alkyl, d-Cr-alkoxy-d-Cr-alkyl, amino-CT-C^alkyl and/or (N'-mono- or N',N'-di-(C1-C7-alkyl)-amino-C1-C7-alkyl)-carbamoyl, pyrrolidin-1- carbonyl, piperidin-1-carbonyl, piperazin-1-carbonyl, 4-(C1-C7-alkyl)piperazin-1-carbonyl, morpholin-1-carbonyl, thiomorpholin-1-carbonyl, S-oxo- or S,S-dioxothiomorpholin-1- carbonyl, sulfo, d-C^alkanesulfonyl, d-C7-alkanesulfinyl, sulfamoyl, N-mono- or N,N-di- (CTCT-alkylJ-sulfamoyl, morpholinosulfonyl, thiomorpholinosulfonyl, cyano and nitro, R2 is phenyl, naphthyl, indanyl, pyridyl, oxo-1H-pyridyl, pyrazolyl, thiophenyl, indolyl, dihydroindolyl, oxo-dihydroindolyl, quinolinyl, isoquinolinyl or 7/-/-benzoimidazolyl, each of which is bound via a ring carbon atom and is unsubstituted or substituted by one to three moieties independently selected from the group consisting of d-Cy-alkyl, hydroxyl-C^C6- alkyl, amino-d-C7-alkyl, halo-CrCjr-alkyl, N-d-CT-a^anoylamino-d-d-alkyl, N-C1-C7-
Figure imgf000004_0001
C7-alkyl, pyrrolidino-C^C^alkyl, oxo-pyrrolidino-CrCr-alkyl, piperidino-C^C^alkyl, piperazin- 1-yl-d-C7-alkyl, 4-(d-C7-alkyl, CTC^alkoxy-d-Cralkyl, halo-C^Cralkyl or C3-C10- cycloalkylJ-piperazin-i-yl-CrC^alkyl, 4-(amino-C1-C7-alkyl)-piperazin-1-yl-C1-C7-alkyl, 4-[N- mono- or N,N-di-(C1-C7-alkylamino)-C1-C7-alkyl]-piperazin-1-yl-C1-C7-alkyl, 4-(C1-C7- alkanoyO-piperazin-i-yl-CrC^alkyl, oxo-piperazin-1-yl-d-C7-alkyl, pyrrolidin-1-carbonyl-d- C7-alkyl, piperidin-i-carbonyl-C^Cr-alkyl, piperazin-i-carbonyl-CTC^alkyl, 4-(C1-C7- alkyl)piperazin-1-carbonyl-d-C7-alkyl, morpholin-1-carbonyl-C^C^alkyl, thiomorpholin-1- carbonyl-CrCT-alkyl, S-oxo- or S.S-dioxothiomorpholin-i-carbonyl-d-Cr-alkyl, C1-C^aIkOXy- CrCT-alkyl, halo-C^C^alkyl, d-C7-alkanoyl or Cs-C^-cycloalkyO-oxo-piperazin-i-yl-d-CTr- alkyl, morpholino-CrC^alkyl, thiomorpholino-CrCT-alkyl, S-mono- or S,S-dioxo- thiomoφholino-CrC^alkyl, imidazol-1-yl-d-C7-alkyl, carbamoyl-d-C7-alkyl, [N-mono- or N,N-di-(C1-C7-alkyl)-carbamoyl]-C1-C7-alkyl, CrCz-alkanesulfinyl-d-CT-alkyl, C1-C7- alkanesulfonyl-d-C7-alkyl, halo, hydroxyl, C1-C^aIkOXy, amino, N-mono- or N,N-di-(d-C7- alkyl)-amino, d-C7-alkanoylamino, pyrrolidino, oxo-pyrrolidino, piperidino, piperazin-1-yl, 4- - A -
(d-C7-alkyl, Ci-C7-alkoxy-d-C7-alkyl, halo-d-C7-alkyl or C3-Ci0-cycloalkyl)-piperazin-1-yl, A-
(amino-d-C7-alkyl)-piperazin-1-yl, 4-[N-mono- or N,N-di-(C1-Cτ-alkylamino)-C1-C7-alkyl]- piperazin-1-yl, morpholino, thiomorpholino, S-oxo- or S,S-dioxothiomorphplino, d-C7-alkane- sulfonylamino, carbamoyl, N-mono- or N,N-di-(C1-C7-alkyl, d-C7-alkoxy-d-C7-alkyl, amino- d-C7-alkyl and/or (N'-mono- or N^'-dHd-C^alkyO-amino-d-d-alkyO-carbamoyl, pyrrolidin-1-carbonyl, piperidin-1-carbonyl, piperazin-1-carbonyl, 4-(C1-C7-alkyl)piperazin-1- carbonyl, morpholin-1-carbonyl, thiomorpholin-1-carbonyl, S-oxo- or S,S-dioxothiomorpholin-
1-carbonyl, sulfo, d-d-alkanesulfonyl, d-d-alkanesulfinyl, sulfamoyl, N-mono- or N,N-di-
(d-C7-alkyl)-sulfamoyl, moφholinosulfonyl, thiomorpholinosulfonyl, S-oxo- thiomorpholinosulfonyl, S,S-dioxothiomorpholinosulfonyl, cyano and nitro, preferably in the meta, (more preferably once) in the meta (preferably up to once) and the para or in the para position
R3 is cyano, hydroxyl, d-C7-alkyl, amino, N-mono- or N.N-di-d-C^alkyl or preferably hydrogen; and
R4 is hydroxyl, amino or preferably hydrogen;
or a pharmaceutically acceptable salt thereof.
Also preferably, the invention relates to a compound of the formula I wherein X is CR5 or N, wherein R5 is halo, cyano, hydroxyl, C1-C^aIkOXy, C1-C7-alkyl, amino, N- mono- or N.N-di-d-Cr-alkyl or preferably hydrogen;
R1 is unsubstituted or substituted aryl or is unsubstituted or substituted heterocyclyl, R2 is phenyl, naphthyl, pyridyl, pyrazolyl, thiophenyl, quinolinyl, isoquinolinyl or fH-benzo- imidazolyl, each of which is bound via a ring carbon atom and is unsubstituted or substituted by one to three moieties independently selected from the group consisting of d-C7-alkyl, amino-d-C7-alkyl, N-d-CT-alkanoylamino-d-d-alkyl, N-d-C7-alkanesulfonyl-amino-d-C7- alkyl, carbamoyl-d-C7-alkyl, [N-mono- or N,N-di-(C1-C7-alkyl)-carbamoyl]-C1-C7-alkyl, Ci-C7- alkanesulfinyl-Ci-C7-alkyl, d-Cralkanesulfonyl-d-d-alkyl, halo, hydroxyl, d-C7-alkoxy, amino, N-mono- or N.N-dKd-d-alkyO-amino, CrC7-alkanoylamino, d-C7-alkane- sulfonylamino, carbamoyl, N-mono- or N,N-di-(C,-C7-alkyl)-carbamoyl, sulfo, d-C^alkane- sulfonyl, sulfamoyl, N-mono- or N,N-di-(C1-C7-alkyl)-sulfamoyl, morpholinosulfonyl, thiomorpholinosulfonyl, cyano and nitro, preferably in the meta, (more preferably once) in the meta (preferably up to once) and the para or in the para position R3 is cyano, hydroxyl, d-C7-alkyl, amino, N-mono- or N,N-di-d-C7-alkyl or preferably hydrogen; and
R4 is hydroxyl, amino or preferably hydrogen;
or a pharmaceutically acceptable salt thereof.
More preferably, the invention relates to a compound of the formula I wherein
X is CR5 or N, wherein R5 is halo, cyano, hydroxyl, d-C7-alkoxy, d-C7-alkyl, amino, N- mono- or N,N-di-d-C7-alkyl or preferably hydrogen;
R1 is unsubstituted or substituted aryl or is unsubstituted or substituted heterocyclyl,
R2 is phenyl, naphthyl, pyridyl, pyrazolyl, thiophenyl, quinolinyl, isoquinolinyl or 7H-benzo- imidazolyl, each of which is bound via a ring carbon atom and is unsubstituted or substituted by one to three moieties independently selected from the group consisting of d-C^alkyl, amino-d-d-alkyl, N-C1-C7-alkanoylamino-C1-C7-alkyl, N-d-d-alkanesulfonyl-amino-d-d- alkyl, carbamoyl-d-C^alkyl, [N-mono- or N.N-dHd-d-alkyO-carbamoyrj-d-d-alkyl, C1-C7- alkanesulfinyl-C,-C7-alkyl, d-Cralkanesulfonyl-d-d-alkyl, halo, hydroxyl, d-C7-alkoxy, amino, N-mono- or N,N-di-(C1-C7-alkyl)-amino, d-C^alkanoylamino, C1-C7- alkanesulfonylamino, carbamoyl, N-mono- or N,N-di-(C1-C7-alkyl)-carbamoyl, sulfo, C1-C7- alkanesulfonyl, sulfamoyl, N-mono- or N,N-di-(C1-C7-alkyl)-sulfamoyl, morpholinosulfonyl, thiomorpholinosuJfonyl, cyano and nitro,
R3 is cyano, hydroxyl, d-C^alkyl, amino, N-mono- or N,N-di-d-C7-alkyl or preferably hydrogen; and
R4 is hydroxyl, amino or preferably hydrogen;
or a pharmaceutically acceptable salt thereof.
The invention relates very especially to a compound of the formula I, wherein X is CH or N;
R1 is phenyl, (especially 3,4,5-)trimethoxyphenyr, (especially 3,4- or 3,5-)dimethoxyphenyl*, (especially 4-)morpholinophenyl, (especially 4-) N-(2-methoxyethyl)-carbamoylphenyr, or (especially 4-)N,N-(2-dimethylamino-ethyl)-carbamoylphenyr, (especially 4-)dimethylamino- carbonyl-(especially 3-)methyl-phenyl*, (especially 4-)-(preferably 4-)-(2-methoxy-ethyl- piperazin-(especially 1 -)yl-(especially 3-)-methyl-phenyl, (especially 4-)pyrrolidin-1 -carbonyl- (especially 3-)methyl-phenyl*, (especially 3-)methyl-(especially 4-)-4-methylpiperazin-1- carbonyl-phenyl, (especially 3- or 4-)4-methyl-piperazin-1-yl-phenyr, (especially 4-)-4-ethyl- piperazin-1-yl-(especially 3-)methyl-phenyl*, (especially 4-)-4-methyl-piperazin-1-yl- (especially 3-)cyano-phenyl, (especially 4-)-piperazin-1-yl-phenyl, (especially 4-)-4- cyclopropyl-piperazin-1 -yl-phenyl, (especially 4-)-4-(2-dimethylaminoethyl)-piperazin-1 -yl- (especially 3-)methyl-phenyl*, (especially 4-)4-isopropyl-piperazin-1-yl)-(especially 3-)methyl- phenyl*, (especially 4-)N,N-diethylaminocarbonyl-(especially 3-)methyl-phenyl*. (especially 4- )4-ethylpiperazin-1-carbonyl-(especially 3-)methyl-phenyl, (especially 4-)-(4-ethylpiperazin-1- ylmethyl)-(espedally 3-)methyl-phenyl, (especially 4-)N-methylaminocarbonyl-(especially 3- )methylphenyl, (especially 4-)-4-(3,3,3-trifluoropropyl)-piperazin-1 -yi-(especially 3-)methyl- phenyl, (especially 4-)-4-(2-(N',N'-dimethylamino)ethyl-aminocarbonyl-(especially 3-)methyl- phenyl, (especially 4-)-methanesulfonyl-phenyl*. (especially 4-)[(especially 2-)-oxo-pyrrolidin- 1-yl]-phenyl, (especially 4-)N,N-diethylaminocarbonyl-(especially 3-)methoxyphenyl, (especially 3-)-4-methylpiperazin-1-yl-(especially 4-)methyl-phenyl, (especially 3-)-4- methylpiperazin-1-yl-(especially 4-)methoxy-phenyr, (especially 3- or 4-)-morpholinomethyl- (especially 4- or 3-)methyl-phenyl, (especially 2-)acetylamino-indan-(especially 5-)yl, (especially 2-)oxo-2,3-dihydroindol-(especially 5-)yl, (especially 4-)methylsulfinylphenyl, (especially 4-)methoxyphenyl, (especially 4-)methyl-(especially 3-)methoxyphenyl, (especially 4-)-N-(2-methoxyethyl)-aminocarbonyl-phenyl, (especially 4-)N,N-dimethylcarbamoyl-phenyl, (especially 3-)methanesulfonylamino-phenyl, (especially 4-)methoxycarbonyl-(especially 3- )methoxy-phenyl, (especially 4-)N,N-dimethylcarbamoyl-(especially 3-)methoxy-phenyl, (especially 4-)-(4-cyclopropyl-piperazin-1-yl)-(especially 3-)methyl-phenyl*, (especially 4-}-N- (2-(N',N'-dimethylaminoethyl)-N-methyl-carbamoyl-(especially 3->methyl-phenyr, 1,3- dimethyl-oxo-1H-pyridine-5-yl, (especially 3- or 4-)morpholino-(especially 4- or 3-)methyl- phenyl*, (especially 4-)morpholinomethyl-(especially 3-)methyl-phenyl, (especially 4- )morpholin-1-carbonyl-(especially 3-)methyl-phenyl, (especially 4-)-N-2- (methoxyethyl)aminocarbonyl-(especially 3-)methyl-phenyI, (especially 4-)-N-(3-N'.N'- dimethylaminopropyl)amino-carbonyl-(especially 3-)methyl-phenyl, (especially 5-)-methyl- (especially 6-)methoxy-pyridin-3-yl, (especially 4-)dimethylcarbamoyl-(especially 3,5- )dimethyl-phenyl, (especially 4-)dimethylcarbamoyl-(especially 3-)ethyl-phenyl, (especially A- (4-)N,N-dimethylcarbamoyl-(especially 3-)methyl-phenyl or (especially 4-)morpholino- (especially 3-)cyano-phenyl;
R2 is phenyl, (especially 4-)methylphenyl, (especially 3-)methylphenyl, (especially 2-)me- thylphenyl, (especially 4-)-hydroxymethyl-phenyl, (especially 4-)aminomethyl-phenyl, (especially 3-)aminomethyl-phenyl, (especially 4-)acetylaminomethyl-phenyl*, (especially 4- )methanesulfonylaminomethyl-phenyl, (especially 3-)acetylaminomethyl-phenyl, (especially 3-)methanesulfonylaminomethyl-phenyl*, (especially 4-)methanesulfonylaminomethyl-phenyl, (especially 4-)(N-methylcarbamoyl)-methylphenyr, (especially 4-)methanesulfinylmethyl- phenyl, (especially 4-)methanesulfonylmethylphenyl, (especially 3-)chlorophenyl, (especially 3-)hydroxyphenyl, (especially 4-)methoxyphenyl, (especially 3-)methoxyphenyl*, (especially 2-)methoxyphenyl, (especially 4-)aminophenyl, (especially 3-)aminophenyl, (especially 2- )aminophenyl, (especially 3-)N-methylamino-phenyl, (especially 4-)N,N-dimethylamino- phenyl*. (especially 4-)acetylamino-phenyl, (especially 3-)acetylamino-phenyl, (especially 4- )methanesulfonylamino-phenyl*, (especially 4-)methanesulfonylamino-phenyl, (especially 3-) methanesulfonylamino-phenyl*, (especially 4-)carbamoylphenyl, (especially 3-)carbamoyl- phenyl, (especially 4-)(N-methyl-carbamoyl)-phenyl, (especially 4-)(N,N-dimethyl-carbamoyl)- phenyl, (especially 4-)methanesulfonylphenyl*. (especially 3-)methanesulfonylptienyl, (especially 4-)sulfamoylphenyl*, (especially 4-)(N-methylsulfamoyl)-phenyr, (especially 4- )[N,N-(dimethyl)-sulfamoyl]-phenyl, (especially 4-)morpholinosulfonylphenyl, (especially A- )cyanophenyl, (especially 3-)cyanophenyl, (especially 3-)nitrophenyl, (especially 3-)amino-4- methyl-phenyl, (especially 3-)amino-4-methoxyphenyl, (especially 3-)amino-4-chlorophenyl, (especially 4-)methoxy-3-nitrophenyl, (especially 4-)morpholin-4-ylmethyl-phenyl, (especially 3-)methyl-(especially 4-)morpholin-4-ylmethyl-phenyr, (especially 3-)fluoro-(especially 4- )morpholin-4-ylmethyl-phenyr, (especially 4-)S,S-dioxothiomorpholin-4-ylmethylphenyl*, (especially 3,5-)difluoro-(especially 4-) morpholin-4-ylmethyl-phenyl, (especially 3-)fluoro- (especially 4-)S,S-dioxothiomorpholin-4-ylmethyl-phenyr, (especially 3,5-)difluoro-(especially 4-)S,S-dioxothiomorpholin-4-ylmethyl-phenyr, (especially 3-)trifluoromethyl-(especially 4- )morpholin-4-ylmethyl-phenyl, (especially 3,5-)difluoro-(especially 4-)[(preferably 4-)acetyl- piperazin-1-yl]methyl-phenyl, (especially 3,5-)difluoro-(especially 4-)(preferably 4-)piperazin- 1-yl]methyl-phenyl, (especially 4-)f (preferably 4-)methyl-piperazin-1-yl]methyl-phenyl, (especially 3,5-)difluoro-(especially 4-)[(especially 3-)oxo-piperazin-1-yl]methyl-phenyl (especially 3,5-)difluoro-(especially 4-)[(preferably 4-)methyl-(especially 3-)oxo-piperazin-1- yl]methyl-phenyl, (especially 4-)imidazol-1-ylmethyl-phenyl, (especially 4-)-4-methylpiperazin- 1-carbonyl-phenyl, (especially 4-)morpholin-4-carbonyl-phenyr, (especially 2- or 3-)fluoro- (especially 4-)morpholin-4-carbonyl-phenyl*, (especially 3-)methyl-(especially 4-)morpholin-4- carbonyl-phenyl*, (especially 3,5-)difluoro-(especially 4-)morpholin-4-carbonyl-phenyl, (especially 4-)S,S-dioxothiomorpholin-4-carbonyl-phenyl, (especially 3-)-fluoro-(especially 4- )S,S-dioxothiomorpholin-4-carbonyl-phenyr, (especially 4-)morpholin-4-carbonylmethyl- phenyl*, (especially 3-)fluoro-(especially 4-)morpholin-4-carbonylmethyl-phenyl*, [(especially 4-)morpholin-4-carbonyl-(1 ,1 ,dimethyl)-methyl]-phenyl, (especially 4-)S,S- dioxothiomorpholin-4-carbonylmethyl-phenyl*, (especially 3-)fluoro-(especially 4-)S,S- dioxothiomorpholin-4-carbonylmethyl-phenyl*, 2H-pyrazol-(especially 3-)yl, (especially 5-)N- methylcarbamoyl-thiophenyl, (especially 4-)pyridyl, (especially 3-)pyridyl, (especially 2-
)pyridyl, (especially 6-)methoxy-pyridin-(especially 3-)yl, 1 H-benzoimidazol-(especially 5-)yl, quinolin-(especially 6-)yl or isoquinolin-(especially 4-)yl,
(where the moieties marked with an asterisk (*) are especially preferred, as are the moieties where the position after "especially" is given) and each of R3 and R4 is hydrogen; or a pharmaceutically acceptable salt thereof.
The invention relates especially also to a compound of the formula I, wherein X is CH or N;
R1 is (especially 3,4,5- )trimethoxyphenyl, (especially 3,4- or 3,5-)dimethoxyphenyl, (especially 4-)morpholinophenyl, (especially 4-) N-(2-methoxyethyl)-carbamoylphenyl, or (especially 4-)N,N-(2-dimethylamino-ethyl)-carbamoylphenyl,
R2 is phenyl, (especially 4-)methylphenyl, (especially 3-)methylphenyl, (especially 2-)me- thylphenyl, (especially 4-)aminomethyl-phenyl, (especially 3-)aminomethyl-phenyl, (especially 4-)acetylaminomethyl-phenyl*, (especially 4-)methanesulfonylaminomethyl- phenyl, (especially 3-)acetylaminomethyl-phenyl, (especially 3-)methanesulfonylamino- methyl-phenyl, (especially 4-)methanesulfonylaminomethyl-phenyl, (especially 4-)(N- methylcarbamoyl)-methylphenyl, (especially 4-)methanesulfinylmethylphenyl, (especially 4- )methanesulfonylmethylphenyl, (especially 3-)chlorophenyl, (especially 3-)hydroxyphenyl, (especially 4-)methoxyphenyl, (especially 3-)methoxyphenyl*, (especially 2-)methoxyphenyl1 (especially 4-)aminophenyl, (especially 3-)aminophenyl, (especially 2-)aminophenyl, (especially 3-)N-methylamino-phenyl, (especially 4-)N,N-dimethylamino-phenyl, (especially 4-)acetylamino-phenyl, (especially 3-)acetylamino-phenyl, (especially 4-)methanesulfonyl- amino-phenyl*. (especially 4-)methanesulfonylamino-phenyl, (especially 3-) methanesulfonyl- amino-phenyl*, (especially 4-)carbamoylphenyl, (especially 3-)carbamoylphenyl, (especially 4-)(N-methyl-carbamoyl)-phenyl, (especially 4-)(N,N-dimethyl-carbamoyl)-phenyl, (especially 4-)methanesulfonylphenyl*. (especially 3-)methanesulfonylphenyl, (especially 4-)sulf- amoylphenyl*, (especially 4-)(N-methylsulfamoyl)-phenyl, (especially 4-)[N,N-(dimethyl)- sulfamoylj-phenyl, (especially 4-)morpholinosulfonylphenyl, (especially 4-)cyanophenyl, (especially 3-)cyanophenyl, (especially 3-)nitrophenyl, (especially 3-)amino-4-methyl-phenyl, (especially 3-)amino-4-methoxyphenyl, (especially 3-)amino-4-chlorophenyl, (especially 4- )methoxy-3-nitrophenyl, 2H-pyrazol-(especially 3-)yl, (especially 5-)N-methylcarbamoyl- thiophenyl, (especially 4-)pyridyl, (especially 3-)pyridyl, (especially 2-)pyridyl, (especially 6- )methoxy-pyridin-(especially 3-)yl, 1 H-benzoimidazol-(especially 5-)yl, quinolin-(especially 6- )yl or isoquinolin-(especially 4-)yl, (where the moieties marked with an asterisk (*) are especially preferred, as are the moieties where the position after "especially" is given) and each of R3 and R4 is hydrogen; or a pharmaceutically acceptable salt thereof.
The invention relates especially to the compounds of the formula I given in the Examples, as well as the methods of manufacture described therein.
The general terms used hereinbefore and hereinafter preferably have within the context of this disclosure the following meanings, unless otherwise indicated (where one or more up to all more general expressions in embodiments characterized as preferred above or below can be replaced with a more specific definition, thus leading to a more preferred embodiment of the invention, respectively):
Where the plural form (e.g. compounds, salts) is used, this includes the singular (e.g. a single compound, a single salt). "A compound" does not exclude that (e.g. in a pharmaceutical formulation) more than one compound of the formula I (or a salt thereof) is present.
Any asymmetric carbon atoms (for example in compounds of formula I carrying a substituent with an 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 as pure isomers, preferably as enantiomer-pure diastereomers.
The invention also relates to tautomers where tautomeric forms are possible.
The prefix "lower" or "Ci-C7" denotes a radical having up to and including a maximum of 7, especially up to and including a maximum of 4 carbon atoms, the radicals in question being either linear or branched with single or multiple branching.
CrCy-alkyl is preferably alkyl with from and including 1 up to and including 7, preferably from and including 1 to and including 4, and is linear or branched; preferably, lower alkyl is butyl, such as n-butyl, sec-butyl, isobutyl, tert-butyl, propyl, such as n-propyl or isopropyl, ethyl or preferably methyl.
Halogen (or halo) is especially fluorine, chlorine, bromine, or iodine, especially fluorine, chlorine, or bromine.
That a heterocyclyl is bound via a ring carbon atom means that it is not bound via a nitrogen atom to the rest of the molecule in formula I (that is, to the 7-position of the central benzoxazole ring or if X is N to the 4-position of the central oxazolopyridine ring).
In unsubstituted or substituted aryl, aryl is preferably an aromatic moiety with 6 to 14 ring carbon atoms, more preferably with 6 to 10 ring carbon atoms, such as phenyl or naphthyl, which is unsubstituted or substituted by one or more, preferably up to three, more preferably up to two substituents independently selected from the group consisting of unsubstituted or substituted heterocyclyl as described below, especially pyrrolidinyl, such as pyrrolidino, oxopyrrolidinyl, such as oxopyrrolidino, C1-C7-alkyl-pyrrolidinyl, 2,5-di-(C1-C7alkyl)pyrrolidinyl, such as 2,5-di-(C1-C7alkyl)-pyrrolidino, tetrahydrofuranyl, thiophenyl, C1-C7-alkylpyrazolidinyl, pyridinyl, d-C7-alkylpiperidinyl, piperidino, piperidino substituted by amino or N-mono- or N,N-di-[lower alkyl, phenyl, d-Cr-alkanoyl and/or phenyl-lower alkyl)-amino, unsubstituted or N-lower alkyl substituted piperidinyl bound via a ring carbon atom, piperazino, lower alkyl- piperazino, morpholino, thiomorpholino, S-oxo-thiomorpholino or S,S-dioxothiomorpholino; d-d-alkyl, amino-Ci-C7-alkyl, N-d-d-alkanoylamino-d-Cr-alkyl, N-d-C7-alkanesulfonyl- amino-C,-C7-alkyl, carbamoyl-C1-C7-alkyl, [N-mono- or N,N-di-(C1-C7-alkyl)-carbamoyl]-C1- C7-alkyl, d-d-alkanesulfinyl-d-d-alkyl, d-d-alkanesulfonyl-d-d-alkyl, phenyl, naphthyl, mono- to tri-[d-C7-alkyl, halo and/or cyano]-phenyl or mono- to tri-[d-C7-alkyl, halo and/or cyano]-naphthyl; C3-C8-cycloalkyl, mono- to tri-[d-d-alkyl and/or hydroxy]-C3-Ca-cycloalkyl; halo, hydroxy, lower alkoxy, lower-alkoxy-lower alkoxy, (lower-alkoxy)-lower alkoxy-lower alkoxy, halo-d-d-alkoxy, phenoxy, naphthyloxy, phenyl- or naphthyl-lower alkoxy; amino- d-C7-alkoxy, lower-alkanoyloxy, benzoyloxy, naphthoyloxy, formyl (CHO), amino, N-mono- or N,N-di-(d-C7-alkyl)-amino, d-d-alkanoylamino, d-C7-alkanesulfonylamino, carboxy, lower alkoxy carbonyl, e.g.; phenyl- or naphthyl-lower alkoxycarbonyl, such as benzyloxy- carbonyl; d-C7-alkanoyl, such as acetyl, benzoyl, naphthoyl, carbamoyl, N-mono- or N1N- disubstituted carbamoyl, such as N-mono- or N,N-di-substituted carbamoyl wherein the substitutents are selected from lower alkyl, (lower-alkoxy)-lower alkyl and hydroxy-lower alkyl; amidino, guanidino, ureido, mercapto, lower alkylthio, phenyl- or naphthylthio, phenyl- or naphthyl-lower alkylthio, lower alkyl-phenylthio, lower alkyl-naphthylthio, halogen-lower alkylmercapto, sulfo (-SO3H), lower alkanesulfonyl, phenyl- or naphthyl-sulfonyl, phenyl- or naphthyl-lower alkylsulfonyl, alkylphenylsulfonyl, halogen-lower alkylsulfonyl, such as trifluoromethanesulfonyl; sulfonamido, benzosulfonamido, azido, azido-d-C7-alkyl, especially azidomethyl, d-Cr-alkanesulfonyl, sulfamoyl, N-mono- or N,N-di-(d-C7-alkyl)- sulfamoyl, morpholinosulfonyl, thiomorpholinosulfonyl, cyano and nitro; where each phenyl or naphthyl (also in phenoxy or naphthoxy) mentioned above as substituent or part of a substituent of substituted alkyl (or also of substituted aryl, heterocyclyl etc. mentioned herein) is itself unsubstituted or substituted by one or more, e.g. up to three, preferably 1 or 2, substituents independently selected from halo, especially fluoro, chloro, bromo or iodo, halo- lower alkyl, such as trifluoromethyl, hydroxy, lower alkoxy, azido, amino, N-mono- or N,N-di- (lower alkyl and/or d-d-alkanoy^-amino-, nitro, carboxy, lower-alkoxycarbonyl, carbamoyl, cyano and/or sulfamoyl.
In a preferred embodiment, the substituents of substituted aryl are up to three substituents independently selected from the group consisting of d-C7-alkyl, hydroxyl-d-C7-alkyl, C1-C7- alkoxy-d-C7-alkyl, amino-d-C7-alkyl, halo-d-C7-alkyl, N-C1-C7-alkanoylamino-C1-C7-alkyl, N-C1-C7-alkanesulfonyl-amino-C1-C7-alkyl, pyrrolidino-d-C7-alkyl, oxo- pyrrcHidino-d-Cr- alkyl, piperidino-CrC7-alkyl, piperazin-1-yl-d-Cτ-alkyl, 4-(C1-C7-alkyl, C1-C7^aIkOXy-C1-C7- alkyl, halo-CrC7-alkyl or Qrdo-cycloalkyO-piperazin-i-yl-d-d-alkyl, 4-(amino-C1-C7-alkyl)- piperazin-1-yl-d-C7-alkyl, 4-[N-mono- or N.N-dHd-d-alkylaminoJ-d-d-alkylj-piperazin-i- yl-C1-C7-alkyl, (C1-C7-alkoxy-C1-C7-alkyl, d-C7-alkanoyl or C3-C10-cycIoalkyl)-oxo-piperazin- 1-yl-d-C7-alkyl, morpholino-d-Cr-alkyt, thiomorpholino-d-CT-alkyl, S-mono- or S.S-dioxo- thiomorpholino-d-C7-alkyl, 4-(C1-C7-alkanoyl)-piperazin-1 -yl-d-C7-alkyl, oxo-piperazin-1 -yl- Ct-Cy-alkyl, imidazol-1-yl-CrCτ-alkyl, pyrrolidin-1-carbonyl-d-d-alkyl, pipehdin-1-carbonyl- d-Cτ-alkyl, piperazin-1-carbonyl-d-C^alkyl, 4-(C1-C7-alkyl)piperazin-1-carbonyl-C1-C7-alkyl, morpholin-i-carbonyl-d-d-alkyl, thiomorpholin-i-carbonyl-CTCTr-alkyl, S-oxo- or S1S- dioxothiomorpholin-1-carbonyl-d-Cr-alkyl, carbamoyl-C^CT-alkyl, [N-mono- or N1N^i-(C1- C^alkylJ-carbamoylj-d-d-alkyl, CrCT-alkanesulfinyl-CTC^alkyl, d-C^alkanesulfonyl-C,- C7-alkyl, halo, hydroxyl, C1-C^aIkOXy, amino, N-mono- or N,N-di-(d-C7-alkyl)-amino, C1-C7- alkanoylamino, pyrrolidino, oxo-pyrrolidino, piperidino, piperazin-1-yl, 4-(C1-C7-alkyl, C1-C7- alkoxy-C^CT-alkyl, halo-d-C7-alkyl or C3-C10-cycloalkyl)-piperazin-1-yl, 4-(amino-C!-C7- alkyl)-piperazin-1-yl, 4-[N-mono- or N,N-di-(C1-C7-alkylamino)-C1-C7-alkyl]-piperazin-1-yl, morpholino, thiomorpholino, S-oxo- or S,S-dioxothiomorpholino, d-C7-alkanesulfonylamino, carbamoyl, N-mono- or N,N-di-(C,-C7-alkyl, C1-C7-alkoxy-C1-C7-alkyl, amino-d-Cy-alkyl and/or (N'-mono- or N',N'-di-(C1-C7-alkyl)-amino-Ci-C7-alkyl)-carbamoyl, pyrrolidin-1- carbonyl, piperidin-1-carbonyl, piperazin-1-carbonyl, 4-(d-C7-alkyl)piperazin-1-carbonyl, morpholin-1-carbonyl, thiomorpholin-1-carbonyl, S-oxo- or S,S-dioxothiomorpholin-1- carbonyl, sulfo, d-C7-alkanesulfonyl, d-C7-alkanesulfinyl, sulfamoyl, N-mono- or N,N-di- (C1-C7-alkyl)-sulfamoyl, morpholinosulfonyl, thiomorpholinosulfonyl, S-oxo- thiomorpholinosulfonyl, S,S-dioxothiomorpholinosulfonyl, cyano and nitro.
In the case of R1, unsubstituted or substituted aryl is preferably phenyl, naphthyl or indanyl, each of which is unsubstituted or substituted by one to three moieties independently selected from the group consisting of d-C^alky!, ammo-d-d-alky!, halo-CrC7-alkyl, N-C1-C7-alkanoylamino-C1-C7-alkyl, N-C1-C7-alkanesulfonyl-amino-C1-C7- alkyl, pyrrolidino-C1-C7-alkyl, oxo- pyrrolidino-d-C^alkyl, piperidino-d-C^alkyl, piperazin-1- yl-d-C7-alkyl, 4-(C1-C7-alkyl, d-C^alkoxy-d-C^alkyl, halo-d-C7-alkyl or C3-C1o-cycloalkyl)- piperazin-1-yl-d-C7-alkyl, 4-(amino-C1-C7-alkyl)-piperazin-1-yl-C1-C7-alkyl, 4-[N-mono- or N.N-dKd-C^alkylaminoy-d-C^alkyfJ-piperazin-i-yl-d-Cr-alkyl, morpholino-Ci-CT-alkyl,
Figure imgf000013_0001
S-mono- or S.S-dioxo-thiomorpholino-d-CT'-alkyl, carbamoyl-Ci- Cτ-alkyl, [N-mono- or N,N-di-(C1-C7-alkyl)-carbamoyl]-C1-C7-alkyl, C^CT-alkanesulfinyl-d-CT-- alkyl, Ci-d-alkanesulfonyl-Ci-C^alkyl, halo, hydroxyl, Ci-C7-alkoxy, amino, N-mono- or N1N-
Figure imgf000013_0002
pyrrolidino, oxo-pyrrolidino, piperidino, piperazin-1-yl, 4-(C1-C7-alkyl, d-C^alkoxy-CrC^alkyl, halo-d-C^alkyl or C3-C10-cycloalkyl)- piperazin-1-yl, 4-(amino-C1-C7-alkyl)-piperazin-1-yl, 4-[N-mono- or N,N-di-(C1-C7-alkylamino)- Ct-C^alkylJ-piperazin-i-yl, morpholino, thiomorpholino, S-oxo- or S,S-dioxothiomorpholino,
Figure imgf000013_0003
alkyl, amino-d-C7-alkyl and/or (N'-mono- or N',N'-di-(C1-d-alkyl)-amino-Ci-d-alkyl)- carbamoyl, pyrrolidin-1-carbonyl, piperidin-1-carbonyl, piperazin-1-carbonyl, 4-(C1-C7- alkyl)piperazin-1-carbonyl, morpholin-1-carbonyl, thiomorpholin-1-carbonyl, S-oxo- or S1S- dioxothiomorpholin-1-carbonyl, sulfo,
Figure imgf000013_0004
sulfamoyl, N-mono- or N,N-di-(C1-C7-alkyl)-sulfamoyl, morpholinosulfonyl, thiomorpholinosulfonyl, cyano and nitro; for example, it can bpreferably be phenyl or naphthyl that is substituted by one or more, especially one to four substituents independently selected from the group consisting of d-C7-alkoxy, carbamoyl, N-mono- or N,N-di-(C1-C7-alkyl and/or d-C^alkyloxy- d-C7-alkyl)-carbamoyl, N-mono- or N,N-di-{[unsubstituted, N'-mono- or N',N'-di-(d-C7- alkyl)-substituted]-carbamoyl, sulfamoyl, N-mono- or N,N-di-(C1-C7-alkyl)-sulfamoyl, piperidino, piperazino, N-d-C7-alkylpiperazino, morpholino, thiomorpholino, S- oxothiomorpholino and S,S-dioxothiomorpholino, in the case of R2, unsubstituted or substituted aryl is preferably phenyl or naphthyl that is unsubstituted or substituted by one or more, especially up to three, more especially up to two, substituents, preferably not in ortho- position, more preferably with not more than one substituent in meta-position, most preferably with one substituent in meta- and/or one substituent in para position, most preferably with one substituent in meta-position or especially one in para-position, where the substituents are independently selected from the group consisting of d-C7-alkyl, amino-d- C7-alkyl, N-d-C^alkanoylamino-d-CT-alkyl, N-d-CT-alkanesulfonyl-amino-d-C^alkyl,
Figure imgf000014_0001
[N-mono- or N,N-di-(C1-C7-alkyl>-carbamoyl}-C1-C7-alkyl, C1-C7- alkanesulfinyl-d-C7-alkyl, d-d-alkanesulfonyl-d-d-alkyl, halo, hydroxyl, C1-C^aIkOXy, amino, N-mono- or N,N-di-(C1-C7-alkyl)-amino, d-C7-alkanoylamino, CrC7-alkanesulfonyl- amino, carbamoyl, N-mono- or N,N-di-(C1-C7-alkyl)-carbamoyl, d-C7-alkanesulfonyl, sulfamoyl, N-mono- or N,N-di-(C1-C7-alkyl)-sulfamoyl, morpholinosulfonyl, thiomorpholino- sulfonyl, cyano and nitro.
In the case of unsubstituted or substituted aryl R2, R2 is preferably phenyl which is unsubstituted or substituted by one to three moieties independently selected from the group consisting of d-d-alkyl, hydroxyl-C1-C7-alkyl, amino-d-C7-alkyl, halo-d-C7-alkyl, N-C1-C7- alkanoylamino-d-d-alkyl, N-d-d-alkanesulfonyl-amino-d-d-alkyl, d-C7-alkanesulfonyl- d-C^alkyl, d-d-alkenesulfinyl-d-CT-alkyl, pyrrolidino-C^CT-alkyl, OXO-Py^oIJdJnO-C1-C7- alkyl, piperidino-CTC^alkyl, piperazin-i-yl-CrCTr-alkyl, 4-(d-C7-alkyl, C1-C^aIkOXy-C1-C7- alkyl,
Figure imgf000014_0002
4-(amino-C1-C7-alkyl)- piperazin-1-yl-C^C^alkyl, 4-[N-mono- or N,N-di-(C1-C7-alkylamino)-C1-C7-alkyl]-piperazin-1- yl-C^C^alkyl, 4-(C1-C7-alkanoyl)-piperazin-1-yl-C1-C7-alkyl, oxo-piperazin-i-yl-d-CTr-alkyl, pyrrolidin-1-carbonyl-C^C^alkyl, piperidin-1-carbonyl-C^C^alkyl, piperazin-i-carbonyl-C^ C^alkyl, 4-(C1-C7-alkyl)piperazin-1-carbonyl-C1-C7-alkyl, morpholin-1-carbonyl-Crd-alkyl, thiomorpholin-i-carbonyl-CrCT-alkyl, S-oxo- or S.S-dioxothiomorpholin-i-carbonyl-C^C^ alkyl,
Figure imgf000014_0003
halo-d-CT-alkyl, CrC7-alkanoyl or C3-C10-cycloalkyl)-oxo- piperazin-1-yl-C^C^alkyl, morpholino-C^C^alkyl, thiomorpholino-CrCT-alkyl, S-mono- or S,S-dioxo-thiomorpholino-d-C7-alkyl, imidazol-1-yl-C^C^alkyl, carbamoyl-CrC^alkyl, [N- mono- or N,N-di-(C1-C7-alkyl)-carbamoyl]-C1-C7-alkyl, d-CT-alkanesulfinyl-CrC^alkyl, C1- C7-alkanesulfonyl-d-d-alkyl, halo, hydroxyl, d-d-alkoxy, amino, N-mono- or N,N-di-(Cr C7-alky!)-amino, d-d-alkanoylamino, pyrrolidino, oxo-pyrrolidino, piperidino, piperazin-1-yl, 4-(d-C7-alkyl, d-d-alkoxy-d-d-alkyl, halo-d-C7-alkyl or C3-C10-cycloalkyl)-piperazin-1-yl, 4-(amino-C1-C7-alkyl)-piperazin-1-yl, 4-[N-mono- or N,N-di-(C1-C7-alkylamino)-Ci-C7-alkyl]- piperazin-1-yl, morpholino, thiomorpholino, S-oxo- or S,S-dioxothiomorpholino, d-C7-alkane- sulfonylamino, carbamoyl, N-mono- or N,N-di-(C1-C7-alkyl, d-d-alkoxy-d-C7-alkyl, amino- CrC7-alkyl and/or (N'-mono- or N'.N'-dKd-d-alkylJ-amino-d-d-alkyO-carbamoyl, pyrrolidin-1-carbonyl, piperidin-1-carbonyl, piperazin-1-carbonyl, 4-(C1-C 7-alkyl)piperazin-1- carbonyl, morpholin-1-carbonyl, thiomorpholin-1-carbonyl, S-oxo- or S,S-dioxothiomorpholin- 1-carbonyl, sulfo, d-C7-alkanesulfonyl, d-C7-alkanesulfinyl, sulfamoyl, N-mono- or N,N-di- (d-C7-alkyl)-sulfamoyl, morpholinosulfonyl, thiomorpholinosulfonyl, S-oxo- thiomorpholinosulfonyl, S,S-dioxothiomorpholinosulfonyl, cyano and nitro, preferably in the meta, (more preferably once) in the meta (preferably up to once) and the para or in the para position.
In unsubstituted or substituted- heterocyclyl, heterocyclyl is preferably a heterocyclic radical that is unsaturated (= carrying the highest possible number of conjugated double bonds in the ring(s)), saturated or partiaHy saturated and is preferably a monocyclic or in a broader aspect of the invention bicyclic or tricyclic ring; and has 3 to 24, more preferably 4 to 16, most preferably 4 to 10 and most preferably 6 ring atoms; wherein one or more, preferably one to four, especially one or two carbon ring atoms are replaced by a heteroatom selected from the group consisting of nitrogen, oxygen and sulfur, the bonding ring preferably having 4 to 12, especially 5 to 7 ring atoms; which heterocyclic radical (heterocyclyl) is unsubstituted or substituted by one or more, especially 1 to 3, substituents independently selected from the group consisting of the substituents defined above for substituted alkyl; and where heterocyclyl is especially a heterocyclyl radical selected from the group consisting of oxiranyl, azirinyl, aziridinyl, 1 ,2-oxathiolanyl, thienyl (= thiophenyl), furanyl, tetrahydrofuryl, pyranyl, thiopyranyl, thianthrenyl, isobenzofuranyl, benzofuranyl, chromenyl, 2H-pyrrolyl, pyrrolyl, pyrrolinyl, pyrrolidinyl, imidazolyl, imidazolidinyl, benzimidazolyl, pyrazolyl, pyrazinyl, pyrazolidinyl, thiazolyl, isothiazolyl, dithiazolyl, oxazolyl, isoxazolyl, pyridyl, pyrazinyl, pyrimidinyl, piperidinyl, piperazinyl, pyridazinyl, morpholinyl, thiomorpholinyl, (S-oxo or S1S- dioxo)-thiomorpholinyl, indolizinyl, azepanyl, diazepanyl, especially 1 ,4-diazepanyl, isoindolyl, 3H-indolyl, indolyl, benzimidazolyl, cumaryl, indazolyl, triazolyl, tetrazolyl, purinyl, 4H-quinoli- zinyl, isoquinolyl, quinolyl, tetrahydroquinolyl, tetrahydroisoquinolyl, decahydroquinolyl, octahydroisoquinolyl, benzofuranyl, dibenzofuranyl, benzothiophenyl, dibenzothiophenyl, phthalazinyl, naphthyridinyl, quinoxalyl, quinazolinyl, quinazolinyl, cinnolinyl, pteridinyl, carbazolyl, beta-carbolinyl, phenanthridinyl, acridinyl, perimidinyl, phenanthrolinyl, furazanyl, phenazinyl, phenothiazinyl, phenoxazinyl, chromenyl, isochromanyl, chromanyl, benzo[1 ,3]- dioxol-5-yl and 2,3-dihydro-benzo[1 ,4]dioxin-6-yl, each of these radicals being un- substituted or substituted by one or more, preferably up to three, substituents selected from those mentioned above for substituted aryl and/or from oxo.
In the case of R1, unsubstituted or substituted heterocyclyl is preferably pyrrolyl, oxo-pyrrolyl, 2,3-dihydroindolyl, 2-oxo-2,3-dihydroindolyl or 1H-pyridin-2-onyl, each of which is unsubstituted or substituted by one to three substituents independently selected from those mentioned above for unsubstituted or substituted aryl R1.
In the case of R2, unsubstituted or substituted heterocyclyl is preferably pyridyl, pyrazolyl, thiophenyl, quinolinyl, isoquinolinyl or fH-benzoimidazofyl, each of which is unsubstituted or substituted by one to three moieties independently selected from those mentioned above as substituents for aryl R2, or especially from the group consisting of d-Cτ-alkyl, amino-d-d- alkyl, N-d-Cr-alkanoylamino-d-Cr-alkyl, N-d-CT-alkanesulfonyl-amino-d-CT-alkyl, carba- moyl-d-Cy-alkyl, [N-mono- or N,N-di-(C1-C7-alkyl)-carbamoyl]-C1-C7-alkyl, d-C7-alkane- sulfinyl-d-Cτ-alkyl, C1-C7-alkanesulfonyl-C1-d-alkyl, halo, hydroxyl, d-d-alkoxy, amino, N- mono- or N,N-di-(d-d-alkyl)-amino, d-d-alkanoylamino, d-d-alkanesulfonylamino, carbamoyl, N-mono- or N,N-di-(C1-d-alkyl)-carbamoyl, Ci-C7-alkanesulfonyl, sulfamoyl, N- mono- or N,N-di-(C1-C7-alkyl)-sulfamoyl, morpholinosulfonyl, thiomorpholinosulfonyl, cyano and nitro.
X is preferably CH (especially for JAK2 inhibitors of the formula I) or preferably N (especially for JAK3 inhibitors of the formula I).
In R2, preferably not more than one substituent (if a substituent is present at all) is present in ortho-position and in meta position. That is, the substituent or substituents is or are present preferably in para-position and not more than one is present in ortho- and meta-position.
As R3and R4, hydrogen is especially preferred, respectively. Treatment" includes both prophylactic and therapeutic treatment.
Protein tyrosine kinase (especially JAK2 and/or JAK3 kinase) mediated diseases are especially such disorders that respond in a beneficial way (e.g. amelioration of one or more symptoms, delay of the onset of a disease, up to temporary or complete cure from a disease) to the inhibition of a protein tyrosine kinase, especially inhibition of a JAK (preferably JAK2 and/or JAK3) kinase or TYK2, more especially inhibition of JAK2 kinase (where among the diseases to be treated, especially proliferative diseases such as tumor diseases, leukaemias, polycythemia vera, essential thrombocythemia, and myelofibrosis with myeloid metaplasia may be mentioned) and/or of JAK3 kinase (where preferably the treatment (e.g. by immunosuppression) of diseases such as organ transplant rejection, lupus erythema- todes, multiple sclerosis, rheumatoid arthritis, psoriasis, dermatitis, Crohn's disease, type-1 diabetes and complications from type-1 diabetes are to be mentioned as preferred.
Salts (which, what is meant by "or salts thereof or "or a salt thereof, can be present alone or in mixture with free compound of the formula I) are preferably pharmaceutically acceptable salts.
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, e.g., carboxylic acids or sulfonic acids, such as fumaric acid or methansulfonic acid.
For isolation or purification purposes it is also possible to use pharmaceutically unacceptable salts, for example picrates or perchlorates. For therapeutic use, only pharmaceutically acceptable salts or free compounds are employed (where applicable in the form of pharmaceutical preparations), and these are therefore preferred.
In view of the close relationship between the novel compounds in free form and those in the form of their salts, including those salts that can be used as intermediates, for example in the purification or identification of the novel compounds, any reference to the free compounds hereinbefore and hereinafter is to be understood as referring also to the corresponding salts, as appropriate and expedient. The compounds of formula I thereof have valuable pharmacological properties, as described hereinbefore and hereinafter.
They inhibit various protein tyrosine kinases, and especially JAK2 and/or JAK3-receptor tyrosine kinase.
The efficacy of the compounds of the invention as inhibitors of JAK2-receptor tyrosine kinase activity can be demonstrated as follows ("alternative method to that given in the examples"):
Baculovirus including the amino acid domain ASP751-VAL1129 of the JAK2 protein is obtainable by ProQinase, Freiburg, Germany. The virus is scaled up as following: Virus containing media is collected from the transfected cell culture and used for infection to increase its titer. Virus containing media obtained after two rounds of infection is used for large-scale protein expression. For large-scale protein expression 100 cm2 round tissue culture plates are seeded with 5 x 107 ceHs/plate and infected with 1 m!_ of virus-containing media (approx. 5 MOIs). After 3 days the cells are scraped off the plate and centrifuged at 500 rpm for 5 min. Cell pellets from 10-20, 100 cm2 plates, are re-suspended in 50 mL of ice-cold lysis buffer (25mMTris-HCI, pH7.5, 2mMEDTA, 1%NP-40, 1mM DTT, 1mMP MSF). The cells are stirred on ice for 15 min and then centrifuged at 5000 rpms for 20 min. The protein is purified by loading the centrifuged ceH lysate 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 (1 mL each) of 25 mM Tris-HCI, pH 7.5, 10 mM reduced-glutathione, 100 mM NaCI, 1 mM DTT, 10 % Glycerol and stored at -700C.
The activity of JAK2 is assayed in the presence or absence of inhibitor measuring the incorporation of 33P from [γ33P]ATP into appropriate substrates [Garcia-Echeverria C, Pearson MA, Marti A, et al (2004) In vivo antitumor activity of NVP-AEW541 - A novel, potent, and selective inhibitor of the IGF-IR kinase. Cancer Cell; 5: 231 -2391. The test compound is dissolved in DMSO (10 mM) and stored at -20°C. Serial dilutions are freshly made in DMSO and are 1000 times concentrated than test solutions ("pre-dilution plates"). They are further diluted with pure water to yield "master plates" containing 3 times concentrated test solutions in 3% DMSO. The final volume of the assay is 30μL containing 10μl_ of test solution (1% DMSO), 10μl_ assay mix including the assay components descry- bed by Garcia-Echeverria (2004) and in the following section as well as 10μL enzyme. The pipetting steps can be programmed to be performed either on the MultiPROBE lix, MultiPROBE MLx or HamiltonSTAR robots in the 96 well format.
The protein kinase assays are carried as described in details by Garcia-Echeverria (see above). The assay for JAK2 is carried out in 96-well plates at ambient temperature for 10 min (filter-biding method) or 30 min (flash plates) in a finial volume of 30 μl_ including the following components: 300 ng of GST-JAK2, 20 mM Tris-HCI, pH 7.5, 1.0 mM MnCI2, 10 mM MgCI2, 1 mM DTT, 3 μg/mL poiy(Glu,Tyr) 4:1 , 1 % DMSO and 1.0 μM ATP (γ-[33PJ-ATP 0.1 μCi); The assays are terminated by the addition of 20 μl of 125 mM EDTA. The capturing of the phosphorylated- peptides by the filter-binding method is performed as following: 40 μl_ of the reaction mixture are transferred onto Immobilon-PVDF membranes previously soaked for 5 min with methanol, rinsed with water, then soaked for 5 min with 0.5 % H3PO4 and mounted on vacuum manifold with disconnected vacuum source. After spotting all samples, vacuum is connected and each well rinsed with 200 μl 0.5 % H3PO4. Free membranes are removed and washed 4 x on a shaker with 1.0 % H3PO4, once with ethanol. Membranes are counted after drying at ambient temperature, mounting in Packard TopCount 96-well frame, and addition of 10 μl/well of Microscint. The plates are eventually sealed and counted in a microplate scintillation counter (TopCount NXT, TopCount NXT HTS, PerkinElmer, Brussels, Belgium).
The assays for the flash plate method is carried out in a total volume of 30 μl_ at RT in conventional 96-well flash plates. The reaction is stopped after 30 min by the addition of 20μL of 125 mM EDTA The assay plates are then washed three times with PBS and dried at room temperature. The plates are sealed and counted in a microplate scintillation counter (TopCount NXT, TopCount NXT HTS). IC50 values are calculated by linear regression analysis of the percentage inhibition of the compound either in duplicate, at four concentrations (usually 0.01 , 0.1 , 1 and 10 μM) or as 8 single point IC50 starting at 10μM following by 1 :3 dilutions. With compounds according to the invention, IC50 values in the range from 5 nM to 5 μM can be found with compounds of the formula I.
Alternatively, the following assays are made: 1. JAK kinase assays ("lance assays")
JAK-2 or JAK-3 enzymatic activity is determined using a time-resolved fluorescence energy transfer technology. The phosphorylation of a synthetic biotinylated peptide substrate (GGEEEEYFELVKKKK, SEQ ID NO: 3)) by either JAK-2 or JAK-3 in the presence of ATP is quantified using Europium labeled anti phosphotyrosine antibody and Streptavidin- Allophycocyanin Both JAK-2 and JAK-3 enzymes used in these assays contain the kinase domain (JH-1 domain) of the full length proteins and are used as glutathione S-transferase (GST) fusion proteins.
Inhibitors are dissolved in dimethylsulfoxide (DMSO). Dilutions are prepared in 90% DMSO followed by additional dilutions steps as required to perform a 8-point concentration- response.
The reaction mix consists of 5 μL of diluted compound, 10 μl_ of assay buffer and 5 μl_ of enzyme dilution. After incubation for 60 minutes at room temperature the reaction is stopped by the addition of EDTA. For detection of the product anti-phosphotyrosine antibody and Streptavidin-APC are added and after 60 minutes the samples are measured in an EnVision 2102 Multilabel Reader (Perkin Elmer, Inc., Wellesley, MA, USA, in the following mentioned as "PerkinElmer") with excitation wavelength of 320nm and emission at 665nm.
Alternatively, the kinase assays are performed as described in details by Garcia-Echeverria et al [(2004), Cancer Cell; 5: 231-239] in 96-well plates at ambient temperature for 10 min (filter-biding method) or 30 min (flash plates) in a final volume of 30 μL including the following components: GST-JAK-2 or GST-JAK-3, 20 mM Tris-HCI, pH 7.5, 0-1.0 mM MnCI2, 1-10 mM MgCI2, 1 mM dithiothreitol (DTT), 3 μg/mL poly(Glu.Tyr) 4:1, 1 % DMSO and 1.0 μM ATP (γ-[33P]-ATP 0.1 μCi); The assays are terminated by the addition of 20 μl of 125 mM ethylendiamine tetraacetate (EDTA). The capturing of the phosphorylated peptides by the filter-binding method is performed as following: 40 μL of the reaction mixture are transferred onto Immobilon-PVDF membranes previously soaked for 5 min with methanol, rinsed with water, then soaked for 5 min with 0.5 % H3PO4 and mounted on vacuum manifold with disconnected vacuum source. After spotting all samples, vacuum is connected and each well rinsed with 200 μl 0.5 % H3PO4. Free membranes are removed and washed 4 x on a shaker with 1.0 % H3PO4, once with ethanol. Membranes are counted after drying at ambient temperature, mounting in Packard TopCount 96-well frame (now PerkinElmer), and addition of 10 μl/well of Microscint (PerkinElmer). The plates are eventually sealed and counted in a microplate scintillation counter (TopCount NXT, TopCount NXT HTS, PerkinElmer, Brussels, Belgium).
In these assays, the compounds of the invention have IC50 values of from ca. 0.1 -1000 nM.
2. JAK-2 and JAK-3 assays (filter binding/flash plate kinase assays):
Enzyme activities: Enzyme activities are measured by mixing 10 μL of a 3-fold concentrated compound solution with 10 μL of the corresponding substrate mixture (peptidic substrate,
ATP and [γ^3p]ATP) and the reactions are initiated by the addition of 10 μL of a 3-fold concentrated solution of GST-JAK-2 and GST-JAK-3 respectively, in assay buffer. The enzymatic reactions are stopped by the addition of 20 μL of 125 mM EDTA. The incorporation of
33p jnto the substrates is quantified by either filter binding (FB) or flash-plate (FP) method:
Kinase reaction: The assays are carried out in 96-well plates at room temperature for 10 min
(FB) in a finial volume of 30 μL including the following components:
JAK-2: 200 ng GST-JAK-2, 20 mM Tris-HCI, pH 7.5, 10 mM MgCI2, 1.0 mM MnCI2, 1 mM
DTT, 3 μg/mL poly-EY, 1% DMSO and 1.0μM ATP (γ-[33P]-ATP 0.1 μCi);
JAK-3: 15 ng GST-JAK-3, 20 mM Tris-HCI, pH 7.5, 10 mM MgCI2, 1 mM DTT, 1% DMSO,
3 μg/mL poly-EY and 3.0μM ATP (γ-[33P]-ATP 0.1 μCi);
Filter binding method: The capturing of the phosphorylated peptides by the FB method is performed as following: 40 μL of the stopped reaction mixture were transferred onto Immobilon-PVDF (Millipore, Eschbom, Germany) membranes previously soaked for 5 min with methanol, rinsed with water, soaked for 5 min with 0.5% H3PO4 and mounted on vacuum manifold with disconnected vacuum source. After spotting, vacuum is connected and each well rinsed with 200 μL 0.5% H3PO4. Free membranes are removed and washed 4 times on a shaker with 1% H3PO4 and once with ethanol. Membranes are counted after drying, mounting in Packard TopCount 96-well frame, and addition of 10 μL/well of Microscint™. The plates are eventually sealed and counted in a microplate scintillation counter (TopCount NXT, TopCount NXT HTS).
Flash plate method: For the capturing of the phosphorylated substrates (60 min, RT), 96-well standard FPs (i.e. polystyrene microplates in which the interior of each well is permanently coated with a thin layer of polystyrene-based scintillant) are used. The assay plates are then washed three times with PBS and dried at room temperature. The plates are sealed and counted in a microplate scintillation counter (TopCount NXT, TopCount NXT HTS).
Calculation of IC50 1S: A 4 Parameter logistic equation is used to calculate IC50 values (IDBS XLfit) of the percentage inhibition of each compound at 8 concentrations (usually 10, 3.0, 1.0, 0.3, 0.1 , 0.030,0.010 and 0.003 μM).
Preferably, the test system as described in Example 186 is used.
Preferably, IC50 values in the range from 0.1 nM to 10 μM, e.g. from less than 3 nM to 5 μM, most preferably from 0.1 nM to 1000 nM can be found in the above-mentioned test systems.
The activity of the compounds of the formula I can also be determined in vivo:
JAK-2 in vivo
The assay can be performed as described by G. Wernig, T. Mercher, R. Okabe, R.L. Levine, B. H. Lee, D.G. Gilliland-, Blood First Edition paper, published online February 14, 2006; DOI 10, 1182/blood-2005-12-4824.
On the basis of these studies, a compound of formula I according to the invention shows therapeutic efficacy especially against disorders dependent on protein kinase, especially proliferative diseases mediated JAK2 kinase activity.
In addition, further protein kinases can be inhibited by compounds of the present invention, such as Tyk 2, c-src, Flt-3, KDR and others, for each of which test systems are known in the art.
The dosage of the active ingredient to be applied to a warm-blooded animal depends upon a variety of factors including type, species, age, weight, sex and medical condition of the patient; the severity of the condition to be treated; the route of administration; the renal and hepatic function of the patient; and the particular compound employed. A physician, clinician or veterinarian of ordinary skill can readily determine and prescribe the effective amount of the drug required to prevent, counter or arrest the progress of the condition. Optimal precision in achieving concentration of drug within the range that yields efficacy without toxicity requires a regimen based on the kinetics of the drug's availability to target sites. This involves a consideration of the distribution, equilibrium, and elimination of a drug. The dose of a compound of the formula I or a pharmaceutically acceptable salt thereof to be administered to warmblooded animals, for example humans of approximately 70 kg body weight, is preferably from approximately 3 mg to approximately 5 g, more preferably from approximately 10 mg to approximately 1.5 g per person per day, divided preferably into 1 to 3 single doses which may, for example, be of the same size. Usually, children receive half of the adult dose.
A compound of formula I can be administered alone or in combination with one or more other therapeutic agents, possible combination therapy taking the form of fixed combinations or the administration αf a compound of the invention and one or more other therapeutic agents being staggered or given independently of one another, or the combined administration of fixed combinations and one or more other therapeutic agents. A compound of formula I can besides or in addition be administered especially for tumor therapy in combination with chemotherapy, radiotherapy, immunotherapy, surgical intervention, or a combination of these. Long-term therapy is equally possible as is adjuvant therapy in the context of other treatment strategies, as described above. Other possible treatments are therapy to maintain the patient's status after tumor regression, or even chemopreventive therapy, for example in patients at risk.
Thus, a compound of the formula I may be used to advantage in combination with other antiproliferative compounds. Such antiproliferative compounds include, but are not limited to aromatase inhibitors; antiestrogens; topoisomerase I inhibitors; topoisomerase Il inhibitors; microtubule active compounds; alkylating compounds; histone deacetylase inhibitors; compounds which induce cell differentiation processes; cyclooxygenase inhibitors; MMP inhibit- tors; 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; bisphospho- nates; biological response modifiers; antiproliferative antibodies; heparanase inhibitors; inhibitors of Ras oncogenic isoforms; telomerase inhibitors; proteasome inhibitors; compounds used in the treatment of hematologic malignancies; compounds which target, decrease or inhibit the activity of Flt-3; Hsp90 inhibitors such as 17-AAG (17-allylaminogeldanamycin, NSC330507), 17-DMAG (17-dimethylaminoethylamino-17-demethoxy-geldanamycin, NSC707545), IPI-504, CNF1010, CNF2024, CNF1010 from Conforma Therapeutics; temo- zolomide (TEMODAL®); kinesin spindle protein inhibitors, such as SB715992 or SB743921 from GlaxoSmithKline, or pentamidine/chlorpromazine from CombinatoRx; MEK inhibitors such as ARRY142886 from Array PioPharma, AZD6244 from AstraZeneca, PD181461 from Pfizer, leucovorin, EDG binders, antileukemia compounds, ribonucleotide reductase inhibit- tors, S-adenosylmethionine decarboxylase inhibitors, antiproliferative antibodies or other chemotherapeutic compounds. Further, alternatively or in addition they may be used- in combination with other tumor treatment approaches, including surgery, ionizing radiation, photo- dynamic therapy, implants, e.g. with corticosteroids, hormones, or they may be used as radiosensitizers. Also, in anti-inflammatory and/or antiproliferative treatment, combination with anti-inflammatory drugs is included. Combination is also possible with antihistamine drug substances, bronchodilatatory drugs, NSAID or antagonists of chemokine receptors.
The term "aromatase inhibitor" as used herein relates to a compound which inhibits the estrogen production, i.e. the conversion of the substrates androstenedione and testosterone to estrone and estradiol, respectively. The term includes, but is not limited to steroids, especially atame- stane, exemestane and formestane and, in particular, non-steroids, especially aminogluteth- imide, roglethimide, pyridoglutethimide, trilostane, testolactone, ketokonazole, vorozole, fadro- zole, anastrozole and letrozole. Exemestane can be administered, e.g., in the form as it is marketed, e.g. under the trademark AROMASIN. Formestane can be administered, e.g., in the form as it is marketed, e.g. under the trademark LENTARON. Fadrozole can be administered, e.g., in the form as it is marketed, e.g. under the trademark AFEMA. Anastrozole can be administered, e.g., in the form as it is marketed, e.g. under the trademark ARIMIDEX. Letrozole can be administered, e.g., in the form as it is marketed, e.g. under the trademark FEMARA or FEMAR. Aminoglutethimide can be administered, e.g., in the form as it is marketed, e.g. under the trademark ORIMETEN. A combination of the invention comprising a chemotherapeutic agent which is an aromatase inhibitor is particularly useful for the treatment of hormone receptor positive tumors, e.g. breast tumors.
The term "antiestrogen" as used herein relates to a compound which antagonizes the effect of estrogens at the estrogen receptor level. The term includes, but is not limited to tamoxifen, ful- vestrant, 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.
The term "anti-androgen" as used herein relates to any substance which is capable of inhibiting the biological effects of androgenic hormones and includes, but is not limited to, bicalutamide (CASODEX), which can be formulated, e.g. as disclosed in US 4,636,505. The term "gonadorelin agonist" as used herein includes, but is not limited to abarelix, goserelin and goserelin acetate. Goserelin is disclosed in US 4,100,274 and can be administered, e.g., in the form as it is marketed, e.g. under the trademark ZOLADEX. Abarelix can be formulated, e.g. as disclosed in US 5,843,901.
The term "topoisomerase I inhibitor" as used herein includes, but is not limited- to topotecan, gimatecan, irinotecan, camptothecian and its analogues, 9-nitrocamptothecin and the macromolecular camptothecin conjugate PNU-166148 (compound A1 in WO99/ 17804). Irinotecan can be administered, e.g. in the form as it is marketed, e.g. under the trademark CAMPTOSAR. Topotecan can be administered, e.g., in the form as it is marketed, e.g. under the trademark HYCAMTIN.
The term "topoisomerase Il inhibitor" as used herein includes, but is not limited to the an- thracyclines such as doxorubicin (including liposomal formulation, e.g. CAELYX), daunorubicin, epirubicin, idarubicin and nemorubicin, the anthraquinones mitoxantrone and losoxantrone, and the podophillotoxines etoposide and teniposide. Etoposide can be administered, e.g. in the form as it is marketed, e.g. under the trademark ETOPOPHOS. Teniposide can be administered, e.g. in the form as it is marketed, e.g. under the trademark VM 26-BRISTOL. Doxorubicin can be administered, e.g. in the form as it is marketed, e.g. under the trademark ADRIBLASTIN or ADRIAMYCIN. Epirubicin can be administered, e.g. in the form as it is marketed, e.g. under the trademark FARMORUBICIN. Idarubicin can be administered, e.g. in the form as it is marketed, e.g. under the trademark ZAVEDOS. Mitoxantrone can be administered, e.g. in the form as it is marketed, e.g. under the trademark NOVANTRON.
The term "microtubule active compound" relates to microtubule stabilizing, microtubule destabilizing compounds and microtublin polymerization inhibitors including, but not limited to taxanes, e.g. paclitaxel and docetaxel, vinca alkaloids, e.g., vinblastine, especially vinblastine sulfate, vincristine especially vincristine sulfate, and vinorelbine, discodermolides, cochicine and epothilones and derivatives thereof, e.g. epothilone B or D or derivatives thereof. Paclitaxel may be administered e.g. in the form as it is marketed, e.g. TAXOL. Docetaxel can be administered, e.g., in the form as it is marketed, e.g. under the trademark TAXOTERE. Vinblastine sulfate can be administered, e.g., in the form as it is marketed, e.g. under the trademark VINBLASTIN R.P.. Vincristine sulfate can be administered, e.g., in the form as it is marketed, e.g. under the trademark FARMISTIN. Discodermolide can be obtained, e.g., as disclosed in US 5,010,099. Also included are Epothilone derivatives which are disclosed in WO 98/10121 , US 6,194,181 , WO 98/25929, WO 98/08849, WO 99/43653, WO 98/22461 and WO 00/31247. Especially preferred are Epothilone A and/or B.
The term "alkylating compound" as used herein includes, but is not limited to, cyclophosphamide, ifosfamide, melphalan or nitrosourea (BCNU or Gliadel). Cyclophosphamide can be administered, e.g., in the form as it is marketed, e.g. under the trademark CYCLOSTIN. Ifosfamide can be administered, e.g., in the form as it is marketed, e.g. under the trademark HOLOXAN.
The term "histone deacetylase inhibitors" or "HDAC inhibitors" relates to compounds which inhibit the histone deacetylase and which possess antiproliferative activity. This includes compounds disclosed in WO 02/22577, especially N-hydroxy-3-[4-[[(2-hydroxyethyl)[2-(1H-indol- 3-yl)ethyl]-amino]methyl]phenyl]-2E-2-propenamide, N-hydroxy-3-[4-[[[2-(2-methyl-1H-indol-3- yl)-ethyl]-amino]methyl]phenyl]-2£-2-propenamide and pharmaceutically acceptable salts thereof. It further especially includes Suberoylanilide hydroxamic acid (SAHA). The term "antineoplastic antimetabolite" includes, but is not limited to, 5-Fluorouracil or 5-FU, capecitabine, gemcitabine, DNA demethylating compounds, 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 term "platin compound" as used herein includes, but is not limited to, carboplatin, cis-platin, cisplatinum and oxaliplatin. Carboplatin can be administered, e.g., in the form as it is marketed, e.g. under the trademark CARBOPLAT. Oxaliplatin can be administered, e.g., in the form as it is marketed, e.g. under the trademark ELOXATIN.
The term "compounds targeting/decreasing a protein or lipid kinase activity"; or a "protein or lipid phosphatase activity"; or "further anti-angiogenic compounds" as used herein includes, but is not limited to, protein tyrosine kinase and/or serine and/or threonine kinase inhibitors or lipid kinase inhibitors, e.g., a) compounds targeting, decreasing or inhibiting the activity of the platelet-derived growth factor-receptors (PDGFR), such as compounds which target, decrease or inhibit the activity of PDGFR, especially compounds which inhibit the PDGF receptor. e.g. a N-phenyl-2-pyrimidine-amine derivative, e.g. imatinib, SU101 , SU6668 and GFB-111 ; b) compounds targeting, decreasing or inhibiting the activity of the fibroblast growth factor-receptors (FGFR); c) compounds targeting, decreasing or inhibiting the activity of the insulin-like growth factor receptor I (IGF-IR), such as compounds which target, decrease or inhibit the activity of IGF-IR, especially compounds which inhibit the kinase activity of IGF-I receptor, such as those compounds disclosed in WO 02/092599, or antibodies that target the extracellular domain of IGF-I receptor or its growth factors; d) compounds targeting, decreasing or inhibiting the activity of the Trk receptor tyrosine kinase family, or ephrin B4 inhibitors; e) compounds targeting, decreasing or inhibiting the activity of the AxI receptor tyrosine kinase family; f) compounds targeting, decreasing or inhibiting the activity of the Ret receptor tyrosine kinase; g) compounds targeting, decreasing or inhibiting the activity of the Kit/SCFR receptor tyrosine kinase, e.g. imatinib; h) compounds targeting, decreasing or inhibiting the activity of the C-kit receptor tyrosine kinases - (part of the PDGFR family), such as compounds which target, decrease or inhibit the activity of the c-Kit receptor tyrosine kinase family, especially compounds which inhibit the c-Kit receptor, e.g. imatinib; i) compounds targeting, decreasing or inhibiting the activity of members of the c-Abl family, their gene-fusion products (e.g. BCR-AbI kinase) and mutants, such as compounds which target decrease or inhibit the activity of c-Abl family members and their gene fusion products, e.g. a N-phenyl-2-pyrimidine-amine derivative, e.g. imatinib or nilotinib (AMN107); PD180970; AG957; NSC 680410; PD173955 from ParkeDavis; or dasatinib (BMS-354825) j) compounds targeting, decreasing or inhibiting the activity of members of the protein kinase C (PKC) and Raf family of serine/threonine kinases, members of the MEK. SRC, JAK, FAK, PDK1 , PKB/Akt, and Ras/MAPK family members, and/or members of the cyclin-dependent kinase family (CDK) and are especially those staurosporine derivatives disclosed in US 5,093,330, e.g. midostaurin; examples of further compounds include e.g. UCN-01 , safingol, BAY 43-9006, Bryostatin 1 , Perifosine; llmofosine; RO 318220 and RO 320432; GO 6976; lsis 3521 ; LY333531/LY379196; isochinoline compounds such as those disclosed in WO 00/09495; FTIs; PD184352 or QAN697 (a P13K inhibitor) or AT7519 (CDK inhibitor); k) compounds targeting, decreasing or inhibiting the activity of protein-tyrosine kinase inhibitors, such as compounds which target, decrease or inhibit the activity of protein-tyrosine kinase inhibitors include imatinib mesylate (GLEEVEC) or tyrphostin. A tyrphostin is preferably a low molecular weight (Mr < 1500) compound, or a pharmaceutically acceptable salt thereof, especially a compound selected from the benzylidenemalonitrile class or the S-arylbenzenemalonirile or bisubstrate quinoline class of compounds, more especially any compound selected from the group consisting of Tyrphostin A23/RG-50810; AG 99; Tyrphostin AG 213; Tyrphostin AG 1748; Tyrphostin AG 490; Tyrphostirv 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);
I) compounds targeting, decreasing or inhibiting the activity of the epidermal growth factor family of receptor tyrosine kinases (EGFR, ErbB2, ErbB3, ErbB4 as homo- or heterodimers) and their mutants, such as compounds which target, decrease or inhibit the activity of the epidermal growth factor receptor family are especially compounds, proteins or antibodies which inhibit members of the EGF receptor tyrosine kinase family, e.g. EGF receptor, ErbB2, ErbB3 and ErbB4 or bind to EGF or EGF related ligands, and are in particular those compounds, proteins or monoclonal antibodies generically and specifically disclosed in WO 97/02266, e.g. the compound of ex. 39, or in EP 0 564 409, WO 99/03854, EP 0520722, EP 0 566 226, EP 0 787 722, EP 0 837 063, US 5,747,498, WO 98/10767, WO 97/30034, WO 97/49688, WO 97/38983 and, especially, WO 96/30347 (e.g. compound known as CP 358774), WO 96/33980 (e.g. compound ZD 1839) and WO 95/03283 (e.g. compound ZM105180); e.g. trastuzumab (Herceptin™), cetuximab (Erbitux™), Iressa, Tarceva, OSI-774, Cl- 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 ; and m) compounds targeting, decreasing or inhibiting the activity of the c-Met receptor, such as compounds which target, decrease or inhibit the activity of c-Met, especially compounds which inhibit the kinase activity of c-Met receptor, or antibodies that target the extracellular domain of c-Met or bind to HGF.
Further anti-angiogenic compounds include compounds having another mechanism for their activity, e.g. unrelated to protein or lipid kinase inhibition e.g. thalidomide (THALOMID) and TNP-470.
Compounds which target, decrease or inhibit the activity of a protein or lipid phosphatase are e.g. inhibitors of phosphatase 1 , phosphatase 2A, or CDC25, e.g. okadaic acid or a derivative thereof.
Compounds which induce cell differentiation processes are e.g. retinoic acid, α- γ- or δ- tocopherol or α- γ- or δ-tocotrienol.
The term cyclooxygenase inhibitor as used herein includes, but is not limited to, e.g. Cox-2 inhibitors, 5-alkyl substituted 2-arylaminophenylacetic acid and derivatives, such as celecoxib (CELEBREX), rofecoxib (VIOXX), etoricoxib, valdecσxib or a 5-alkyl-2- arylaminophenylacetic acid, e.g. 5-methyl-2-(2'-chloro-6'-fluoroanilino)phenyl acetic acid, lumiracoxib.
The term "bisphosphonates" as used herein includes, but is not limited to, etridonic, clodronic, tiludronic, pamidronic, alendronic, ibandronic, risedronic and zoledronic acid. "Etridonic acid" can be administered, e.g., in the form as it is marketed, e.g. under the trademark DIDRONEL. "Clodronic acid" can be administered, e.g., in the form as it is marketed, e.g. under the trademark BONEFOS. "Tiludronic acid" can be administered, e.g., in the form as it is marketed, e.g. under the trademark SKELID. "Pamidronic acid" can be administered, e.g. in the form as it is marketed, e.g. under the trademark AREDIA™. "Alendronic acid" can be administered, e.g., in the form as it is marketed, e.g. under the trademark FOSAMAX. "Ibandronic acid" can be administered, e.g., in the form as it is marketed, e.g. under the trademark BONDRANAT. "Risedronic acid" can be administered, e.g., in the form as it is marketed, e.g. under the trademark ACTONEL. "Zoledronic acid" can be administered, e.g. in the form as it is marketed, e.g. under the trademark ZOMETA. The term "mTOR inhibitors" relates to compounds which inhibit the mammalian target of rapamycin (mTOR) and which possess antiproliferative activity such as siroh'mus (RapamuneCD), everolimus (Certican™), CCI-779 and ABT578.
The term "heparanase inhibitor" as used herein refers to compounds which target, decrease or inhibit heparin sulfate degradation. The term includes, but is not limited to, PI-88. The term " biological response modifier" as used herein refers to a lymphokine or interferons, e.g. interferon γ. The term "inhibitor of Ras oncogenic isoforms", e.g. H-Ras, K-Ras, or N-Ras, as used herein refers to compounds which target, decrease or inhibit the oncogenic activity of Ras e.g. a
"farnesyl transferase inhibitor" e.g. L-744832, DK8G557 or R115777 (Zarnestra).
The term "telomerase inhibitor" as used herein refers to compounds which target, decrease or inhibit the activity of telomerase. Compounds which target, decrease or inhibit the activity of telomerase are especially compounds which inhibit the telomerase receptor, e.g. telomestatin.
The term "methionine aminopeptidase inhibitor" as used herein refers to compounds which target, decrease or inhibit the activity of methionine aminopeptidase. Compounds which target, decrease or inhibit the activity of methionine aminopeptidase are e.g. bengamide or a derivative thereof.
The term "proteasome inhibitor" as used herein refers to compounds which target, decrease or inhibit the activity of the proteasome. Compounds which target, decrease or inhibit the activity of the proteasome include e.g. Bortezomid (Velcade™)and MLN 341.
The term "matrix metalloproteinase inhibitor" or ("MMP" inhibitory as used herein includes, but is not limited to, collagen peptidomimetic and nonpeptidomimetic inhibitors, tetracycline derivatives, e.g. hydroxamate peptidomimetic inhibitor batimastat and its orally bioavailable analogue marimastat (BB-2516), prinomastat (AG3340), metastat (NSC 683551 ) BMS-
279251 , BAY 12-9566, TAA211, MMI270B or AAJ996.
The term "compounds used in the treatment of hematologic malignancies" as used herein includes, but is not limited to, FMS-like tyrosine kinase inhibitors e.g. compounds targeting, decreasing or inhibiting the activity of FMS-like tyrosine kinase receptors (Flt-3R); interferon,
1-b-D-arabinofuransylcytosine (ara-c) and bisulfan; and ALK inhibitors e.g. compounds which target, decrease or inhibit anaplastic lymphoma kinase.
Compounds which target, decrease or inhibit the activity of FMS-like tyrosine kinase receptors (Flt-3R) are especially compounds, proteins or antibodies which inhibit members of the Flt-3R receptor kinase family, e.g. PKC412, midostaurin, a staurosporine derivative,
SU11248 and MLN518.
The term "HSP90 inhibitors" as used herein includes, but is not limited to, compounds targeting, decreasing or inhibiting the intrinsic ATPase activity of HSP90; degrading, targeting, decreasing or inhibiting the HSP90 client proteins via the ubiquitin proteosome pathway. Compounds targeting, decreasing or inhibiting the intrinsic ATPase activity of
HSP90 are especially compounds, proteins or antibodies which inhibit the ATPase activity of HSP90 e.g., 17-allylamino,17-demethoxygeldanamycin (17AAG), a geldanamycin derivative; other geldanamycin related compounds; radicicol and HDAC inhibitors. The term "antiproliferative antibodies" as used herein includes, but is not limited to, trastuzumab (Herceptin™), Trastuzumab-DMI.erbitux, bevacizumab (Avastin™), rituximab (Rituxan®), PRO64553 (anti-CD40) and 2C4 Antibody. By antibodies is meant e.g. intact monoclonal antibodies, polyclonal antibodies, multispecific antibodies formed from at least 2 intact antibodies, and antibodies fragments so long as they exhibit the desired biological activity.
For the treatment of acute myeloid leukemia (AML), compounds of formula (I) can be used in combination with standard leukemia therapies, especially in combination with therapies used for the treatment of AML. In particular, compounds of formula (I) can be administered in combination with, e.g., farnesyf transferase inhibitors and/or other drugs useful for the treatment of AML, such as Daunorubicin, Adriamycin, Ara-C, VP-16, Teniposide, Mitoxantrone, Idarubicin, Carboplatinum and PKC412.
The term "antileukemic compounds" includes, for example, Ara-C, a pyrimidine analog, which is the 2 -alpha-hydroxy ribose (arabinoside) derivative of deoxycytidine. Also included is the purine analog of hypoxanthine, 6-mercaptopurine (6-MP) and fludarabine phosphate. Compounds which target, decrease or inhibit activity of histone deacetylase (HDAC) inhibitors such as sodium butyrate and suberoylanilide hydroxamic acid (SAHA) inhibit the activity of the enzymes known as histone deacetylases. Specific HDAC inhibitors include MS275, SAHA, FK228 (formerly FR901228), Trichostatin A and compounds disclosed in US 6,552,065, in particular, Λ/-hydroxy-3-[4-[[[2-(2-methyl-1H-indol-3-yl)-ethyl]-amino]me- thyl]phenyl]-2E-2-propenamide, or a pharmaceutically acceptable salt thereof and Λ/-hydro- xy-3-[4-[(2-hydroxyethyl){2-(1/-/-indol-3-yl)ethyl]-amino]methyl]phenyl]-2E-2-propenamide, or a pharmaceutically acceptable salt thereof, especially the lactate salt. Somatostatin receptor antagonists as used herein refers to compounds which target, treat or inhibit the somatostatin receptor such as octreotide, and SOM230. Tumor cell damaging approaches refer to approaches such as ionizing radiation. The term "ionizing radiation" referred to above and hereinafter means ionizing radiation that occurs as either electromagnetic rays (such as X-rays and gamma rays) or particles (such as alpha and beta particles). Ionizing radiation is provided in, but not limited to, radiation therapy and is known in the art. See Hellman, Principles of Radiation Therapy, Cancer, in Principles and Practice of Oncology, Devita et al., Eds., 4th Edition, Vol. 1 , pp. 248-275 (1993). The term "EDG binders" as used herein refers a class of immunosuppressants that modulates lymphocyte recirculation, such as FTY720.
The term "ribonucleotide reductase inhibitors" refers to pyrimidine or purine nucleoside analogs including, but not limited to, fludarabine and/or cytosine arabinoside (ara-C),
6-thioguanine, 5-fluorouracil, cladribine, 6-mercaptopurine (especially in combination with ara-C against ALL) and/or pentostatin. Ribonucleotide reductase inhibitors are especially hydroxyurea or 2-hydroxy-1/-/-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 Oncologics, Vol. 33, No. 8, pp. 953-961 (1994).
The term "S-adenosylmethionine decarboxylase inhibitors" as used herein includes, but is not limited to the compounds disclosed in US 5,461 ,076.
Also included are in particular those compounds, proteins or monoclonal antibodies of VEGF disclosed in WO 98/35958, e.g. 1-(4-chloroanilino)-4-(4-pyridylmethyl)phthalazine or a pharmaceutically acceptable salt thereof, e.g. the succinate, or in WO 00/09495,
WO 00/27820, WO 00/59509, WO 98/11223, WO 00/27819 and EP 0 769 947; those as described by Prewett et al, Cancer Res, Vol. 59, pp. 5209-5218 (1999); Yuan et al.,
Proc Natl Acad Sci U S A, Vol. 93, pp. 14765-14770 (1996); Zhu et al.. Cancer Res, Vol. 58, pp. 3209-3214 (1998); and Mordenti et al., Toxicol Pathol, Vol. 27, No. 1, pp. 14-21 (1999); in WO 00/37502 and WO 94/10202; ANGIOSTATIN, described by O'Reilly et al., Cell,
Vol. 79, pp. 315-328 (1994); ENDOSTATIN, described by O'Reilly et al., Cell, Vol. 88, pp. 277-285 (1997); anthranilic acid amides; ZD4190; ZD6474; SU5416; SU666&; bevacizumab; or anti-VEGF antibodies or anti-VEGF receptor antibodies, e.g. rhuMAb and
RHUFab, VEGF aptamer e.g. Macugon; FLT-4 inhibitors, FLT-3 inhibitors, VEGFR-2 IgGI antibody, Angiozyme (RPI 4610) and Bevacizumab (Avastin™).
Photodynamic therapy as used herein refers to therapy which uses certain chemicals known as photosensitizing compounds to treat or prevent cancers. Examples of photodynamic therapy includes treatment with compounds, such as e.g. VISUDYNE and porfimer sodium. Angiostatic steroids as used herein refers to compounds which block or inhibit angiogenesis, such as, e.g., anecortave, triamcinolone, hydrocortisone, 11-α-epihydrocotisol, cortexolone, 17α-hydroxyprogesterone, corticosterone, desoxycorticosterone, testosterone, estrone and dexamethasone.
Implants containing corticosteroids refers to compounds, such as e.g. fluocinolone, dexamethasone. "Other chemotherapeutic compounds" include, but are not limited to, plant alkaloids, hormonal compounds and antagonists; biological response modifiers, preferably lymphokines or interferons; antisense oligonucleotides or oligonucleotide derivatives; shRNA or siRNA; or miscellaneous compounds or compounds with other or unknown mechanism of action.
The compounds of the invention are also useful as co-therapeutic compounds for use in combination with other drug substances such as anti-inflammatory, bronchodilatory or antihistamine drug substances, particularly in the treatment of inflammatory diseases such as those mentioned hereinbefore, for example as potentiators of therapeutic activity of such drugs or as a means of reducing required dosaging or potential side effects of such drugs. A compound of the invention may be mixed with the other drug substance in a fixed pharmaceutical composition or it may be administered separately, before, simultaneously with or after the other drug substance. Accordingly the invention includes a combination of a compound of the invention as hereinbefore described with an anti-inflammator or antihistamine drug substance, said compound of the invention and said drug substance being in the same or different pharmaceutical composition.
Suitable anti-inflammatory drugs include steroids, in particular glucocorticosteroids such as budesonide, beclamethasone dipropionate, fluticasone propionate, ciclesonide or mometasone furcate, or steroids described in WO 02/88167, WO 02/12266, WO 02/100879, WO 02/00679 (especially those of Examples 3, 11 , 14, 17, 19, 26, 34, 37, 39, 51 , 60, 67, 72, 73, 90, 99 and 101), WO 03/035668, WO 03/048181 , WO 03/062259, WO 03/064445, WO 03/072592, non-steroidal glucocorticoid receptor agonists such as those described in WO 00/00531 , WO 02/10143, WO 03/082280, WO 03/082787, WO 03/104195, WO 04/005229; LTB4 antagonists such LY293111 , CGS025019C, CP-195543, SC-53228, BIIL 284, ONO 4057, SB 209247 and those described in US 5451700; LTD4 antagonists such as montelu- kast and zafirlukast; PDE4 inhibitors such cilomilast (Ariflo® GlaxoSmithKline), Roflumilast (Byk Gulden),V-11294A (Napp), BAY19-8004 (Bayer), SCH-351591 (Schering-Plough), Arofylline (Almirall Prodesfarma), PD189659 / PD168787 (Parke-Davis), AWD-12-281 (Asta Medica), CDC-801 (Celgene), SeICID(TM) CC-10004 (Celgene), VM554/UM565 (Vernalis), T-440 (Tanabe), KW-4490 (Kyowa Hakko Kogyo), and those disclosed in WO 92/19594, WO 93/19749, WO 93/19750, WO 93/19751 , WO 98/18796, WO 99/16766, WO 01/13953, WO 03/104204, WO 03/104205, WO 03/39544, WO 04/000814, WO 04/000839, WO 04/005258, WO 04/018450, WO 04/018451 , WO 04/018457, WO 04/018465, WO 04/ 018431 , WO 04/018449, WO 04/018450, WO 04/018451 , WO 04/018457, WO 04/018465, WO 04/019944, WO 04/019945, WO 04/045607 and WO 04/037805; A2a agonists such as those disclosed in EP 409595A2, EP 1052264, EP 1241176, WO 94/17090, WO 96/02543, WO 96/02553, WO 98/28319, WO 99/24449, WO 99/24450, WO 99/24451 , WO 99/38877, WO 99/41267, WO 99/67263, WO 99/67264, WO 99/67265, WO 99/67266, WO 00/23457, WO 00/77018, WO 00/78774, WO 01/23399, WO 01/27130, WO 01/27131 , WO 01/60835, WO 01/94368, WO 02/00676, WO 02/22630, WO 02/96462, WO 03/086408, WO 04/ 039762, WO 04/039766, WO 04/045618 and WO 04/046083; A2b antagonists such as those described in WO 02/42298; and beta-2 adrenoceptor agonists such as albuterol (salbutamol), metaproterenol, terbutaline, salmeterol fenoterol, procaterol, and especially, formoterol and pharmaceutically acceptable salts thereof, and compounds (in free or salt or solvate form) of formula I of WO 0075114, which document is incorporated herein by reference, preferably compounds of the Examples thereof, especially a compound of formula
Figure imgf000034_0001
OH and pharmaceutically acceptable salts thereof, as well as compounds (in free or salt or solvate form) of formula I of WO 04/16601 , and also compounds of WO 04/0334t2. Suitable bronchodilatory drugs include anticholinergic or antimuscarinic compounds, in particular ipratropium bromide, oxitropium bromide, tiotropium salts and CHF 4226 (Chiesi), and glycopyrrolate, but also those described in WO 01/04118, WO 02/51841 , WO 02/53564, WO 03/00840, WO 03/87094, WO 04/05285, WO 02/00652, WO 03/53966, EP 424021 , US 5171744, US 3714357, WO 03/33495 and WO 04/018422.
Suitable antihistamine drug substances include cetirizine hydrochloride, acetaminophen, clemastine fumarate, promethazine, loratidine, desloratidine, diphenhydramine and fexofenadine hydrochloride, activastine, astemizole, azelastine, ebastine, epinastine, mizolastine and tefenadine as well as those disclosed in WO 03/099807, WO 04/026841 and JP 2004107299.
Other useful combinations of compounds of the invention with anti-inflammatory drugs are those with antagonists of chemokine receptors, e.g. CCR-1 , CCR-2, CCR-3, CCR-4, CCR-5, CCR-6, CCR-7, CCR-8, CCR-9 and CCR10, CXCR1 , CXCR2, CXCR3, CXCR4, CXCR5, particularly CCR-5 antagonists such as Schering-Plough antagonists SC-351125, SCH- 55700 and SCH-D, Takeda antagonists such as N-[[4-[[[6,7-dihydro-2-(4-methylphenyl)-5H- benzo-cyclohepten-8-yl]carbonyl]amino]phenyl]-methyl]tetrahydro-N,N-dimethyl-2H-pyran-4- amin-ium chloride (TAK-770), and CCR-5 antagonists described in US 6166037 (particularly claims 18 and 19), WO 00/66558 (particularly claim 8), WO 00/66559 (particularly claim 9), WO 04/018425 and WO 04/026873.
Therapeutic agents for possible combination are especially one or more antiproliferative, cytostatic or cytotoxic compounds, for example one or several agents selected from the group which includes, but is not limited to, an inhibitor of polyamine biosynthesis, an inhibitor of a protein kinase, especially of a serine/threonine protein kinase, such as protein kinase C, or of a tyrosine protein kinase, such as the EGF receptor tyrosine kinase, e.g. Iressa®, the VEGF receptor tyrosine kinase, e.g. PTK787 or Avastin®, or the PDGF receptor tyrosine kinase, e.g. STI571 (Glivec®), a cytokine, a negative growth regulator, such as TGF-β or IFN-β, an aromatase inhibitor, e.g. letrozole (Femara®) or anastrozole, an inhibitor of the interaction of an SH2 domain with a phosphorylated protein, antiestrogens, topoisomerase I inhibitors, such as irinotecan, topoisomerase Il inhibitors, microtubule active agents, e.g. paclitaxel or an epothilone, alkylating agents, antiproliferative antimetabolites, such as gemcitabine or capecitabine, platin compounds, such as carboplatin or cis-platin, bisphosphonates, e.g. AREDIA®or ZOMETA®, and monoclonal antibodies, e.g. against HER2, such as trastuzumab.
The structure of the active agents identified by code nos., generic or trade names may be taken from the actual edition of the standard compendium "The Merck Index" or from databases, e.g. Patents International (e.g. IMS World Publications). The corresponding content thereof is hereby incorporated by reference.
The above-mentioned compounds, which can be used in combination with a compound of the formula I, can be prepared and administered as described in the art, such as in the documents cited above.
By "combination", there is meant either a fixed combination in one dosage unit form, or a kit of parts for the combined administration where a compound of the formula (I) and a combination partner may be administered independently at the same time or separately within time intervals, especially where these time intervals allow that the combination partners show a cooperative, e.g. synergistic effect.
The invention also provides a pharmaceutical preparation, comprising a compound of formula I as defined herein, or a pharmaceutically acceptable salt of such a compound, or a hydrate or solvate thereof, and at least one pharmaceutically acceptable carrier.
The compounds of the invention may be administered by any conventional route, in particular parenterally, for example in the form of injectable solutions or suspensions, enterally, e.g. orally, for example in the form of tablets or capsules, topically, e.g. in the form of lotions, gels, ointments or creams, or in a nasal or a suppository form. Topical administration is e.g. to the skin. A further form of topical administration is to the eye. Pharmaceutical compositions comprising a compound of the invention in association with at least one pharmaceutical acceptable carrier or diluent may be manufactured in conventional manner by mixing with a pharmaceutically acceptable carrier or diluent.
The invention relates also to pharmaceutical compositions comprising an effective amount, especially an amount effective in the treatment of one of the above-mentioned diseases (= disorders), of a compound of formula I or a pharmaceutically acceptable salt thereof together with one or more pharmaceutically acceptable carriers that are suitable for topical, enteral, for example oral or rectal, or parenteral administration and that may be inorganic or organic, solid or liquid. There can be used for oral administration especially tablets or gelatin capsules that comprise the active ingredient together with diluents, for example lactose, dextrose, mannitol, and/or glycerol, and/or lubricants and/or polyethylene glycol. Tablets may also comprise binders, for example magnesium aluminum silicate, starches, such as corn, wheat or rice starch, gelatin, methylcellulose, sodium carboxymethylcellulose and/or polyvinylpyrrolidone, and, if desired, disintegrators, for example starches, agar, alginic acid or a salt thereof, such as sodium alginate, and/or effervescent mixtures, or adsorbents, dyes, flavorings and sweeteners. It is also possible to use the pharmacologically active compounds of the present invention in the form of parenterally administrable compositions or in the form of infusion solutions. The pharmaceutical compositions may be sterilized and/or may comprise excipients, for example preservatives, stabilisers, wetting compounds and/or emulsifiers, so- lubilisers, salts for regulating the osmotic pressure and/or buffers. The present pharmaceutical compositions, which may, if desired, comprise other pharmacologically active substances are prepared in a manner known per se, for example by means of conventional mixing, granulating, confectionning, dissolving or lyophilising processes, and comprise approximately from 1% to 99%, especially from approximately 1% to approximately 20%, active ingredients).
Additionally, the present invention provides a compound of formula I or an N-oxide or a tau- tomer thereof, or a pharmaceutically acceptable salt of such a compound, for use in a method for the treatment of the human or animal body, especially for the treatment of a disease mentioned herein, most especially in a patient requiring such treatment.
Furthermore, the invention relates to a method for the treatment of a disease which responds to an inhibition of JAK-2 and/or Jak-3 kinase, which comprises administering a compound of formula I or a pharmaceutically acceptable salt thereof, wherein the radicals and symbols have the meanings as defined above, especially in a quantity effective against said disease, to a warm-blooded animal requiring such treatment.
Furthermore, the invention relates to a pharmaceutical composition for treatment of a disease, e.g. of solid or liquid tumours in warm-blooded animals, including humans, comprising a dose effective in the treatment of said disease of a compound of the formula I as described above or a pharmaceutically acceptable salt of such a compound together with a pharmaceutically acceptable carrier (= carrier material).
Processes of Manufacture
A compound of the formula I may be prepared by processes that, though not applied hitherto for the new compounds of the present invention where they thus form new processes, are known per se: preferably, a process for the manufacture of a compound of the formula I comprises either
a) reacting a compound of the formula II,
Figure imgf000037_0001
(II) wherein X, R1, R3 and R4 are as defined for a compound of the formula I and Hal is halo, especially bromo, under Suzuki coupling conditions with a boronic acid of the formula III,
R2-B(OH)2 (III)
wherein R2 is as defined for a compound of the formula I, or a reactive derivative thereof, or b) reacting a compound of the formula II,
Figure imgf000038_0001
wherein X, R1, R3 and R4 are as defined for a compound of the formula I and Hal is halo, especially bromo, under Stitle coupling conditions with an organotin compound of the formula
R2-Sn(alk)3 (III*)
wherein R2 is as defined for a compound of the formula I and alk is alkyl, preferably C1-C7 alkyl,
and, if desired, converting an obtainable compound of the formula I into a different compound of the formula I, converting an obtainable salt of a compound of the formula I into a different salt thereof, converting an obtainable free compound of the formula I into a salt thereof, and/or separating an obtainable isomer of a compound of the formula I from one or more different obtainable isomers of the formula I.
The reaction a) preferably takes place under Suzuki(-Miyaura) conditions, that is, by palladium-catalyzed crosscoupling of organoboranes, by reacting the halo- carrying compound of the formula Il with the boronic acid of the formula III, or a reactive derivative thereof. A reactive derivative of a boronic acid of the formula II! is preferably one wherein instead of the hydroxyl groups at the boron atom an aryl, alkenyl or especially alkyl moiety is present, or wherein the OH groups are present in bridged form, e.g. , together with the boron atom, forming a group of the formula (A)
Figure imgf000039_0001
The reaction preferably takes place in a mixture of a polar aprotic solvent, such as dimethyl- formamide (DMF) or tetrahydrofurane, and water in the presence of a catalyst for the cross- coupling, especially a noble metal catalyst, preferably a palladium catalyst, such as palla- dium(ll) complex, for example bis(triphenylphosphine)palladium (II) dichloride, in the presence of a base, such as potassium carbonate, sodium hydroxide or sodium carbonate, at a preferred temperature in the range from 60 0C to 130 0C, e.g. at about 80 0C; or according to a another preferred method in an ether solvent, e.g. tetrahydrofurane or 1 ,2- dimethoxyethane, in the presence of a catalyst for the cross coupling, especially a noble metal catalyst, preferably a palladium (0) complex, for example tris(dibenzylideneacetone)- dipalladium(O) or tetrakis (triphenylphosphin)palladium(O), in the presence of a base, such as sodium hydroxide, potassium carbonate of sodium carbonate, if desired in the presence of an appropriate ligand, such as 2-dicyclohexylphosphino-2',6'-dimethoxybiphenyl (SPhos), at a preferred temperature in the range from 60 to 150 °C, preferably from 70 to 110 0C; if required conducting the reaction in a sealed vessel (e.g. a seal reactor) if the boiling point of the reaction mixture is exceeded and especially if the heating is effected by microwave excitation. Preferably, oxygen is excluded, e.g. by the presence of an inert gas, such as nitrogen or especially argon.
The reaction b) given above is wherein in formula III* alk is alkyl, preferably d-Cz-alkyl, more preferably methyl, is preferably conducted under Stille coupling conditions, or in analogy thereto, preferably in an appropriate polar solvent, such as N,N-dimethylacetamide or N,N-dimethylformamide, an ether, such as tetrahydrofurane or dimethoxy-ethane, and/or a mixture of two or more such solvents, in the presence of a a palladium catalyst, especially a palladium (0) complex, for example tetrakistriphenylpalladium, e.g. at temperatures in the range from 80 to 160 0C, if required conducting the reaction in a sealed vessel (e.g. a seal reactor or a microwave vessel) if the boiling point of the reaction mixture is exceeded and/or especially if (as is a preferred embodiment) the heating is effected by microwave excitation.
Where temperatures are given hereinbefore or hereinafter, "about" has to be added, as minor deviations from the numeric values given, e.g. variations of ±10 %, are tolerable.
Protecting groups
If one or more other functional groups, for example carboxy, hydroxy, amino, sulfhydryl or the like are or need to be protected in a starting material of the formula Il or III or any other precursor, because they should not take part in the reaction or disturb the reaction, these are such groups as are usually used in the synthesis of peptide compounds, and also of cephalosporins and penicillins, as well as nucleic acid derivatives and sugars. Protecting groups are such groups that are no longer present in the final compounds once they are removed, while groups that remain as substituents are not protecting groups in the sense used here which is groups that are added at a starting material or intermediate stage and removed to obtain a final compound. Also in the case of conversions of a compound of the formula I into a different compound of the formula I, protecting groups may be introduced and removed, if useful or required.
The protecting groups may already be present in precursors and should protect the functional groups concerned against unwanted secondary reactions, such as acylations, etheri- fications, esterifications, oxidations, solvolysis, and similar reactions. It is a characteristic of protecting groups that they lend themselves readily, i.e. without undesired secondary reactions, to removal, typically by acetolysis, protonolysis, solvolysis, reduction, photolysis or also by enzyme activity, for example under conditions analogous to physiological conditions, and that they are not present in the end-products. The specialist knows, or can easily establish, which protecting groups are suitable with the reactions mentioned above and below.
The protection of such functional groups by such protecting groups, the protecting groups themselves, and their removal reactions are described for example in standard reference works, such as J. F. W. McOmie, "Protective Groups in Organic Chemistry", Plenum Press, London and New York 1973, in T. W. Greene, "Protective Groups in Organic Synthesis", Third edition, Wiley, New York 1999, in "The Peptides"; Volume 3 (editors: E. Gross and J. Meienhofer), Academic Press, London and New York 1981 , in "Methoden der organischen Chemie" (Methods of organic chemistry), Houben Weyl, 4th edition, Volume 15/1, Georg Thieme Verlag, Stuttgart 1974, in H. -D. Jakubke and H. Jescheit, "Aminosauren, Peptide, Proteine" (Amino acids, peptides, proteins), Verlag Chemie, Weinheim, Deerfield Beach, and Basel 1982, and in Jochen Lehmann, "Chemie der Kohtenhydrate: Monosaccharide und Derivate" (Chemistry of carbohydrates: monosaccharides and derivatives), Georg Thieme Verlag, Stuttgart 1974.
Optional Reactions and Conversions
A compound of the formula I may be converted into a different compounds of the formula I.
For example, in a compound of the formula I wherein R1 or especially R2 carries an amino or amino-Ci-Cy-alkyl substituent, the amino can be converted into acylamino, e.g. C1-C7- alkanoylamino or Ci-Cy-alkanesulfonylamino, by reaction with a corresponding C1-C7^l- kanoylhalogenide or Ci-Cr-alkanesulfonylhalogenide, e.g. a corresponding chloride, in the presence of a tertiary nitrogen base, such as triethylamine or pyridine, in the absence or presence of an appropriate solvent, such a methylene chloride, for example at temperatures in the range from -20 to 50 0C, e.g. at about room temperature.
In a compound of the formula I wherein R1 or especially R2 carries a cyano substituent, the cyano may be converted to an aminomethyl group, e.g. by hydrogenation in the presence of an appropriate metal catalyst, such as Raney Nickel or Raney Cobalt, in an appropriate solvent, e.g. a lower alkanol, such as methanol and/or ethanol, for example at temperatures in the range from -20 to 50 0C, e.g. at about room temperature.
In a compound of the formula I wherein R1 or especially R2 carries a carboxyl (COOH) substituent, the latter can be converted into an amide group, e.g. an N-C^Cr-alkyl-carba- moyl group, by reaction with the corresponding amine, e.g. in the presence of a coupling agent, that forms a preferred reactive derivative of the carboxyl group in situ, for example dicyclohexylcarbodiimide/i-hydroxybenzotriazole (DCC/ HOBT); bis(2-oxo-3-oxaz- olidinyl)phosphinic chloride (BOPCI); O-(1 ,2-dihydro-2-oxo-1-pyridyl)-Λ/,Λ/,Λ/',/V-tetramethyl- uronium tetrafluoroborate (TPTU); O-benzotriazol-1-yl)-N,N,N',N'-tetramethyluronium tetrafluoroborate (TBTU); (benzotriazol-1 -yloxy)-tripyrrolidinophosphonium-hexafluoro- phosphate (PyBOP), O-(1 H-6-chlorobenzotriazole-1-yl)-1 ,1 ,3,3-tetramethyluronium hexafluorophosphate, 1 -(S-dimethylaminopropylJ-S-ethylcarbodiimide hydrochloride/hy- droxybenzotriazole or/1-hydroxy-7-azabenzotriazole (EDC/HOBT or EDC/HOAt) or HOAt alone, or with (1-chloro-2-methyl-propenyl)-dimethylamine. For review of some other possible coupling agents, see e.g. Klauser; Bodansky, Synthesis (1972), 453-463. The reaction mixture is preferably stirred at a temperature. of between approximately -20 and 50 °C, especially between 0 0C and 30 0C, e.g. at room temperature.
In a compound of the formula I wherein R1 or especially R2 carries two vicinal amino groups, the two nitrogen atoms of the two amino groups can be bridged by a -CH= group (thus forming, together with the two carbon atoms that bind the original amino groups and the bond between them, an 7H-imidazolo ring annelated to R1 or R2; for example, (vicinal diamino)- phenyl can be converted into benzoimidazolyl according to this method. The reaction preferably takes place by first reacting the compound of the formula I carrying the two vicinal amino groups with formic acid, e.g. in the presence of a coupling agent as mentioned in the preceding paragraph, such as EDC hydrochloride, a base, such as N,N-dimethylaminopyridine (DMAP) and preferably an appropriate solvent, such as methylene chloride, e.g. at temperatures in the range from -20 to 50 0C, e.g. at about room temperature, thus converting one (especially a para-positioned) of the vicinal amino groups into a formylamino group. In a second step, the amino and formylamino group are then reacted to -N=C-N- by heating in the presence of an acid, especially acetic acid, e.g. at temperatures in the range from 50 to 110 0C, for example at about 100 "C.
Note that the intermediate with the formylamino group obtainable by the first reaction in the preceding paragraph is also a compound of the formula I, so that this first reaction also is a conversion reaction according to the invention.
Salts of a compound of formula I with a salt-forming group may be prepared in a manner known per se. Acid addition salts of compounds of formula I may thus be obtained by treatment with an acid or with a suitable anion exchange reagent. A salt with two acid molecules (for example a dihalogenide of a compound of formula I) may also be converted into a salt with one acid molecule per compound (for example a monohalogenide); this may be done by heating to a melt, or for example by heating as a solid under a high vacuum at elevated temperature, for example from 130 to 1700C, one molecule of the acid being expelled per molecule of a compound of formula I. Salts can usually be converted to free compounds, e.g. by treating with suitable basic compounds, for example with alkali metal carbonates, alkali metal hydrogencarbonates, or alkali metal hydroxides, typically potassium carbonate or sodium hydroxide.
Stereoisomeric mixtures, e.g. mixtures of diastereomers, can be separated into their corresponding isomers in a manner known per se by means of suitable separation methods. Dia- stereomeric mixtures for example may be separated into their individual diastereomers by means of fractionated crystallization, chromatography, solvent distribution, and similar procedures. This separation may take place either at the level of a starting compound or in a compound of formula I itself. Enantiomers may be separated through the formation of dia- stereomeric salts, for example by salt formation with an enantϊomer-pure chiral acid, or by means of chromatography, for example by HPLC, using chromatographic substrates with chiral ligands.
It should be emphasized- that reactions analogous to the conversions mentioned in this chapter may also take place at the level of appropriate intermediates (and are thus useful in the preparation of corresponding starting materials).
Starting materials:
The starting materials of the formulae Il and III, as well as other starting materials mentioned herein, e.g. below, can be prepared according to or in analogy to methods that are known in the art, are known in the art and/or are commercially available. Novel starting- materials, as" well as processes for the preparation thereof, are likewise an embodiment of the present invention. In the preferred embodiments, such starting materials are used and the reaction chosen are selected so as to enable the preferred compounds to be obtained.
In the starting materials (including intermediates), which may also be used and/or obtained as salts where appropriate and expedient, R1, R2, R3, R4 and X are preferably as defined for a compound of the formula I. Hal is halogen, especially chloro or bromo.
A compound of the formula Il can, for example, be obtained by reacting a thiourea compound of the formula IV,
Figure imgf000044_0001
under cyclization in the presence of an organic sulfonyl chloride, such as toluene-4-sulfonyl chloride, in an appropriate solvent, e.g. a cyclic ether, such as tetrahydrofurane, in the presence of water and a base, such as sodium hydroxide, at temperatures in the range from -20 to 50 0C, e.g. at about room temperature.
A thiourea compound of the formula IV can, for example, be prepared from an amino phenol of the formula V1
Figure imgf000044_0002
by reacting it with an isothiocyanate of the formula Vl,
R1-N=C=S (Vl)
e.g. in an appropriate solvent, e.g. a cyclic ether, such as tetrahydrofurane, at temperatures e.g. in the range from -20 to 50 0C, e.g. at about room temperature.
A compound of the formula V may, for example, be prepared by reducing a nitro compound of the formula VII,
Figure imgf000044_0003
(VII) e.g. with hydrogen in the presence of a catalyst, such as Raney-Nickel (Ra-Ni) or Raney Cobalt or the like, in an appropriate solvent, such as an alcohol, e.g. methanol, and/or a cyclic ether, such as tetrahydrofurane, at temperatures e.g. in the range from -20 to 50 0C, e.g. at about room temperature.
Alternatively, a compound of the formula Il can be prepared by reacting a methyl sulfanyl compound of .the formula VIII,
Figure imgf000045_0001
with an amine of the formula IX,
R1NH- (IX)
e.g. without solvent (e.g. in a melt or by dissolving the compound of the formula VIII in the amine of the formula IX) at elevated temperatures e.g. in the range from 50 to 150 0C, e.g. at about 100 °C. This reaction can preferably be conducted, in the presence of an agent capable of oxidising the methanesulfanyl at the oxazole ring to the methanesulfinyl, e.g. in the presence of a peroxide, such as m-chloroperbenzoic acid, and appropriate solvent, such as dichloromethane, preferably at temperatures in the range from 0 to 50 0C, e.g. at about room temperature.
A compound of the formula VIII can, for example, be prepared by reacting a thiol compound of the formula X,
Figure imgf000045_0002
(X) in an appropriate solvent, e.g. a dialkyl carboxylic acid amide, such as dimethylformamide, in the presence of a base, e.g. an alkali metal (such as potassium) carbonate, at temperatures e.g. in the range from -20 to 50 0C, e.g. at about room temperature, with a methyl halogenide, e.g. methyl iodide.
A compound of the formula X can, for example, be prepared from a compound of the formula V described (and obtainable as) above by reacting it with an alkali metal ethyl xanthoge- nate, such as potassium ethyl xanthogenate, in an appropriate solvent, such as an alcohol, e.g. ethanol, preferably at elevated temperatures, e.g. in the range from 50 0C to the reflux temperature of the reaction mixture, e.g. under reflux.
A compound of the formula III* can, for example, be prepared from a compound of the formula Xl,
R2-Hal (Xl)
wherein Hal is halσ, especially bromo, by reaction with a hexa-alkyl tin, especially hexa-C,- C7-alkyl-tin, e.g. hexamethyltin, in an appropriate solvent, such as toluene, and a customary noble metal catalyst, such as tetrakis(triphenylphosphine)palladium, preferably at elevated temperatures e.g. in the range from 50 to 150 0C.
Other starting materials, e.g. those of the formula Vl, VII, IX, X and Xl, are known in the art, can be prepared according to or in analogy to methods that are known in the art or in analogy to methods described in the Examples, and/or they are commercially available.
The following examples serve to illustrate the invention without limiting the scope thereof.
If not indicated otherwise, reactions take place at room temperature. Abbreviations used are:
Ac acetate
Ahx aminohexanoic acid
Brij 35 Polyoxyethylene(23)-laurylether (trademark of ICI
Americas, Inc.)
BSA bovine serum albumine
DMAP N,N-dimethylaminopyridine DMF dimethyl formamide
DTT Di-thiothreitol
DMSO dimethyl sulfoxide
EDC 1-(3-dimethylaminopropyl)-3-ethylcarbodiimide
EDTA ethylenediamine tetraacetate
Et3N triethylamine
EtOAc ethyl acetate
FITC fluoresceine-isothiocyanate derived fluoresceine moiety h hour(s)
Hepes 4-(2-hydroxyethyl)-1 -piperazineethanesulfonic acid
HOBT 1 -hydroxybenzotriazole
MeOH methanol min minute(s)
MS Mass Spectrometry
Ra-Ni Raney-Nickel
Rt Retention time
RT room temperature sat. saturated (at RT)
TFA trifluoro acetic acid
THF tetra hyd rof u ra ne
Tween 20 Polyoxyethylen(20)-sorbitan-monolaurate (ICI Americas,
Inc.)
Example 1 : (7-m-Tolyl-benzooxazol-2-ylH3,4,5-trimethoxy-phenyl)-amine.
Figure imgf000047_0001
0.12g (0.316mmol) (7-bromo-benzooxazol-2-yl)-(3,4,5-trimethoxy-phenyl)-amine and 0.048g (0.353mmol) 3-tolyl boronic acid are dissolved in 4ml 1 ,2-dimethoxy-ethane, a solution of 0.1g (0.95mmol) Na2CO3 (in 0.5ml water) is added and a stream of argon is bubbled through the mixture in order to exclude oxygen from the reaction mixture. Tetrakis (triphenylphosphine) palladium (76mg, 0.064mmol) is added and the reaction mixture is stirred at 1000C or 3h. After that the reaction mixture is poured on water and extracted 3x with EtOAc. The combined organic layers are washed with water, dried over MgSO4, filtered and the filtrate is concentrated in vacuo. The residue is purified by chromatography (silicagel, hexane: EtOAc = 3:1 => 1 :1) to afford the title compound as an off-white solid, m.p. 153-155°C. R, = 2.54 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min). MS: 391 (M+1)+ ; m.p. 155-1580C.
The starting materials can be prepared as follows:
a ) (7-Bromo-benzooxazol-2-yl )-(3,4.5-trimethoxy-phenyl )-amine .
To a suspension of 0.798mg (1.93mmol) 1-(3-bromo-2-hydroxy-phenyl)-3-(3,4,5-trimethoxy- phenyl)-thiourea in 8ml THF, 0.197g (4.8mmol) NaOH (in 5ml water) and 0.419g (2.12mmol) toluene-4-sulfonyl chloride are added. The reaction mixture is stirred at room temperature for 1.5h. Then the reaction mixture is concentrated in vacuo. To the residue EtOAc and sat. NaCI-solution are added and the layers are separate. The water layer is extracted 3x with EtOAc. The combined organic layers are washed with water, dried over MgSO4, filtered and the filtrate is concentrated in vacuo. The residue is stirred in diethyl ether, filtered and dried to afford the title compound.
bM-(3-Bromo-2-hydroxy-phenyl)-3-(3,4,5-trirnethoxy-phenyl)-thiourea.
A mixture of 0.376g (2.0mmol) 2-amino-6-bromo-phenol and 0.46g (2.0mmol) 3,4,5- trimethoxy-isothiocyanate in 10ml THF is stirred at room temperature for ca. 2Oh. The reaction mixture is concentrated in vacuo, followed by the addition of toluene and concentration. The toluene addition and evaporation is repeated one more time to afford the title compound as a brown solid.
c) 2-Amino-6-bromo-phenol.
5g (22.9mmol) 2-nitro-6-bromo-phenol (Fluka 67211) is hydrogenated in the presence of 0.1g Ra-Ni (B113W EtOH, Degussa) in 100ml of THF:MeOH = 1 :1. for 4h. The reaction mixture is filtered (2 glass fiber filters used) and the filtrate is concentrated in vacuo. The residue is purified by chromatography (silicagel, hexane: EtOAc = 2:1 ) to afford the title compound as a reddish oil which slowly solidifies.
Using the same synthetic methods as described in example 1 , reaction of (7-bromo- benzooxazol-2-yl)-(3,4,5-trimethoxy-phenyl)-amine and the appropriate boronic acid derivative leads to the following examples:
Example 2: (7-Phenyl-benzooxazol-2-yl)-(3,4,5-trimethoxy-phenyl)-amine.
R, = 2.47 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 377 (M+1 )+ ; m.p. 182-185°C.
Example 3: (7-Pyridin-3-yl-benzooxazol-2-ylH3.4,5-trimethoxy-phenyl)-amine.
R, = 1.80 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 378 (M+1)+ ; m.p. 165-1700C.
Example 4: f7-(3-Methoxy-phenyl)-benzooxazol-2-yll-(3,4,5-trimethoxy-phenyl)-amine.
R, = 2.45 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate LOrrtf/min); MS: 407 (M+1)+ ; m.p. 167-1700C.
Example 5: f7-(2-Methoxy-phenyl)-benzooxazol-2-yll-(3,4.5-trimethoxy-phenyl)-amine.
Rt = 2.40 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 407 (M+1)+ ; m.p. 185-187°C.
Example 6: f7-(3-Hvdroxy-phenyl)-benzooxazol-2-yll-(3,4,5-trimethoxy-phenyl)-amine.
R, = 2.22 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 393 (M+1)+ .
Example 7: [7-(4-Methoxy-phenyl)-benzooxazol-2-yll-(3,4,5-trimethoxy-phenyl)-amine. R, = 2.44 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.Oml/min); MS: 407 (M+ 1 )+ ; m.p. 179-182°C.
Example 8: (7-lsoquinolin-4-γl-benzooxazol-2-yl)-(3.4.5-trimethoxy-phenyl)-amine.
R, = 1.90 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1. Oml/min); MS: 428 (M+1 )+ ; m.p. 201-2030C.
Example 9: f7-(3-Chloro-phenyl)-benzooxazol-2-vn-(314.5-trimethoxy-phenyl)-amine.
R1 = 2.60 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1. Oml/min); MS: 411 (M+1 , 35CI)+ ; m.p. 199-2010C.
Example 10: f7-(3-Amino-phenyl)-benzooxazol-2-yll-(3,4,5-trimethoxy-phenyl)-amine.
R, = 1.88 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1. Oml/min); MS: 392 (M+1 )+ ; m.p. 145-1700C.
Example 11 : f7-(4-Amino-phenyl)-benzooxazol-2-yll-(3,4,5-trimethoxy-phenyl)-amine.
R, = 1.86 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1. Oml/min); MS: 392 (M+1 )+ ; m.p. 192-196°C.
Example 12: [7-(6-Methoxy-pyridin-3-yl)-benzooxazol-2-yll-(3,4.5-trimethoxy-phenyl)-amine.
R1 = 2.31 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1. Oml/min); MS: 408 (M+1 )+ ; m.p. 192-193°C.
Example 13: f7-(3-Amino-4-methyl-phenyl)-benzooxazol-2-vn-(3,4,5-trimethoxy-phenyl)- amine.
Rt = 1.94 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1. Oml/min); MS: 406 (M+1)+ ; m.p. 188-1900C. Example 14: f7-(2-Amino-phenyl)-benzooxazol-2-yll-(3,4.5-trimethoxy-phenyl)-amine.
R, = 2.06 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 392 (M+1)+
Example 15: (7-Quinolin-6-yl-benzooxazol-2-yl)-(3.4,5-trimetrιoxy-phenyl)-amine.
R, = 1.91 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 428 (M+1)+ ; m.p. 187-1900C.
Example 16: 4-f2-(3,4,5-Trimethoxy-phenylamino)-benzooxazol-7-vπ-benzamide.
R, = 2.08 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 420 (M+1 )+ ; m.p. >280°C.
Example 17: f7-(4-Methanesulfonyl-phenyl)-benzooxazol-2-ylH3,4,5-trimethoxy-phenyl)- amine.
R, = 2.26 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 455 (M+1)+ ; m.p. 205-2070C.
Example 18: 4-f2-(3,4,5-Trimethoxy-phenylamino)-benzooxazol-7-yll-benzenesulfonamide.
R, = 2.16 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 456 (M+1)+ ; m.p. 247-252°C.
Example 19: 4-f2-(3,4,5-Trimethoxy-phenylamino)-benzooxazol-7-vn-benzonitrile.
Rt = 2.42 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 402 (M+1>+ ; m.p. 222-223°C.
Example 20: f7-(2H-Pvrazol-3-vl)-benzooxazol-2-vn-(3.4.5-trimethoxv-phenyl)-amine. R, = 2.00 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 367 (M+1 )+ ; m.p. 185-189°C.
Example 21 : N-Methyl-4-[2-(3,4,5-trimethoxy-phenylamino)-benzooxazol-7-yll- benzenesulfonamide.
R, = 2.26 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 470 (M+1 )+ ; m.p. 260-2620C.
Example 22: N,N-Dimethyl-4-[2-(3,4,5-trimethoxy-phenylamino)-benzooxazol-7-vn- benzenesulfonamide.
Rt = 2.39x min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 484 (M+1)+ ; m.p. 230-2330C.
Example 23: {7-f4-(Morpholine-4-sulfonyl)-phenyl1-benzooxazol-2-ylH3,4,5-trimethoxy- phenyQ-amine.
R, = 2.36 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 526 (M+1)+ ; m.p. 201-2030C.
Example 24: N-Methyl-4-f2-(3,4,5-trimethoxy-phenylamino)-benzooxazol-7-yl1-benzamide.
R, = 2.13 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 434 (M+1)+ ; m.p. 258-259°C.
Example 25: N,N-Dimethyl-4-f2-(3,4.5-trimethoxy-phenylamino)-benzooxazol-7-yl1-benz- amide.
R, = 2.19 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 448 (M+1)+ ; m.p. >280°C.
Example 26: N-{4-f2-(3,4I5-Trimethoxy-phenylamino)-benzooxazol-7-vn-phenyl>- methanesulfonamide. A mixture of 0.12g (0.307mmol) [7-(4-amino-phenyl)-benzooxazol-2-yl]-(3,4,5-trimethoxy- pheny!)-amine (example 11 ) and 0.041 g (0.36mmol) methanesulfonyl chloride in 6ml pyridine is stirred for 1.5h at room temperature. Then the reaction mixture is poured on water and extracted 3x with EtOAc. The combined organic layers are washed with 0.1 N NaOH solution and water, dried over MgSO4, filtered and the filtrate is concentrated in vacuo. The residue is purified by crystallisation from dichloromethane/diethyl ether to afford the title compound as off-white crystals. Rt = 2.22 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 470 (M+1 )+ ; m.p. 222-225°C.
Example 27: N-{4-f2-(3,4,5-Trimethoxy-phenylamino)-benzooxazol-7-vn-phenyl)-acetamide.
A mixture of 0.075g (0.1927mmol) t7-(4-amino-phenyl)-benzooxazol-2-yl]-(3,4,5-trimethoxy- phenyl)-amine (example 11), 0.027 ml triethylamine and 0.016g (0.199mmol) acetyl chloride in 4ml dichloromethane is stirred for 1h at room temperature. Then the reaction mixture is poured on water and extracted 3x with EtOAc. The combined organic layers are washed with water and saturated NaCI solution, dried over MgSO4, filtered and the filtrate is concentrated in vacuo. The residue is purified by chromatography (silicagel, EtOAc) to afford the title compound as orange crystals. Rt = 2.16 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 434 (M+1 )+ ; m.p. 265-2700C.
Using the same reaction conditions the following example is prepared from [7-(3-amino- phenyl)-benzooxazol-2-yl]-(3,4,5-trimethoxy-phenyl)-amine (Example 10):
Example 28: N-{3-f2-(3,4.5-Trimethoxy-phenylamino)-benzooxazol-7-vπ-phenyl)-acetamide.
R, = 2.21 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 434 (M+1)+ ; m.p. 170-1720C.
Example 29: r7-(4-Aminomethvl-phenvl)-benzooxazol-2-vlM3,4,5-trimethoxv-phenvl)-amine. To a solution of 0.117g (0.291mmol) 4-[2-(3,4,5-trimethoxy-phenylamino)-benzooxazol-7-yl]- benzonitrile (example 19) in 5ml MeOH (MeOH contains 5% NH3), 5ml THF and 30mg Ra-Ni (B113W EtOH, Degussa) are added. Then this mixture is hydrogenated under normal pressure for 2Oh at room temperature. The reaction mixture is filtered (2 glass fiber filters used) and the filtrate is concentrated in vacuo. The residue is purified by crystallization from dichloromethane/diethyl ether to afford the title compound as a light grey solid. R1 = 1.85 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 406 (M+1)+ ; m.p. 208-2110C.
Example 30: N-{4-[2-(3,4,5-Trimethoxy-phenylaminoVbenzooxazol-7-yll-benzyl)-acetamide.
A mixture of 0.07Og (0.173mmol) [7-(4-aminomethyl-phenyl).-benzooxazol-2-yl]-(3,4,5- trimethoxy-phenyl)-amine (example 29), 1ml pyridine and 0.016g (0.199mmol) acetyl chloride is stirred for 1.5h at room temperature. Then the reaction mixture is poured on water and extracted 2x with EtOAc. The combined organic layers are washed with water and 0.1 N NaOH solution, dried over MgSO4, filtered and the filtrate is concentrated in vacuo and co- evaporated twice with toluene. The residue is purified by crystallization from dichloromethane/diethyl ether to afford the title compound as a light brown solid. R1 = 2.12 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 448 (M+1)+ ; m.p. 237-239°C.
Example 31 : N-{4-[2-(3,4,5-Trimethoxy-phenylamino)-benzooxazol-7-vπ-benzyl)- methanesulfonamide.
A mixture of 0.1Og (0.247mmol) [7-(4-aminomethyl-phenyl)-benzooxazol-2-yl]-(3,4,5- trimethoxy-phenyl)-amine (example 29) and 0.037g (0.32mmol) methanesulfonyl chloride in 4ml pyridine is stirred for 1.5h at room temperature. Then the reaction mixture is poured on water and extracted 3x with EtOAc. The combined organic layers are washed with 0.1 N NaOH solution and water, dried over MgSO4, filtered and the filtrate is concentrated in vacuo and co-evaporated twice with toluene. The residue is purified by crystallisation from dichloromethane to afford the title compound as off-white crystals. R, = 2.20 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 474 (M+1)+. Using the synthetic methods described in examples 29 and 30, the following examples are prepared:
Example 32: r7-(3-Aminomethyl-phenyl)-benzooxazol-2-ylH3,4,5-trimethoxy-phenyl)-amine.
R, = 1.86 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 406 (M+1 )+ ; m.p. 145-1500C.
Example 33: N-{3-f2-(3,4,5-Trimethoxγ-phenylamino)-benzooxazol-7-yll-benzyl>-acetamide.
R, = 2.16 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 448 (M+1 )+ ; m.p. 203-2040C.
Example 34: 4-f2-(3,4,5-Trimethoxy-phenylaminoV-benzooxazol-7-vπ-thiophene-2-carboxylic acid methylamide.
A mixture of 0.04g (0.083mmol) 4-[2-(3,4,5-trimethoxy-phenylamino)-benzooxazol-7-ylJ- thiophene-2-carboxylic acid, 0.025g (0.127mmol) EDC-HCI, 0.016g (0.128mmol) DMAP, 0.83ml (1.7mmol) methylamine THF solution (2M), 0.012g (0.088mmol) HOBt and 4ml dichloromethane is stirred at room temperature for 72h. Then EtOAc is added to the reaction mixture and the organic layer is washed with water (2x), dried over MgSO4, filtered and the filtrate is concentrated in vacuo. The residue is purified by chromatography (silicagel, hexane:EtOAc = 1 :2), followed by crystallization from dichloromethane/diethyl ether to afford the title compound as off-white crystals. Rt = 2.15 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 440 (M+1)+ ; m.p. 260-262°C.
The starting material 4-[2-(3,4,5-trimethoxy-phenylamino)-benzooxazol-7-yl]-thiophene-2- carboxylic acid is prepared from 2-carboxythiophene-4-boronic acid pinacol ester as described in Example 1.
Example 35: f7-(1 H-Benzoimidazol-5-yl)-benzooxazol-2-yn-(3,4,5-trimethoxy-phenyl)-amine. A solution of 0.182g (0.243mmol) N-{2-amino-4-[2-(3,4,5-trimethoxy-phenylamino)- benzooxazol-7-yl]-phenyl}-formamide in 3ml acetic acid is stirred at 1000C for 1h. Then EtOAc is added to the reaction mixture and the organic layer is washed with 4N NaOH solution (2x) and with water (2x), dried over MgSO4, filtered and the filtrate is concentrated in vacuo. The residue is purified by chromatography (silicagel, EtOAc => EtOAc:MeOH = 95:5) to afford the title compound as an orange solid. R1 = 1.86 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 417 (M+1 )+ ; m.p. 150-1560C.
The starting material N-{2-amino-4-[2-(3,4,5-trimethoxy-phenylamino)-benzooxazol-7-yl]- phenyl}-formamide is prepared as follows:
A mixture of 0.16g (0.228mmol) 4-[2-(3,4,5-trimethoxy-phenylamino)-benzooxazol-7-yl]- benzene-1 ,2-diamine , 0.068g (0.346mmo)l EDC-HCI, 0.043g (0.345mmol) DMAP, 0.012g (0.27mmol) formic acid), and 7ml dichloromethane is stirred at room temperature for 2Oh. Then the reaction mixture is concentrated in vacuo. The residue is purified by chromatography (silicagel, EtOAc: MeOH = 9:1) to afford ca. the title compound. 4-[2-(3,4,5-trimethoxy-phenylamino>-benzooxazol-7-yl]-benzene-1 ,2-diamine is prepared from 4-(4,4,5,5-tetramethyl-[1 , 3, 2}dioxaborolan-2-yl)-benzene-1 ,2-diamine as described in Example 1.
4-(4,4,5,5-tetramethyl-[1 ,3,2]dioxaborolan-2-yl)-benzene-1 ,2-diamine is prepared from the commercially available 2-nitro-4-(4,4,5,5-tetramethyl-[1 ,3,2]dioxaborolan-2-yl)-phenylamine using the Ra-Ni catalyzed nitro reduction method described in Example 1 (step c).
Example 36: f4-(4-Amino-phenyl)-oxazolo[5,4-clpyridin-2-yll-(3,4,5-trimethoxy-phenyl)- amine.
0.082g (0.209mmol) (4-bromo-oxazolo[5,4-c]pyridin-2-yl)-(3,4,5-trimethoxy-phenyl)-amine and 0.04g (0.22mmol) (4-aminophenyl) boronic acid are dissolved in 3ml 1 ,2-dimethoxy- ethane, a solution of 0.044g (0.425mmol) Na2CO3 (in 0.6ml water) is added and a stream of argon is bubbled through the mixture in order to exclude oxygen from the reaction mixture. Tetrakis (triphenylphosphine) palladium (0.0259g, 0.021 mmol) is added and the reaction mixture is stirred at 1000C or 83h. After that the reaction mixture is poured on water and extracted 3x with EtOAc. The combined organic layers are washed with water and saturated NaCI solution, dried over MgSO4, filtered and the filtrate is concentrated in vacuo. The residue is purified by chromatography (silicagel, hexane => EtOAc) and recrystallisation from dichloromethane/diethyl ether to afford the title compound. Rt = 1.70 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 393 (M+1 )+ ; m.p. 148-1510C.
The starting materials can be prepared as follows:
a> (4-Bromo-oxazolof5,4-cipyhdin-2-γl)-(3,4,5-trimethoxy-phenyl)-amine.
0.6g (2.61 mmol) 4-bromo-2-methylsulfanyl-oxazolo[5,4-c]pyridine are heated to 1000C until it liquefies, then 0.977g (5.23mmol) 3,4,5-trimethoxyanilin is added in small portions with stirring. Stirring is continued for 2h. The reaction mixture is cooled to room temperature and purified by chromatography (silicagel, hexane => EtOAc) to afford the title compound.
b) 4-Bromo-2-methylsulfanyl-oxazolo[5,4-clpyridine
A mixture of 1.21g (5.24mmol) 4-bromo-oxazolo[5,4-c]pyridine-2-thiol, 0.8g (5.76mmol) K2CO3, 0.9g (6.28mmol) MeI in 12ml DMF is stirred at room temperature for 1h. The reaction mixture is then poured on water and extracted 2x with EtOAc. The combinded organic layers are washed with water and saturated NaCI solution, dried over MgSO4, filtered and the filtrate is concentrated in vacuo to afford the title compound in quantitative yiekJ.
c) 4-Bromo-oxazolo[5,4-clpyridine-2-thiol
1.34g (7.09mmol) 4-amino-2-bromo-pyridin-3-ol are dissolved in 13ml EtOH and 1.86g (11.3mmol) potassium ethyl xanthogenate are added. This mixture is stirred for 18h at reflux temperature. After cooling to room temperature, the reaction mixture is concentrated in vacuo and 5ml water are added. With the addition of acetic acid, a pH of 5 is adjusted. The product starts to crystallize and is filtered off, washed 2x with water and dried to afford the title compound.
d) 4-Amino-2-bromo-pyridin-3-ol A solution of 5.3g (22.7mmol) 2-bromo-4-nitro-pyridin-3-oI in 100ml MeOH:THF = 1 :2 is hydrogenated in the presence of 0.5g Pt/C (5%, Engelhard 4709). The reaction mixture is filtered (2 glass fiber filters used) and the filtrate is concentrated in vacuo to afford the crude title compound as a brown solid.
e) 2-Bromo-4-nitro-pyridin-3-ol
To a solution of 13g (73.2mmol) 2-bromo-3-pyridinol (Fluka 18292) in 40ml of cone, sulfuric acid 5.1 ml (74mmol) of nitric acid (65%) are added at 00C. The reaction mixture is stirred at 00C for 12h, then poured on water and extracted 2x with EtOAc. The combinded organic layers are washed with water and saturated NaCI solution, dried over MgSO4, filtered and the filtrate is concentrated in vacuo. The residue is purified by chromatography (silicagel, EtOAc) to afford 5.3g of 2-bromo-4-nitro-pyridin-3-ol and 2g of 2-bromo-6-nitro-pyridin-3-ol.
Using the same synthetic methods as described in Example 36, reaction of (4-bromo- oxazolo[5,4-c]pyridin-2-yl)-(3,4,5-trimethoxy-phenyl)-amine and the appropriate boronic acid derivative leads to the following examples:
Example 37: f4-(3-Amino-phenyl)-oxazolof5,4-c1pyridin-2-yll-(3,4,5-trimethoxy-phenyl)- amine.
R, = 1.63 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 393 (M+1)+ ; m.p. 120-1260C.
Example 38: 4-f2-(3,4.5-Trimethoxy-phenylamino)-oxazolof5,4-clpyridin-4-yll- benzenesulfonamide.
R, = 1.71 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 457 (M+1 )+ ; m.p. >280°C.
Using the appropriately substituted isothiocyanate derivative in the synthetic methods described in example 1 (step b), the following derivatives are obtained: Example 39: [7-(3-Methoxy-phenyl)-benzooxazol-2-yll-(4-morpholin-4-yl-phenyl)-amine. (from 4-morpholinophenyl-isothiocyanate)
R, = 2.14 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 402 (M+1 )+ ; m.p. 180-1820C.
Example 40: r7-(4-Amino-phenyl)-benzooxazol-2-yll-(4-morpholin-4-yl-prienyl)-amine. (from 4-morpholinophenyl-isothiocyanate)
Rt = 1.65 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 387 (M+1 )+ ; m.p. 241-2430C.
Example 41 : N-{4-[2-(4-Morpholin-4-yl-phenylamino)-benzooxazol-7-vn-phenyl}-methane- sulfonamide. (from 4-morpholinophenyl-isothiocyanate)
Rt = 1.94 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 387 (M+1)+ ; m.p. 258-265°C.
Example 42: f7-(3-Amino-phenyl)-benzooxazol-2-vn-(6-morpholin-4-yl-pyridin-3-yl)-amine. (from 4-(5-lsothiocyanato-pyridin-2-yl)-morpholine)
Rt = 1.576 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 388 (M+ 1 )* ; m.p. 235-237°C.
The following compounds are prepared in analogy to the methods described herein:
Example 43: f7-(3-Amino-phenyl)-Denzooxazol-2-yll-(4-morpholin-4-yl-phenyl)-amine:
Figure imgf000059_0001
R, = 1.66 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 387 (M+1 )+ . Example 44: (7-o-To)yl-benzooxazol-2-yl)-(3,4.5-trimethoxy-phenyl)-amine:
Figure imgf000060_0001
R, = 2.53 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 391 (M+1 )+ .
Example 45: 3-f2-(3,415-Trimethoxy-phenylamino)-benzooxazol-7-vπ-benzonitrile:
Figure imgf000060_0002
R, = 2.43 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 402 (M+ 1 )+ .
Example 46: (7-Pyridin-2-yl-benzooxazol-2-ylM3,4,5-trimethoxy-phenyl)-amine:
Figure imgf000060_0003
R, = 1.87 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 378 (M+1 >+ .
Example 47: 3-[2-(3,415-Trimethoxy-phenylamino)-benzooxazol-7-yll-benzamide:
Figure imgf000060_0004
Rt = 2.12 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.Oml/min); MS: 420 (M+1 )+ . Example 48: f7-(3-Nitro-phenyl)-benzooxazol-2-vn-(3,4,5-trimethoxy-phenyl)-amine:
Figure imgf000061_0001
R, = 2.49 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 422 (M+1 )+ .
Example 49: f7-(3-Methanesulfonyl-phenyl)-benzooxazol-2-vn-(3.4,5-trimethoxy-phenyl)- amine:
Figure imgf000061_0002
R, = 2.28 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 455 (M+1)+ .
Example 50: N-Methyl-3-f2~(3,4.5-trimethoxy-phenylamino)-benzooxazol-7-yll-benzene- sulfonamide:
Figure imgf000061_0003
R, = 2.29 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.Oml/min); MS: 470 (M+1 )+ .
Example 51 : f7-(4-Methoxy-3-nitro-phenγl)-benzooxazol-2-ylH3 A5-trimethoxy-phenyl)- amine:
Figure imgf000062_0001
R, = 2.45 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 452 (M+1 )+ .
Example 52: f7-(3-Amino-4-chloro-phenyl)-benzooxazol-2-ylH3A5-trimethoxy-phenyl)- amine:
Figure imgf000062_0002
R, = 2.4 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 426 (M+1)+ .
Example 53: r7-(3-Amino-4-methoxy-phenyl)-benzooxazol-2-yll-(3,4,5-trimethoxy-phenyl)- amine:
Figure imgf000062_0003
R, = 1.91 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 422 (M+1)+ .
Example 54: [7-(4-Dimethylamino-phenyl)-benzooxazol-2-vn-(3,4,5-trimethoxy-phenyl>- amine:
Figure imgf000063_0001
Rt = 1.98 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 420 (M+1)+ .
Example 55: N-{3-f2-(3.4,5-Trimethoxy-phenylamino)-benzooxazol-7-yll-phenyl>- methanesulfonamide:
Figure imgf000063_0002
R, = 2.24 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 470 (M+1)+ .
The following Examples are prepared in analogy to the procedures described above:
Example 56: N-(2-Methoxy-ethyl)-4-r7-(4-sulfamoyl-phenyl)-benzooxazol-2-ylamino1- benzamide:
Figure imgf000063_0003
R, = 2.00 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 467 (M+1 )+ .
Example 57: N-(2-Dimethylamino-ethyl)-4-f7-(4-sulfamoyl-phenyl)-benzooxazol-2-ylamino1- benzamide:
Figure imgf000064_0001
R, = 1.77 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 480 (M+1 )\
Example 58: 4-[2-(3,4-Dimethoxγ-phenylamino)-benzooxazol-7-vn-benzenesulfonamide:
Figure imgf000064_0002
Rt = 2.07 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 426 (M+1)+ .
Example 59: 4-[2-(3,4-Dimethoxy-phenylamino)-benzooxazol-7-vH-N-methyl- benzenesulfonamide:
Figure imgf000064_0003
R, = 2.19 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 440 (M+1 )+ .
Example 60: 4-f2-(3,5-Dimethoxy-phenylamino)-benzooxazol-7-vn-benzenesulfonamide:
Figure imgf000065_0001
R, = 2.26 min (Waters Symmetry C8, 2.1x5Omm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 426 (M+1 )+ .
Example 61 : N-Methyl-2-{4-[2-(3 A5-trimethoxy-phenylamino)-benzooxazol-7-yl]-phenyl)- acetamide:
Figure imgf000065_0002
R, = 2.1 1 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.Oml/min); MS: 448 (M+1 )+ .
Example 62: N-{4-[2-(3,4,5-Trimethoxy-phenylamino)-oxazolof5,4-c1pyridin-4-vπ-benzyl>- methanesulfonamide:
Figure imgf000065_0003
R, = 1.73 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1. Oml/min); MS: 485332 (M+1 )+ .
Example 63: f7-(4-Methanesulfinylmethyl-phenyl)-benzooxazol-2-vH-(3,4,5-trimethoxy- phenvD-amine:
Figure imgf000066_0001
R, = 2.08 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 453 (M+1 )+ .
Example 64: 4-f7-(3-Fluoro-4-morpholin-4-ylmethyl-phenyl)-benzooxazol-2-ylamino1-2,N,N- trimethyl-benzamide:
Figure imgf000066_0002
0.1g (0.267mmol) 4-(7-bromo-benzooxazol-2-ylamino)-2,N,N-trimethyl-benzamide and 0.17g (0.305mmol) 4-(2-fluoro-4-trimethylstannanyl-benzyl)-morpholine are dissolved in 2ml 1 ,2- dimethoxy-ethane and a stream of argon is bubbled through the mixture in order to exclude oxygen from the reaction mixture. Tetrakis (triphenylphosphine) palladium (0.02Og, 0.016mmol) is added and the reaction mixture is stirred at 1500C for 1h. After that the reaction mixture is poured on water and extracted 3x with EtOAc. The combined organic layers are washed with water and saturated NaCI solution, dried over MgSO4, filtered and the filtrate is concentrated in vacuo. The residue is purified by chromatography (silicagel, EtOAc : MeOH = 95:5; column chromatography followed by thick-layer chromatography) to afford the title compound. Rt = 1.67 min (Waters Symmetry C8, 2.1x50mm, detection 210- 25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 375 (M+1 )\
Preparation of starting materials:
a) 4-(7-Bromo-benzooxazol-2-ylamino)-2,N,N-trimethyl-benzamide: To a solution of 0.49g (2.01 mmol) 7-bromo-2-methylsulfanyl-benzooxazole in 30ml dichloromethane, 3-chloroperbenzoic acid (0.494g, 2.01 mmol) is added. This mixture is stirred for 1h at room temperature. Then, 0.325g (1.82mmol) 4-amino-2,N,N-trimethyl- benzamide is added and the reaction mixture is heated to 400C with stirring continued for 2Oh. The reaction mixture is concentrated in vacuo and the residue is purified by chromatography (silicagel, hexane : EtOAc = 1 :1 => 1 :4) to afford the title compound as an off-white crystalline solid.
b) 7-Bromo-2-methylsulfanyl-benzooxazole:
A mixture of 6g (26.1 mmol) 7-bromo-benzooxazole-2-thiol, 7.28g (52.2mmol) K2CO3, 4.1g (28.7mmol) MeI in 80ml DMF is stirred at room temperature for 1h. The reaction mixture is then poured on water and extracted 2x with EtOAc. The combined organic layers are washed with water and saturated NaCI solution, dried over MgSO4, filtered and the filtrate is concentrated in vacuo to afford the title compound.
c) 7-Bromo-benzooxazole-2-thiol:
5g (26.6mmol) 2-amino-6-bromo-phenol are dissolved in 20ml EtOH and 6.52g (39.9mmol) potassium ethyl xanthogenate are added. This mixture is stirred for 6h at reflux temperature. After cooling to room temperature, the reaction mixture is concentrated in vacuo and 50ml water are added. With the addition of acetic acid, a pH of 5 is adjusted. The product starts to crystallize and is filtered off, washed 2x with water and dried to afford the title compound.
d) 2-Amino-6-bromo-phenol:
A solution of 5.3g (22.7mmol) 2-bromo-6-nitro-phenol in 100ml MeOH:THF = 1 :1 is hydrogenated in the presence of 0.2g Ra-Ni (in EtOH, Degussa B113W). The reaction mixture is filtered (2 glass fiber filters used) and the filtrate is concentrated in vacuo to afford the crude title compound.
f) 4-Amino-2,N,N-trimethyl-benzamide:
A solution of 1.7g (8.16mmol) 2,N,N-trimethyl-4-nitro-benzamide in 45ml MeOHTHF = 1 :1 is hydrogenated in the presence of 0.2g 10% Pd/C (Fluka 75990). The reaction mixture is filtered (2 glass fiber filters used) and the filtrate is concentrated in vacuo to afford the crude title compound.
g) 2,N,N-Trimethyl-4-nitro-benzamide:
A solution of 2g (10.7mmol) 2-methyl-4-nitro-benzoic acid, 1.32ml (11.8mmol) N-methyl- morpholine, 2.36g (12mmol) EDC-HCI and 2.36g (12mmol) HOBt in 50ml dichloromethane is stirred at RT for 45 min, then dimethylamine solution (5.9ml, 33% in EtOH) is added and the reaction mixture is heated to 400C and stirred at this temperature for 2Oh. Another 1.32ml (11.8mmol) N-methyl-morpholine, 2.36g (12mmol) EDC-HCI and 2.36g (12mmol) HOBt (hydroxyl-benzotriazole) are added and stirred for 45 min, the again dimethylamine solution (5.9ml, 33% in EtOH) is added and the reaction mixture is stirred at 400C for 35h. After that the reaction mixture is cooled to RT and poured onto EtOAc/water. The organic layer is washed with sat. NAHCO3 solution and with water, dried over MgSO4, filtered and the filtrate is concentrated in vacuo. The residue is purified by chromatography (silicagel, hexane : EtOAc = 1:1 => EtOAc) to afford the title compound as a yellowish oil.
h) 4-(2-Fluoro-4-trimethylstannanyl-benzyl)-morpholine:
A solution of 1g (3.65mmol) 4-(4-bromo-2-fluoro-benzyl)-morpholine and 0.95ml (4.56mmol) hexamethylditin in 15ml toluene is prepared and a stream of argon is bubbled through the mixture in order to exclude oxygen from the reaction mixture. Tetrakis (triphenylphosphine) palladium (0.221 g, 0.185mmol) is added and the reaction mixture is stirred at 1100C for 5h. The reaction mixture is allowed to cool to RT and is filtered through a layer of Hyflo. The filtrate is concentrated in vacuo and further dried under high vacuum for 2Oh to afford the title compound as a yellowish oil.
Example 65: 4-(7-f4-(1 ,1-Dioxo-thiomorpholin-4-ylmethyl)-3,5-difluoro-phenvπ-benzooxazol- 2-ylamino}-2,N,N-trimethyl-benzamide:
Figure imgf000069_0001
A solution of 0.1g (0.267mmol) 4-(7-bromo-benzooxazol-2-ylamino)-2,N,N-trimethyl- benzamide, 0.114g (0.294mmol) 4-f2,6-difluoro-4-(4,4,5,5-tetramethyl-[1 ,3,2]dioxaborolan-2- yl)-benzyl]-thiomorpholine 1 ,1-dioxid and 174mg K3PO4 are dissolved in 5ml 1 ,2-dimethoxy- ethane, water (0.1ml) is added and a stream of argon is bubbled through the mixture in order to exclude oxygen from the reaction mixture. Tetrakis (triphenylphosphine) palladium (0.0095g, 0.0082mmol) is added and the reaction mixture is stirred at 1000C for 8h. After that the reaction mixture is poured on water and extracted 3x with EtOAc. The combined organic layers are washed with water and saturated NaCI solution, dried over MgSO4, filtered and the filtrate is concentrated in vacuo. The residue is purified by chromatography (silicagel, EtOAc), followed by recrystallisation diethylether/methanol to afford the title compound as white crystals. R1 = 2.13 min (Waters Symmetry C8, 2.1x50mm, detection 210- 25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 555 (M+1 )+.
Preparation of starting materials:
a) 4-f2.6-difluoro-4-(4,4,5,5-tetramethyl-f1 ,3,21dioxaborolan-2-γl)-benzvπ-thiomorpholine 1.1- dioxide:
A solution (degassed with argon) of 1.02g (3.0mmol) 4-(4-bromo-2,6-difluoro-benzyl)- thiomorpholine 1 ,1 -dioxide in 4ml dimethylacetamide is added to a solution (degassed with argon) of 0.855g (3.3mmol) bis-(pinacolato)-diboron and 0.594g (Θ.Ommol) dried KOAc in 4ml dimethylacetamide. After that 0.076g (0.092mmol) Pd(dppf)CI2-CH2CI2 is added. The reaction mixture is heated to 800C and stirred at this temperature for 4h. Then the reaction mixture is cooled to room temperature and poured on water and extracted 3x with EtOAc. The combined organic layers are washed with water and saturated NaCI solution, dried over MgSO4, filtered and the filtrate is concentrated in vacuo to afford the title compound as a dark brown oil. Note: in some cases, (partial) hydrolysis to the free boronic acid derivative occurs, however, this is no problem as the free boronic acid (or a mixture of free boronic acid with the tetramethyl[1 ,3,2Jdioxaborolane derivative work equally well in the Suzuki coupling reaction.
b) 4-(4-Bromo-2.6-difluoro-benzyl)-thiomorpholine 1 ,1-dioxide:
A solution of 3.8g (11.3mmol) 5-bromo-2-bromomethyl-1 ,3-difluoro-benzene, 1.83g (13.6mmol) thiomorpholine dioxide and 1.88ml (13.6mmol) triethylamine in 40ml dichlorometbane is stirred at room temperature for 2Oh. Then the reaction mixture is poured on water and extracted 3x with EtOAc. The combined organic layers are washed with water and saturated NaCI solution, dried over MgSO4, filtered and the filtrate is concentrated in vacuo. The residue is taken up in diethylether, vigorously stirred, and the title compound is obtained after filtration as a white crystalline solid.
c) 5-Bromo-2-bromomethyl-1 ,3-difluoro-benzene:
To a cooled (0°C) solution of 4g (17.6mmol) 4-bromo-2,6-difluorobenzyl alcohol in 50ml THF, 7g (26.4mmol) triphenylphosphine and 8.83g (26.4mmoi) carbon tetrabromide are added with stirring. Stirring is continued for 10min at 00C and for 1h at room temperature. After that the reaction mixture is filtered and the filtrate is concentrated in vacuo. The residue is purified by chromatography (silicagel, hexane) to afford the title compound as yellowish oil.
Example 66: r4-(2-(4-f4-(2-Methoxy-ethyl)-piperazin-1-vn-3-methyl-phenylamino}- benzooxazol-7-yl)-2-methyl-phenvπ-morpholin-4-yl-methanone:
Figure imgf000070_0001
The title compound is prepared from (7-bromo-benzooxazol-2-yl)-{4-[4-(2-methoxy-ethyl)- piperazin-1 -yl]-3-methyl-phenyl}-amine and [2-methyl-4-(4,4,5,5-tetramethyl-[1 ,3,2Jdioxa- borolan-2-yl)-phenyl]-morpholin-4-yl-methanone using methodology described in the preparation of example 65. The title compound is obtained as an off-white foam. Rt = 2.00 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 570 (M+1)+.
Preparation of starting materials:
a) (7-Bromo-benzooxazol-2-yl)-{4-[4-(2-methoxy-ethyl)-piperazin-1-yl]-3-methyl-phenyl}- amine:
The title compound is prepared from 4-[4-(2-methoxy-ethyl)-piperazin-1-yl]-3-methyl- phenylamine and 7-bromo-2-methylsulfanyl-benzooxazole using methodology described in the preparation of example 64.
b) 4-[4-(2-Methoxy-ethyl)-piperazin-1-yl]-3-methyl-phenylamine:
A solution of 0.85g (3.04mmol) 1-(2-methoxy-ethyl)-4-(2-methyl-4-nitro-phenyl)-piperazine in 20ml MeOH:THF = 1 :1 is hydrogenated in the presence of 0.2g 10% Pd/C (Engelhard 4505). The reaction mixture is filtered (2 glass fiber filters used-) and the filtrate is concentrated in vacuo to afford the title compound as an oil.
c) 1 -(2-Methoxy-ethyl)-4-(2-methyl~4-nitro-phenyl)-piperazine:
A solution of 0.95g (6.06mmol) 2-fluoro-5-nitrotoluene and 0.99g (6.67mmol) 1-(2- methoxyethyl)piperazine in 10ml dimethylacetamide is stirred at 1200C for 2Oh. After that the reaction mixture is poured on water and extracted 3x with EtOAc. The combined organic layers are washed with water and saturated NaCI solution, dried over MgSO4, filtered and the filtrate is concentrated in vacuo. The residue is purified by chromatography (silicagel, EtOAc) to afford the title compound as an oil.
d) (4-Bromo-2-methyl-phenyl)-morpholin-4-yl-methanone A solution of 10.6 ml (47mmol) oxalyl chloride in 30 ml dichloromethane is added dropwise to a iced-cooled solution of 5.21 g (23.5mmol) 4-bromo-2-methyl-benzoic acid and 0.0087ml DMF in 100 ml CH2CI2. After complete addition, the cooling bath is removed and stirring maintained for 2h at RT. The solvent is evaporated to dryness and the residue is dried in vacuo and dissolved in 100ml dichloromethane and 8.21ml N-ethyl-diisopropylamine is added. To this solution 2.5ml (28mmol) morpholine is added slowly. Stirring is continued for 0.5h, then the reaction mixture is poured on water and extracted 3x with EtOAc. The combined organic layers are washed with water and saturated NaCI solution, dried over MgSO4, filtered and the filtrate is concentrated in vacuo to afford the title compound which is used in the next step without further purification.
e) f2-Methyl-4-(4,4,5,5-tetramethyl-f1 ,3,2ldioxaborolan-2-yl)-phenyll-morpholin-4-yl- methanone:
A solution (degassed with argon) of 3.Og (10.6mmol) (4-bromo-2-methyl-phenyl)-morpholin- 4-yl-methanone in 15ml dimethylacetamide is added to a solution (degassed with argon) of 3.01 g (11.6mmol) bis-(pinacolato)-diboron and 2.09g (21.1mmol) dried KOAc in 15ml dimethylacetamide. After that 0.261 g (0.317mmol) Pd(dppf)CI2-CH2CI2 is added. The reaction mixture is heated- to 800C and stirred at this temperature for 4h. After that the reaction mixture is cooled to room temperature and poured on water and extracted 3x with EtOAc. The combined organic layers are washed with water and saturated NaCI solution, dried over MgSO4, filtered and the filtrate is concentrated in vacuo and further dried under high vavuum to afford the title compound as a dark brown oil.
Using the reaction conditions described for the preparation of the previously listed examples, especially as described in examples 64, 65 and 66, the following examples can be prepared. The starting materials are either commercially available or can be prepared from commercially available reagents using synthetic methodology as described in "preparation of starting materials" of examples 64, 65 and 66:
Example 67: (4-f7-(3-Fluoro-4-morpholin-4-ylmethyl-phenyl)-benzooxazol-2-ylamino]-2- methyl-phenyl)-pyrrolidin-1-yl-methanone:
Figure imgf000073_0001
R, = 1.96 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 515 (M+1 )+ .
Example 68: 4-f7-(3,5-Difluoro-4-morpholin-4-ylmethvt-phenyl)-benzooxazol-2-ylaminol- 2,N,N-trimethyl-benzamide:
Figure imgf000073_0002
R, = 1.90 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.Oml/min); MS: 507 (M+1)+ .
Example 69: 2.N,N-Trimethyl-4-{7-[4-(morpholine-4-carbonyl)-phenvn-benzooxazol-2- ylamino}-benzamide:
Figure imgf000073_0003
R, = 1.87 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.Oml/min); MS: 485 (M+1)+ .
Example 70: 21N,N-Trimethyl-4-{7-f4-(2-moφholin-4-yl-2-oxo-ethyl)-phenylVbenzooxazol-2- ylamino)-benzamide:
Figure imgf000074_0001
R, = 1.896 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.Oml/min); MS: 499 (M+1 )+ .
Example 71 : 4-[7-(4-Methanesulfonylmethyl-phenyl)-benzooxazol-2-ylaminoT-N,N-dimetrιyl- benzamide:
Figure imgf000074_0002
R, = 1.89 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1. Oml/min); MS: 450 (M+1 )+ .
Example 72: 2-(4-{2-f3-Methyl-4-(4-methyl-piperazine-1-carbonyl)-phenylamino1- benzooxazol-7-yl}-phenγl)-1-morpholin-4-yl-ethanone:
Figure imgf000074_0003
R, = 1.855 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1. Oml/min); MS: 554 (M+1 )+ . Example 73: 2-(2-Fluoro-4-(2-[3-methyl-4-(4-methyl-piperazine-1-carbonyl)-phenylaminol- benzooxazol-7-yl}-phenyl)-1-morpholin-4-yl-ethanone:
Figure imgf000075_0001
R, = 1.697 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 572 (M+1 )+ .
Example 74: 2-(4-{2-f4-(4-Methyl-piperazin-1-yl)-phenylamino1-benzooxazol-7-yl)-phenyl)-1- morpholin-4-yl-ethanone:
Figure imgf000075_0002
R, = 1.65 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 512 (M+1 )\
Example 75: (2-Fluoro-4-{2-f4-(4-methyl-piperazin-1 -yl)-phenylamino1-benzooxazol-7-yl}- phenyl)-morpholin-4-yl-methanone:
Figure imgf000075_0003
R, = 1.90 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 516 (M+1 )+ .
Example 76: 4-(7-f2-Fluoro-4-(morpholine-4-carbonyl)-phenvn-benzooxazol-2-ylaminoV- 2,N,N-trimethyl-benzamide:
Figure imgf000076_0001
R, = 1.892 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 503 (M+1)+ .
Example 77: 4-(7-{4-f2-(1.1-Dioxo-1lambda*6*-thiomorpholin-4-yl)-2-oxo-ethyll-phenyl>- benzooxazol-2-ylamino)-2, N , N-trimethyl-benzamide :
Figure imgf000076_0002
R, = 1.88 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 547 (M+1)+ .
Example 78: 4-{7-[3-Fluoro-4-(morpholine-4-carbonyl)-phenyn-benzooxazol-2-ylamino)- 2, N . N-trimethyl-benzamide :
Figure imgf000077_0001
Rt = 1.935 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 503 (M+1 )+ .
Example 79: 4-(7-f4-(1,1-Dioxo-1lambda*6*-thiomorpholin-4-ylmethyl)-phenyll-benzooxazol- 2-ylamino}-2,N1N-trimethyl-benzamide:
Figure imgf000077_0002
R, = 1.71 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 519 (M+1 )+ .
Example 80: H J-Dioxo-1lambda*6Mhiomorpholin-4-ylH2-fluoro-4-{2-r4-(4-methyl- piperazin-1-yl)-phenylamino^-benzooxazol-7-yl)-phenyl)-methanone:
Figure imgf000077_0003
R, = 1.64 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 564 (M+1 )+ . Example 81 : 4-[7-(4-Methanesulfinylmethyl-phenyl)-benzooxazol-2-ylaminol-2,N.N-trimethyl- benzamide:
Figure imgf000078_0001
S -
R, = 1.79 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 448 (M+1)+ .
Example 82: (2-Methyl-4-{2-f4-(4-methyl-piperazin-1-yl)-phenylaminol-benzooxazol-7-yl}- phenyl)-morpholin-4-yl-methanone:
Figure imgf000078_0002
R, = 1.88 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.Oml/min); MS: 512 (M+1)+ .
Example 83: 4-(7-(4-f2-(1 ,1-Dioxo-1 lambda*6*-thiomorpholin-4-yl)-2-oxo-ethvH-3-fluoro- phenyl)-benzooxazol-2-ylarnino)-2,N,N-trimethyl-benzamide:
Figure imgf000078_0003
R, = 1.924 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 565 (M+1 )\
Example 84: 4-{7-f4-(1 ,1-Dioxo-1lambda*6Mhiomorpholin-4-ylmethyl)-3-fluoro-phenvπ- benzooxazol-2-ylamino>-2.N,N-trimetriyl-benzamide:
Figure imgf000079_0001
R, = 1.786 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 537 (M+1 )+ .
Example 85: 2,N,N-Trimethyl-4-{7-f3-methyl-4-(morpholine-4-carbonyl)-phenvπ-benzooxazol- 2-ylaminol-benzamide:
Figure imgf000079_0002
Rt = 1.914 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 499 (M+1 )+ .
Example 86: 2,N1N-Trimethyl-4-f7-(4-morpholin-4-ylmethyl-3-trifluoromethyl-phenyl)- benzooxazol-2-ylaminol-benzamide:
Figure imgf000080_0001
R, = 1.75 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 539 (M+1 )+ .
Example 87: f4-(4-Ethγl-piperazin-1-yl)-3-methyl-phenvn-f7-(3-fluoro-4-morpholin-4-γlmethyl- phenyl)-benzooxazol-2-vH-amine:
Figure imgf000080_0002
R, = 1.57 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 530 (M+1 )+ .
Example 88: (7-[4-(1,1-Dioxo-1 lambda *6*-thiomorpholin-4-ylmethyl)-3-fluoro-phenyll- benzooxazol-2-ylH4-(4-ethyl-pιperazin-1-yl)-3-methyl-phenyπ-amine:
Figure imgf000080_0003
R, = 1.64 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 578 (M+1 )+ . Example 89: (7-f4-(1 ,1-Dioxo-1 lambda*6Mhiomorpholin-4-ylmethyl)-phenyl}-benzooxazol-2- ylH4-(4-methyl-piperazin-1-yl)-phenγn-amine:
Figure imgf000081_0001
R, = 1.52 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 532 (M+1 )+ .
Example 90: 5-(7-[3-Fluoro-4-(morpholine-4-caronyl)-phenyll-benzooxazol-2-γlamino}-2-(4- methyl-piperazin-1-yl)-benzonitrile:
Figure imgf000081_0002
R, = 1.76 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 1.76 (M+1 )+ .
Example 91 : {2-Fluoro-4-f2-(4-piperazin-1-yl-phenylamino)-benzooxazol-7-ylT-phenyl}- morpholin-4-yl-methanone:
Figure imgf000081_0003
R, = 1.89 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 502 (M+1)+ . Example 92: r4-(4-Ethyl-piperazin-1-yl)-3-methyl-phenylH7-(4-methanesulfonyl-phenyl)- benzooxazol-2-vπ-amine:
Figure imgf000082_0001
Rt = 2.00 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 491 (M+1 )+ .
Example 93: 4-{2-f4-(4-Methyl-piperazin-1-vU-phenylamino1-benzooxazol-7-yl>- benzenesulfonamide:
Figure imgf000082_0002
R1 = 1.81 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 464 (M+1}\
Example 94: (4-{2-f4-(4-Ethyl-piperazin-1 -yl)-3-methyl-phenylaminol-benzooxazol-7-yl}-2- fluoro-phenyl)-morpholin-4-yl-methanone:
Figure imgf000082_0003
R, = 2.00 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 544 (M+1)+ .
Example 95: (7-[4-(1 ,1-Dioxo-1lambda*6*-thiomorpholin-4-ylmethyl)-3,5-difluoro-phenyll- benzooxazol-2-ylH4-(4-methyl-piperazin-1-yl)-phenyll-amine:
Figure imgf000083_0001
R, = 1.50 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 568 (M+1)\
Example 96: (4-{7-f4-(1 ,1-Dioxo-1lambda*6*-thiomorpholin-4-ylmethyl)-3,5-difluoro-phenyll- benzooxazol-2-ylamino)-2-methyl-phenyl)-pyrrolidin-1-yl-methanone:
Figure imgf000083_0002
R, = 2.20 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 581 (M+1 )+ .
Example 97: (4-(2-[4-(4-Cyclopropyl-piperazin-1 -yl)-phenylaminol-benzooxazol-7-yl)-2- methyl-phenyl)-morpholin-4-yl-methanone:
Figure imgf000084_0001
R, = 2.00 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 538 (M+1)+ .
Example 98: f7-(3,5-Difluoro-4-morpholin-4-ylmethyl-phenyl)-benzooxazol-2-ylH4-(4-ethyl- piperazin-1-yl)-3-methyl-phenyll-amine:
Figure imgf000084_0002
Rt = 1.58 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 548 (M+1)+ .
Example 99: {4-f7-(3,5-Difluoro-4-morpholin-4-ylmethyl-phenyl)-benzooxazol-2-ylaminol-2- methyl-phenyl}-pyrrolidin-1-yl-methanone:
Figure imgf000084_0003
R, = 1.96 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 533 (M+1 )+ . Example 100: {4-f4-(2-Dimethylamino-ethγl)-piperazin-1 -vπ-3-methyl-phenylM7-(4- methanesulfonyl-phenyl)-benzooxazol-2-vπ-amine:
Figure imgf000085_0001
R, = 1.86 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 534 (M+1}+ .
Example 101 : {7-f4-(1 ,1-Dioxo-1lambda*6*-thiomorpholin-4-ylmethyl)-3,5-clifluoro-phenyll- benzooxazol-2-ylH4-(4-isopropγl-piperazin-1-yl)-3-methyl-phenvπ-amine:
Figure imgf000085_0002
R, = 2.02 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 610 (M+1 )+ .
Example 102: [4-(2-{4-r4-(2-Dimethylamino-ethyl)-piperazin-1-vn-3-methyl-phenylamino>- benzooxazol-7-yl)-2-methyl-phenyn-morpholin-4-yl-methanone:
Figure imgf000086_0001
R, = 1.86 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 583 (M+1 )+ .
Example 103: 4-[7-(3,5-Difluoro-4-morpholin-4-ylmethγl-phenyl>-benzooxazol-2-ylamino1- N , N-diethyl-2-meth yl-benzamide:
Figure imgf000086_0002
R, = 2.04 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.Oml/min); MS: 535 (M+ 1 }+ .
Example 104: {4-r7-(3,5-Difluoro-4-morpholin-4-ylmethyl-phenyl)-benzooxazol-2-ylarnino1-2- methyl-phenyl)-(4-ethyl-piperazin-1-yl)-methanone:
Figure imgf000086_0003
R, = 1.71 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 576 (M+1 )\
Example 105: f4-(4-lsopropyl-piperazin-1 -yl)-3-methyl-phenylH7-(4-methanesulfonyl- phenyl)-benzooxazol-2-yri-amine:
Figure imgf000087_0001
R, = 2.04 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 505 (M+1 )+ .
Example 106: (4-{2-f4-(4-lsopropyl-piperazin-1 -yl)-3-methyl-phenylaminol-benzooxazol-7-yl>- 2-methyl-phenyl)-morpholin-4-yl-methanone:
Figure imgf000087_0002
R, = 2.01 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 554 (M+1)+ .
Example 107: (4-f4-(2-Dimethylamino-ethyl)-piperazin-1-vπ-3-methyl-phenylH7-f4-(1.1- dioxo-1lambda*6*-thiomorpholin-4-ylmethyl)-3,5-difluoro-phenvπ-benzooxazol-2-yl>-amine:
Figure imgf000088_0001
R, = 1.87 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 639 (M+1 )+ .
Example 108: (7-f4-(1 ,1-Dioxo-1lambda*6Mhiomorpholin-4-ylmethyl)-3.5-difluoro-phenylI- benzooxazol-2-ylH4-f4-(2-methoxy-ethyl)-piperazin-1-vπ-3-methyl-phenyl>-amine:
Figure imgf000088_0002
Rt = 2.01 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 626 (M+1)+ .
Example 109: (4-{2-f4-(4-Ethyl-piperazin-1 -yl)-3-methyl-phenylamino1-benzooxazol-7-yl)-2,6- difluoro-phenyl)-morpholin-4-yl-methanone:
Figure imgf000088_0003
Rt = 2.04 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 562 (M+1)+ .
Example 110: {2-Methyl-4-f7-(3-methyl-4-morpholin-4-ylmethyl-phenyl)-benzooxazol-2- ylamino1-phenyl}-pyrrolidin-1-yl-methanone:
Figure imgf000089_0001
R, = 1.98 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate LOml/min); MS: 511 (M+1)+ .
Example 111 : 2-(4-{2-[4-(4-Ethyl-piperazin-1-yl)-3-methyl-phenylaminol-benzooxazol-7-yl>- phenyl)-2-methyl-1-morpholin-4-yl-propan-1-one:
Figure imgf000089_0002
R, = 2.08 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 568 (M+1)+ .
Example 1 12: 4-(2-f4-(4-Ethyl-piperazin-1-yl)-3-methyl-phenylamino1-benzooxazol-7-yl>- benzenesulfonamide:
Figure imgf000090_0001
R, = 1.92 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to- 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.Oml/min); MS: 492 (M+1 )+ .
Example 113: f4-(4-Cyclopropyl-piperazin-1-yl)-phenylH7-(4-methanesulfonyl-phenyl)- benzooxazol-2-vH-amine:
Figure imgf000090_0002
R, = 1.95 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.Oml/min); MS: 489 (M+1)+ .
Example 114: r4-(4-Cyclopropyl-piperazin-1-ylH>henylH7-f4-(1 ,1-dioxo-1lambda*6*- thiomorpholin-4-ylmethyl)-3,5-difluoro-phenyll-benzooxazol-2-yl}-amine:
Figure imgf000090_0003
R1 = 1.95 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1. Oml/min); MS: 594 (M+1 )+ . Example 115: f7-(4-Methanesulfonγl-phenyl)-benzooxazol-2-yll-{4-[4-(2-methoxy-ethyl)- piperazin-1-yll-3-methyl-phenyl)-amine:
Figure imgf000091_0001
Rt = 2.02 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 521 (M+1 )\
Example 116: (1 ,1-Dioxo-1lambda*6*-thiomorpholin-4-yl)-(4-{2-f4-(4-ethyl-piperazin-1-yl)-3- methyl-phenylamino|-benzooxazol-7-yl)-phenyl)-methanone:
Figure imgf000091_0002
R1 = 1.94 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA1 flow rate 1.Oml/min); MS: 574 (M+1 )+ .
Example 117: 4-(7-f4-(4-Acetyl-pipβrazin-1-ylmethyl)-3,5-difluoro-phenvn-benzooxazol-2- ylamino)-N,N-diethyl-2-methyl-benzamide:
Figure imgf000092_0001
Rt = 2.014 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 576 (M+1)+ .
Example 118: 4-f7-(3,5-Difluoro-4-piperazin-1-γlmethyl-phenyl)-benzooxazol-2-ylaminol-N,N- diethyl-2-methyl-benzamicle:
Figure imgf000092_0002
R, = 1.96 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 534 (M+1 )+ .
Example 118 is prepared from 4-{4-[2-(4-diethylcarbamoyl-3-methyl-phenylamino)- benzooxazol-7-yl]-2,6-difluoro-benzyl}-piperazine-1-carboxylic acid tert-butyl ester as follows:
A mixture of 0.146g (0.224mmol) 4-{4-[2-(4-diethylcarbamoyl-3-methyl-phenylamino)- benzooxazol-7-yl]-2,6-difluoro-benzyl}-piperazine-1-carboxylic acid tert-butyl ester, 2ml trifluoroacetic acid and 10ml dichloromethane is stirred at room temperature for 2h. Then the reaction mixture is poured on water and extracted 3x with EtOAc. The combined organic layers are washed with water and saturated NaCI solution, dried over MgSO4, filtered and the filtrate is concentrated in vacuo. The residue is purified by chromatography (silicagel, 100% EtOAc => EtOAc : MeOH = 1:1 + 1% triethylamine) to afford 0.04g of the title compound as a white solid.
Example 119: (4-{2-f4-(4-Ethyl-piperazin-1-ylmethyl)-3-methyl-phenylamino1-benzooxazol-7- yl)-2-fluoro-phenyl)-morpholin-4-yl-methanone:
Figure imgf000093_0001
Rt = 1.85 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 558 (M+1)+ .
The 1-ethyl-4-(2-methyl-4-nitro-benzyl)-piperazine used for the preparation of Example 119 is prepared as follows:
a) (2-Methyl-4-nitro-phenyl)-methanol:
To a solution of 5.08g (27.2mmol) 2-methyl-4-nitrobenzoic acid in 50ml dry THF, 41ml (41mmol) borane-THF complex (1M solution in THF) is added drop-wise at 00C. After completion of the borane addition, the reaction mixture is stirred at room temperature for 2Oh. After that a K2CO3 solution (1.33g in 49ml water) is slowly added under stirring. Then the reaction mixture is poured on water and extracted 3x with EtOAc. The combined organic layers are washed with water and saturated NaCI solution, dried over MgSO4, filtered and the filtrate is concentrated in vacuo. The residue is taken up in diethylether, vigorously stirred, and the title compound is obtained after filtration as a yellow crystalline solid.
b) 1 -Bromomethyl-2-methyl-4-nitro-benzene:
To a solution of 4.55g (27.2mmol) (2-methyl-4-nitro-phenyl)-methanol 10.8g (40.8mmol) triphenylphosphine and 13.7g (40.8mmol) carbon tetrabromide is added at 0°C. The reaction mixture is stirred at room temperature for 1h. After that the reaction mixture is filtered and the filtrate is concentrated in vacue. The residue is purified by chromatography (silicagel, 100% hexane => 100% EtOAc) to afford the title compound as an oil.
c) 1 -Ethyl-4-(2-methyl-4-nitro-benzyl)-piperazine:
A solution of 1g (3.78mmol) 1-bromomethyl-2-methyJ-4-nitro-benzene, 0.539ml (4.16mmol) 1 -ethylpiperazine and 0.63ml (4.54mmol) triethylamine in 15ml dichloromethane is stirred at room temperature for 0.5h. Then the reaction mixture is poured on water and extracted 3x with EtOAc. The combined organic layers are washed with water and saturated NaCI solution, dried over MgSO4, filtered and the filtrate is concentrated in vacuo. The residue is purified by chromatography (silicagel, 100% EtOAc => EtOAc : MeOH = 7:3) to afford the title compound as a solid.
Example 120: 4-f7-(3,5-Difluoro-4-morpholin-4-ylmethyl-phenyl)-benzooxazol-2-ylaminol- 2,N-dimethyl-benzamide:
Figure imgf000094_0001
R, = 1.83 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 493 (M+1)+ .
Example 121 : f7-(3,5-Difluoro-4-morpholin-4-ylmethyl-phenyl)-benzooxazol-2-ylH3-methyl-4- r4-(3,3,3-trifluoro-propyl)-piperazin-1-vπ-phenyl}-amine:
Figure imgf000095_0001
R, = 1.90 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 616 (M+1)+ .
Example 122: 4-[7-(3,5-Difluoro-4-morpholin-4-ylmethyl-phenyl)-benzooxazol-2-ylamino]-N- (2-dimethylamino-ethyl)-2-methyl-benzamide:
Figure imgf000095_0002
R, = 1.71 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 550 (M+1 )\
Example 123: 4-(7-(4-f2-(1 J-Dioxo-1lambda*6Mhiomorpholin-4-yl)-2-oxo-ethyl^-pr)enyl)- benzooxazol-2-ylamino).-N,N-diethyl-2-methoxy-benzarnide:
Figure imgf000095_0003
R, = 2.02 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 591 (M+1 )+ .
Example 124: 2-{4-f2-(4-Methanesulfonγl-phenylamino)-benzooxazol-7-vπ-phenyl}-1 - morpholin-4-yl-ethanone:
Figure imgf000096_0001
R, = 1.97 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.Oml/min); MS: 492 (M+1)* .
Example 125: 1-(4-(7-f4-(1 ,1-Dioxo-1lambda*6*-thiomorpholin-4-ylmethyl)-3,5-difluoro- phenvn-benzooxazol-2-ylamino>-phenyl)-pyrrolidin-2-one:
Figure imgf000096_0002
R, = 2.144 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1. Oml/min); MS: 553 (M+1 )+ .
Example 126: 1-{4-[7-(3,5-Difluoro-4-morpholin-4-ylmethyl-phenyl)-benzooxazol-2-ylamino1- phenyl)-pyrrolidin-2-one:
Figure imgf000097_0001
Rt = 1.93 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.Oml/min); MS: 505 (M+1)+ .
Example 127: (4-(2-f4-Methoxy-3-(4-methyl-piperazin-1 -yl)-phenylamino1-benzooxazol-7-yl)- 2-methyl-phenyl)-morpholin-4-yl-methanone:
Figure imgf000097_0002
R, = 1.92 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1. Oml/min); MS: 542 (M+1 )+ .
Example 128: (7-f4-(1 ,1-Dioxo-1lambda*6*-thiomorpholin-4-ylmethyl)-3,5-difluoro-phenylV benzooxazol-2-yl}-[3-(4-methyl-piperazin-1-yl)-phenvπ-amine:
N N -
Figure imgf000097_0003
R, = 1.94 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 568 (M+1 )+ .
Example 129: (2-Methyl-4-{2-f3-(4-methyl-piperazin-1-yl)-phenylaminol-benzooxazol-7-yl}- phenyl)-morpholin-4-yl-methanone:
Figure imgf000098_0001
R, = 1.94 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 512 (M+1 )+ .
Example 130: (7-f4-(1 ,1-Dioxo-1lambda*6*-thiomorpholin-4-ylmethyl)-3,5-difluoro-phenyn- benzooxazol-2-ylM4-methoxy-3-(4-methyl-piperazin-1-yl)-phenyll-amine:
Figure imgf000098_0002
R, = 1.92 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 598 (M+1 )+ .
Example 131 : (2-Fluoro-4-f2-(3-methyl-4-morpholin-4-ylmethyl-phenylamino)-benzooxazol-7- yll-phenyl)-morpholin-4-yl-methanone:
Figure imgf000099_0001
R, = 1.95 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.Oml/min); MS: 531 (M+1 )+ .
The 4-(2-methyl-4-nitro-benzyl)-morpholine used in the preparation of example 131 is prepared as described in example 119 by using morpholine instead of 1-ethyl-piperazine.
Example 132: N-{5-f7-(3-Fluoro-4-morpholin-4-ylmethyl-phenyl)-benzooxazol-2-ylaminol- indan-2-yl)-acetamide:
Figure imgf000099_0002
R, = 1.64 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1. Oml/min); MS: 501 (M+1 )+ .
Example 133: N-(5-(7-f4-(1.1-Dioxo-1lambda*6*-thiomorpholin-4-ylmethyl)-phenyll- benzooxazol-2-ylamino)-indan-2-yl)-acetamide:
Figure imgf000099_0003
R, = 1.67 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 530 (M+1 )+ .
Example 134: 5-f7-(3,5-Difluoro-4-morpholin-4-ylmethyl-phenyl)-benzooxazol-2-ylaminol-1 ,3- dihydro-indol-2-one:
Figure imgf000100_0001
R, = 1.78 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 477 (M+1)+ .
Example 135: 2-{4-f2-(4-Methanesulfinγl-phenylamino)-benzooxazol-7-yl]-phenyl)-1 - morpholin-4-yl-ethanone:
Figure imgf000100_0002
R, = 1.82 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 476 (M+1 )+ .
4-Methanesulfinyl-phenylamine that is needed for the preparation of example 135 can be prepared as described by C. Almansa et al. in Journal of Medicinal Chemistry (2003), 46(16), 3463-3475. Example 136: r7-(4-lmidazol-1-ylmethyl-phenyl)-benzooxazol-2-ylH3A5-trimethoxy-phenyl)- amine:
Figure imgf000101_0001
R, = 1.93 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.Oml/min); MS: 457 (M+1)+ .
Example 137: (7-[4-(4-Methyl-piperazin-1 -ylmethyl)-phenvπ-benzooxazol-2-ylH3,4,5- trimethoxy-phenyl famine :
Figure imgf000101_0002
R, = 1.80 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1. Oml/min); MS: 489 (M+1)+ .
Example 138: [7-(4-Morpholin-4-ylmethyl-phenyl)-benzooxazol-2-yll-(3,4,5-trimethoxy- phenyQ-amine:
Figure imgf000101_0003
R, = 1.904 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM1 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 476 (M+1 )+ .
Example 139: 4-f2-(4-Methoxy-phenylamino)-benzooxazol-7-yπ-N-methyl- benzenesulfonamide:
Figure imgf000102_0001
R, = 2.28 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 410 (M+1)+ .
Example 140: 1 -Morpholin-4-yl-2-(4-f2-(3,4.5-trimethoxy-phenylamino>-benzooxazol-7-vn- phenyD-ethanone:
Figure imgf000102_0002
Rt = 2.186 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 504 (M+1)+ .
Example 141 : Morpholin-4-yl-(4-f2-(3,4,5-trimethoxy-phenylamino)-benzooxazol-7-yll- phenyll-methanone:
Figure imgf000103_0001
R, = 2.176 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 490 (M+1 )\
Example 142: (4-Methyl-piperazin-1-ylH4-f2-(3A5-trimethoxy-phenylamino)-benzooxazol-7- vπ-phenyl)-methanone:
Figure imgf000103_0002
R1 = 1.87 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.Oml/min); MS: 503 (M+1 )+ .
Example 143: (4-f2-(3,4,5-Trimethoxy-phenylamino)-benzooxazol-7-yll-phenyl)-methanol:
Figure imgf000103_0003
R, = 2.16 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 407 (M+1 )+ .
Example 144: 4-f2-(3-Methoxy-4-methyl-phenylamino)-benzooxazol-7-vπ-N-methyl- benzenesulfonamide:
Figure imgf000104_0001
R, = 2.44 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 424 (M+1 )\
Example 145: N-(2-Methoxy-ethyl)-4-{7-f4-(morpholine-4-carbonyl)-phenyll-benzooxazol-2- ylaminoj-benzamide:
Figure imgf000104_0002
R, = 2.035 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 501 (M+1 )+ .
Example 146: N,N-Dimethyl-4-f7-(4-sulfamoyl-phenyl)-benzooxazol-2-ylaminol-benzamide:
Figure imgf000104_0003
R, = 2.02 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 437 (M+1 )+ .
Example 147: N-(2-Methoxy-ethyl)-4-{7-[4-(2-morpholin-4-yl-2-oxo-ethyl)-phenyri- benzooxazol-2-ylamino)-benzamide:
Figure imgf000105_0001
Rt = 2.04 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 515 (M+1)+ .
Example 148: N-(3-(7-r4-(Morpholine-4-carbonyl)-phenyn-benzooxazol-2-ylamino)-phenyl)- methanesulfonamide:
Figure imgf000105_0002
R, = 2.07 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 493 (M+1 )+ .
Example 149: 2-Methoxy-4-{7-f4-(morpholine-4-carbonyl)-phenyll-benzooxazol-2-ylamino)- benzoic acid methyl ester:
Figure imgf000106_0001
R, = 2.04 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 488 (M+1 )+ .
Example 150: 2-Methoxy-N,N-dimethyl-4-(7-[4-(morpholine-4-carbonyl)-phenyll- benzooxazol-2-ylamino)-benzamide:
Figure imgf000106_0002
R, = 1.87 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 501 (M+1 )+ .
Example 151 : 2-Methyl-2-(4-{2-r4-(4-methyl-piperazin-1-yl)-phenylamino1-benzooxazol-7-yl)- phenvD-1 -morpholin-4-γl-propan-1 -one:
Figure imgf000106_0003
R, = 2.06 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 540 (M+1 )+ . Example 152: f4-(3,5-Difluoro-4-morpholin-4-ylmethyl-phenyl)-oxazolof5,4-c1pyridin-2-yll-f4- (4-ethyl-piperazin-1-yl)-3-methγl-phenvH-amine:
Figure imgf000107_0001
Rt = 1.58 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 549 (M+1)+ .
Example 153: (4-f4-(1 l1-Dioxo-Hambda*6*-thiomorpholin-4-ylmethylV3,5-difluoro-phenyll- oxazolof5,4-c1pyridin-2-yl)-f4-(4-ethyl-piperazin-1-yl)-3-methyl-prienyπ-amine:
Figure imgf000107_0002
R, = 1.71 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 597 (M+1 )+ .
Example 154: f4-(4-Cyclopropyl-piperazin-1-yl)-3-methyl-phenylH7-[4-(1 ,1-dioxo- 1 lambda*6*-thiomorpholin-4-ylmethyl)-3,5-difluoro-phenvn-benzooxazol-2-yl}-amine:
Figure imgf000107_0003
R, = 2.02 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 608 (M+1 )+ .
Example 155: (4-(2-f4-(4-Cvclopropyl-piperazin-1 -yl)-3-methyl-phenylaminol-benzooxazol-7- yl)-2-methyl-phenyl)-morpholin-4-yl-methanone:
Figure imgf000108_0001
R, = 2.01 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.Oml/min); MS: 552 (M+1)+ .
Example 156: (4-{2-f4-(4-Cyclopropyl-piperazin-1 -yl)-3-methyl-phenylamino1-benzooxazol-7- Vl>-2-fluoro-phenyl)-morpholin-4-yl-methanone:
Figure imgf000108_0002
R, = 2.01 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1. Oml/min); MS: 556 (M+1 )+ .
Example 157: 2-(4-{2-f4-(4-Cyclopropyl-piperazin-1 -yl)-3-methyl-phenylamino1-benzooxazol- 7-yl}-phenyl)-2-methyl-1 -morpholin-4-yl-propan-1 -one:
Figure imgf000109_0001
Rt = 2.11 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.Oml/min); MS: 580 (M+1 )+ .
Example 158: 4-f7-(3.5-Difluoro-4-morpholin-4-ylmethyl-phenyl)-benzooxazol-2-ylaminol-N- (2-dimethylamino-ethyl)-2,N-dimethyl-benzamide:
Figure imgf000109_0002
R1 = 1.72 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1. Oml/min); MS: 564 (M+1 )+ .
Example 159: N-(2-Dimethylamino-ethyl)-4-{7-f4-( 1 , 1 -dioxo- 1 lambda*6*-thiomorpholin-4- ylmethyl)-3,5-difluoro-phenvπ-benzooxazol-2-ylamino}-2,N-dimethyl-benzamide:
Figure imgf000109_0003
R, = 1.91 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 612 (M+1 )\
Example 160: f4-(4-Cvclopropyl-piperazin-1 -yl)-3-methyl-phenyl1-f7-(3.5-difluoro-4- morpholin-4-ylmethyl-phenyl)-benzooxazol-2-vn-amine:
Figure imgf000110_0001
R, = 1.82 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 560 (M+1 )+ .
Example 161 : 5-|7-(3,5-Diflυoro-4-morpholin-4-ylmethyl-phenyl)-benzooxazol-2-ylamino1-1 ,3- dimethyl-1 H-pyridin-2-one:
Figure imgf000110_0002
R, = 1.72 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 467 (M+1 )+ .
The 5-amino-1 ,3-dimethyl-1 H-pyridin-2-one needed for the preparation of the title compound is as follows:
a) 5-Amino-1 ,3-dimethyl-1 H-pyridin-2-one:
A solution of 1.0g (5.77mmol) 1 ,3-dimethyl-5-nitro-1 H-pyridin-2-one in 40ml MeOHTHF = 1 :1 is hydrogenated in the presence of 0.18g 10% Pd/C (Engelhard 4505). The reaction mixture is filtered (2 glass fiber filters used) and the filtrate is concentrated in vacuo to afford the crude title compound as an oil.
b) 1 ,3-Dimethyl-5-nitro-1 H-pyridin-2-one:
A mixture of 1g (6.49mmol) 1-hydroxy-3-methyl-5-nitropyridine, 0.197g (7.8mmol) NaH and 0.61ml (9.73mmol) MeI in 20ml DMF is stirred at room temperature for 2Oh. Then the reaction mixture is poured on water and extracted 3x with EtOAc. The combined organic layers are washed with water and saturated NaCI solution, dried over MgSO4, filtered and the filtrate is concentrated in vacuo to afford the title compound as off-white crystals.
Example 162: (4-{2-f4-(4-Cvclopropyl-piperazin-1 -yl)-3-methyl-phenylamino1-benzooxazol-7- yl)-2-methyl-phenylH1.1-dioxo-1lambda*6*-thiomorpholin-4-yl)-methanone:
Figure imgf000111_0001
R, = 2.03 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA1 flow rate 1.0ml/min); MS: 600 (M+1)\
Example 163: f7-(3,5-Difluoro-4-morpholin-4-ylmethyl-phenyl)-benzooxazol-2-vn-(3-methyl-4- morpholin-4-yl-phenyl)-amine:
Figure imgf000111_0002
R, = 1.92 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM1 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 521 (M+1)+ .
Example 164: (7-f4-(1 ,1-Dioxo-1lambda*6*-thiomorpholin-4-γlmethyl)-3,5-difluoro-phenyll- benzooxazol-2-ylH3-methγl-4-morpholin-4-yl-phenyl)-amine:
Figure imgf000112_0001
R, = 2.02 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.Oml/min); MS: 569 (M+1>+ .
Example 165: (1 J-Dioxo-1lambda*6Mhiomorpholin-4-ylH4-f2-(3-methyl-4-morpholin-4- ylmethyl-phenylamino)-benzc>oxazol-7-vπ-phenyl}-methanone:
Figure imgf000112_0002
R, = 1.89 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1. Oml/min); MS: 561 (M+1 )+ .
Example 166: (2-Methyl-4-f2-(3-methyl-4-morpholin-4-ylmethyl-phenylamino)-benzooxazol-7- vn-phenyl)-morpholin-4-yt-methanone:
Figure imgf000113_0001
R, = 1.92 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 527 (M+1 )+ .
Example 167: 2-Methyl-2-(4-f2-(3-methyl-4-morpholin-4-ylmethyl-phenylamino)-benzooxazol- 7-vπ-phenyl}-1-morpholin-4-yl-propan-1-one:
Figure imgf000113_0002
R, = 2.06 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 555 (M+1)+ .
Example 168: 4-{4-f4-(1 ,1-Dioxo-1 lambda*6Mhiomorpholin-4-ylmethyl)-3,5-difluoro-phenyl1- oxazolof5,4-clpyridin-2-ylamino}-2.N,N-trimethyl-benzamide:
Figure imgf000113_0003
R, = 1.80 min (Waters Symmetry C8, 2.1x50mm, detection 210-25OnM, 5% to 100% CH3CN in H2O in 2min + 0.05% TFA, flow rate 1.0ml/min); MS: 556 (M+1)+ .
Example 169: 4-(7-r3,5-Difluoro-4-(3-oxo-piperazin-1 -ylmethyl)-phenvπ-benzooxazol-2- ylamino)-2,N,N-trirnethyl-benzamide:
Figure imgf000114_0001
Example 170: 4-{2,6-Difluoro-4-f2-(3-methyl-4-morpholin-4-yl-phenylamino)-benzooxazol-7- ylT-benzyl}-piperazin-2-one:
Figure imgf000114_0002
Example 171 : 4-(4-(2-f4-(4-Cyclopropyl-piperazin-1-yl)-3-methyl-phenylaminol-benzooxazol- 7-yl)-2,6-difluoro-benzyl)-piperazin-2-one:
Figure imgf000114_0003
Example 172: 4-(7-r3,5-Difluoro-4-(4-methyl-3-oxo-piperazin-1 -ylmethyQ-phenyH- benzooxazol-2-ylamino>-2,N,N-trinnethyl-benzamide:
Figure imgf000115_0001
Example 173: 4-{2,6-Difluoro-4-f2-(3-methyl-4-morpholin-4-yl-phenylamino)-benzooxazol-7- yl]-benzyl)-1-methyl-piperazin-2-one:
Figure imgf000115_0002
Example 174: 4-(4-{2-f4-(4-Cvclopropyl-piperazin-1-yl)-3-methyl-phenylaminol-benzooxazol- 7-yl}-2,6-difluoro-benzyl)-1-methyl-piperazin-2-one:
Figure imgf000115_0003
Example 175: 4-f7-(4-Methanesulfinylmethyl-3-methyl-phenyl)-benzooxazol-2-ylaminol- 2,N.N-trimethyl-benzamide:
Figure imgf000116_0001
Example 176: f4-(4-Cvclopropyl-piperazin-1-yl)-3-methyl-phenvπ-f7-(4- methanesulfonylmethyl-3-methyl-phenyl)-benzooxazol-2-vH-amine:
Figure imgf000116_0002
Example 177: r7-(4-Methanesulfinylmethyl-3-methyl-phenyl)-benzooxazol-2-yll-(4-methyl-3- morpholin-4"-yl-phenyl)-amine:
Figure imgf000116_0003
Example 178: (4-[7-(3,5-Difluoro-4-morpholin-4-ylmethyl-phenyl)-benzooxazol-2-ylamino1-2- methyl-phenyl)-morpholin-4-yl-methanone:
Figure imgf000116_0004
Example 179: 4-[7-(3,5-Difluoro-4-morpholin-4-ylmethγl-phenyl)-benzooxazol-2-ylaminol-N- (2-methoxy-ethyl)-2-methγl-benzamide:
Figure imgf000117_0001
Example 180: 4-f7-(315-Difluoro-4-morpholin-4-ylmethyl-phenyl)-benzooxazol-2-ylaminol-N- (3-dimethylamino-propyl)-2-methyl-benzamide:
Figure imgf000117_0002
Example 181 : f7-(3,5-Difluoro-4-morpholin-4-ylmethyl-phenyl)-benzooxazol-2-vπ-(6-methoxy- 5-methyl-pyridin-3-yl)-amine:
Figure imgf000117_0003
Example 182: 4-f7-(3,5-Difluoro-4-morpholin-4-ylmethyl-phenyl)-benzooxazol-2-ylamino|- 2,6,N,N-tetramethyl-benzamide:
Figure imgf000118_0001
Example 183: 4-f7-(3,5-Difluoro-4-morphohn-4-ylmethyl-phenyl)-benzooxazol-2-ylamino]-2- ethyl-N.N-dimethyl-benzamide:
Figure imgf000118_0002
Example 184: 4-f7-(3,5-Difluoro-4-morpholin-4-ylmethyl-phenyl)-benzooxazol-2-ylamino1-N- (2-methoxy-ethyl)-2,N-dimethyl-benzamide:
Figure imgf000118_0003
Example 185: 4-f7-(3,5-Difluoro-4-morpholin-4-ylnnethyl-phenyl)-benzooxazol-2-ylaminol-2- morpholin-4-yl-benzonitrile:
Figure imgf000118_0004
Example 186: Soft capsules
5000 soft gelatin capsules, each comprising as active ingredient 0.05 g of one of the compounds of formula I mentioned in the preceding Examples, are prepared as follows:
Composition
Active ingredient 250 g
Lauroglycol 2 litres
Preparation process: The pulverized active ingredient is suspended in Lauroglykol® (propylene glycol laurate, Gattefosse S.A., Saint Priest, France) and ground in a wet pulverizer to produce a particle size of about 1 to 3 μm. 0.419 g portions of the mixture are then introduced into soft gelatin capsules using a capsule-filling machine.
Example 187: EPK JAK/TYK-kinase family profiling assays
The efficacy of the compounds of the invention as inhibitors of JAK/TYK kinase activity can be demonstrated as follows:
All four kinases of the JAK/TYK-kinase family are used as purified recombinant GST-fusion proteins, containing the active kinase domains. GST-JAK1 (866- 1154), GST-JAK3(811- 1124), and GST-TYK2(888-1187) are expressed and purified by affinity chromatography. GST-JAK2(808-1132) is purchased from Invitrogen (Carlsbad, USA, #4288).
The kinase assays are based on the Caliper mobility shift assay using the LabChip 3000 systems. This technology is similar to capillary electrophoresis and uses charge driven separation of substrate and product in a microfluidic chip.
All kinase reactions are performed in 384 well microtiter plates in a total reaction volume of 18 μl. The assay plates are prepared with 0.1 μl per well of test compound in the appropriate test concentration, as described under the section "preparation of compound dilutions". The reactions are started by combining 9 μl of substrate mix (consisting of peptide and ATP) with 9 μl of kinase dilution. The reactions are incubated for 60 minutes at 300C and stopped by adding 70 μl of stop buffer (100 mM Hepes, 5% DMSO, 0.1% Coating reagent, 10 mM EDTA, O.αi5% Brij 35).
Fluorescently labeled synthetic peptides are used as substrates in all reactions. A peptide derived from the sequence of IRS-1 (IRS-1 peptide, FITC-AhX-KKSRGDYMTMQIG-NH2 (SEQ ID NO: 1 ); see J. Biol. Chem. 268(33), 25146-51 (1993)) is used for JAK1 and TYK2 and a peptide named JAK3tide (FITC-GGEEEEYFELVKKKK-NH2 (SEQ ID NO: 2); Upstate (Millipore), Temecula, California, USA)) for JAK2 and JAK3. Specific assay conditions are described in Table 1 : GGEEEYFELVKKKK
Table 1: Assay conditions of individual kinase assays
Kinase JAK1 JAK2 JAK3 TΫK2
Buffer 50 mM Hepes 50 mM Hepes 50 mM Hepes 50 mM Hepes pH 7.5, pH 7.5, pH 7.5, pH 7.5,
0.02% Tween 0.02% Tween 0.02% Tween 0.02% Tween 20, 1 mM DTT, 20, 1 mM DTT, 20, 1 mM DTT, 20, 1 mM DTT, 0.02% BSA, 0.02% BSA, 0.02% BSA, 0.02% BSA,
12 mM MgCI2 9 mM MgCI2 1.S mM MgCI2 9 mM MgCl2
DMSO 0.6 % 0.6 % 0.6 % 0.6 %
Kinase cone. 50 nM 1.8 nM 6 nM 40 nM
Substrate peptide 5 μM 2 μM 2 μM 5 μM cone. ATP cone. 40 μM 20 μM 80 μM 30 μM
The terminated reactions are transferred to a Caliper LabChip 3000 reader (Caliper Life Siciences, Mountain View, California, USA) and the turnover of each reaction is measured by determining the substrate/product ratio.
Preparation of compound dilutions
Test compounds are dissolved in DMSO (10 mM) and transferred into 1.4mL flat bottom or V-shaped Matrix tubes carrying a unique 2D matrix chip by individual compound hubs. The numbers of these chips are distinctively linked to the individual compound identification numbers. The stock solutions are stored at -200C if not used immediately. For the test procedure the vials are defrosted and identified by a scanner whereby a working sheet is generated that guides the subsequent working steps.
Compound dilutions are made in 96 well plates. This format enabled the assay of maximally 40 individual test compounds at 8 concentrations (single points) including 4 reference compounds. The dilution protocol includes the production of pre-dilution plates, master plates and assay plates:
Pre-dilution plates: 96 polypropylene well plates are used as pre-dilution plates. A total of 4 pre-difution plates are prepared including 10 test compounds each on the plate positions A1- A10, one standard compound at A11 and one DMSO control at A12. All dilution steps are done on a Hamilton STAR robot (Hamilton, Co.., Reno, NV, USA).
Master plates: 100μl_ of individual compound dilutions including standard compound and controls of the 4 "pre-dilution plates" are transferred into a 384 "master plate" including the following concentrations 1'820, 564, 182, 54.6, 18.2, 5.46, 1.82 and 0.546μM, respectively in 90 % of DMSO.
Assay plates: Identical assay plates are then prepared by pipetting 100 nl_ each of compound dilutions of the master plates into 384-well "assay plates". In the following the compounds are mixed with 9μl_ of assays components plus 9μL enzyme corresponding to a 1 :181 dilution steps enabling the final concentration of 10, 3.0, 1.0, 0.3, 0.1 , 0.03, 0.01 and 0.003μM, respectively. The preparation of the master plates are handled by the Matrix PlateMate Plus robot (Thermo Fisher Scientific, Handforth, Cheshire, United Kingdom) and replication of assay plates by the HummingBird robot (Genomic Solutions, Inc., Ann Arbor, Michigan, USA).
On the basis of these studies, a compound of the invention shows therapeutic efficacy especially against disorders dependent on protein kinase, especially proliferative diseases mediated by JAK/TYK kinase activity.
Exemplified compounds show JAK2 enzyme inhibitory activities with IC50-values of 0.1 to 1000 nM as shown in the following table:
Figure imgf000122_0001
Figure imgf000123_0001
Figure imgf000124_0001
Figure imgf000125_0001
Figure imgf000126_0001
Figure imgf000127_0001
Figure imgf000128_0001
Figure imgf000129_0001
Figure imgf000130_0001

Claims

Claims:
1. A compound of the formula I,
Figure imgf000131_0001
wherein
X is N or CR5, wherein R5 is halo, cyano, hydroxyl, d-C7-alkoxy, d-Cy-alkyl, amino, N- mono- or N1N-Oi-C1-C7-BlRyI or preferably hydrogen;
R1 is unsubstituted or substituted aryl or is unsubstituted or substituted heterocyclyl; and R2 is unsubstituted or substituted aryl or is unsubstituted or substituted heterocyclyl which is bound via a ring carbon atom;
R3 is cyano, hydroxyl, d-C7-alkyl, amino, N-mono- or N,N-di-CrC7-alkyl or preferably hydrogen; and
R4 is hydroxyl, amino or preferably hydrogen; or salts thereof.
2. A compound of the formula I according to claim 1 , where, if mentioned, unsubstituted or substituted aryl is an aromatic moiety with 6 to 14 ring carbon atoms, more preferably with 6 to 10 ring carbon atoms, such as phenyl or naphthyl, which is unsubstituted or substituted by one or more, preferably up to three, more preferably up to two substituents independently selected from the group consisting of unsubstituted or substituted heterocyclyl as described below, especially pyrrolidinyl, such as pyrrolidino, oxopyrrolidinyl, such as oxo- pyrrolidino, d-Cr-alkyl-pyrrolidinyl, 2,5-di-(C1-C7alkyl)pyrrolidinyl, such as 2,5-di-(d-C7alkyl)- pyrrolidino, tetrahydrofuranyl, thiophenyl, d-C7-alkylpyrazolidinyl, pyridinyl, d-C7-alkyl- piperidinyl, piperidino, piperidino substituted by amino or N-mono- or N,N-di-[lower alkyl, phenyl, d-C7-alkanoyl and/or phenyl-lower alkyl)-amino, unsubstituted or N-lower alkyl substituted piperidinyl bound via a ring carbon atom, piperazino, lower alkylpiperazino, mor- pholino, thiomorpholino, S-oxo-thiomorpholino or S,S-dioxothiomorpholino; d-C7-alkyf, amino-d-CT-alkyl, N-d-d-alkanoylamino-d-CT-alkyl, N-d-d-alkanesulfonyl-amino-d-C;-- alkyl, carbamoyl-d-d-alkyl, [N-mono- or N,N-di-(d-C7-alkyl)-carbamoyl]-CrC7-alkyl, C1-C7- alkanesulfinyl-d-C7-alkyl, d-CTr-alkanesulfonyl-d-d-alkyl, phenyl, naphthyl, mono- to tri- [Ci-C7-alkyl, halo and/or cyano]-phenyl or mono- to tri-[Ci-C7-alkyl, halo and/or cyano]- naphthyl; C3-C8-cycloalkyl, mono- to tri-[d-C7-alkyl and/or hydroxy]-C3-C8-cycloalkyl; halo, hydroxy, lower alkoxy, lower-alkoxy-lower alkoxy, (lower-alkoxy)-lower alkoxy-lower alkoxy, halo-d-C7-alkoxy, phenoxy, naphthyloxy, phenyl- or naphthyl-lower alkoxy; amino-d-C6- alkoxy, lower-alkanoyloxy, benzoyloxy, naphthoyloxy, formyl (CHO), amino, N-mono- or N1N- di-(Ci-d-alkyl)-amino, d-C7-alkanoylamino, CrCralkanesulfonylamino, carboxy, lower alkoxy carbonyl, e.g.; phenyl- or naphthyl-lower alkoxycarbonyl, such as benzyloxycarbonyl; d-C7-alkanoyl, such as acetyl, benzoyl, naphthoyl, carbamoyl, N-mono- or N,N-disubstituted carbamoyl, such as N-mono- or N,N-di-substituted carbamoyl wherein the substitutents are selected from lower alkyl, (lower-alkoxy)-lower alkyl and hydroxy-lower alkyl; amidino, guani- dino, ureido, mercapto, lower alkylthio, phenyl- or naphthylthio, phenyl- or naphthyl-lower alkylthio, lower alkyl-phenylthio, lower alkyl-naphthylthio, halogen-lower alkylmercapto, sulfo, lower alkanesulfonyl, phenyl- or naphthyl-sulfonyl, phenyl- or naphthyl-lower alkylsulfonyl, alkylphenylsulfonyl, halogen-lower alkylsulfonyl, such as trifluoromethanesulfonyl; sulfon- amido, benzosulfonamido, azido, azido-CrC7-alkyl, especially azidomethyl, d-C^alkanesul- fonyl, sulfamoyl, N-mono- or N,N-di-(C1-C7-alkyl)-sulfamoyl, morpholinosulfonyl, thiomorpho- linosulfonyl, cyano and nitro; where each phenyl or naphthyl (also in phenoxy or naphthoxy) mentioned above as substituent or part of a substituent of substituted alkyl (or also of substituted aryl, heterocyclyl etc. mentioned herein) is itself unsubstituted or substituted by one or more, e.g. up to three, preferably 1 or 2, substituents independently selected from halo, especially fluoro, chloro, bromo or iodo, halo-lower alkyl, such as trifluoromethyl, hydroxy, lower alkoxy, azido, amino, N-mono- or N,N-di-(lower alkyl and/or d-C7-alkanoyl)-amino, nitro, carboxy, lower-alkoxycarbonyl carbamoyl, cyano and/or sulfamoyl;
unsubstituted or substituted heterocyclyl is a heterocyclic radical that is unsaturated (= carrying the highest possible number of conjugated double bonds in the ring(s)), saturated or partially saturated and is preferably a monocyclic or in a broader aspect of the invention bicyclic or tricyclic ring; and has 3 to 24, more preferably 4 to 16, most preferably 4 to 10 and most preferably 6 ring atoms; wherein one or more, preferably one to four, especially one or two carbon ring atoms are replaced by a heteroatom selected from the group- consisting of nitrogen, oxygen and sulfur, the bonding ring preferably having 4 to 12, especially 5 to 7 ring atoms; which heterocyclic radical (heterocyclyl) is unsubstituted or substituted by one or more, especially 1 to 3, substituents independently selected from the group consisting of the substituents defined above for substituted alkyl; and where heterocyclyl is especially a heterocyclyl radical selected from the group consisting of oxiranyl, azirinyl, aziridinyl, 1 ,2- oxathiolanyl, thienyl (= thiophenyl), furanyl, tetrahydrofuryl, pyranyl, thiopyranyl, thianthrenyl, isobenzofuranyl, benzofuranyl, chromenyl, 2H-pyrrolyl, pyrrolyl, pyrrolinyl, pyrrolidinyl, imidazolyl, imidazolidinyl, benzimidazolyl, pyrazolyl, pyrazinyl, pyrazolidinyl, thiazolyl, isothiazolyl, dithiazolyl, oxazolyl, isoxazolyl, pyridyl, pyrazinyl, pyrimidinyl, piperidinyl, piperazinyl, pyridazinyl, morpholinyl, thiomorpholinyl, (S-oxo or S,S-dioxo)-thiomorpholinyl, indolizinyl, azepanyl, diazepanyl, especially 1 ,4-diazepanyl, isoindolyl, 3H-indolyl, indolyl, benzimidazolyl, cumaryl, indazolyl, triazolyl, tetrazolyl, purinyl, 4H-quinolizinyl, isoquinolyl, quinolyl, tetrahydroquinolyl, tetrahydroisoquinolyl, decahydroquinolyl, octahydroisoquinolyl, benzofuranyl, dibenzofuranyl, benzothiophenyl, dibenzothiophenyl, phthalazinyl, naphthyri- dinyl, quinoxalyl, quinazolinyl, quinazolinyl, cinnolinyl, pteridinyl, carbazolyl, beta-carbolinyl, phenanthridinyl, acridinyl, perimidinyl, phenanthrolinyl, furazanyl, phenazinyl, phenothiazinyl, phenoxazinyl, chromenyl, isochromanyl, chromanyl, benzo[1 ,3]dioxol-5-yl and 2,3-dihydro- benzo[1 ,4]dioxin-6-yl, each of these radicals being unsubstituted or substituted by one or more, preferably up to three, substituents selected from those mentioned above for substituted aryl, and X, R3 and R4 are as defined in claim 1. or a (preferably pharmaceutically acceptable) salt thereof.
3. A compound of the formula I according to claim 1 , wherein, if mentioned,
unsubstituted or substituted aryl is phenyl, naphthyl or indanyl that is unsubstituted or substituted by one to three substituents independently selected from the group consisting of d-d-alkyl, hydroxyl-d-C7-alkyl, d-d-alkoxy-d-d-alkyl, amino-d-C7-alkyl, halo-C,-C7- alkyl, N-d-d-alkanoylamino-d-d-alkyl, N-d-d-alkanesulfonyl-amino-d-d-alkyl, pyrrolidino-CVCr-alkyl, oxo- pyrrolidino-d-C7-alkyl, piperidino-C,-C7-alkyl, piperazin-1-yl-d- C7-alkyl, 4-(C1-C7-alkyl, d-C7-alkoxy-d-C7-alkyf, halo-CrC7-alkyl or C3-C10-cycloalkyl)- piperazin-1-yl-d-C7-alkyl, 4-(amino-C1-C7-alkyl)-piperazin-1-yl-C1-C7-alkyl, 4-[N-mono- or N.N-dKCi-C^alkylaminoJ-CrC^alkylJ-piperazin-i-yl-CTCT-alkyl, (d-C7-alkoxy-d-d-alkyl, d-Cy-alkanoyl or C3-C10-cycloalkyl)-oxo-piperazin-1-yl-C1-C7-alkyl, morpholino-C1-C7-alkyl, thiomorpholino-d-C7-alkyl, S-mono- or S,S-dioxo-thiomorpholino-Crd-alkyl, 4-(C1-C7- alkanoyl)-piperazin-1-yl-C1-C7-alkyl, oxo-piperazin-i-yl-d-Cy-alkyl, imidazol-1-yl-Ct-Cτ-alkyl, pyrrolidin-i-carbonyl-C^C^alkyl, piperidin-i-carbonyl-d-CTr-alkyl, piperazin-1-carbonyt-C,- C7-alkyl, 4-(C1-C7-alkyl)piperazin-1-carbonyl-C,-C7-alkyl, morpholin-i-carbonyl-CTC^alkyl, thiomorpholin-1-carbonyl-C1-C7-alkyl, S-oxo- or S.S-dioxothiomorpholin-i-carbonyl-CrCr- alkyl, carbamoyl-CrCT-alkyl, [N-mono- or N.N-dKCrCralkyO-carbamoylJ-CrC^alkyl, C1-C7- alkanesulfinyl-Ci-C7-a!kyl, CrCr-alkanesulfonyMIVC^alkyl, halo, hydroxyl, d-CT-alkoxy, amino, N-mono- or N.N-di-^T-C^alkylJ-amino, C^C^alkanoylamino, pyrrolidino, oxo- pyrrolidino, piperidino, piperazin-1-yl, 4-(C1-C7-alkyl, C1-C7-alkoxy-C1-C7-alkyl, MaIo-C1-C7- alkyl or C3-C10-cycloalkyl)-piperazin-1-yl, 4-(amino-C1-C7-alkyl)-piperazin-1-yl, 4-[N-mono- or N.N-di^CrC^alkylaminoVCi-CT-alkylj-piperazin-i-yl, morpholino, thiomorpholino, S-oxo- or S,S-dioxothiomorpholino, d-C^alkanesulfonylamino, carbamoyl, N-mono- or N1N^i-(C1-C7- alkyl, C1-C7-alkoxy-C1-C7-alkyl, amino-CrC7-alkyl and/or (N'-mono- or N',N'-di-(C1-C7-alkyl)- amino-CT-C^alkylJ-carbamoyl, pyrrolidin-1-carbonyl, piperidin-1-carbonyl, piperazin-1- carbonyl, 4-(C1-C7-alkyl)piperazin-1-carbonyl, morpholin-1-carbonyl, thiomorpholin-1- carbonyl, S-oxo- or S.S-dioxothiomorpholin-i-carbonyl, sulfo, C1-C7-alkanesulfonyl, C1-C7- alkanesulfinyl, sulfamoyl, N-mono- or N,N-di-(C1-C7-alkyl)-sulfamoyl, morpholinosulfonyl, thiomorpholinosulfonyl, S-oxo-thiomorpholinosulfonyl, S,S-dioxothiomorpholinosulfonyl, cyano and nitro;
and unsubstituted or substituted heterocyclyl is preferably pyrrolyl, oxo-pyrrolyl, 2,3- dihydroindolyl, 2-oxo-2,3-dihydroindolyl or 1 H-pyridinonyl, each of which is unsubstituted or substituted by one to three moieties independently selected from those just mentioned for substituted aryl,
or a salt thereof.
4. A compound of the formula I according to claim 1 , wherein
X is CR5 or N, wherein R5 is halo, cyano, hydroxyl, C1-C^aIkOXy, C^C^alkyl, amino, N- mono- or N.N-di-CrCTr-alkyl or preferably hydrogen;
R1 is unsubstituted or substituted aryl or is unsubstituted or substituted heterocyclyl, especially R1 is phenyl, naphthyl, indanyl, pyridyl, oxo-1H-pyridyl, indolyl, dihydroindolyl or oxo-dihydroindolyl, each of which is bound via a ring carbon atom and is unsubstituted or substituted by one to three moieties independently selected from the group consisting of C1-
C7-alkyl, amino-C^CT-alkyl, halo-CrC7-alkyl, N-C1-C7-alkanoylamino-C1-C7-alkyl, N-C1-C7- alkanesulfonyl-amino-Ci-C^alkyl, pyrrolidino-CrC^alkyl, oxo- pyrrolidino-C^C^alkyl,
Figure imgf000134_0001
halo-d-C^alkyl or Ca-C^-cycloalkyO-piperazin-i-yl-d-C^alkyl, 4-(amino-C1-C7-alkyl)- piperazin-1-yl-CrC7-alkyl, 4-[N-mono- or N,N-di-(C1-C7-alkylamino)-C1-C7-alkyl]-piperazin-1- yl-CrC7-alkyl, morpholino-d-C7-alkyl, thiomorpholino-C1-C7-alkyl, S-mono- or S,S-dioxo- thiomorpholino-d-C^alkyl, carbamoyl-d-CT-alkyl, [N-mono- or N,N-di-(d-C7-alkyl)- carbamoyl]-C1-C7-alkyl, d-C7-alkanesulfinyl-d-C7-alkyl, C1-C7-alkanesulfonyl-C1-C7-alkyl, halo, hydroxyl, C1-C7-alkoxy, amino, N-mono- or N,N-di-(d-C7-alkyl)-amino, C1-C7- alkanoylamino, pyrrolidino, oxo-pyrrolidino, piperidino, piperazin-1-yl, 4-(C1-C7-alkyl, C1-C7- alkoxy-d-C7-alkyl, halo-d-C7-alkyl or C3-C10-cycloalkyl)-piperazin-1-yl, 4-(amino-d-C7- alkyl)-piperazin-1-yl, 4-[N-mono- or N,N-dir(C1-C7-alkylamino)-C1-C7-alkyl]-piperazin-1-yl, morpholino, thiomorpholino, S-oxo- or S,S-dioxothiomorpholino, d-C7-alkanesυlfonylamino, carbamoyl, N-mono- or N,N-di-(d-C7-alkyl, d-d-alkoxy-d-d-alkyl, amino-C^C^alkyl and/or (N '-mono- or N',N'-di-(C1-C7-alkyl)-amino-C1-Cτ-alkyl)-carbamoyl, pyrrolidin-1- carbonyl, piperidin-1-carbonyl, piperazin-1-carbonyl, 4-(C1-C7-alkyl)piperazin-1-carbonyl, morpholin-1-carbonyl, thiomorpholin-1-carbonyl, S-oxo- or S,S-dioxothiomorpholin-1- carbonyl, sulfo, d-C7-alkanesulfonyl, CrC7-alkanesulfiny!, sulfamoyl, N-mono- or N,N-di- (d-C7-alkyl)-sulfamoyl, morpholinosυlfonyl, thiomorpholinosulfonyl, cyano and nitro, R2 is phenyl, naphthyl, indanyl, pyridyl, oxo-1H-pyridyl, pyrazolyl, thiophenyl, indolyl, dihydroindolyl, oxo-dihydroindolyl, quinolinyl, isoquinolinyl or fH-benzoimidazolyl, each of which is bound via a ring carbon atom and is unsubstituted or substituted by one to three moieties independently selected from the group consisting of d-C7-alkyl, hydroxyl-d-C7- alkyl, amino-d-C7-alkyl, halo-d-C^alky!, N-d-CTr-alkanoylamino-d-CT-alkyl, N-C1-C7- alkanesulfonyl-amino-d-C7-alkyl, d-C^alkanesulfonyl-Ci-CTr-alkyl, d-Cr-alkanesulfinyl-C^ C7-alkyl, pyrrolidino-Ci-CT-alkyl, oxo-pyrrolidino-C1-C7-alkyl, piperidino-C^C^alkyl, piperazin- i-yl-CTC^alkyl, 4-(C1-C7-alkyl, C^CT-alkoxy-CTC^alkyl, halo-CrC7-alkyl or C3-C10- cycloalkyl)-piperazin-1 -yl-d-C7-alkyl, 4-(amino-CrC7-alkyl)-piperazin-1 -yl-C^C^alkyl, 4-[N- mono- or N.N-dKd-C^alkylaminoJ-Ci-C^alkyll-piperazin-i-yl-CrCT-alkyl, 4-(C1-C7- alkanoyl)-piperazin-1-yl-C1-C7-alkyl, oxo-piperazin-1-yl-C^C^alkyl, pyrrolidin-1-carbonyl-d- C^alkyl, piperidin-1-carbonyl-C^C^alkyl, piperazin-i-carbonyl-CrC^alkyl, 4-(C1-C7- alkyl)piperazin-1 -carbonyl-d-C^alkyl, morpholin-1 -carbonyl-CrC^alkyl, thiomorpholin-1 - carbonyl-C1-C7-alkyl, S-oxo- or S.S-dioxothiomorpholin-i-carbonyl-d-CTr-alkyl, d-C7-alkoxy- d-C7-alkyl, halo-d-C7-alkyl, CrC7-alkanoyl or Cs-CKj-cycloalkyO-oxo-piperazin-i-yl-CTCT-- alkyl, morpholino-d-d-alkyl, thiomorpholino-CTC^alkyl, S-mono- or S,S-dioxo- thiomorpholino-d-Cralkyl, imidazol-1-yl-CrC7-alkyl, carbamoyl-CrCT-alkyl, [N-mono- or N,N-di-(CrC7-alkyl)-carbamoyl]-d-C7-alkyl, d-d-alkanesulfinyl-d-C^alkyl, C1-C7- alkanesulfonyl-d-C7-alkyl, halo, hydroxyl, d-C7-alkoxy, amino, N-mono- or N,N-di-(C>-C7- alkyl)-amino, d-C7-alkanoylamino, pyrrolidine oxo-pyrrolidino, piperidino, piperazin-1-yl, 4- (d-CT-alkyl, d-Cτ-alkoxy-d-C7-alkyl, halo-d-C7-alkyl or Crdo-cycloalkyO-piperazin-i-yl, 4- (amino-d-C7-alkyl)-piperazin-1-yl, 4-[N-mono- or N,N-di-(d-C7-alkylamino)-d-C7-alkyl]- piperazin-1-yl, morpholino, thiomorpholino, S-oxo- or S,S-dioxothiomorpholino, d-C7-alkane- sulfonylamino, carbamoyl, N-mono- or N,N-di-(d-C7-alkyl, d-C7-alkoxy-d-C7-alkyl, amino- d-Cτ-alkyl and/or (N'-mono- or N'.N'-dKd-d-alky^-amino-d^-alkyO-carbamoyl, pyrrolidin-1-carbonyl, piperidin-1-carbonyl, piperazin-1-carbonyl, 4-(C1-C7-alkyl)piperazin-1- carbonyl, morpholin-1-carbonyl, thiomorpholin-1-carbonyl, S-oxo- or S,S-dioxothiomorpholin- 1-carbonyl, sulfo, d-C7-alkanesulfonyl, d-C7-alkanesulfinyl, sulfamoyl, N-mono- or N,N-di- (d-C7-alkyl)-sulfamoyl, morpholinosulfonyl, thiomorpholinosulfonyl, S-oxo- thiomorpholinosulfonyl, S,S-dioxothiomorpholinosulfonyl, cyano and nitro, preferably in the meta, (more preferably once) in the meta (preferably up to once) and the para or in the para position
R3 is cyano, hydroxyl, d-C7-alkyl, amino, N-mono- or N,N-di-d-C7-alkyl or preferably hydrogen; and R4 is hydroxyl, amino or preferably hydrogen;
or a pharmaceutically acceptable salt thereof.
5. A compound of the formula I according to claim 1 , wherein
X is CR5 or N, wherein R5 is halo, cyano, hydroxyl, d-C7-alkoxy, d-CValkyl, amino, N- mono- or N,N-di-Ci-C7-alkyl or preferably hydrogen;
R1 is unsubstituted or substituted aryl or is unsubstituted or substituted heterocyclyl,
R2 is phenyl, naphthyl, pyridyl, pyrazolyl, thiophenyl, quinolinyl, isoquinolinyl or fH-benzo- imidazolyl, each of which is bound via a ring carbon atom and is unsubstituted or substituted by one to three moieties independently selected from the group consisting of d-C7-alkyl, amino-d-d-alkyl, N-d-d-alkanoylamino-d-d-alkyl, N-d-C7-alkanesulfonyl-amino-d-C7- alkyl, carbamoyl-C1-C7-alkyl, [N-mono- or N.N-dKd-d-alkyO-carbamoyll-d-d-alkyl, C1-C7- alkanesulfinyl-d-C7-alkyl, C1-C7-alkanesulfonyl-C1-C7-alkyl, halo, hydroxyl, d-C7-alkoxy, amino, N-mono- or N,N-di-(Ci-C7-alkyl)-amino, C^C^alkanoylamino, C1-C7- alkanesulfonylamino, carbamoyl, N-mono- or N,N-di-(C1-C7-alkyl)-carbamoyl, sulfo, C1-C7- alkanesulfonyl, sulfamoyl, N-mono- or N,N-di-(d-C7-alkyl)-sulfamoyl, morpholinosulfonyl, thiomorpholinosulfonyl, cyano and nitro, R3 is cyano, hydroxyl, d-Cy-alkyl, amino, N-mono- or N.N-di-d-Cr-alkyl or preferably hydrogen; and
R4 is hydroxyl, amino or preferably hydrogen; or a pharmaceutically acceptable salt thereof.
6. A compound of the formula I according to claim 1 , wherein
X is CR5 or N, wherein R5 is halo, cyano, hydroxyl, Ci-C7-alkoxy, d-d-alkyl, amino, N- mono- or N,N-di-d-C7-alkyl or preferably hydrogen;
R1 is unsubstituted or substituted aryl or is unsubstituted or substituted heterocyclyl,
R2 is phenyl, naphthyl, pyridyl, pyrazolyl, thiophenyl, quinolinyl, isoquinolinyl or 1H-benzo- imidazolyl, each of which is bound via a ring carbon atom and is unsubstituted or substituted by one to three moieties independently selected from the group consisting of Ci-C7-alkyl, amino-d-Cr-alkyl, N-d-d-alkanoylamino-d-CT-alkyl, N-C1-C7-alkanesulfonyl-amino-C1-C7- alkyl, cart>amoyl-d-C7-alkyl, [N-mono- or N,N-di-(C1-C7-alkyl)-carbamoyl]-C1-C7-alkyl, C1-C7- alkanesulfinyl-d-C7-alkyl, C1-C7-alkanesulfonyl-C1-C7-alkyl, halo, hydroxyl, d-Cr-alkoxy, amino, N-mono- or N.N-dHd-CT-alkyO-amino, d-CTr-alkanoylamino, C1-C7- alkanesulfonylamino, carbamoyl, N-mono- or N.N-dHd-CT-alkyO-carbamoyl, sulfo, C1-C7- alkanesulfonyl, sulfamoyl, N-mono- or N,N-di-(C1-C7-alkyl)-sulfamoyl, morpholinosulfonyl, thiomorpholinosulfonyl, cyano and nitro, preferably in the meta, the meta and para or the para position;
R3 is cyano, hydroxyl, Ci-C7-alkyl, amino, N-mono- or N,N-di-d-Cτ-alkyl or preferably hydrogen; and
R4 is hydroxyl, amino or preferably hydrogen; or a pharmaceutically acceptable salt thereof.
7. A compound of the formula I according to claim 1 , wherein X is CH or N;
R1 is (especially 3,4,5-)trimethoxyphenyl or is 4-morpholinophenyl, (especially 3,4- or 3,5)- dimethoxyphenyl, (especially 4-) N-(2-methoxyethy!)-carbamoylphenyl, or (especially 4-)-
N,N-(2-dimethylamino-ethyl)-carbarnoylphenyl;
R2 is phenyl, 4-methylphenyl, 3-methylphenyl, 2-methylphenyl, 4-aminomethyl-phenyl, 3- aminomethyl-phenyl, 4-acetylaminomethyl-phenyl, 4-methanesulfonylarninomethyl-phenyl, 3- acetylaminomethyl-phenyl, 3-methanesulfonylaminomethyl-phenyl, 4-methanesulfonyl- aminomethyl-phenyl, 4-(N-methylcarbamoyl)methyl-phenyl, 4-methanesulfinylmethyl-phenyl, 4-methanesulfonylmethyl-phenyl, 3-chlorophenyl, 3-hydroxyphenyl, 4-methoxyphenyl, 3- methoxyphenyl, 2-methoxyphenyl, 4-aminophenyl, 3-aminophenyl, 2-aminophenyl, 3-N- methylamino-phenyl, 4-N,N-dimethylamino-phenyl, 4-acetylamino-phenyl, 3-acetylamino- phenyl, 4-methanesulfonylamino-phenyl, 4-methanesulfonylamino-phenyl, 3- methanesulfonylamino-phenyl, 4-carbamoylphenyl, 3-carbamoylphenyl, 4-(N-methyl-carba- moyl)-phenyl, 4-(N,N-dimethyl-carbamoyl)-phenyl, 4-methanesulfonylphenyl, 3-methanesul- fonylphenyl, 4-sulfamoylphenyr, 4-(N-methylsulfamoyl)-phenyl, 4-[N,N-(dimethyl)-sulfamoyl}- phenyl, 4-morpholinosulfonylphenyl, 4-cyanophenyl, 3-cyanophenyl, 3-nitrophenyl, 3-amino- 4-methyl-phenyl, 3-amino-4-methoxyphenyl, 3-amino-4-chlororphenyl, 4-methoxy-3-nitro phenyl, 2H-pyrazol-3-yl, 5-N-methylcarbamoyl-thiophenyl, 4-pyridyl, 3-pyridyl, 2-pyridyl, 6- methoxy-pyridin-3-yl, 1H-benzoimidazol-5-yl, quinolin-6-yl or isoquinolin-4-yl, and each of R3 and R4 is hydrogen; or a pharmaceutically acceptable salt thereof.
8. A compound of the formula I according to claim 1 , wherein X is CH or N;
R1 is phenyl, (especially 3,4,5-)trimethoxyphenyr, (especially 3,4- or 3,5-)dimethoxyphenyl*, (especially 4-)morpholinophenyl, (especially 4-) N-(2-methoxyethyl)-carbamoylphenyr, or (especially 4-)N,N-(2-dimethylamino-ethyl)-carbamoylphenyr, (especially 4-)dimethylamino- carbonyl-(especially 3-)methyl-phenyl*, (especially 4-)-(preferably 4-)-(2-methoxy-ethyI- piperazin-(especially 1-)yl-(especially 3-)-methyl-phenyl, (especially 4-)pyrrolidin-1-carbonyl- (especially 3-)methyl-phenyl*, (especially 3-)methyl-(especially 4-)-4-methylpiperazin-1- carbonyl-phenyl, (especially 3- or 4-)4-methyl-piperazin-1-yl-phenyl*, (especially 4-)-4-ethyl- piperazin-1-yl-(especially 3-)methyl-phenyl*, (especially 4-)-4-methyl-piperazin-1-yl- (espedally 3-)cyano-phenyl, (especially 4-)-piperazin-1-yl-phenyl, (especially 4-)-4- cyclopropyl-piperazin-1-yl-phenyl, (especially 4- )-4-(2-dimethylaminoethyl)-piperazin-1-yl- (especially 3-)methyl-phenyr, (especially 4-)4-isopropyl-piperazin-1-yl)-(especially 3-)methyl- phenyl*. (especially 4-)N,N-diethylaminocarbonyl-(especially 3-)methyl-phenyl*. (especially 4- )4-ethylpiperazin-1-carbonyl-(especially 3-)methyl-phenyl, (especially 4-)-(4-ethylpiperazin-1- ylmethyl)-(especially 3-)methyl-phenyl, (especially 4-)N-methylaminocarbonyl-(especially 3- )methylphenyl, (especially 4-)-4-(3,3,3-trifluoropropyl)-piperazin-1-yl-(especially 3-)methyl- phenyl, (especially 4-)-4-(2-(N',N'-dimethylamino)ethyl-aminocarbonyl-(especially 3-)methyl- phenyl, (especially 4-)-methanesulfonyl-phenyl*, (especially 4-)[(especially 2-)-oxo-pyrrolidin- 1-yl]-phenyl, (especially 4-)N,N-diethylaminocarbonyl-(especially 3-)methoxyphenyl, (especially 3-)-4-methylpiperazin-1-yl-(especially 4-)methyl-phenyl, (especially 3-)-4- methylpiperazin-1-yl-(especially 4-)methoxy-phenyr, (especially 3- or 4-)-morpholinomethyl- (especially 4- or 3-)methyl-phenyl, (especially 2-)acetylamino-indan-(especially 5-)yl, (especially 2-)oxo-2,3-dihydroindol-(especially 5-)yl, (especially 4-)methylsulfinylphenyl, (especially 4-)methoxyphenyl, (especially 4-)methyl-(especially 3-)methoxyphenyl, (especially 4-)-N-(2-methoxyethyl)-aminocarbonyl-phenyl, (especially 4-)N,N-dimethylcarbamoyl-phenyl, (especially 3-)methanesulfonylamino-phenyl, (especially 4-)methoxycarbonyl-(especially 3- )methoxy-phenyl, (especially 4-)N,N-dimethylcarbamoyl-(especially 3-)methoxy-phenyl, (especially 4-)-(4-cyclopropyl-piperazin-1-yl)-(especially 3-)methyl-phenyr, (especially 4-)-N- (2-(N',N'-dimethylaminoethyl)-N-methyl-carbamoyl-(especially 3-)methyl-phenyl*, 1 ,3- dimethyl-oxo-1 H-pyridine-5-yl, (especially 3- or 4-)morpholino-(especially 4- or 3-)methyl- phenyl*, (especially 4-)morpholinomethyl-(especially 3-)methyl-phenyl, (especially 4- )morpholin-1-carbonyl-(especially 3-)methyl-phenyl, (especially 4-)-N-2-(methoxyethyl)ami- nocarbonyl-(espedally 3-)methyl-phenyl, (especially 4-)-N-(3-N'.N'-dimethylaminopropyl)- amino-carbonyl-(especially 3-)methyl-phenyl, (especially 5-)-methyl-(especially 6-)methoxy- pyridin-3-yl, (especially 4-)dimethylcarbamoyl-(especially 3,5-)dimethyl-phenyl, (especially 4- )dimethylcarbamoyl-(especially 3-)ethyl-phenyl, (especially 4-(4-)N,N-dimethylcarbamoyl- (espedally 3-)methyl-phenyl or (especially 4-)morpholino-(especially 3-)cyano-phenyl; R2 is phenyl, (especially 4-)methylphenyl, (especially 3-)methylphenyl, (especially 2-)me- thylphenyl, (especially 4-)-hydroxymethyl-phenyl, (especially 4-)aminomethyl-phenyl, (especially 3-)aminomethyl-phenyl, (especially 4-)acetylaminomethyl-phenyl*, (especially 4- )methanesulfonylaminomethyl-phenyl, (especially 3-)acetylaminomethyl-phenyl, (especially 3-)methanesulfonylaminomethyl-phenyr, (especially 4-)methanesulfonylaminomethyl-phenyl, (especially 4-)(N-methylcarbamoyl)-methylphenyr> (especially 4-)methanesulfinytmethyl- phenyl, (especially 4-)methanesulfonylmethylphenyl, (especially 3-)chlorophenyl, (especially 3-)hydroxyphenyl, (especially 4-)methoxyphenyl, (especially 3-)methoxyphenyl*, (especially 2-)methoxyphenyl, (especially 4-)aminophenyl, (especially 3-)aminophenyl, (especially 2- )aminophenyl, (especially 3-)N-methylamino-phenyl, (especially 4-)N,N-dimethylamino- phenyl*, (especially 4-)acetylamino-phenyl, (especially 3-)acetylamino-phenyl, (especially 4- )methanesulfonylamino-phenyr, (especially 4-)methanesulfonylamino-phenyl, (especially 3-) methanesulfonylamino-phenyl*, (especially 4-)carbamoylphenyl, (especially 3-)carbamoyl- phenyl, (especially 4-)(N-methyl-carbamoyl)-phenyl, (especially 4-)(N,N-dimethyl-carbamoyl)- phenyl, (especially 4-)methanesulfonylphenyr, (especially 3-)methanesulfonylphenyl, (especially 4-)sulfamoylphenyr, (especially 4-)(N-methylsulfamoyl)-phenyr, (especially 4- )[N,N-(dimethyl)-sulfamoyl]-phenyl, (especially 4-)morpholinosulfonylphenyl, (especially A- )cyanophenyl, (especially 3-)cyanophenyl, (especially 3-)nitrophenyl, (especially 3-)amino-4- methyl-phenyl, (especially 3-)amino-4-methoxyphenyl, (especially 3-)amino-4-chforophenyl, (especially 4-)methoxy-3-nitrophenyl, (especially 4-)morpholin-4-ylmethyl-phenyl, (especially 3-)methyl-(especially 4-)morpholin-4-ylmethyl-phenyr, (especially 3-)fluoro-(especially A- )morpholin-4-ylmethyl-phenyl*, (especially 4-)S,S-dioxo-thiomorpholin-4-ylmethylphenyr, (especially 3,5-)difluoro-(especially 4-) morpholin-4-ylmethyl-phenyl, (especially 3-)fluoro- (especially 4-)S,S-dioxothiomorpholin-4-ylmethy(-phenyl*, (especially 3,5-)difluoro-(especially 4-)S,S-dioxothiomorpholin-4-ylmethyl-phenyr, (especially 3-)trifluoromethyl-(especially A- )morpholin-4-ylmethyl-phenyl, (especially 3,5-)difluoro-(especially 4-)[(preferably 4-)acetyl- piperazin-1-yl]methyl-phenyl, (especially 3,5-)difiuoro-(especially 4-)(preferably 4-)piperazin- 1-yl]methyl-phenyl, (especially 4-)[(preferably 4-)methyl-piperazin-1-yl]methyI-phenyl, (especially 3,5-)difluoro-(especially 4-)[(especially 3-)oxo-piperazin-1-yl]methyl-phenyl (especially 3,5-)difluoro-(espedally 4-)[(preferably 4-)methyl-(especially 3-)oxo-piperazin-1- yljmethyl-phenyl, (especially 4-)imidazol-1-ylmethyl-phenyl, (especially 4-)-4-methylpiperazin- 1-carbonyl-phenyl, (especially 4-)morpholin-4-carbonyl-phenyr, (especially 2- or 3-)fluoro- (especially 4-)morpholin-4-carbonyl-phenyr, (especially 3-)methyl-(especially 4-)morpholin-4- carbonyl-phenyl*, (especially 3,5-)difluoro-(especially 4-)morpholin-4-carbonyl-phenyl, (especially 4-)S,S-dioxothiomorpholin-4-carbonyl-phenyl, (especially 3-)-fluoro-(especially A- )S,S-dioxothiomorpholin-4-carbonyl-phenyl*, (especially 4-)morpholin-4-carbonylmethyl- phenyl*, (especially 3-)fluoro-(especially 4-)morpholin-4-carbonylmethyl-phenyr, [(especially 4-)morpholin-4-carbonyl-(1 ,1 ,dimethyl)-methyl]-phenyl, (especially 4-)S,S- dioxothiomorpholin-4-carbonylmethyl-phenyl*, (especially 3-)fluoro-(especially 4-)S,S- dioxothiomorpholin-4-carbonylmethyl-phenyl*, 2H-pyrazol-(especially 3-)yl, (especially 5-)N- methylcarbamoyl-thiophenyl, (especially 4-)pyridyl, (especially 3-)pyridyl, (especially 2- )pyridyl, (especially 6-)methoxy-pyridin-(especially 3-)yl, 1 H-benzoimidazol-(especially 5-)yl, quinolin-(especially 6-)yl or isoquinolin-(especially 4-)yl,
(where the moieties marked with an asterisk (*) are especially preferred, as are the moieties where the position after "especially" is given) and each of R3 and R4 is hydrogen; or a pharmaceutically acceptable salt thereof.
9. A compound of the formula I according to any one of claims 1 to 8, selected from the group of compounds with the following names: (7-m-tolyl-benzooxazol-2-yl)-(3,4,5-trimethoxy-phenyl)-amine;
(7-phenyl-benzooxazol-2-yl)-(3,4,5-trimethoxy-phenyl)-amine;
(7-pyridin-3-yl-benzooxazol-2-yl)-(3,4,5-trimethoxy-phenyl)-amine;
[7-(3-methoxy-phenyl)-benzooxazol-2-yl}-(3,4,5-trimethoxy-phenyl)-amine;
[7-(2-methoxy-phenyl)-benzooxazol-2-yl]-(3,4,5-trimethoxy-phenyl)-amine;
[7-(3-hydroxy-phenyl)-benzooxazol-2-yl]-(3,4,5-trinnethoxy-phenyl)-amine;
[7-(4-methoxy-phenyl)-benzooxazol-2-yl]-(3,4,5-trimethoxy-phenyl)-amine;
(7-isoquinolin-4-yl-benzooxazol-2-yl)-(3,4,5-trimethoxy-phenyl)-amine;
[7-(3-chloro-phenyl)-benzooxazol-2-yl]-(3,4,5-trimethoxy-phenyl)-amine;
[7-(3-amino-phenyl)-benzooxazol-2-yl]-(3,4,5-trimethoxy-phenyl)-amine;
[7-(4-amino-phenyl)-benzooxazol-2-yl]-(3,4,5-trimethoxy-phenyl)-amine;
[7-(6-methoxy-pyridin-3-yl>-benzooxazol-2-yl]-(3,4,5-trimethoxy-phenyl)-amine;
[7-(3-amino-4-methyl-phenyl)-benzooxazol-2-yl]-(3,4,5-trimethoxy-phenyl)-amine;
[7-(2-amino-phenyl)-benzooxazol-2-yl]-(3,4,5-trimethoxy-phenyl)-amine;
(7-quinolin-6-yl-benzooxazol-2-yl)-(3,4,5-trimethoxy-phenyl)-amine;
4-[2-(3,4,5-trimethoxy-phenylamino)-benzooxazol-7-yl]-benzamide;
7-(4-methanesulfonyl-phenyl)-benzooxazol-2-yl]-(3,4,5-trimethoxy-phenyl)-amine;
4-[2-(3,4,5-trimethoxy-phenylamino)-benzooxazol-7-yl]-benzenesulfonamide;
4-[2-(3,4,5-trimethoxy-phenylamino)-benzooxazol-7-yl]-benzonitrile;
[7-(2H-pyrazol-3-yt)-benzooxazol-2-yl]-(3,4,5-trimethoxy-phenyl)-amine;
N-methyl-4-[2-(3,4,5-trimethoxy-phenylamino)-benzooxazol-7-yl]-benzenesulfonamide;
N,N-dimethyl-4-[2-(3,4,5-trimethoxy-phenylamino)-benzooxazol-7-yl]-benzenesulfonamide;
{7-[4-(morpholine-4-sulfonyl)-phenyl]-benzooxazol-2-yl}-(3,4,5-trimethoxy-phenyl)-amine;
N-methyl-4-[2-(3,4,5-trimethoxy-phenylamino)-benzooxazol-7-yl]-benzamide;
N,N-dimethyl-4-[2-(3,4,5-trimethoxy-phenylamino)-benzooxazol-7-yl]-benzamide;
N-{4-[2-(3,4,5-trimethoxy-phenylamino)-benzooxazol-7-ylJ-phenyl}-methanesulfonamide;
N-{4-[2-(3,4,5-trimethoxy-phenylamino)-benzooxazol-7-yl]-phenyl}-acetamide;
N-{3-[2-(3,4,5-trimethoxy-phenylamino)-benzooxazol-7-yl]-phenyl}-acetamide;
[7-(4-aminomethyl-phenyl)-benzooxazol-2-yl]-(3,4,5-trimethoxy-phenyl)-amine;
N-{4-[2-(3,4,5-trimethoxy-phenylamino)-benzooxazol-7-yf}-benzyl}-acetamide;
N-{4-[2-(3,4,5-trimethoxy-phenylamino)-benzooxazol-7-yl]-benzyl}-methanesulfonamide;
[7-(3-aminomethyl-phenyl)-benzooxazol-2-yl]-(3,4,5-trimethoxy-phenyl)-amine;
N-{3-[2-(3,4,5-trimethoxy-phenylamino)-benzooxazol-7-yl]-benzyl}-acetamide; 2-(3,4,5-trimethoxy-phenylamino)-benzooxazot-7-yl]-thiophene-2-carboxylic acid methylamide;
[7-(1 H-benzoimidazol-5-yl)-benzooxazol-2-yl]-(3,4,5-trimethoxy-phenyl)-amine; [4-(4-amino-phenyl)-oxazolo[5,4-c]pyridin-2-yl]-(3,4,5-trimethoxy-phenyl)-amine; [4-(3-amino-phenyl)-oxazolo[5,4-c]pyridin-2-yl]-(3,4,5-trimethoxy-phenyl)-amine; 4-[2-(3,4,5-trimethoxy-phenylamino)-oxazolo[5,4-c]pyridin-4-yl]-benzenesulfonamide; [7-(3-methoxy-phenyl)-benzooxazol-2-yl]-(4-morpholin-4-yl-phenyl)-amine; [7-(4-amino-phenyl)-benzooxazol-2-yl]-(4-morpholin-4-yl-phenyl)-amine; N-{4-[2-(4-morpholin-4-yl-phenytamino)-benzooxazol-7-yl]-phenyl}-methane-sulfonamide; [7-(3-amino-phenyl)-benzooxazol-2-yl]-(6-morpholin-4-yl-pyridin-3-yl)-amine; N-(2-methoxy-ethyl)-4-[7-(4-sulfamoyl-phenyl)-benzooxazol-2-ylamino]-benzamide N-(2-dimethylamino-ethyl)-4-[7-(4-sulfamoyl-phenyl)-benzooxazol-2-ylamino]-benzamide 4-[2-(3,4-dimethoxy-phenylamino)-benzooxazol-7-yl]-benzenesulfonamide 4-[2-(3,4-dimethoxy-phenylamino)-benzooxazol-7-yl]-N-methyl-benzenesulfonamide 4-[2-(3,5-dimethoxy-phenylamino)-benzooxazol-7-yl]-benzenesulfonamide N-methyl-2-{4-[2-(3,4,5-trimethoxy-phenylamino)-benzooxazol-7-yl]-phenyl}-acetamide N-{4-[2-(3,4,5-trimethoxy-phenylamino)-benzooxazol-7-yl}-benzyl}-methanesulfonamide; and [7-(4-methanesulfinylmethyl-phenyl)-benzooxazol-2-yl]-(3,4,5-trimethoxy-phenyl>-amine; K^-Amino-phenyO-oxazolofS^-cJpyridin^-yQ-CS^.S-trimethoxy-phenyO-amine: (7-o-Tolyl-benzooxazol-2-yl)-(3,4,5-trimethoxy-phenyl)-amine: 3-[2-(3>4,5-Trimethoxy-phenylamino)-benzooxazol-7-yl]-benzonitrile: (7-Pyridin-2-yl-benzooxazol-2-yl)-(3,4,5-trimethoxy-phenyl)-amine: 3-[2-(3,4,5-Trimethoxy-phenylamino)-benzooxazol-7-yl]-benzamide: [7-(3-Nitro-phenyl)-benzooxazol-2-yl]-(3,4,5-trimethoxy-phenyl)-amine: [7-(3-Methanesulfonyl-phenyl)-benzooxazol-2-yl]-(3,4,5-trimethoxy-phenyl)-amine: N-Methyl-3-[2-(3,4,5-trimethoxy-phenylamino)-benzooxazol-7-yl]-benzenesulfonamide: [7-(4-Methoxy-3-nitro-phenyI)-benzooxazol-2-yl]-(3,4,5-trimethoxy-phenyl)-amine: [7-(3-Amino-4-chloro-phenyl)-benzooxazol-2-yl]-(3,4,5-trimethoxy-phenyl)-amine: [7-(3-Amino-4-methoxy-phenyl)-benzooxazol-2-yl]-(3,4,5-trimethoxy-phenyl)-amine: [7-(4-Dimethylamino-phenyl)-benzooxazol-2-yl]-(3,4,5-trimethoxy-phenyl)-amine: N-{3-[2-(3,4,5-Trimethoxy-phenylamino)-benzooxazol-7-yl]-phenyl}-methanesulfonamide:
or a pharmaceutically acceptable salt thereof.
10. A compound of the formula I according to any one of claims 1 to 9, selected from the group of compounds with the following names:
[7-(3-amino-phenyl)-benzooxazol-2-yl}-(4-morpholin-4-yhphenyl)-amine:;
N-{4-[2-(3,4,5-trimethoxy-phenylamino)-oxazolo[5,4-c]pyridin-4-yl]-benzyl}- methanesulfonamide;
4-[7-(3-fluoro-4-morpholin-4-ylmethyl-phenyl)-benzooxazol-2-ylamino]-2,N,N-trimethyl- benzamide;
4-{7-[4-(1 ,1-dioxo-thiomorpholin-4-ylmethyl)-3,5-difluoro-phenyl]-benzooxazol-2-ylamino}-
2,N,N-trimethyl-benzamide;
[4-(2-{4-[4-(2-methoxy-ethyl)-piperazin-1-yl]-3-methyl-phenylamino}-benzooxazol-7-yl)-2- methyl-phenyl]-morpholin-4-yl-methanone;
{4-[7-(3-fluoro-4-morpholin-4-ylmethyl-phenyl)-benzooxazol-2-ylamino]-2-methyl-phenyl}- pyrrolidin-1 -yl-methanone;
4-[7-(3,5-Difluoro-4-morphoHn-4-ylmethyl-phenyl)-benzooxazol-2-ylamino]-2,N,N-trimethyl- benzamide;
2,N,N-trimethyl-4-{7-[4-(morpholine-4-carbonyl)-phenyl]-benzooxazol-2-ylamino}-benzamide;
2,N,N-trimethyl-4-{7-[4-(2-morpholin-4-yl-2-oxo-ethyl)-phenyl]-benzooxazol-2-ylamino}- benzamide;
4-[7-(4-methanesulfonylmethyl-phenyl)-benzooxazol-2-ylamino]-N,N-dimethyl-benzamide;
2-(4-{2-[3-methyl-4-(4-methyl-piperazine-1-carbonyl)-phenylamino}-benzooxazol-7-yl}- phenyl)-1-morpholin-4-yl-ethanone;
2-(2-fluoro-4-{2-[3-methyl-4-(4-methyl-piperazine-1-carbonyl)-phenylamino]-benzooxazol-7- yl}-phenyl)-1-morpholin-4-yl-ethanone;
2-(4-{2-[4-(4-methyl-piperazin-1-yl)-phenylamino]-benzooxazol-7-yl}-phenyl)-1-morpholin-4- yl-ethanone;
(2-fluoro-4-{2-[4-(4-methyl-piperazin-1-yl)-phenylamino]-benzooxazol-7-yl}-phenyl)- morpholin-4-yl-methanone;
4-{7-[2-fluoro-4-(morpholine-4-carbonyl)-phenyl]-benzooxazol-2-ylamino}-2,N,N-trimethyl- benzamide;
4-(7-{4-[2-(1 ,1-dioxo-1lambda*6*-thiomorpholin-4-yl)-2-oxo-ethyl]-phenyl}-benzooxazol-2- ylamino)-2,N,N-trimethyl-benzamide;
4-{7-[3-fluoro-4-(morpholine-4-carbonyl)-phenyl]-benzooxazol-2-ylamino}-2,N,N-trimethyl- benzamide; 4-{7-[4-(1 ,1-dioxo-1lambda*6*-thiomorpholin-4-ylmethyl)-phenyl]-benzooxazol-2-ylamino}-
2,N,N-trimethyl-benzamide;
(1 ,1-dioxo-1lambda*6*-thiomorpholin-4-yl)-(2-fluoro-4-{2-[4-(4-methyl-piperazin-1-yl)- phenylamino]-benzooxazol-7-yl}-phenyl)-methanone;
4-[7-(4-methanesulfinylmethyl-phenyl)-benzooxazol-2-ylamino]-2,N,N-trimethyl-benzamide;
(2-methyl-4-{2-[4-(4-methyl-piperazin-1-yl)-phenylamino]-benzooxazol-7-yl}-phenyl)- morpholin-4-yl-methanone;
4-(7-{4-[2-(1 ,1 -dioxo-1 lambda*6*-thiomorpholin-4-yl)-2-oxo-ethyl]-3-fluoro-phenyl}- benzooxazol-2-ylamino)-2,N,N-trimethyl-benzamide;
4-{7-[4-(1 ,1 -dioxo-1 lambda*6*-thiomorpholin-4-ylmethyl)-3-fluoro-phenyl]-benzooxazol-2- ylamino}-2,N,N-trimethyl-benzamide;
2,N,N-trimethyl-4-{7-[3-methyl-4-(morpholine-4-carbonyl)-phenyl]-benzooxazol-2-ylamino}- benzamide;
2,N,N-trimethyl-4-[7-(4-morpholin-4-ylmethyl-3-trifluoromethyl-phenyl)-benzooxazol-2- ylaminoj-benzamide;
[4-(4-ethyl-piperazin-1-yl)-3-methyl-phenyl]-[7-(3-fluoro-4-morpholin-4-ylmethyl-phenyl)- benzooxazol-2-yl]-amine;
{7-[4-(1 ,1 -dioxo-1 lambda*6*-thiomorpholin-4-ylmethyl)-3-fluoro-pher>yl]-benzooxazol-2-yl}-[4-
(4-ethyl-piperazin-1-yl)-3-methyl-phenyl]-amine;
{7-[4-(1 , 1 -dioxo-1 lambda *6*-thiomorphoIin-4-ylmethyl)-phenyl]-benzooxazo]-2-yl}-[4-(4- methyt-piperazin-1-yl)-phenylj-amine;
5-{7-[3-fluoro-4-(morpholine-4-caronyl)-phenyll-benzooxazol-2-ylamino}-2-(4-methyl- piperazin-1-yl)-benzonitrile;
{2-fluoro-4-[2-(4-piperazin-1-yl-phenylamino)-benzooxazol-7-yl]-phenyl}-morpholin-4-yl- methanone;
[4-(4-ethyl-piperazin-1-yl)-3-methyl-phenyl]-[7-(4-methanesulfonyl-phenyl)-benzooxazol-2-yl]- amine;
4-{2-[4-(4-methyl-piperazin-1-yl)-phenylamino]-benzooxazol-7-yl}-benzenesulfonamide;
(4-{2-[4-(4-ethyl-piperazin-1-yl)-3-methyl-phenylamino]-benzooxazol-7-yl}-2-fluoro-phenyl)- morpholin-4-yl-methanone;
{7-[4-(1 ,1 -dioxo-1 lambda*6*-thiomorpholin-4-ylmethyl)-3,5-difluoro-phenyl]-benzooxazol-2- yl}-[4-(4-methyl-piperazin-1-yl)-phenyl]-amine;
(4-{7-[4-(1 , 1 -dioxo-1 lambda*6*-thiomorpholin-4-ylmethyl)-3,5-difluoro-phenyl]-benzooxazol-
2-ylamino}-2-methyl-phenyl)-pyrrolidin-1-yl-methanone; (4-{2-[4-(4-cyclopropyl-piperazin-1-yl)-phenylamino]-benzooxazol-7-yl}-2-methyl-phenyl)- morpholin-4-yl-methanone;
[7-(3,5-difluoro-4-morpholin-4-ylmethyl-phenyl)-benzooxazol-2-yl]-[4-(4-ethyl-piperazin-1-yl)-
3-methyl-phenyl]-amine;
{4-[7-(3,5-difluoro-4-morpholin-4-ylmethyl-phenyl)-benzooxazol-2-ylamino]-2-methyl-phenyl}- pyrrolidin-1 -yl-methanone;
{4-[4-(2-dimethylamino-ethyl)-piperazin-1-yl]-3-methyl-phenyl}-[7-(4-methanesulfonyl- phenyl)-benzooxazol-2-yl]-amine;
{7-[4-(1 ,1 -dioxo-1 lambda*6*-thiomorpholin-4-ylmethyl)-3,5-difluoro-phenyl]-benzooxazol-2- yl}-[4-(4-isopropyl-piperazin-1-yl)-3-methyl-phenyl]-amine;
[4-(2-{4-[4-(2-dimethylamino-ethyl)-piperazin-1-yl]-3-methyl-phenylamino}-benzooxazol-7-yl)-
2-methyl-phenyl]-morpholin-4-yl-methanone;
4-[7-(3,5-difluoro-4-morpholin-4-ylmethyl-phenyl)-benzooxazol-2-ylamino]-N,N-diethyl-2- methyl-benzamide;
{4-[7-(3,5-difluoro-4-rnorpholin-4-ylmethyl-phenyl)-benzooxazol-2-yfamino]-2-methyl-phenyl}-
(4-ethyl-pi perazi n- 1 -yl )-metha none ;
[4-(4-isopropyl-piperazin-1-yl)-3-methyl-phenyl]-[7-(4-methanesulfonyl-phenyl)-benzooxazol-
2-yl]-amine;
(4-{2-[4-(4-isopropyl-piperazin-1-yl)-3-methyl-phenylamino}-benzooxazol-7-yl}-2-methyl- phenyl )-morpholin-4-yl-methanone;
{4-[4-(2-dimethylamino-ethyl)-piperazin-1-yl]-3-methyl-phenyl}-{7-[4-(1 ,1 -dioxo-1 lambda*6*- thiomorpholin-4-ylmethyl)-3,5-difluoro-phenyl]-benzooxazol-2-yl}-amine;
{7-[4-(1 , 1 -dioxo-1 lambda*6*-thiomorpholin-4-ylmethyl)-3,5-difluoro-phenyl]-benzooxazol-2- yl}-{4-[4-(2-methoxy-ethyl)-piperazin-1-yl]-3-methyl-phenyl}-amine;
(4-{2-[4-(4-ethyl-piperazin-1-yl)-3-methyl-phenylamino]-benzooxazol-7-yl}-2,6-difluoro- phenyl )-morpholin-4-yl-methanone;
{2-methyl-4-[7-(3-methyl-4-morpholin-4-ylmethyl-phenyl)-benzooxazol-2-ylamino]-phenyl}- pyrrolidin-1 -yl-methanone;
2-(4-{2-[4-(4-ethyl-piperazin-1-yl)-3-methyl-phenylamino]-benzooxazol-7-yl}-phenyl)-2- methyl-1-morpholin-4-yl-propan-1-one;
4-{2-[4-(4-ethyl-piperazin-1-yl)-3-methyl-phenylamino]-benzooxazol-7-yl}- benzenesulfonamide;
[4-(4-cyclopropyl-piperazin-1-yl)-phenyl]-[7-(4-methanesulfonyl-phenyl)-benzooxazol-2-yl]- amine; [4-(4-cyclopropyl-piperazin-1-yl)-phenyl]-{7-[4-(1 ,1-dioxo-1lambda*6*-thiomorpholin-4- ylmethyl)-3,5-difluoro-phenyl]-benzooxazol-2-yl}-amine;
[7-(4-methanesulfonyl-phenyl)-benzooxazol-2-yl]-{4-[4-(2-methoxy-ethyl)-piperazin-1-yl}-3- methyl-phenyl}-amine;
(1 ,1-dioxo-1lambda*6*-thiomorpholin-4-yl)-(4-{2-[4-(4-ethyl-piperazin-1-yl)-3-methyl- phenylamino]-benzooxazol-7-yl}-phenyl)-methanone;
4-{7-[4-(4-acetyl-piperazin-1-ylmethyl)-3,5-difluoro-phenyl]-benzooxazol-2-ylamino}-N,N- diethyl-2-methyl-benzamide;
4-[7-(3,5-difluoro-4-piperazin-1-ylmethyl-phenyl)-benzooxazol-2-ylamino]-N,N-diethyl-2- methyl-benzamide;
(4-{2-[4-(4-ethyl-piperazin-1-ylmethyl)-3-methyl-phenylamino]-benzooxazol-7-yl}-2-fluoro- pheny!)-morpholin-4-yl-methanone;
4-[7-(3,5-difluoro-4-morpholin-4-ylmethyl-phenyl)-benzooxazol-2-ylamino]-2,N-dimethyl- benzamide;
[7-(3,5-difluoro-4-morpholin-4-ylmethyl-phenyl)-benzooxazol-2-ylH3-methyl-4-[4-(3,3,3- trifluoro-propyl)-piperazin-1-yl]-phenyl}-amine;
4-[7-(3,5-difluoro-4-morpholin-4-ylmethyl-phenyl)-benzooxazol-2-ylamino]-N-(2- dimethylamino-ethyl)-2-methyl-benzamide;
4-(7-{4-[2-(1 ,1-dioxo-1lambda*6*-thiomorpholin-4-yl)-2-oxo-ethyl]-phenyl}-benzooxazol-2- ylamino)-N,N-diethyl-2-methoxy-benzamide;
2-{4-[2-(4-methanesulfonyl-phenylamino)-benzooxazol-7-yl]-phenyl}-1-morpholin-4-yt- ethanone;
1-(4-{7-[4-(1 ,1-dioxo-1lambda*6*-thiomoφholin-4-ylmethyl)-3,5-difluoro-phenyl]- benzooxazol-2-ylamino)-phenyl)-pyrrolidin-2-one;
1-{4-[7-(3,5-difluoro-4-morpholin-4-ylmethyl-phenyl)-benzooxazol-2-ylamino]-phenyl}- pyrrolidin-2-one;
(4-{2-[4-methoxy-3-(4-methyl-piperazin-1-yl)-phenylamino]-benzooxazol-7-yl}-2-methyl- phenyl)-morpholin-4-yl-methanone;
{7-[4-(1 ,1-dioxo-1lambda*6*-thiomorpholin-4-ylmethyl)-3,5-difluoro-phenyl]-benzooxazol-2- yl}-[3-(4-methyl-piperazin-1-yl)-phenyl]-amine;
(2-methyl-4-{2-[3-(4-methyl-piperazin-1-yl)-phenylamino]-benzooxazol-7-yl}-phenyl)- morpholin-4-yl-methanone;
{7-[4-(1 ,1 -dioxo-1 lambda*6*-thiomorpholin-4-ylmethyl)-3,5-difluoro-phenyI]-benzooxazol-2- yI}-[4-methoxy-3-(4-methyl-piperazin-1-yl)-phenyl]-amine; {2-fluoro-4-[2-(3-methyl-4-morpholin-4-ylmethyl-phenylamino)-benzooxazol-7-yl]-phenyl}- morpholin-4-yl-methanone;
N-{5-[7-(3-fluoro-4-morpholin-4-ylmethyl-phenyl)-benzooxazol-2-yfamino]-indan-2-yl}- acetamide;
N-(5-{7-[4-(1 ,1-dioxo-1 lambda*6*-thiomorpholin-4-ylmethyl>-phenyl]-benzooxazol-2-ylamino}- indan-2-yl)-acetamide;
5-[7-(3,5-difluoro-4-morpholin-4-ylmethyl-phenyl)-benzooxazol-2-ylamino]-1 ,3-dihydro-indol-
2-one;
2-{4-[2-(4-methanesulfinyl-phenylamino)-benzooxazol-7-yl]-phenyl}-1-morpholin-4-yl- ethanone;
[7-(4-imidazol-1-ylmethyl-phenyl)-benzooxazol-2-yl]-(3,4,5-trimethoxy-phenyl)-amine;
{7-[4-(4-methyl-piperazin-1-ylmethyl)-phenyl]-benzooxazol-2-yl}-(3,4,5-trimethoxy-phenyl)- amine;
[7-(4-morpholin-4-ylmethyl-phenyl)-benzooxazol-2-yl]-(3,4,5-trimethoxy-phenyl)-amine;
4-[2-(4-methoxy-phenylamino)-benzooxazol-7-yl]-N-methyl-benzenesulfonamide;
1-morpholin-4-yl-2-{4-[2-(3,4,5-trimethoxy-phenylamino)-benzooxazol-7-yl]-phenyl}- ethanone; morpholin-4-yl-{4-[2-(3,4,5-trimethoxy-phenylamino)-benzooxazol-7-yl}-phenyl}-methanone;
(4-methyl-piperazin-1-yl)-{4-[2-(3,4,5-trimethoxy-phenylamino)-benzooxazol-7-yl]-phenyl}- methanone;
{4-[2-(3,4,5-trimethoxy-phenylamino)-benzooxazol-7-yl]-phenyl}-methanol;
4-[2-(3-methoxy-4-methyl-phenylamino)-benzooxazo!-7-yl]-N-methyl-benzenesulfonamide;
N-(2-methoxy-ethyl)-4-{7-[4-(moφhoHne-4-carbonyl)-phenyl]-benzooxazol-2-ylamino}- benzamide;
N,N-dimethyl-4-[7-(4-sulfamoyl-phenyl)-benzooxazol-2-ylamino]-benzamide;
N-(2-methoxy-ethyl)-4-{7-[4-(2-morpholin-4-yl-2-oxo-ethyl)-phenyl]-benzooxazol-2-ylamino}- benzamide;
N-(3-{7-[4-(morpholine-4-carbonyl)-phenyl]-benzooxazol-2-ylamino}-phenyl)- methanesulfonamide;
2-methoxy-4-{7-[4-(morpholine-4-carbonyl)-phenyl]-benzooxazol-2-ylamino}-benzoic acid methyl ester;
2-methoxy-N,N-dimethyl-4-{7-[4-(morpholine-4-carbonyl)-phenyl]-benzooxazol-2-ylamino}- benzamide; 2-methyl-2-(4-{2-[4-(4-methyl-piperazin-1-yl)-phenylamino]-benzooxazol-7-yl}-phenyl)-1- morpholin-4-yl-propan-1 -one;
[4-(3,5-difluoro-4-morpholin-4-ylmethyl-phenyl)-oxazolo[5,4-c]pyridin-2-yl]-[4-(4-ethyl- piperazin-1-yl)-3-methyl-phenyl]-amine;
{4-[4-(1 ,1-dioxo-1lambda*6*-thiomorpholin-4-ylmethyl)-3,5-clifluoro-pheny(]-oxazolo[5,4- c]pyridin-2-yl}-[4-(4-ethyl-piperazin-1-yl)-3-methyl-phenyl]-amine;
[4-(4-cyclopropyl-piperazin-1-yl)-3-methyl-phenyl]-{7-[4-(1 ,1-dioxo-1lambda*6*-thiomorpholin-
4-ylmethyl)-3,5-difluoro-phenyl]-benzooxazol-2-yl}-amine;
(4-{2-[4-(4-cyclopropyl-piperazin-1-yl)-3-methyl-phenylamino}-benzooxazol-7-yl}-2-methyl- phenyl )-morpholin-4-yl-methanone;
(4-{2-[4-(4-cyclopropyl-piperazin-1-yl)-3-methyl-phenylamino]-benzooxazol-7-yl}-2-fluoro- phenyl)-morpholin-4-yl-methanone;
2-(4-{2-[4-(4-cyclopropyl-piperazin-1-yl)-3-methyl-phenylamino]-benzooxazol-7-yl}-phenyl)-2- methyl-1-morpholin-4-yl-propan-1-one;
4-[7-(3,5-difluoro-4-morpholin-4-ylmethyl-phenyl)-benzooxazol-2-ylamino]-N-(2- dimethylamino-ethyl)-2,N-dimethyl-benzamide;
N-(2-dimethylamino-ethyl)-4-{7-[4-(1 ,1-dioxo-1lambda*6*-thiomorpholin-4-ylmethyl)-3,5- difluoro-phenyl]-benzooxazol-2-ylamino}-2,N-dimethyl-benzamide;
[4-(4-cyclopropyl-piperazin-1-yl)-3-methyl-phenyl]-[7-(3,5-difluoro-4-morpholin-4-ylmethyl- phenyl)-benzooxazol-2-yl]-amine;
5-[7-(3,5-difluoro-4-morpholin-4-ylmethyl-phenyl)-benzooxazol-2-ylamino]-1,3-dimethyl-1H- pyridin-2-one;
(4-{2-[4-(4-cyclopropyl-piperazin-1-yl)-3-methyl-ptienylamino]-benzooxazol-7-yl}-2-methyl- phenyl )-(1 , 1 -dioxo-1 lambda*6*-thiomorpholin-4-yl)-methanone;
[7-(3,5-difluoro-4-morpholin-4-ylmethyl-phenyl)-benzooxazol-2-yl]-(3-methyl-4-morpholin-4- yl-phenyl)-amine;
{7-[4-(1 ,1 -dioxo-1 lambda*6*-thiomorpholin-4-ylmethyl)-3,5-difluoro-phenyl]-benzooxazol-2- yl}-(3-methyl-4-morpholin-4-yl-phenyl)-amine;
(1 ,1 -dioxo-1 lambda*6*-thiomorpholin-4-yl)-{4-[2-(3-methyl-4-morpholin-4-ylmethyl- phenylamino)-benzooxazol-7-yl]-phenyl}-methanone;
{2-methyl-4-[2-(3-methyl-4-morpholin-4-ylmethyl-phenylamino)-benzooxazol-7-yl]-phenyl}- morpholin-4-yl-methanone;
2-methyl-2-{4-[2-(3-methyl-4-morpholin-4-ylmethyl-phenylamino)-benzooxazol-7-yl]-phenyl}-
1 -morpholin-4-yl-propan-1 -one; 4-{4-[4-(1 ,1-dioxo-1lambda*6*-thiomorpholin-4-ylmethyl)-3,5-difluoro-phenyl]-oxazolo[5,4- c]pyridin-2-ylamino}-2,N,N-trimethyl-benzamide;
4-{7-[3,5-difluoro-4-(3-oxo-piperazin-1-ylmethyl)-phenyl]-benzooxazol-2-ylamino}-2,N,N- trimethyl-benzamide;
4-{2,6-difluoro-4-[2-(3-methyl-4-morpholin-4-yl-phenylamino)-benzooxazol-7-yl]-benzyl}- piperazin-2-one;
4-(4-{2-[4-(4-cyclopropyl-piperazin-1-yl)-3-methyl-phenylamino]-benzooxazol-7-yl}-2,6- difluoro-benzyl)-piperazin-2-one;
4-{7-[3,5-difluoro-4-(4-methyl-3-oxo-piperazin-1-ylmethyl)-phenyl]-benzooxazol-2-ylamino}-
2,N,N-trimethyl-benzamide;
4-{2,6-difluoro-4-[2-(3-methyl-4-morpholin-4-yl-phenylamino)-benzooxazol-7-yl]-benzyl}-1- methyl-piperazin-2-one;
4-(4-{2-[4-(4-cyclopropyl-piperazin-1-yl)-3-methyl-phenylamino]-benzooxazol-7-yl}-2,6- difluoro-benzyl)-1 -methyl-piperazin-2-one;
4-[7-(4-methanesulfinylmethyl-3-methyl-phenyl)-benzooxazol-2-ylamino]-2,N,N-trimethyl- benzamide;
[4-(4-cyclopropyl-piperazin-1-yl)-3-methyl-phenyl]-[7-(4-methanesu!fonylmethyl-3-methyl- phenyl )-benzooxazol-2-yl]-amine;
[7-(4-methanesulfinylmethyl-3-methyl-phenyl)-benzooxazol-2-yl]-(4-methyl-3-morpholin-4-yl- phenyl)-amine;
{4-[7-(3,5-difluoro-4-morpholin-4-ylmethyl-phenyl)-benzooxazol-2-ylamino]-2-methyl-phenyl}- morpholin-4-yl-methanone;
4-[7-(3,5-difluoro-4-morpholin-4-ylmethyl-phenyl)-benzooxazol-2-ylamino]-N-(2-methoxy- ethyl)-2-methyl-benzamide;
4-[7-(3,5-difluoro-4-morpholin-4-ylmethyl-phenyl)-benzooxazol-2-ylamino]-N-(3- dimethylamino-propyl)-2-methyl-benzamide;
[7-(3,5-difluoro-4-morpholin-4-ylmethyl-phenyl)-benzooxazol-2-yl]-(6-methoxy-5-methyl- pyridin-3-yl)-amine;
4-[7-(3,5-difluoro-4-morpholin-4-ylmethyl-phenyl)-benzooxazol-2-ylamino]-2,6,N,N- tetramethyl-benzamide;
4-[7-(3,5-difluoro-4-morpholin-4-ylmethyl-phenyl)-benzooxazol-2-ylamino]-2-ethyl-N,N- dimethyl-benzamide;
4-[7-(3,5-difluoro-4-morpholin-4-ylmethyl-phenyl)-benzooxazol-2-ylamino]-N-(2-methoxy- ethyl)-2,N-dimethyl-benzamide; 4-[7-(3,5-difluoro-4-morpholin-4-ylmethyl-phenyl)-benzooxazol-2-ylamino]-2-morpholin-4-yl- benzonitrile; or a pharmaceutically acceptable salt thereof.
11. A compound of the formula I according to any one of claims 1 to 10 for use in the treatment of the animal, especially human, body.
12. A compound of the formula I according to claim 11 where the use is in the treatment of a JAK2 and/or Jak3 kinase mediated disease.
13. A pharmaceutical composition comprising a compound according to any one of claims 1 to 10 and a pharmaceutically acceptable carrier material.
14. A pharmaceutical composition according to claim 13 for use in the treatment of a JAK2 and/or JAK 3 kinase mediated disease.
15. The use of a compound of the formula I according to any one of claims 1 to 10 for the manufacture of a pharmaceutical preparation for use in the treatment of a JAK2- and/or JAK3-kinase mediated disease.
16. A process for the manufacture of a compound of the formula I according to any one of claims 1 to 10, comprising either a) reacting a compound of the formula II,
Figure imgf000150_0001
wherein X, R1, R3 and R4 are as defined for a compound of the formula I in claim 1 or any one or more of claims 2 to 7, and Hal is halo, especially bromo, under Suzuki coupling conditions with a boronic acid of the formula III,
R2-B(OH)2 (III) wherein R2 is as defined for a compound of the formula I, or a reactive derivative thereof, or b) reacting a compound of the formula II,
Figure imgf000151_0001
wherein X, R1, R3 and R4 are as defined for a compound of the formula I and Hal is halo, especially bromo, under Stille coupling conditions with an organotin compound of the formula
R -Sn(alk)3 (HI*)
wherein R is as defined for a compound of the formula I and alk is alkyl, preferably C1-C7- alkyl,
and, if desired, converting an obtainable compound of the formula I into a different compound of the formula I, converting an obtainable salt of a compound of the formula I into a different salt thereof, converting an obtainable free compound of the formula I into a salt thereof, and/or separating an obtainable isomer of a compound of the formula I from one or more different obtainable isomers of the formula I.
PCT/EP2007/007983 2006-09-15 2007-09-13 Benzoxazoles and oxazolopyridines being useful as janus kinases inhibitors WO2008031594A1 (en)

Priority Applications (16)

Application Number Priority Date Filing Date Title
JP2009527741A JP5325105B2 (en) 2006-09-15 2007-09-13 Benzoxazole and oxazolopyridine useful as Janus kinase inhibitors
MX2009002812A MX2009002812A (en) 2006-09-15 2007-09-13 Benzoxazoles and oxazolopyridines being useful as janus kinases inhibitors.
US12/440,298 US8629168B2 (en) 2006-09-15 2007-09-13 Benzoxazoles and oxazolopyridines being useful as janus kinases inhibitors
EA200900388A EA200900388A1 (en) 2006-09-15 2007-09-13 BENZOXASOLES AND OXAZOZOLYPIRIDINES APPLICABLE AS AN INHIBITOR KINAZ JANUS
CN2007800339907A CN101516860B (en) 2006-09-15 2007-09-13 Benzoxazoles and oxazolopyridines being useful as FANUS kinases inhibitors
CA002660987A CA2660987A1 (en) 2006-09-15 2007-09-13 Benzoxazoles and oxazolopyridines being useful as janus kinases inhibitors
AU2007296916A AU2007296916B2 (en) 2006-09-15 2007-09-13 Benzoxazoles and oxazolopyridines being useful as Janus kinases inhibitors
BRPI0716841-1A2A BRPI0716841A2 (en) 2006-09-15 2007-09-13 benzoxazoles and oxazolopyrindins being possible as janus kinase inhibitors
AT07802300T ATE532774T1 (en) 2006-09-15 2007-09-13 BENZOXAZOLES AND OXAZOLOPYRIDINES AS JANUS KINASE INHIBITORS
PL07802300T PL2066647T3 (en) 2006-09-15 2007-09-13 Benzoxazoles and oxazolopyridines being useful as janus kinases inhibitors
ES07802300T ES2377148T3 (en) 2006-09-15 2007-09-13 Benzoxazoles and oxazolopyridines useful as Janus kinase inhibitors
EP07802300A EP2066647B1 (en) 2006-09-15 2007-09-13 Benzoxazoles and oxazolopyridines being useful as janus kinases inhibitors
IL196860A IL196860A0 (en) 2006-09-15 2009-02-02 Benzoxazoles and oxazolopyridines being useful as janus kinases inhibitors
TN2009000070A TN2009000070A1 (en) 2006-09-15 2009-02-27 Benzoxazoles and oxazolopyridines being useful as janus kinases inhibitors
SM200900023T SMP200900023B (en) 2006-09-15 2009-04-09 Benzoxazoles and oxazolopyridines useful as inhibitors of kinase janus
NO20091469A NO20091469L (en) 2006-09-15 2009-04-15 Benzoxazoles and oxazole pyridines useful as Janus kinase inhibitors

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP06120733A EP1900729A1 (en) 2006-09-15 2006-09-15 Benzoxazoles and oxazolopyridines being useful as Janus kinases inhibitors
EP06120733.8 2006-09-15

Publications (1)

Publication Number Publication Date
WO2008031594A1 true WO2008031594A1 (en) 2008-03-20

Family

ID=37717673

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2007/007983 WO2008031594A1 (en) 2006-09-15 2007-09-13 Benzoxazoles and oxazolopyridines being useful as janus kinases inhibitors

Country Status (30)

Country Link
US (1) US8629168B2 (en)
EP (2) EP1900729A1 (en)
JP (1) JP5325105B2 (en)
KR (1) KR20090064389A (en)
CN (1) CN101516860B (en)
AR (1) AR062786A1 (en)
AT (1) ATE532774T1 (en)
AU (1) AU2007296916B2 (en)
BR (1) BRPI0716841A2 (en)
CA (1) CA2660987A1 (en)
CL (1) CL2007002669A1 (en)
CO (1) CO6150142A2 (en)
CR (1) CR10639A (en)
EA (1) EA200900388A1 (en)
ES (1) ES2377148T3 (en)
GT (1) GT200900056A (en)
IL (1) IL196860A0 (en)
MA (1) MA30723B1 (en)
MX (1) MX2009002812A (en)
NO (1) NO20091469L (en)
PA (1) PA8748101A1 (en)
PE (1) PE20080842A1 (en)
PL (1) PL2066647T3 (en)
PT (1) PT2066647E (en)
SM (1) SMP200900023B (en)
TN (1) TN2009000070A1 (en)
TW (1) TW200819444A (en)
UY (1) UY30587A1 (en)
WO (1) WO2008031594A1 (en)
ZA (1) ZA200900477B (en)

Cited By (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010018874A1 (en) * 2008-08-12 2010-02-18 Takeda Pharmaceutical Company Limited Amide compound
WO2010061971A1 (en) 2008-11-28 2010-06-03 興和株式会社 Pyridine-3-carboxyamide derivative
WO2011078360A1 (en) 2009-12-24 2011-06-30 武田薬品工業株式会社 Amide compound
CN103102359A (en) * 2011-11-15 2013-05-15 江苏先声药物研究有限公司 Pyrimidine compounds and application thereof
US8476282B2 (en) 2008-11-03 2013-07-02 Intellikine Llc Benzoxazole kinase inhibitors and methods of use
RU2503680C2 (en) * 2008-07-15 2014-01-10 Санофи-Авентис OXAZOLOPYRIMIDINES AS AGONISTS OF Edg-1 RECEPTOR
US8993580B2 (en) 2008-03-14 2015-03-31 Intellikine Llc Benzothiazole kinase inhibitors and methods of use
RU2557246C2 (en) * 2010-01-13 2015-07-20 Санофи 2,5,7-substituted oxazolpyrimidine derivatives
US9096611B2 (en) 2008-07-08 2015-08-04 Intellikine Llc Kinase inhibitors and methods of use
WO2015117147A1 (en) 2014-02-03 2015-08-06 Quadriga Biosciences, Inc. Beta-substituted beta-amino acids and analogs as chemotherapeutic agents
WO2015117146A1 (en) 2014-02-03 2015-08-06 Quadriga Biosciences, Inc. Beta-substituted gamma-amino acids and analogs as chemotherapeutic agents
US9359349B2 (en) 2007-10-04 2016-06-07 Intellikine Llc Substituted quinazolines as kinase inhibitors
WO2017024009A1 (en) 2015-08-03 2017-02-09 Quadriga Biosciences, Inc. Beta-substituted beta-amino acids and analogs as chemotherapeutic agents and uses thereof
US9724354B2 (en) 2013-03-22 2017-08-08 Millennium Pharmaceuticals, Inc. Combination of catalytic mTORC1/2 inhibitors and selective inhibitors of Aurora A kinase
WO2022251597A1 (en) 2021-05-28 2022-12-01 Verge Analytics, Inc. Methods of treating neurological disorders with modulators of ribosomal protein s6 kinase alpha-1 (rsk1) and ribosomal protein s6 kinase alpha-3 (rsk2)

Families Citing this family (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AR085397A1 (en) 2011-02-23 2013-09-25 Intellikine Inc COMBINATION OF QUINASA INHIBITORS AND THEIR USES
US8907093B2 (en) * 2011-07-07 2014-12-09 Sanofi Carboxylic acid derivatives having an oxazolo[4,5-c]pyridine ring
US8580816B2 (en) * 2011-07-07 2013-11-12 Sanofi Carboxylic acid derivatives having an oxazolo[5,4-b]pyridine ring
US9089574B2 (en) 2011-11-30 2015-07-28 Emory University Antiviral JAK inhibitors useful in treating or preventing retroviral and other viral infections
WO2014075318A1 (en) * 2012-11-19 2014-05-22 江苏先声药业有限公司 Pyrimidine compounds and use thereof
CN103664759A (en) * 2013-12-06 2014-03-26 常熟市联创化学有限公司 Preparation method of 3-hydroxy-2-nitropyridine
WO2016047678A1 (en) * 2014-09-25 2016-03-31 武田薬品工業株式会社 Heterocyclic compound

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004099204A1 (en) * 2003-05-09 2004-11-18 Astrazeneca Ab Imidazo and thiazolopyridines as jak3 kinase inhibitors
WO2007025897A2 (en) * 2005-09-02 2007-03-08 F. Hoffmann-La Roche Ag Benzooxazole, oxazolopyridine, benzothiazole and thiazolopyridine derivatives

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1195755C (en) * 1999-12-10 2005-04-06 辉瑞产品公司 Pyrrolo [2,3-d] pyrimidine compounds
DE10344223A1 (en) * 2003-09-24 2005-04-21 Merck Patent Gmbh New 2-anilino-1,3-benzoxazole derivatives, are inhibitors of kinases, especially tyrosine- or Raf-kinases, useful e.g. for treating solid tumors, angiogenesis, diabetic retinopathy, inflammation or psoriasis
GB0402140D0 (en) * 2004-01-30 2004-03-03 Smithkline Beecham Corp Novel compounds
EP1674467A1 (en) * 2004-12-22 2006-06-28 4Sc Ag 2,5- and 2,6-disubstituted benzazole derivatives useful as protein kinase inhibitors
JP2009544592A (en) * 2006-07-21 2009-12-17 ノバルティス アクチエンゲゼルシャフト 2,4-Di (arylamino) -pyrimidine-5-carboxamide compounds as JAK kinase inhibitors

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004099204A1 (en) * 2003-05-09 2004-11-18 Astrazeneca Ab Imidazo and thiazolopyridines as jak3 kinase inhibitors
WO2007025897A2 (en) * 2005-09-02 2007-03-08 F. Hoffmann-La Roche Ag Benzooxazole, oxazolopyridine, benzothiazole and thiazolopyridine derivatives

Cited By (29)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9359349B2 (en) 2007-10-04 2016-06-07 Intellikine Llc Substituted quinazolines as kinase inhibitors
US8993580B2 (en) 2008-03-14 2015-03-31 Intellikine Llc Benzothiazole kinase inhibitors and methods of use
US9637492B2 (en) 2008-03-14 2017-05-02 Intellikine Llc Benzothiazole kinase inhibitors and methods of use
US9828378B2 (en) 2008-07-08 2017-11-28 Intellikine Llc Kinase inhibitors and methods of use
US9096611B2 (en) 2008-07-08 2015-08-04 Intellikine Llc Kinase inhibitors and methods of use
RU2503680C2 (en) * 2008-07-15 2014-01-10 Санофи-Авентис OXAZOLOPYRIMIDINES AS AGONISTS OF Edg-1 RECEPTOR
WO2010018874A1 (en) * 2008-08-12 2010-02-18 Takeda Pharmaceutical Company Limited Amide compound
US8476431B2 (en) 2008-11-03 2013-07-02 Itellikine LLC Benzoxazole kinase inhibitors and methods of use
US8476282B2 (en) 2008-11-03 2013-07-02 Intellikine Llc Benzoxazole kinase inhibitors and methods of use
US9345706B2 (en) 2008-11-03 2016-05-24 Intellikine, Llc Benzoxazole kinase inhibitors and methods of use
WO2010061971A1 (en) 2008-11-28 2010-06-03 興和株式会社 Pyridine-3-carboxyamide derivative
WO2011078360A1 (en) 2009-12-24 2011-06-30 武田薬品工業株式会社 Amide compound
RU2557246C2 (en) * 2010-01-13 2015-07-20 Санофи 2,5,7-substituted oxazolpyrimidine derivatives
CN103102359A (en) * 2011-11-15 2013-05-15 江苏先声药物研究有限公司 Pyrimidine compounds and application thereof
US9724354B2 (en) 2013-03-22 2017-08-08 Millennium Pharmaceuticals, Inc. Combination of catalytic mTORC1/2 inhibitors and selective inhibitors of Aurora A kinase
US9861599B2 (en) 2014-02-03 2018-01-09 Quadriga Biosciences, Inc. Beta-substituted beta-amino acids and analogs as chemotherapeutic agents
WO2015117147A1 (en) 2014-02-03 2015-08-06 Quadriga Biosciences, Inc. Beta-substituted beta-amino acids and analogs as chemotherapeutic agents
US9394237B2 (en) 2014-02-03 2016-07-19 Quadriga Biosciences, Inc. β-substituted β-amino acids and analogs as chemotherapeutic agents
US9682921B2 (en) 2014-02-03 2017-06-20 Quadriga Biosciences, Inc. β-substituted γ-amino acids and analogs as chemotherapeutic agents
US9394236B2 (en) 2014-02-03 2016-07-19 Quadriga Biosciences, Inc. β-substituted γ-amino acids and analogs as chemotherapeutic agents
US10245246B2 (en) 2014-02-03 2019-04-02 Quadriga Biosciences, Inc. β-substituted β-amino acids and analogs as chemotherapeutic agents
WO2015117146A1 (en) 2014-02-03 2015-08-06 Quadriga Biosciences, Inc. Beta-substituted gamma-amino acids and analogs as chemotherapeutic agents
US10034847B2 (en) 2014-02-03 2018-07-31 Quadriga Biosciences, Inc. Beta-substituted beta-amino acids and analogs as chemotherapeutic agents
US9937139B2 (en) 2014-02-03 2018-04-10 Quadriga Biosciences, Inc. β-substituted γ-amino acids and analogs as chemotherapeutic agents
US10017459B2 (en) 2015-08-03 2018-07-10 Quadriga Biosciences, Inc. β-substituted β-amino acids and analogs as chemotherapeutic agents and uses thereof
WO2017024009A1 (en) 2015-08-03 2017-02-09 Quadriga Biosciences, Inc. Beta-substituted beta-amino acids and analogs as chemotherapeutic agents and uses thereof
US9783487B2 (en) 2015-08-03 2017-10-10 Quadriga Biosciences, Inc. Beta-substituted beta-amino acids and analogs as chemotherapeutic agents and uses thereof
US10246406B2 (en) 2015-08-03 2019-04-02 Quadriga Biosciences, Inc. Beta-substituted beta-amino acids and analogs as chemotherapeutic agents and uses thereof
WO2022251597A1 (en) 2021-05-28 2022-12-01 Verge Analytics, Inc. Methods of treating neurological disorders with modulators of ribosomal protein s6 kinase alpha-1 (rsk1) and ribosomal protein s6 kinase alpha-3 (rsk2)

Also Published As

Publication number Publication date
CN101516860A (en) 2009-08-26
EP1900729A1 (en) 2008-03-19
ES2377148T3 (en) 2012-03-22
NO20091469L (en) 2009-04-15
JP5325105B2 (en) 2013-10-23
CN101516860B (en) 2012-06-27
CA2660987A1 (en) 2008-03-20
EP2066647B1 (en) 2011-11-09
SMP200900023B (en) 2009-07-14
SMAP200900023A (en) 2009-05-11
CO6150142A2 (en) 2010-04-20
CL2007002669A1 (en) 2008-05-09
EP2066647A1 (en) 2009-06-10
US20100009978A1 (en) 2010-01-14
AR062786A1 (en) 2008-12-03
TN2009000070A1 (en) 2010-08-19
AU2007296916B2 (en) 2011-10-06
TW200819444A (en) 2008-05-01
PL2066647T3 (en) 2012-04-30
PT2066647E (en) 2012-01-17
MX2009002812A (en) 2009-03-31
MA30723B1 (en) 2009-09-01
EA200900388A1 (en) 2009-08-28
ATE532774T1 (en) 2011-11-15
UY30587A1 (en) 2008-05-02
AU2007296916A1 (en) 2008-03-20
BRPI0716841A2 (en) 2013-10-01
PE20080842A1 (en) 2008-08-11
ZA200900477B (en) 2010-01-27
KR20090064389A (en) 2009-06-18
PA8748101A1 (en) 2009-08-26
JP2010503629A (en) 2010-02-04
US8629168B2 (en) 2014-01-14
GT200900056A (en) 2011-11-08
CR10639A (en) 2009-07-03
IL196860A0 (en) 2009-11-18

Similar Documents

Publication Publication Date Title
EP2066647B1 (en) Benzoxazoles and oxazolopyridines being useful as janus kinases inhibitors
US7795273B2 (en) Pyrazolo[1,5-a]pyridine-3-carboxylic acids as EphB and VEGFR2 kinase inhibitors
KR101257158B1 (en) Derivatives of quinolines and quinoxalines as protein tyrosine kinase inhibitors
EP1687305B1 (en) 1h-imidazoquinoline derivatives as protein kinase inhibitors
EP2247591A1 (en) Pyrrolo [2, 3-d]pyridines and use thereof as tyrosine kinase inhibitors
KR20100019489A (en) Substituted imidazopyridazines as pi3k lipid kinase inhibitors
KR20090073121A (en) Pyrazolopyrimidines as pi3k lipid kinase inhibitors
EP1689747A1 (en) 1h-imidazo [4,5-c] quinoline derivatives in the treatment of protein kinase dependent diseases
KR20100016460A (en) 3,6-disubstituted-imidazo[1,2-b]pyridazines and 3,5-disubstituted pyrazolo[1,5-a]pyrimidines as phosphatidylinositol-3-kinase inhibitors
WO2009023978A1 (en) Pyrazolo [3,4 -d] pyrimidine compounds and their use as modulators of protein kinase
KR20120028882A (en) Heterocyclic compounds as mek inhibitors
US8389526B2 (en) 3-heteroarylmethyl-imidazo[1,2-b]pyridazin-6-yl derivatives
US20090275606A1 (en) Heterocyclic Compounds as MEK Inhibitors
US20120245209A1 (en) Heterocyclic sulfonamide derivatives

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 200780033990.7

Country of ref document: CN

121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 07802300

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 577/DELNP/2009

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: 574430

Country of ref document: NZ

Ref document number: 2007802300

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 196860

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: 2007296916

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2660987

Country of ref document: CA

Ref document number: 12009500318

Country of ref document: PH

WWE Wipo information: entry into national phase

Ref document number: CR2009-010639

Country of ref document: CR

ENP Entry into the national phase

Ref document number: 2007296916

Country of ref document: AU

Date of ref document: 20070913

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 12440298

Country of ref document: US

WWE Wipo information: entry into national phase

Ref document number: 09024347

Country of ref document: CO

WWE Wipo information: entry into national phase

Ref document number: 11158

Country of ref document: GE

WWE Wipo information: entry into national phase

Ref document number: 1020097005248

Country of ref document: KR

Ref document number: MX/A/2009/002812

Country of ref document: MX

ENP Entry into the national phase

Ref document number: 2009527741

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 200900388

Country of ref document: EA

WWE Wipo information: entry into national phase

Ref document number: DZP2009000207

Country of ref document: DZ

ENP Entry into the national phase

Ref document number: PI0716841

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20090316