WO2006075152A1 - 4- (1h-indol-3-yl) -pyrimidin-2-ylamine derivates and their use in therapy - Google Patents

4- (1h-indol-3-yl) -pyrimidin-2-ylamine derivates and their use in therapy Download PDF

Info

Publication number
WO2006075152A1
WO2006075152A1 PCT/GB2006/000087 GB2006000087W WO2006075152A1 WO 2006075152 A1 WO2006075152 A1 WO 2006075152A1 GB 2006000087 W GB2006000087 W GB 2006000087W WO 2006075152 A1 WO2006075152 A1 WO 2006075152A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
pyrimidin
indol
amine
formula
Prior art date
Application number
PCT/GB2006/000087
Other languages
French (fr)
Inventor
Peter Martin Fischer
Shudong Wang
Christopher Keith Meades
Martin James Inglis Andrews
Darren Gibson
Kenneth Duncan
Original Assignee
Cyclacel Limited
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Cyclacel Limited filed Critical Cyclacel Limited
Priority to EP06701760A priority Critical patent/EP1836194A1/en
Priority to US11/794,449 priority patent/US20090318446A1/en
Priority to MX2007008373A priority patent/MX2007008373A/en
Priority to AU2006205710A priority patent/AU2006205710A1/en
Priority to CA002592723A priority patent/CA2592723A1/en
Priority to JP2007549955A priority patent/JP2008526824A/en
Priority to BRPI0606313-6A priority patent/BRPI0606313A2/en
Publication of WO2006075152A1 publication Critical patent/WO2006075152A1/en
Priority to IL184313A priority patent/IL184313A0/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/14Drugs for dermatological disorders for baldness or alopecia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • 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
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/10Antimycotics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/20Antivirals for DNA viruses
    • A61P31/22Antivirals for DNA viruses for herpes viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P33/00Antiparasitic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P33/00Antiparasitic agents
    • A61P33/02Antiprotozoals, e.g. for leishmaniasis, trichomoniasis, toxoplasmosis
    • A61P33/06Antimalarials
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/48Drugs for disorders of the endocrine system of the pancreatic hormones
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/04Inotropic agents, i.e. stimulants of cardiac contraction; Drugs for heart failure
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/08Vasodilators for multiple indications
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/14Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing three or more hetero rings

Definitions

  • the present invention relates to substituted pyrimidine derivatives.
  • the invention relates to 4-(lH-indol-3-yl)-pyrimidin-2-ylamines and their use in therapy. More specifically, but not exclusively, the invention relates to compounds that are capable of inhibiting one or more protein kinases.
  • the eukaryotic protein kinase family is one of the largest in the human genome, comprising some 500 genes [1,2].
  • the majority of kinases contain a 250-300 amino acid residue catalytic domain with a conserved core structure. This domain comprises a binding pocket for ATP (less frequently GTP), whose terminal phosphate group the kinase transfers covalently to its macromolecular substrates.
  • the phosphate donor is always bound as a complex with a divalent ion (usually Mg 2+ or Mn 2+ ).
  • Another important function of the catalytic domain is the binding and orientation for phosphotransfer of the macromolecular substrate.
  • the catalytic domains present in most kinases are more or less homologous.
  • CDKs cyclin-dependent kinases
  • the present invention seeks to provide further substituted heteroaryl-substituted pyrimidine derivatives. More specifically, the invention relates to compounds that have broad therapeutic applications in the treatment of a number of different diseases and/or that are capable of inhibiting one or more protein kinases.
  • a first aspect of the invention relates to 4-(lH-indol-3-yl)-pyrimidin-2-ylamines. More specifically, the invention relates to compounds of formula I, or pharmaceutically acceptable salts thereof,
  • R 3 , R 4 , R 5 , R 6 , R 7 , R 8 , R 9 , and R 10 are each independently H, R 11 or R 12 ;
  • R 1 and R 2 are each independently H, R 11 or R 12 ; or R 1 and R 2 are linked to form a cyclic group together with the nitrogen to which they are attached, and wherein said cyclic group is optionally substituted with one or more R 11 or R 12 groups;
  • each R 11 is independently a hydrocarbyl group optionally substituted by one or more R 12 substituents;
  • each R 12 is independently selected from OR 13 , COR 13 , COOR 13 , CN, CONR 13 R 14 ,
  • each R 13 and each R 14 are independently H or (CHb) n R 15 , where n is O 5 1, 2, or 3; and each R 15 is independently selected from alkyl, cycloalkyl, aryl, heteroaryl, aryl and an alicyclic group; with the proviso that the compound is other than:
  • the present invention provides compounds that are capable of inhibiting various othe ⁇ protein kinases, including aurora kinase [75], FMS-like tyrosine kinase 3 (FLT3) [76], cyclin-dependent kinases (CDKs) [77], and glycogen synthase kinase 3 (GSK3) [78].
  • aurora kinase [75]
  • CDKs cyclin-dependent kinases
  • GSK3 glycogen synthase kinase 3
  • a second aspect of the invention relates to a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of formula I as defined above, or a pharmaceutically acceptable salt thereof, admixed with a pharmaceutically acceptable diluent, excipient or carrier.
  • a third aspect of the invention relates to the use of a compound of formula Ia, or a pharmaceutically acceptable salt thereof,
  • R 3 , R 4 , R 5 , R 6 , R 7 , R 8 , R 9 , and R 10 are each independently H 5 R 1 ⁇ or R 12 ;
  • R 1 and R 2 are each independently H, R 11 or R 12 ; or R 1 and R 2 are linked to form a cyclic group together with the nitrogen to which they are attached, and wherein said cyclic group is optionally substituted with one or more R 11 or R 12 groups; each R 11 is independently a hydrocarbyl group optionally substituted by one or more R 12 substituents; each R 12 is independently selected from OR 13 , COR 13 , COOR 13 , CN, CONR 13 R 14 ,
  • each R 13 and each R 14 are independently H or (CH 2 ) n R 15 , where n is 0, 1, 2, or 3; and each R 1S is independently selected from alkyl, cycloalkyl, aryl, heteroaryl, aryl and an alicyclic group; with the proviso that the compound is other than [4-(lH-indol-3-yl)-pyrimidin-2-yl]-[3-
  • R 3 , R 4 , R 5 , R 6 , R 7 , R 8 , R 9 , and R 10 are each independently H 5 R 11 or R 12 ;
  • R 1 and R 2 are each independently H, R 11 or R 12 ; or R 1 and R 2 are linked to form a cyclic group together with the nitrogen to which they are attached, and wherein said cyclic group is optionally substituted with one or more R 11 or R 12 groups; each R 11 is independently a hydrocarbyl group optionally substituted by one or more R 12 substituents; each R 12 is independently selected from OR 13 , COR 13 , COOR 13 , CN, CONR 13 R 14 ,
  • each R 13 and each R 14 are independently H or (CH 2 ) n R 15 , where n is 0, 1, 2, or 3; and each R 15 is independently selected from alkyl, cycloalkyl, aryl, heteroaryl, aryl and an alicyclic group; in the preparation of a medicament for treating one or more of the following: a viral disorder; a CNS disorder; a stroke; ; ' a microbial infection; a fungal disorder; a parasitic disorder; an inflammatory disorder; a cardiovascular disorder; alopecia; and diabetes.
  • Another aspect of the invention relates to the use of a compound of formula Ib as defined above, or a pharmaceutically acceptable salt thereof, in an assay for identifying further candidate compounds capable of inhibiting one or more of a cyclin dependent kinase, GSK, aurora kinase, FLT3 and a PLK enzyme.
  • Another aspect of the invention relates to compounds of formula I as defined above, or pharmaceutically acceptable salts thereof, for use in medicine.
  • a further aspect of the invention relates to a process for preparing compounds according to the invention.
  • hydrocarbyl refers to a group comprising at least C and H. If the hydrocarbyl group comprises more than one C then those carbons need not necessarily be linked to each other. For example, at least two of the carbons may be linked via a suitable element or group. Thus, the hydrocarbyl group may contain heteroatoms. Suitable heteroatoms will be apparent to those skilled in the art and include, for instance, sulphur, nitrogen, oxygen, phosphorus and silicon. Where the hydrocarbyl group contains one or more heteroatoms, the group may be linked via a carbon atom or via a heteroatom to another group, i.e. the linker atom may be a carbon or a heteroatom.
  • the hydrocarbyl group is an aryl, heteroaryl, alkyl, cycloalkyl, aralkyl, alicyclic, heteroalicyclic or alkenyl group. More preferably, the hydrocarbyl group is an aryl, heteroaryl, alkyl, cycloalkyl, aralkyl or alkenyl group.
  • the hydrocarbyl group may be optionally substituted
  • alkyl includes both saturated straight chain and branched alkyl groups which may be substituted (mono- or poly-) or unsubstituted.
  • the alkyl group is a C 1-2O alkyl group, more preferably a C 1 . ⁇ , more preferably still a C 1-12 alkyl group, more preferably still, a C 1-6 alkyl group, more preferably a C 1 ⁇ alkyl group.
  • Particularly preferred alkyl groups include, for example, methyl, ethyl, propyl, isopropyl, butyl, isobutyl, tert-butyl, pentyl and hexyl.
  • Suitable substituents include, for example, one or more R 12 groups.
  • the alkyl group is unsubstituted.
  • cycloalkyl refers to a cyclic alkyl group which may be substituted (mono- or poly-) or unsubstituted.
  • the cycloalkyl group is a C 3 . 12 cycloalkyl group.
  • Suitable substituents include, for example, one or more R 12 groups.
  • alkenyl refers to a group containing one or more carbon-carbon double bonds, which may be branched or unbranched, substituted (mono- or poly-) or unsubstituted.
  • the alkenyl group is a C 2 . 20 alkenyl group, more preferably a C 2 . 15 alkenyl group, more preferably still a C 2 . 12 alkenyl group, or preferably a C 2 . 6 alkenyl group, more preferably a C 2-3 alkenyl group.
  • Suitable substituents include, for example, one or more R 12 groups as defined above.
  • aryl refers to a C 6-12 aromatic group which may be substituted (mono- or poly-) or unsubstituted. Typical examples include phenyl and naphthyl etc. Suitable substituents include, for example, one or more R 12 groups. ' ⁇
  • heteroaryl refers to a C 2 . 12 aromatic, substituted (mono- or poly- ) or unsubstituted group, which comprises one or more heteroatoms.
  • the heteroaryl group is a C 4-12 aromatic group comprising one or more heteroatoms selected from N, O and S.
  • Suitable heteroaryl groups include pyrrole, pyrazole, pyrimidine, pyrazine, pyridine, quinoline, thiophene, 1,2,3-triazole, 1,2,4-triazole, thiazole, oxazole, iso-thiazole, iso-oxazole, imidazole, furan and the like.
  • suitable substituents include, for example, one or more R groups.
  • alicyclic refers to a cyclic aliphatic group which optionally contains one or more heteroatoms and which may be substituted (mono- or poly-) or unsubstituted.
  • the alicyclic group contains one or more heteroatoms and is thus a heteroalicylic group.
  • Preferred heteroalicyclic groups include piperidinyl, pyrrolidinyl, piperazinyl, thiomorpholinyl and morpholinyl.
  • the heteroalicyclic group is selected from N-piperidinyl, N-pyrrolidinyl, N-piperazinyl, N-thiomorpholinyl and N- morpholinyl.
  • suitable substituents include, for example, one or more R 12 groups.
  • aralkyl includes, but is not limited to, a group having both aryl and alkyl functionalities.
  • the term includes groups in which one of the hydrogen atoms of the alkyl group is replaced by an aryl group, e.g. a phenyl group optionally having one or more substituents such as halo, alkyl, alkoxy, hydroxy, and the like.
  • Typical aralkyl groups include benzyl, phenethyl and the like.
  • aryl-alicyclic includes, but is not limited to, a group having both aryl and alicyclic functionalities.
  • the term includes groups which contain an aryl functionality (for example, a phenyl group) fused to an alicylic group.
  • the alicylic group may contain one or more heteroatoms, i.e. it may be a heteroalicylic group.
  • One preferred embodiment of the invention relates to compounds of formula I, or pharmaceutically acceptable salts thereof, wherein R 3 , R 4 , R 5 , R 6 , R 7 , R 8 , R 9 , and R 10 are each independently H, R 11 or R 12 ; ⁇ : !
  • R 1 and R 2 are each independently H, R 11 or R 12 ; or R 1 and R 2 are linked to form a cyclic group together with the nitrogen to which they are attached, and wherein said cyclic group is optionally substituted with one or more R 11 or R 12 groups; each R 11 is independently a hydrocarbyl group optionally substituted by one or more R 12 substituents; each R 12 is independently selected from OR 13 , COR 13 , COOR 13 , CN, CONR 13 R 14 J
  • R 13 and R 14 are each independently H or (CH 2 ) n R 15 , where n is 0, 1, 2, or 3; and each R 15 is independently selected from alkyl, cycloalkyl, aryl, heteroaryl, aryl and an alicyclic group; with the proviso that the compound is other than:
  • R 1 and R 2 are each independently H, R 11 or R 12 ; or R 1 and R 2 are linked to form a cyclic group together with the nitrogen to which they are attached, wherein said cyclic group contains from two to nine carbon atoms and one or two heteroatoms selected from N, O, and S, and wherein said cyclic group is optionally substituted with one or two substituents selected from R 11 and R 12 .
  • R 1 and R 2 are each independently H, R 11 or R 12 .
  • R and R are each independently H or R 11 .
  • one of R 1 and R 2 is H and the other is R 11 .
  • R and R are both H.
  • R 11 is a hydrocarbyl group containing from 1 to 24 carbon atoms, optionally containing up to six heteroatoms selected from N, O, and S. More preferably, the hydrocarbyl group is optionally substituted by up to six R 12 substituents.
  • R 11 is an aryl group, a heteroaryl group, an aryl-alicyclic group or an alicyclic group, each of which may be optionally substituted by one or more R substituents.
  • R 11 is selected from phenyl, pyridinyl and
  • R 11 is an aryl, heteroaryl or alicyclic group, each of which may be optionally substituted by one or more R 1 substituents.
  • R 11 is a phenyl or pyridinyl group, each of which may be optionally substituted by one or more R substituents.
  • R 3 , R 4 , R 5 , R 6 , R 7 , R 8 , R 9 , and R 10 are each independently H or R 12 .
  • R 3 is H and R 4 is H or R 12 .
  • R 3 and R 4 are both H.
  • R 9 and R 10 are both H.
  • R 5 is H or alkyl, more preferably, H or Me.
  • R 6 is H, alkyl, CO-alkyl or CO-cycloalkyl, and is more preferably, H, Me 5 COMe or CO-cyclopropyl. More preferably still, R 6 is H.
  • R 7 is H, alkyl, alkoxy or halo, more preferably, H, Me, OMe or chloro.
  • R 8 is H, alkoxy or halo, more preferably, H, OMe or F.
  • R 5 , R 6 , R 7 , R 8 , R 9 , and R 10 are all H.
  • each R 15 is independently selected from methyl, ethyl, isopropyl, n-butyl, isobutyl, t-butyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, phenyl, pyridinyl, pyrrolidinyl, pyrrolyl, morpholinyl, piperazinyl, piperidinyl, triazolyl, tetrazolyl and thiazolyl. More preferably, each R 15 is alkyl or aryl.
  • R 15 is Me or phenyl, more preferably, Me.
  • the alicyclic group contains one or more heteroatoms.
  • R is an alicyclic group optionally substituted by one or more R 13 or COR 13 groups.
  • R 12 is a morpholinyl, piperazinyl, thiomorpholinyl or piperidinyl group optionally substituted by one or more R 13 or COR 13 groups.
  • R 12 is a morpholinyl, piperazinyl, thiomorpholinyl or piperidinyl group optionally substituted by one or more alkyl, aralkyl or CO-alkyl groups. More preferably, R 12 is a morpholinyl, piperazinyl, thiomorpholinyl or piperidinyl group optionally substituted by one or more methyl, benzyl or COMe groups.
  • R 12 is selected from the following:
  • each R 12 is independently selected from OR 13 , COR 13 , COOR 13 , CN, CONR 13 R 14 , NR 13 R 14 , SR 13 , SOR 13 , SO 2 R 13 , SO 2 OR 13 , SO 2 NR 13 R 14 , a heteroalicyclic group, halogen, CF 3 , and NO 2 .
  • each R 12 is independently selected from OH, OMe, COMe, CHO, CO 2 Me, COOH, CN, CONH 2 , NHMe, NH 2 , NMe 2 , SH, SMe, SOMe, SO 2 Me, SO 2 NHMe, SO 2 NH 2 , Cl, Br, F, I, CF 3 , NO 2 , N-morpholinyl, N-pyrrolidinyl and N-piperazinyl, N-thiomorpholinyl, 2,6-dimethylmorpholin-4-yl, 4-benzylpiperazin-l-yl, 3,5-dimethylpiperidin-l-yl and 4-acetylpiperazin-l-yl.
  • each R 12 is independently selected from OH, OMe, COMe, CHO, CO 2 Me, COOH, CN, CONH 2 , NHMe, NH 2 , NMe 2 , SH, SMe, SOMe, SO 2 Me, SO 2 NHMe, SO 2 NH 2 , Cl, Br, F, I, CF 3 , NO 2 , N-morpholinyl, N-pyrrolidinyl and N-piperazinyl.
  • R 12 is selected from
  • R 13 is (CEk) n R 15 where n is 0 or 1. More preferably, n is 0.
  • One particularly preferred embodiment of the invention relates to compounds of formula Ic, or pharmaceutically acceptable salts thereof,
  • R 3'10 are as defined above;
  • Z is N or CR 20 ;
  • R 16 - 20 are each independently H, R 11 or R 12 .
  • Z is N.
  • Z is CR 2Q
  • R 16"20 are each independently selected from H and R 12 as defined above.
  • R 16"20 are each independently selected from H, NO 2 , NR 13 R 14 , halogen, alkoxy and an optionally substituted heteroalicyclic group.
  • R 16"20 are each independently selected from H, NO 2 , halogen, alkoxy and a heteroalicyclic group.
  • R 16"20 are each independently selected from H, NO 2 , F, OMe, N-morpholinyl, NH 2 , N-pyrrolidinyl, N-piperazinyl, N-thiomorpholinyl, 2,6- dimethylmorpholin-4-yl, 4-benzylpiperazin-l-yl, 3,5-dimethyl-piperidin-l-yl and 4- acetylpiperazin-1 -yl.
  • R 16"20 are each independently selected from H, NO 2 , F, OMe and N- morpholinyl.
  • the compound of the invention is selected from the following:
  • the compound is selected from the following:
  • the compound of the invention is capable of inhibiting one or more protein kinases selected from CDKl/cyclin B, CDK2/cyclin A, CDK2/cyclin E, CDK4/cyclin Dl, CDK7/cyclin H, CDK9/cyclin Tl, GSK3 ⁇ , aurora kinase, FLT3 and PLKl, as measured by the appropriate assay.
  • protein kinases selected from CDKl/cyclin B, CDK2/cyclin A, CDK2/cyclin E, CDK4/cyclin Dl, CDK7/cyclin H, CDK9/cyclin Tl, GSK3 ⁇ , aurora kinase, FLT3 and PLKl
  • the compound of the invention exhibits an IC 50 value for kinase inhibition of less than about 10 ⁇ M, more preferably less than about 5 ⁇ M, more preferably less than about 1 ⁇ M, more preferably still less than about 0.5 ⁇ M, more preferably less than about 0.1 ⁇ M, even more preferably, less than about 0.01 ⁇ M.
  • IC 50 value for kinase inhibition of less than about 10 ⁇ M, more preferably less than about 5 ⁇ M, more preferably less than about 1 ⁇ M, more preferably still less than about 0.5 ⁇ M, more preferably less than about 0.1 ⁇ M, even more preferably, less than about 0.01 ⁇ M.
  • Compounds falling within each of these preferred embodiments can be identified from Tables 2 and 3, which show the IC 50 values for selected compounds of the invention. Details of the various kinase assays are disclosed in the accompanying Examples section. Compounds (12) and (13) are especially preferred in this regard.
  • the compound of the invention is capable of exhibiting an antiproliferative effect in human cell lines, as measured by a standard 72h MTT cytotoxicity assay.
  • the compound of the invention exhibits an IC 50 value of less than 10 ⁇ M, more preferably less than 5 ⁇ M, even more preferably less than 1 ⁇ M as measured by said MTT assay. More preferably still, the compound exhibits an IC 50 value' of less than 0.5 less ⁇ M, more preferably still less than 0.2 ⁇ M or 0.1 ⁇ M.
  • Compounds falling within each of these preferred embodiments can be identified from Table 4, which show the IC 50 values for selected compounds of the invention. Details of the standard 72h MTT cytotoxicity assay are set forth in the accompanying Examples section. Compound ( 12) is especially preferred in this regard.
  • the compounds of the present invention have been found to possess antiproliferative activity and are therefore believed to be of use in the treatment of proliferative disorders such as cancers, leukaemias and other disorders associated with uncontrolled cellular proliferation such as psoriasis and restenosis.
  • proliferative disorders such as cancers, leukaemias and other disorders associated with uncontrolled cellular proliferation such as psoriasis and restenosis.
  • an antiproliferative effect within the scope of the present invention may be demonstrated. by the ability to inhibit cell proliferation in an in vitro whole cell assay, for example using any of the cell lines A2780, Mia-PaCa-2, A549, HT29 or Saos-2. Using such assays it may be determined whether a compound is antiproliferative in the context of the present invention.
  • a preferred embodiment of the present invention therefore relates to the use of one or more compounds of formula Ia as defined above, or pharmaceutically acceptable salts thereof, in the preparation of a medicament for treating
  • One preferred embodiment of the invention relates to the use of a compound of formula Ia, or a pharmaceutically acceptable salt thereof, wherein R 3 , R 4 , R 5 , R 6 , R 7 , R 8 , R 9 , and R 10 are each independently H, R 11 or R 12 ;
  • R 1 and R 2 are each independently H, R 11 or R 12 ; or R 1 and R 2 are linked to form a cyclic group together with the nitrogen to which they are attached, and wherein said cyclic group is optionally substituted with one or more R 11 or R 12 groups; each R 11 is independently a hydrocarbyl group optionally substituted by one or more R 12 substituents; . . each R 12 is independently selected from OR 13 , COR 13 , COOR 13 , CN, CONR 13 R 14 ,
  • R 13 and R 14 are each independently H or (CH 2 ) n R 15 , where n is 0, 1, 2, or 3; and each R 15 is independently selected from alkyl, cycloalkyl, aryl, heteroaryl, aryl and an alicyclic group; with the proviso that the compound is other than [4-(lH-indol-3-yl)-pyrimidin-2-yl]-[3-
  • preparation of a medicament includes the use of a compound of the invention directly as the medicament in addition to its use in a screening programme for further therapeutic agents or in any stage of the manufacture of such a medicament.
  • the proliferative disorder is a cancer or leukaemia.
  • proliferative disorder is used herein in a broad sense to include any disorder that requires control of the cell cycle, for example cardiovascular disorders such as restenosis ⁇ cardiomyopathy and myocardial infarction, auto-immune disorders such as glomerulonephritis and rheumatoid arthritis, dermatological disorders such as psoriasis, anti-inflammatory, anti-fungal, antiparasitic disorders such as malaria, emphysema, alopecia, and chronic obstructive pulmonary disorder.
  • cardiovascular disorders such as restenosis ⁇ cardiomyopathy and myocardial infarction
  • auto-immune disorders such as glomerulonephritis and rheumatoid arthritis
  • dermatological disorders such as psoriasis, anti-inflammatory, anti-fungal, antiparasitic disorders such as malaria, emphysema, alopecia, and chronic obstructive pulmonary disorder.
  • the compounds of the present invention may induce apopto
  • the compounds of the invention may inhibit any of the steps or stages in the cell cycle, for example, formation of the nuclear envelope, exit from the quiescent phase of the cell cycle (GO), Gl progression, chromosome decondensation, nuclear envelope breakdown, START, initiation of DNA replication, progression of DNA replication, termination of DNA replication, centrosome duplication, G2 progression, activation of mitotic or meiotic functions, chromosome condensation, centrosome separation, microtubule nucleation, spindle formation and function, interactions with microtubule motor proteins, chromatid separation and segregation, inactivation of mitotic functions, formation of contractile ring, and cytokinesis functions.
  • the compounds of the invention may influence certain gene functions such as chromatin binding, formation of replication complexes, replication licensing, phosphorylation or other secondary modification activity, proteolytic degradation, microtubule binding, actin binding, septin binding, microtubule organising centre nucleation activity and binding to components of cell cycle signalling pathways.
  • the compound of the invention is administered in an amount sufficient to inhibit at least one CDK enzyme.
  • the CDK enzyme is CDKl, CDK2, CDK3, CDK4, CDK6, CDK7, CDK8 and/or CDK9.
  • the compound of the invention is administered in an amount sufficient to inhibit at least one of CDK2 and/or CDK4.
  • HCMV human cytomegalovirus
  • HSV-I herpes simplex virus type 1
  • HV-I human immunodeficiency virus type 1
  • VZV varicella zoster virus
  • the invention relates to the use of a compound of formula Ib, or a pharmaceutically acceptable salt thereof, as defined above, wherein R 3 , R 4 , R 5 , R 6 , R 7 , R 8 , R 9 , and R 10 are each independently H, R 1 J or R 12 ;
  • R 1 and R 2 are each independently H, R 11 or R 12 ; or R 1 and R 2 are linked to form a cyclic group together with the nitrogen to which they are attached, and wherein said cyclic group is optionally substituted with one or more R 11 or R 12 groups; each R 11 is independently a hydrocarbyl group optionally substituted by one or more R 12 substituents; each R 12 is independently selected from OR 13 , COR 13 , COOR 13 , CN, CONR 13 R 14 ,
  • R 13 and R 14 are each independently H or (CH 2 ) n R 15 , where n is 0, 1, 2, or 3; and each R 15 is independently selected from alkyl, cycloalkyl, aryl, heteroaryl, aryl and an alicyclic group; in the preparation of a medicament for treating one or more of the following: a viral disorder; a CNS disorder; a stroke; a microbial infection; a fungal disorder; a parasitic disorder; an inflammatory disorder; a cardiovascular disorder; alopecia; and diabetes.
  • the compound of the invention is administered in an amount sufficient to inhibit one or more of the host cell CDKs involved in viral replication, i.e. CDK2, CDK7, CDK8, and CDK9 [23].
  • an anti-viral effect within the scope of the present invention may be demonstrated by the ability to inhibit CDK2, CDK7, CDK8 or CDK9.
  • the invention relates to the use of one or more compounds of the invention in the treatment of a viral disorder which is CDK dependent or sensitive.
  • CDK dependent disorders are associated with an above normal level of activity of one or more CDK enzymes.
  • Such disorders preferably associated with an abnormal level of activity of CDK2, CDK7, CDK8 and/or CDK9.
  • a CDK sensitive disorder is a disorder in which an aberration in the CDK level is not the primary cause, but is downstream of the primary metabolic aberration.
  • CDK2, CDK7, CDK8 and/or CDK9 can be said to be part of the sensitive metabolic pathway and CDK inhibitors may therefore be active in treating such disorders.
  • a further aspect of the invention relates to a method of treating a CDK-dependent disorder, said method comprising administering to a subject in need thereof, a compound of formula Ia or Ib, or a pharmaceutically acceptable salt thereof, as defined above in an amount sufficient to inhibit a cyclin dependent kinase.
  • the CDK-dependent disorder is a viral disorder or a proliferative disorder, more preferably cancer.
  • the compound of the invention is administered in an amount sufficient to inhibit FLT3.
  • FLT3 is known to play an important role in the pathogenesis of acute myeloid leukemia [79].
  • the proliferative disorder is acute myeloid leukemia.
  • Another aspect of the invention relates to a method of treating a FLT3 -dependent disorder, said method comprising administering to a subject in need thereof, a compound of formula Ia or Ib, or a pharmaceutically acceptable salt thereof, in an amount sufficient to inhibit FLT3.
  • Another aspect of the invention relates to the use of a compound of formula Ib as defined above, or a pharmaceutically acceptable salt thereof, in the preparation of a medicament for treating diabetes.
  • the diabetes is type II diabetes.
  • GSK3 is one of several protein kinases that phosphorylate glycogen synthase (GS).
  • GS glycogen synthase
  • the stimulation of glycogen synthesis by insulin in skeletal muscle results from the dephosphorylation and activation of GS.
  • GSK3's action on GS thus results in the latter' s deactivation and thus suppression of the conversion of glucose into glycogen in muscles.
  • Type II diabetes non-insulin dependent diabetes mellitus
  • Hyperglycaemia is due to insulin resistance in the liver, muscles, and other tissues, coupled with impaired secretion of insulin.
  • Skeletal muscle is the main site for insulin-stimulated glucose uptake, there it is either removed from circulation or converted to glycogen..
  • Muscle glycogen deposition is the main determinant in glucose homeostasis and type II diabetics have defective muscle glycogen storage. There is evidence that an increase in GSK3 activity is important in type II diabetes [24]. Furthermore, it has been demonstrated that GSK3 is over-expressed in muscle cells of type II diabetics and that an inverse correlation exists between skeletal muscle GSK3 activity and insulin action [25].
  • GSK3 inhibition is therefore of therapeutic significance in the treatment of diabetes, particularly type II, and diabetic neuropathy.
  • GSK3 is known to phosphorylate many substrates other than GS, and is thus involved in the regulation of multiple biochemical pathways. For example, GSK is highly expressed in the central and peripheral nervous systems.
  • the compound is administered in an amount sufficient to inhibit GSK, more preferably GSK3, more preferably still GSK3 ⁇ .
  • Another aspect of the invention therefore relates to the use of a compound of formula Ib as defined above, or a pharmaceutically acceptable salt thereof, in the preparation of a medicament for treating a CNS disorder, for example neurodegenerative disorders.
  • the CNS disorder is Alzheimer's disease.
  • Tau is a GSK-3 substrate which has been implicated in the etiology of Alzheimer's disease, In healthy nerve cells, Tau co-assembles with tubulin into microtubules. However, in Alzheimer's disease, tau forms large tangles of filaments, which disrupt the microtubule structures in the nerve cell, thereby impairing the transport of nutrients as well as the transmission of neuronal messages.
  • GSK3 inhibitors may be able to prevent and/or reverse the abnormal hyperphosphorylation of the microtubule-associated protein tau that is an invariant feature of Alzheimer's disease and a number of other neurodegenerative diseases, such as progressive supranuclear palsy, corticobasal degeneration and Pick's disease. Mutations in the tau gene cause inherited forms of fronto- temporal dementia, further underscoring the relevance of tau protein dysfunction for the neurodegenerative process [26].
  • Another aspect of the invention therefore relates to the use of a compound of formula Ib as defined above, or a pharmaceutically acceptable salt thereof, in the preparation of a medicament for treating bipolar disorder.
  • Yet another aspect of the invention relates the use of a compound of formula Ib as defined above, or a pharmaceutically acceptable salt thereof, in the preparation of a medicament for treating a stroke.
  • GSK3 as a pro- apoptotic factor in neuronal cells makes this protein kinase an attractive ⁇ therapeutic target for the design of inhibitory drugs to treat these diseases.
  • Yet another aspect of the invention relates the use of a compound of formula Ib as defined above, or a pharmaceutically acceptable salt thereof, in the preparation of a medicament for treating alopecia.
  • a further aspect of the invention relates to a method of treating a GSK3 -dependent disorder, said method comprising administering to a subject in need thereof, a compound of formula Ib, or a pharmaceutically acceptable salt thereof, as defined above in an amount sufficient to inhibit GSK3.
  • the GSK3 -dependent disorder is diabetes.
  • the compound of the invention, or pharmaceutically acceptable salt thereof is administered in an amount sufficient to inhibit GSK3 ⁇ .
  • the compound of the invention is administered in an amount sufficient to inhibit at least one PLK enzyme.
  • PLKs polo-like kinases
  • the polo-like kinases (PLKs) constitute a family of serine/threonine protein kinases.
  • Mitotic Drosophila melanogaster mutants at the polo locus display spindle abnormalities [30] and polo was found to encode a mitotic kinase [31].
  • PLKs In humans, there exist three closely related PLKs [32]. They contain a highly homologous amino-terminal catalytic kinase domain and their carboxyl termini contain two or three conserved regions, the polo boxes. The function of the polo boxes remains incompletely understood but they are implicated in the targeting of PLKs to subcellular compartments [33,34], mediation of interactions with other proteins [35], or may constitute part of an autoregulatory domain [36]. Furthermore, the polo box-dependent PLKl activity is required for proper metaphase/anaphase transition and cytokinesis [37,38].
  • PLKs regulate some fundamental aspects of mitosis [39,40].
  • PLKl activity is believed to be necessary for the functional maturation of centrosomes in late G2/early prophase and subsequent establishment of a bipolar spindle.
  • Depletion of cellular PLKl through the small interfering RNA (siRNA) technique has also confirmed that this protein is required for multiple mitotic processes and completion of cytokinesis [41].
  • the compound of the invention is administered in an amount sufficient to inhibit PLKl.
  • PLKl is the best characterized; it regulates a number of cell division cycle effects, including the onset of mitosis [42,43], DNA-damage checkpoint activation [44,45], regulation of the anaphase promoting complex [46-48], phosphorylation of the proteasome [49], and centrosome duplication and maturation [50].
  • M-phase promoting factor the complex between the cyclin dependent kinase CDKl and B-type cyclins [51].
  • MPF M-phase promoting factor
  • CDC25C triggers the activation of MPF [52].
  • cyclin B localizes to the cytoplasm [53], it then becomes phosphorylated during prophase and this event causes nuclear translocation [54,55].
  • the nuclear accumulation of active MPF during prophase is thought to be important for initiating M-phase events [56].
  • nuclear MPF is kept inactive by WEEl unless counteracted by CDC25C.
  • the phosphatase CDC25C itself, localized to the cytoplasm during interphase, accumulates hi the nucleus in prophase [57- 59].
  • the nuclear entry of both cyclin B [60] and CDC25C [61] are promoted through phosphorylation by PLKl [43]. This kinase is an important regulator of M-phase initiation.
  • the compounds of the invention are ATP- antagonistic inhibitors of PLK 1.
  • ATP antagonism refers to the ability of an inhibitor compound to diminish or prevent PLK catalytic activity, i.e. phosphotransfer from ATP to a macromolecular PLK substrate, by virtue of reversibly or irreversibly binding at the enzyme's active site in such a manner as to impair or abolish ATP binding.
  • the compound of the invention is administered in an amount sufficient to inhibit PLK2 and/or PLK3.
  • PLK2 also known as SNK
  • PLK3 also known as PRK and FNK
  • SNK SNK
  • PLK3 kinase activity appears to peak during late S and G2 phase. It is also activated during DNA damage checkpoint activation and severe oxidative stress.
  • PLK3 also plays an important role in the regulation of microtubule dynamics and centrosome function in the cell and deregulated PLK3 expression results in cell cycle arrest and apoptosis [62].
  • PLK2 is the least well understood homologue of the three PLKs.
  • a further aspect of the invention relates to a method of treating a PLK-dependent disorder, said method comprising administering to a subject in need thereof, a compound of formula Ib, or a pharmaceutically acceptable salt thereof, as defined above in an amount sufficient to inhibit PLK.
  • the PLK-dependent disorder is a proliferative disorder, more preferably cancer.
  • the compound of the invention is administered in an amount sufficient to inhibit aurora kinase.
  • a further aspect of the invention relates to a method of treating an aurora kinase-dependent disorder, said method comprising administering to a subject in need thereof, a compound of formula Ib, or a pharmaceutically acceptable salt thereof, as defined above in an amount sufficient to inhibit aurora kinase.
  • the aurora kinase dependent disorder is a viral disorder as defined above.
  • Another aspect of the invention relates to a pharmaceutical composition
  • a pharmaceutical composition comprising one or more compounds of the invention as defined above admixed with one or more pharmaceutically acceptable diluents, excipients or carriers.
  • the compounds of the present invention can be administered alone, they will generally be administered in admixture with a pharmaceutical carrier, excipient or diluent, particularly for human therapy.
  • the pharmaceutical compositions may be for human or animal usage in human and veterinary medicine.
  • suitable carriers include lactose, starch, glucose, methyl cellulose, magnesium stearate, mannitol, sorbitol and the like.
  • suitable diluents include ethanol, glycerol and water.
  • compositions may comprise as, or in addition to, the carrier, excipient or diluent any suitable binder(s), lubricant(s), suspending agent(s), coating agent(s), solubilising agent(s).
  • Suitable binders include starch, gelatin, natural sugars such as glucose, anhydrous lactose, free-flow lactose, beta-lactose, corn sweeteners, natural and synthetic gums, such as acacia, tragacanth or sodium alginate, carboxymethyl cellulose and polyethylene glycol.
  • Suitable lubricants include sodium oleate, sodium stearate, magnesium stearate, sodium benzoate, sodium acetate, sodium chloride and the like.
  • Preservatives, stabilizers, dyes and even flavoring agents may be provided in the pharmaceutical composition.
  • preservatives include sodium benzoate, sorbic acid and esters of p-hydroxybenzoic acid.
  • Antioxidants and suspending agents may be also used.
  • the compounds of the invention can be present as salts or esters, in particular pharmaceutically acceptable salts or esters.
  • Pharmaceutically acceptable salts of the compounds of the invention include suitable acid addition or base salts thereof.
  • suitable pharmaceutical salts may be found in Berge et al, J Pharm Sci, 66, 1-19 (1977). Salts are formed, for example with strong inorganic acids such as mineral acids, e.g.
  • sulphuric acid, phosphoric acid or hydrohalic acids with strong organic carboxylic acids, such as alkanecarboxylic acids of 1 to 4 carbon atoms which are unsubstituted or substituted (e.g., by halogen), such as acetic acid; with saturated or unsaturated dicarboxylic acids, for example oxalic, malonic, succinic, maleic, fumaric, phthalic or tetraphthalic; with hydroxycarboxylic acids, for example ascorbic, glycolic, lactic, malic, tartaric or citric acid; with aminoacids, for example aspartic or glutamic acid; with benzoic acid; or with organic sulfonic acids, such as or aryl-sulfonic acids which are unsubstituted or substituted (for example, by a halogen) such as methane- or p-toluene sulfonic acid.
  • organic carboxylic acids such as alkanecarboxylic
  • Esters are formed either using organic acids or alcohols/hydroxides, depending on the functional group being esterified.
  • Organic acids include carboxylic acids, such as alkanecarboxylic acids of 1 to 12 carbon atoms which are unsubstituted or substituted (e.g;-; by halogen), such as acetic acid; with saturated or unsaturated dicarboxylic acid, for example oxalic, malonic, succinic, maleic, fumaric, phthalic or tetraphthalic; with hydroxycarboxylic acids, for example ascorbic, glycolic, lactic, malic, tartaric or citric acid; with aminoacids, for example aspartic or glutamic acid; with benzoic acid; or with organic sulfonic acids, such as (C 1 -C 4 )-alkyl- or aryl-sulfonic acids which are unsubstituted or substituted (for example, by a halogen) such as methane- or p-
  • Suitable hydroxides include inorganic hydroxides, such as sodium hydroxide, potassium hydroxide, calcium hydroxide, aluminium hydroxide.
  • Alcohols include alkanealcohols of 1-12 carbon atoms which may be unsubstituted or substituted, e.g. by a halogen).
  • the invention includes, where appropriate all enantiomers and tautomers of compounds of the invention.
  • the man skilled in the art will recognise compounds that possess an optical properties (one or more chiral carbon atoms) or tautomeric characteristics.
  • the corresponding enantiomers and/or tautomers may be isolated/prepared by methods known in the art.
  • Some of the compounds of the invention may exist as stereoisomers and/or geometric isomers - e.g. they may possess one or more asymmetric and/or geometric centres and so may exist in two or more stereoisomeric and/or geometric forms.
  • the present invention contemplates the use of all the individual stereoisomers and geometric isomers of those agents, and mixtures thereof.
  • the terms used in the claims encompass these forms, provided said forms retain the appropriate functional activity (though not necessarily to the same degree).
  • the compounds of the invention may exist in cis or trans forms, either in isolated form, or as mixtures thereof in any ratio.
  • the compounds of the invention contain morpholinyl or piperidinyl substituents
  • the methyl groups on the morpholinyl and piperidinyl rings can be either cis or trans.
  • the present invention also includes all suitable isotopic variations of the agent of pharmaceutically acceptable salt thereof.
  • An isotopic variation of an agent of the present invention or a pharmaceutically acceptable salt thereof is defined as one in which at least one atom is replaced by an atom having the same atomic number but an atomic mass different from the atomic mass usually found in nature.
  • isotopes that can be incorporated into the agent and pharmaceutically acceptable salts thereof include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulphur, fluorine and chlorine such as 2 H, 3 H, 13 C, 14 C, 15 N, 17 O, 18 0, 31 P, 32 P, 35 S, 18 F and 36 Cl, respectively.
  • isotopic variations of the agent and pharmaceutically acceptable salts thereof are useful in drug and/or substrate tissue distribution studies. Tritiated, i.e., 3 H, and carbon-14, i.e., 14 C, isotopes are particularly preferred for their ease of preparation and detectability. Further, substitution with isotopes such as deuterium, i.e., 2 H 5 may afford certain therapeutic advantages resulting from greater metabolic stability, for example, increased in vivo half-life or reduced dosage requirements and hence may be preferred in some circumstances. Isotopic variations of the agent of the present invention and pharmaceutically acceptable salts thereof of this invention can generally be prepared by conventional procedures using appropriate isotopic variations of suitable reagents.
  • the present invention also includes the use of solvate forms of the compounds of the present invention.
  • the terms used in the claims encompass these forms.
  • the invention furthermore relates to the compounds of the present invention in their various crystalline forms, polymorphic forms and (an)hydrous forms. It is well established within the pharmaceutical industry that chemical compounds may be isolated in any of such forms by slightly varying the method of purification and or isolation form the solvents used in the synthetic preparation of such compounds.
  • the invention further includes the compounds of the present invention in prodrug form.
  • prodrugs are generally compounds of the invention wherein one or more appropriate groups have been modified such that the modification may be reversed upon administration to a human or mammalian subject.
  • Such reversion is usually performed by an enzyme naturally present in such subject, though it is possible for a second agent to be administered together with such a prodrug in order to perform the reversion in vivo.
  • Examples of such modifications include ester (for example, any of those described above) ' , wherein the reversion may be carried out be an esterase etc.
  • Other such systems will be well known to those skilled in the art.
  • compositions of the present invention may be adapted for oral, rectal, vaginal, parenteral, intramuscular, intraperitoneal, intraarterial, intrathecal, intrabronchial, subcutaneous, intradermal, intravenous, nasal, buccal or sublingual routes of administration.
  • compositions For oral administration, particular use is made of compressed tablets, pills, tablets, gellules, drops, and capsules. Preferably, these compositions contain from 1 to 250 mg and more preferably from 10-100 mg, of active ingredient per dose.
  • compositions of the present invention may also be in form of suppositories, pessaries, suspensions, emulsions, lotions, ointments, creams, gels, sprays, solutions or dusting powders.
  • the active ingredient can be incorporated into a cream consisting of an aqueous emulsion of polyethylene glycols or liquid paraffin.
  • the active ingredient can also be incorporated, at a concentration of between 1 and 10% by weight, into an ointment consisting of a white wax or white soft paraffin base together with such stabilisers and preservatives as may be required.
  • Injectable forms may contain between 10-1000 mg, preferably between 10-250 mg, of active ingredient per dose.
  • compositions may be formulated in unit dosage form, i.e., in the form of discrete portions containing a unit dose, or a multiple or sub-unit of a unit dose.
  • a person of ordinary skill in the art can easily determine an appropriate dose of one of the instant compositions to administer to a subject without undue experimentation.
  • a physician will determine the actual dosage which will be most suitable for an individual patient and it will depend on a variety of factors including the activity of the specific compound employed, the metabolic stability and length of action of that compound, the age, body weight, general health, sex, diet, mode and time of administration, rate of excretion, drug combination, the severity of the particular condition, and the individual undergoing therapy.
  • the dosages disclosed herein are exemplary of the average case. There can of course be individual instances where higher or lower dosage ranges are merited, and such are within the scope of this invention.
  • the agent may be administered at a dose of from 0.01 to 30 mg/kg body weight, such as from 0.1 to 10 mg/kg, more preferably from 0.1 to 1 mg/kg body weight.
  • one or more doses of 10 to 150 mg/day will be administered to the patient.
  • the one or more compounds of the invention are administered in combination with one or more other therapeutically active agents, for example, existing drugs available on the market.
  • the compounds of the invention may be administered consecutively, simultaneously or sequentially with the one or more other active agents.
  • anticancer drugs in general are more effective when used in combination.
  • combination therapy is desirable in order to avoid an overlap of major toxicities, mechanism of action and resistance mechanism(s).
  • the major advantages of combining chemotherapeutic drugs are that it may promote additive or possible synergistic effects through biochemical interactions and also may decrease the emergence of resistance in early tumor cells which would have been otherwise responsive to initial chemotherapy with a single agent.
  • Beneficial combinations may be suggested by studying the growth inhibitory activity of the test compounds with agents known or suspected of being valuable in the treatment of a particular cancer initially or cell lines derived from that cancer. This procedure can also be used to determine the order of administration of the agents, i.e. before, simultaneously; or after delivery. Such scheduling may be a feature of all the cycle acting agents identified herein.
  • Another aspect of the invention relates to the use of a compound of formula Ib as defined above, or a pharmaceutically acceptable salt thereof, in an assay for identifying further candidate compounds capable of inhibiting one or more protein kinases.
  • Another aspect of the invention relates to the use of a compound of the invention, or a pharmaceutically acceptable salt thereof, in an assay for identifying further candidate compounds capable of inhibiting one or more cyclin dependent kinases, aurora kinase, GSK, FLT3 and PLK.
  • the assay is a competitive binding assay.
  • the competitive binding assay comprises contacting a compound of the invention with a protein kinase and a candidate compound and detecting any change in the interaction between the compound of the invention and the protein kinase.
  • One aspect of the invention relates to a process comprising the steps of:
  • Another aspect of the invention provides a process comprising the steps of:
  • Another aspect of the invention provides a process comprising the steps of:
  • the invention also relates to a ligand identified by the method described hereinabove.
  • Yet another aspect of the invention relates to a pharmaceutical composition
  • a pharmaceutical composition comprising a ligand identified by the method described hereinabove.
  • Another aspect of the invention relates to the use of a ligand identified by the method described hereinabove in the preparation of a pharmaceutical composition for use in the treatment of proliferative disorders, viral disorders, a CNS disorder, stroke, alopecia and diabetes.
  • said candidate compound is generated by conventional SAR modification of a compound of the invention.
  • conventional SAR modification refers to standard methods known in the art for varying a given compound by way of chemical derivatisation.
  • the above methods may be used to screen for a ligand useful as an inhibitor of one or more protein kinases.
  • iV-Unsubstituted 1-H-indoles II may be acylated with acid anhydride or acid halide derivatives of R 4 CH 2 COOH at C-3 to afford the 3-acyl-lH-indole products III ([69], pp. 262-263). If R 6 is other than H, this substituent is next introduced, followed by acylation with appropriate carbonyl compounds containing the group R 3 , to provide the intermediate 1,3-dicarbonyl compounds IV. These can be condensed directly with guanidines VI; alternatively they are first converted to the enaminones V 5 from which 4-(lH-Indol-3-yl)- pyrimidin-2-ylamines I can be obtained [70].
  • a further aspect of the invention therefore relates to a process for preparing a compound of formula Ib as defined above, said process comprising the steps of:
  • the compound of formula IV is prepared by acylating a compound of formula III
  • the compound of formula III is prepared by acylating a compound of formula II with an acid anhydride or acid halide derivative OfR 4 CH 2 COOH
  • said compound of formula III is prepared by a process which comprises treating a compound of formula II as defined above with (i) zinc chloride and ethylmagnesium bromide, and (ii) acetyl chloride.
  • NMR spectra were recorded using a Varian INOVA-500 instrument. Chemical shifts are reported in parts per million relative to internal tetramethylsilane standard. Mass spectra were obtained using a Waters ZQ2000 single quadrupole mass spectrometer with electrospray ionization (ESI). Analytical and preparative RP-HPLC was performed using Vydac 218TP54 (250 x 4.6 mm) and 218TP1022 (250 x 22 mm) columns, respectively. Linear gradient elution using H 2 OMeCN systems (containing 0.1 % CF 3 COOH) at flow rates of 1 mL/min (analytical) and 9 mL/min (preparative) was performed.
  • This compound was the result of a cyclisation with a guanidine containing a 4:1 cis:trans ratio of diastereoisomers.
  • the early fractions from Prep-HPLC contained a 20:1 cisitrans mixture (Batch 01) and later fractions had a 1 :1 mixture (Batch 02).
  • the assay data refers to Batch 01.
  • the tram isomer was obtained from Prep-HPLC (0.1% TFA) on batch 02 of Compound
  • the 4:1 cis:trans mixture obtained was the result of a cyclisation with a guanidine containing a 4:1 cis:trans ratio of diastereoisomers.
  • the compounds from the examples above were investigated for their ability to inhibit the enzymatic activity of various protein kinases. This was achieved by measurement of incorporation of radioactive phosphate from ATP into appropriate polypeptide substrates. Recombinant protein kinases and kinase complexes were produced or obtained commercially. Assays were performed using 96-well plates and appropriate assay buffers (typically 25 mM ⁇ -glycerophosphate, 20 mM MOPS, 5 mM EGTA, 1 mM DTT, 1 mM Na 3 VO 3 , pH 7.4), into which were added 2 - 4 ⁇ g of active enzyme with appropriate substrates.
  • assay buffers typically 25 mM ⁇ -glycerophosphate, 20 mM MOPS, 5 mM EGTA, 1 mM DTT, 1 mM Na 3 VO 3 , pH 7.4
  • the reactions were initiated by addition of Mg/ATP mix (15 mM MgCl 2 + 100 ⁇ M ATP with 30-50 kBq per well of [ ⁇ - 32 P]-ATP) and mixtures incubated as required at 30 0 C. Reactions were stopped on ice, followed by filtration through p81 filterplates or GF/C filterplates (Whatman Polyfiltronics, Kent, UK). After washing 3 times with 75 mM aq orthophosphoric acid, plates were dried, scintillant added and incorporated radioactivity measured in a scintillation counter (TopCount, Packard Instruments, Pangbourne, Berks, UK).
  • CTD peptide substrate biotinyl-Ahx-(Tyr-Ser-Pro-Thr-Ser-Pro-Ser) 4 -NH 2 ; 1 - 2 mg/mL
  • recombinant human CDK7/cyclin H, CDK9/cyclin Tl, or CDK9/cyclin K (0.5 - 2 ⁇ g) were incubated for 45 min at 30 °C in the presence of varying amounts of test compound in 20 mM MOPS pH 7.2, 25mM ⁇ -glycerophosphate, 5 roM EGTA, 1 mM DTT, ImM sodium vanadate, 15 mM MgCl 2 , and 100 ⁇ M ATP (containing a trace amount of 32 PyATP) in a total volume of 25 ⁇ L in a 96-well microtiter plate.
  • the reactions were initiated by addition of MgATP mix (15mM MgCl 2 + lOO ⁇ M ATP with 15-25 kBq per well of [7- 32 P]-ATP) and mixtures incubated for 30 min at 30°C. Reactions were stopped by addition of an equal volume of 75 mM aq orthophosphoric acid, followed by filtration through p81 f ⁇ lterplates (Whatman Polyfiltronics, Kent, UK). After washing 4 times with 75 mM aq orthophosphoric acid, plates were dried, scintillant added and incorporated radioactivity measured in a scintillation counter (TopCount, Packard Instruments, Pangbourne, Berks, UK).
  • the reactions were initiated by addition of MgATP mix (15mM MgCl 2 + lOO ⁇ M ATP with 15-25 kBq per well of [ ⁇ - 32 P]-ATP) and mixtures incubated for 60 min at 3O 0 C. Reactions were stopped by addition of an equal volume of 75 mM aq orthophosphoric acid, followed by filtration through p81 filterplates (Whatman Polyfiltronics, Kent, UK). After washing 4 times with 75 mM aq orthophosphoric acid, plates were dried, scintillant added and incorporated radioactivity measured in a scintillation counter (TopCount, Packard Instruments, Pangbourne, Berks, UK).
  • Aurora-B human, Upstate, Dundee, UK
  • kinase assay in appropriate buffers (2OmM Tris, 25mM ⁇ -glycerophosphate, 5mM EGTA, ImM DTT, ImM sodium vanadate, pH 7.5) by incubating 15 ⁇ g of enzyme with 4 ⁇ g INCENP (Upstate, Dundee, UK) in the presence of MgATP mix (15mM MgCl 2 + lOO ⁇ M ATP) for
  • the reactions were initiated by addition of MgATP mix (15mM MgCl 2 + lOO ⁇ M ATP with 15-25 kBq per well of [ ⁇ - 32 P]-ATP) and mixtures incubated for 30 min at 30°C. Reactions were stopped by addition of an equal volume of 75 mM aq orthophosphoric acid, followed by filtration through p81 filterplates (Whatman Polyfiltronics, Kent, UK). After washing 4 times with 75 mM aq orthophosphoric acid, plates were dried, scintillant added and incorporated radioactivity measured in a scintillation counter (TopCount, Packard Instruments, Pangbourne, Berks, UK).
  • GSK-3 was obtained from New England Biolabs (UK) Ltd., Hitchin, Herts.
  • the recombinant enzyme was isolated from a strain of E. coli that carries a clone expressing GSK-3 ⁇ derived from a rabbit skeletal muscle cDNA library [Wang, Q.M.; Fiol, CJ.; DePaoli-Roach, A. A.; Roach, PJ. J. Biol. Chem., 1994, 269, 14566].
  • Inhibition of GSK-3 function was assessed by measurement of phosphorylation of CREB phosphopeptide KRREILSRRPphosphoSYR in the presence of test compounds.
  • GSK3 (7.5U) was incubated for 30 min at 30 0 C in a total volume of 25 ⁇ L in 20 mM MOPS pH 7.2, 25 mM ⁇ -glycerophosphate, 5 mM EGTA, 1 mM DTT, 1 mM Na 3 VO 3 , 40 ⁇ M CREB peptide, 15 mM MgCl 2 and 100 ⁇ M ATP (containing 0.25 ⁇ Ci [ ⁇ - 32 P]-ATP) in the presence of varying concentrations of test compound.
  • the compounds of the invention were subjected to a standard cellular proliferation assay using human tumour cell lines obtained from the ATCC (American Type Culture Collection, 10801 University Boulevard, Manessas, VA 20110-2209, USA).
  • Standard 72-h MTT thiazolyl blue; 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide assays were performed (Haselsberger, K.; Peterson, D. C; Thomas, D. G.; Darling, J. L. Anti Cancer Drugs 1996, 7, 331-8; Loveland, B. E.; Johns, T. G.; Mackay, I. R.; Vaillant, F.; Wang, Z. X.; Hertzog, P.
  • MTT dye was solubilised with 200 ⁇ L per well of DMSO with agitation. Absorbance was read at 540 nm and data analysed using curve-fitting software (GraphPad Prism version 3.00 for Windows, GraphPad Software, San Diego California USA) to determine IC5 0 values (concentration of test compound which inhibits cell growth by 50 %). Results for representative example compounds are summarised in Table 4.
  • Table 4 MTT Cytoxicity data for cell lines Mia-PaCa-2 and A2780 (IC 50 values in ⁇ M after 96 hours incubation) for selected compounds of the invention

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Oncology (AREA)
  • Virology (AREA)
  • Communicable Diseases (AREA)
  • Cardiology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Diabetes (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Neurosurgery (AREA)
  • Neurology (AREA)
  • Biomedical Technology (AREA)
  • Molecular Biology (AREA)
  • Rheumatology (AREA)
  • Dermatology (AREA)
  • Endocrinology (AREA)
  • Hematology (AREA)
  • Hospice & Palliative Care (AREA)
  • Urology & Nephrology (AREA)
  • Vascular Medicine (AREA)
  • Pain & Pain Management (AREA)
  • Biotechnology (AREA)
  • Obesity (AREA)
  • Emergency Medicine (AREA)
  • Psychiatry (AREA)
  • Immunology (AREA)
  • Orthopedic Medicine & Surgery (AREA)

Abstract

The present invention relates to compounds of formula (I), or pharmaceutically acceptable salts thereof. The present invention seeks to provide further substituted heteroaryl-substituted pyrimidine derivatives. More specifically, the invention relates to compounds that have broad therapeutic applications in the treatment of a number of different diseases and/or that are capable of inhibiting one or more protein kinases.

Description

4- (lH-IND0L-3-YL.) -PYRIMIDIN-2-YLAMINE DBRIVATES AND THEIR USE IN THERAPY
The present invention relates to substituted pyrimidine derivatives. In particular, the invention relates to 4-(lH-indol-3-yl)-pyrimidin-2-ylamines and their use in therapy. More specifically, but not exclusively, the invention relates to compounds that are capable of inhibiting one or more protein kinases.
BACKGROUND TO THE INVENTION
In eukaryotes, all biological functions, including DNA replication, cell cycle progression, energy metabolism, and cell growth and differentiation, are regulated through the reversible phosphorylation of proteins. The phosphorylation state of a protein determines not only its function, subcellular distribution, and stability, but also what other proteins or cellular components it associates with. The balance of specific phosphorylation in the proteome as a whole, as well as of individual members in a biochemical pathway, is thus used by organisms as a strategy to maintain homeostasis in response to an ever-changing environment [71]. The enzymes that carry out these phosphorylation and dephosphorylation steps are protein kinases and phosphatases, respectively. Many kinases have gained importance as drug discovery targets in a variety of therapeutic areas [72]. '
The eukaryotic protein kinase family is one of the largest in the human genome, comprising some 500 genes [1,2]. The majority of kinases contain a 250-300 amino acid residue catalytic domain with a conserved core structure. This domain comprises a binding pocket for ATP (less frequently GTP), whose terminal phosphate group the kinase transfers covalently to its macromolecular substrates. The phosphate donor is always bound as a complex with a divalent ion (usually Mg2+ or Mn2+). Another important function of the catalytic domain is the binding and orientation for phosphotransfer of the macromolecular substrate. The catalytic domains present in most kinases are more or less homologous. A wide variety of molecules capable of inhibiting protein kinase function through antagonising ATP binding are known in the art [3-7]. By way of example, the applicant has previously disclosed 2-anilino-4-heteroaryl-pyrimidine compounds with kinase inhibitory properties, particularly against cyclin-dependent kinases (CDKs) [8-12]. CDKs are serine/threonine protein kinases that associate with various cyclin subunits. These complexes are important for the regulation of eukaryotic cell cycle progression, but also for the regulation of transcription [13,14].
The present invention seeks to provide further substituted heteroaryl-substituted pyrimidine derivatives. More specifically, the invention relates to compounds that have broad therapeutic applications in the treatment of a number of different diseases and/or that are capable of inhibiting one or more protein kinases.
STATEMENT OF INVENTION
A first aspect of the invention relates to 4-(lH-indol-3-yl)-pyrimidin-2-ylamines. More specifically, the invention relates to compounds of formula I, or pharmaceutically acceptable salts thereof,
Figure imgf000003_0001
I
wherein R3, R4, R5, R6, R7, R8, R9, and R10 are each independently H, R11 or R12; R1 and R2 are each independently H, R11 or R12; or R1 and R2 are linked to form a cyclic group together with the nitrogen to which they are attached, and wherein said cyclic group is optionally substituted with one or more R11 or R12 groups; each R11 is independently a hydrocarbyl group optionally substituted by one or more R12 substituents; each R12 is independently selected from OR13, COR13, COOR13, CN, CONR13R14,
NR13R14, SR13, SOR13, SO2R13, SO2OR13, SO2NR13R14, R13, halogen, CF3, NO2 and an alicyclic group itself optionally substituted by one or more R12 or R13 groups; and each R13 and each R14 are independently H or (CHb)nR15, where n is O5 1, 2, or 3; and each R15 is independently selected from alkyl, cycloalkyl, aryl, heteroaryl, aryl and an alicyclic group; with the proviso that the compound is other than:
[4-(lH-indol-3-yl)-pyrimidin-2-yl]-[3-(l,l,2,2-tetrafluoroethoxyphenyl)]-amine;
3-[6-(4-bromophenyl)-2-(l-piperazinyl)-4-pyrimidinyl]-lH-indole;
3 -[6-(4-bromophenyl)-2-( 1 -pyrrolidinyl)-4-pyrimidinyl] - 1 H-indole; or
3-[6-(4-bromophenyl)-2-(4-morpholinyl)-4-pyrimidinyl]-lH-indole.
Several 4-(lH-indol-3-yl)-pyrimidin-2-ylamine compounds are known in the art [73]. However, to date, the only such compound reported to inhibit kinase activity is [4-(1H- indol-3-yl)-pyrimidin-2-yl]-[3-(l,l,2,2-tetrafluoro-ethoxy)-phenyl]-amine [74], which was shown to inhibit PKC-α, PKC-δ and EGF-R.
The present invention provides compounds that are capable of inhibiting various othe^ protein kinases, including aurora kinase [75], FMS-like tyrosine kinase 3 (FLT3) [76], cyclin-dependent kinases (CDKs) [77], and glycogen synthase kinase 3 (GSK3) [78].
A second aspect of the invention relates to a pharmaceutical composition comprising a compound of formula I as defined above, or a pharmaceutically acceptable salt thereof, admixed with a pharmaceutically acceptable diluent, excipient or carrier.
A third aspect of the invention relates to the use of a compound of formula Ia, or a pharmaceutically acceptable salt thereof,
Figure imgf000005_0001
Ia
wherein R3, R4, R5, R6, R7, R8, R9, and R10 are each independently H5 R1 λ or R12;
R1 and R2 are each independently H, R11 or R12; or R1 and R2 are linked to form a cyclic group together with the nitrogen to which they are attached, and wherein said cyclic group is optionally substituted with one or more R11 or R12 groups; each R11 is independently a hydrocarbyl group optionally substituted by one or more R12 substituents; each R12 is independently selected from OR13, COR13, COOR13, CN, CONR13R14,
NR13R14, SR13, SOR13, SO2R13, SO2OR13, SO2NR13R14, R13, halogen, CF3, NO2 and an alicyclic group itself optionally substituted by one or more R12 or R13 groups; and each R13 and each R14 are independently H or (CH2)nR15, where n is 0, 1, 2, or 3; and each R1S is independently selected from alkyl, cycloalkyl, aryl, heteroaryl, aryl and an alicyclic group; with the proviso that the compound is other than [4-(lH-indol-3-yl)-pyrimidin-2-yl]-[3-
(1,1 ,2,2-tetrafluoroethoxyphenyl)] -amine; in the preparation of a medicament for treating a proliferative disorder.
Further aspects of the invention relate to the use of a compound of formula Ib, or a pharmaceutically acceptable salt thereof,
Figure imgf000006_0001
Ib
wherein R3, R4, R5, R6, R7, R8, R9, and R10 are each independently H5 R11 or R12;
R1 and R2 are each independently H, R11 or R12; or R1 and R2 are linked to form a cyclic group together with the nitrogen to which they are attached, and wherein said cyclic group is optionally substituted with one or more R11 or R12 groups; each R11 is independently a hydrocarbyl group optionally substituted by one or more R12 substituents; each R12 is independently selected from OR13, COR13, COOR13, CN, CONR13R14,
NR13R14, SR13, SOR13, SO2R13, SO2OR13, SO2NR13R14, R13, halogen, CF3, NO2 and an alicyclic group itself optionally substituted by one or more R12 or R13 groups; and each R13 and each R14 are independently H or (CH2)nR15, where n is 0, 1, 2, or 3; and each R15 is independently selected from alkyl, cycloalkyl, aryl, heteroaryl, aryl and an alicyclic group; in the preparation of a medicament for treating one or more of the following: a viral disorder; a CNS disorder; a stroke; ; ' a microbial infection; a fungal disorder; a parasitic disorder; an inflammatory disorder; a cardiovascular disorder; alopecia; and diabetes.
Another aspect of the invention relates to the use of a compound of formula Ib as defined above, or a pharmaceutically acceptable salt thereof, in an assay for identifying further candidate compounds capable of inhibiting one or more of a cyclin dependent kinase, GSK, aurora kinase, FLT3 and a PLK enzyme.
Another aspect of the invention relates to compounds of formula I as defined above, or pharmaceutically acceptable salts thereof, for use in medicine.
A further aspect of the invention relates to a process for preparing compounds according to the invention.
DETAILED DESCRIPTION
For the avoidance of doubt, the preferred embodiments described hereinafter refer to all aspects of the present invention.
As used herein, the term "hydrocarbyl" refers to a group comprising at least C and H. If the hydrocarbyl group comprises more than one C then those carbons need not necessarily be linked to each other. For example, at least two of the carbons may be linked via a suitable element or group. Thus, the hydrocarbyl group may contain heteroatoms. Suitable heteroatoms will be apparent to those skilled in the art and include, for instance, sulphur, nitrogen, oxygen, phosphorus and silicon. Where the hydrocarbyl group contains one or more heteroatoms, the group may be linked via a carbon atom or via a heteroatom to another group, i.e. the linker atom may be a carbon or a heteroatom. Preferably, the hydrocarbyl group is an aryl, heteroaryl, alkyl, cycloalkyl, aralkyl, alicyclic, heteroalicyclic or alkenyl group. More preferably, the hydrocarbyl group is an aryl, heteroaryl, alkyl, cycloalkyl, aralkyl or alkenyl group. The hydrocarbyl group may be optionally substituted
10 by one or more R groups. As used herein, the term "alkyl" includes both saturated straight chain and branched alkyl groups which may be substituted (mono- or poly-) or unsubstituted. Preferably, the alkyl group is a C1-2O alkyl group, more preferably a C1.^, more preferably still a C1-12 alkyl group, more preferably still, a C1-6 alkyl group, more preferably a C1^ alkyl group. Particularly preferred alkyl groups include, for example, methyl, ethyl, propyl, isopropyl, butyl, isobutyl, tert-butyl, pentyl and hexyl. Suitable substituents include, for example, one or more R12 groups. Preferably, the alkyl group is unsubstituted.
As used herein, the term "cycloalkyl" refers to a cyclic alkyl group which may be substituted (mono- or poly-) or unsubstituted. Preferably, the cycloalkyl group is a C3.12 cycloalkyl group. Suitable substituents include, for example, one or more R12 groups.
As used herein, the term "alkenyl" refers to a group containing one or more carbon-carbon double bonds, which may be branched or unbranched, substituted (mono- or poly-) or unsubstituted. Preferably the alkenyl group is a C2.20 alkenyl group, more preferably a C2. 15 alkenyl group, more preferably still a C2.12 alkenyl group, or preferably a C2.6 alkenyl group, more preferably a C2-3 alkenyl group. Suitable substituents include, for example, one or more R12 groups as defined above.
As used herein, the term "aryl" refers to a C6-12 aromatic group which may be substituted (mono- or poly-) or unsubstituted. Typical examples include phenyl and naphthyl etc. Suitable substituents include, for example, one or more R12 groups. ' ■
As used herein, the term "heteroaryl" refers to a C2.12 aromatic, substituted (mono- or poly- ) or unsubstituted group, which comprises one or more heteroatoms. Preferably, the heteroaryl group is a C4-12 aromatic group comprising one or more heteroatoms selected from N, O and S. Suitable heteroaryl groups include pyrrole, pyrazole, pyrimidine, pyrazine, pyridine, quinoline, thiophene, 1,2,3-triazole, 1,2,4-triazole, thiazole, oxazole, iso-thiazole, iso-oxazole, imidazole, furan and the like. Again, suitable substituents include, for example, one or more R groups. As used herein, the term "alicyclic" refers to a cyclic aliphatic group which optionally contains one or more heteroatoms and which may be substituted (mono- or poly-) or unsubstituted. Preferably, the alicyclic group contains one or more heteroatoms and is thus a heteroalicylic group. Preferred heteroalicyclic groups include piperidinyl, pyrrolidinyl, piperazinyl, thiomorpholinyl and morpholinyl. More preferably, the heteroalicyclic group is selected from N-piperidinyl, N-pyrrolidinyl, N-piperazinyl, N-thiomorpholinyl and N- morpholinyl. Again, suitable substituents include, for example, one or more R12 groups.
As used herein, the term "aralkyl" includes, but is not limited to, a group having both aryl and alkyl functionalities. By way of example, the term includes groups in which one of the hydrogen atoms of the alkyl group is replaced by an aryl group, e.g. a phenyl group optionally having one or more substituents such as halo, alkyl, alkoxy, hydroxy, and the like. Typical aralkyl groups include benzyl, phenethyl and the like.
As used herein, the term "aryl-alicyclic" includes, but is not limited to, a group having both aryl and alicyclic functionalities. By way of example the term includes groups which contain an aryl functionality (for example, a phenyl group) fused to an alicylic group. The alicylic group may contain one or more heteroatoms, i.e. it may be a heteroalicylic group.
One preferred embodiment of the invention relates to compounds of formula I, or pharmaceutically acceptable salts thereof, wherein R3, R4, R5, R6, R7, R8, R9, and R10 are each independently H, R11 or R12; ■ : !
R1 and R2 are each independently H, R11 or R12; or R1 and R2 are linked to form a cyclic group together with the nitrogen to which they are attached, and wherein said cyclic group is optionally substituted with one or more R11 or R12 groups; each R11 is independently a hydrocarbyl group optionally substituted by one or more R12 substituents; each R12 is independently selected from OR13, COR13, COOR13, CN, CONR13R14J
NR13R14, SR13, SOR13, SO2R13, SO2OR13, SO2NR13R14, an alicyclic group, halogen, CF3,- and NO2; and R13 and R14 are each independently H or (CH2)nR15, where n is 0, 1, 2, or 3; and each R15 is independently selected from alkyl, cycloalkyl, aryl, heteroaryl, aryl and an alicyclic group; with the proviso that the compound is other than:
[4-(lH-indol-3-yl)-pyrimidin-2-yl]-[3-(14,2,2-tetrafluoroemoxyphenyl)]-arnine;
3-[6-(4-bromophenyl)-2-(l-piperazinyl)-4-pyrimidinyl]-lH-indole;
3-[6-(4-bromophenyl)-2-(l-pyπolidinyl)-4-pyrirnidinyl]-lH-indole; or
3-[6-(4-bromophenyl)-2-(4-morpholinyl)-4-pyrimidinyl]-lH-indole.
In one preferred embodiment of the invention, R1 and R2 are each independently H, R11 or R12; or R1 and R2 are linked to form a cyclic group together with the nitrogen to which they are attached, wherein said cyclic group contains from two to nine carbon atoms and one or two heteroatoms selected from N, O, and S, and wherein said cyclic group is optionally substituted with one or two substituents selected from R11 and R12.
In one preferred embodiment of the invention, R1 and R2 are each independently H, R11 or R12.
1 0
In a more preferred embodiment of the invention, R and R are each independently H or R11.
In one particularly preferred embodiment of the invention, one of R1 and R2 is H and the other is R11.
In another particularly preferred embodiment, R and R are both H.
Preferably, R11 is a hydrocarbyl group containing from 1 to 24 carbon atoms, optionally containing up to six heteroatoms selected from N, O, and S. More preferably, the hydrocarbyl group is optionally substituted by up to six R12 substituents.
In one preferrred embodiment, R11 is an aryl group, a heteroaryl group, an aryl-alicyclic group or an alicyclic group, each of which may be optionally substituted by one or more R substituents.
In one preferrred embodiment, R11 is selected from phenyl, pyridinyl and
Figure imgf000011_0001
each of which may be optionally substituted by one or more R substituents.
In one preferrred embodiment, R11 is an aryl, heteroaryl or alicyclic group, each of which may be optionally substituted by one or more R1 substituents.
In an even more preferrred embodiment, R11 is a phenyl or pyridinyl group, each of which may be optionally substituted by one or more R substituents.
In one preferrred embodiment, R3, R4, R5, R6, R7, R8, R9, and R10 are each independently H or R12.
Preferably, R3 is H and R4 is H or R12.
Preferably, R3 and R4 are both H.
Preferably, R9 and R10 are both H.
In another preferred embodiment, R5 is H or alkyl, more preferably, H or Me. In another preferred embodiment, R6 is H, alkyl, CO-alkyl or CO-cycloalkyl, and is more preferably, H, Me5 COMe or CO-cyclopropyl. More preferably still, R6 is H.
In another preferred embodiment, R7 is H, alkyl, alkoxy or halo, more preferably, H, Me, OMe or chloro.
In another preferred embodiment, R8 is H, alkoxy or halo, more preferably, H, OMe or F.
In another preferred, embodiment, R5, R6, R7, R8, R9, and R10 are all H.
In one preferrred embodiment, each R15 is independently selected from methyl, ethyl, isopropyl, n-butyl, isobutyl, t-butyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, phenyl, pyridinyl, pyrrolidinyl, pyrrolyl, morpholinyl, piperazinyl, piperidinyl, triazolyl, tetrazolyl and thiazolyl. More preferably, each R15 is alkyl or aryl.
In one highly preferred embodiment, R15 is Me or phenyl, more preferably, Me.
In one preferrred embodiment, the alicyclic group contains one or more heteroatoms.
10
In one preferrred embodiment, R is an alicyclic group optionally substituted by one or more R13 or COR13 groups.
In a more preferrred embodiment, R12 is a morpholinyl, piperazinyl, thiomorpholinyl or piperidinyl group optionally substituted by one or more R13 or COR13 groups.
In an even more preferrred embodiment, R12 is a morpholinyl, piperazinyl, thiomorpholinyl or piperidinyl group optionally substituted by one or more alkyl, aralkyl or CO-alkyl groups. More preferably, R12 is a morpholinyl, piperazinyl, thiomorpholinyl or piperidinyl group optionally substituted by one or more methyl, benzyl or COMe groups.
More preferably still, R12 is selected from the following:
Figure imgf000013_0001
In one preferrred embodiment, each R12 is independently selected from OR13, COR13, COOR13, CN, CONR13R14, NR13R14, SR13, SOR13, SO2R13, SO2OR13, SO2NR13R14, a heteroalicyclic group, halogen, CF3, and NO2.
In a more preferrred embodiment, each R12 is independently selected from OH, OMe, COMe, CHO, CO2Me, COOH, CN, CONH2, NHMe, NH2, NMe2, SH, SMe, SOMe, SO2Me, SO2NHMe, SO2NH2, Cl, Br, F, I, CF3, NO2, N-morpholinyl, N-pyrrolidinyl and N-piperazinyl, N-thiomorpholinyl, 2,6-dimethylmorpholin-4-yl, 4-benzylpiperazin-l-yl, 3,5-dimethylpiperidin-l-yl and 4-acetylpiperazin-l-yl.
In an even more preferrred embodiment, each R12 is independently selected from OH, OMe, COMe, CHO, CO2Me, COOH, CN, CONH2, NHMe, NH2, NMe2, SH, SMe, SOMe, SO2Me, SO2NHMe, SO2NH2, Cl, Br, F, I, CF3, NO2, N-morpholinyl, N-pyrrolidinyl and N-piperazinyl.
In one highly preferred embodiment, R12 is selected from
Figure imgf000014_0001
NO2, F, OMe, NMe2 and Me.
In one preferred embodiment, R13 is (CEk)nR15 where n is 0 or 1. More preferably, n is 0.
One particularly preferred embodiment of the invention relates to compounds of formula Ic, or pharmaceutically acceptable salts thereof,
Figure imgf000014_0002
Ic wherein
R3'10 are as defined above;
Z is N or CR20; and
R16-20 are each independently H, R11 or R12.
In one preferrred embodiment, Z is N.
In another preferrred embodiment, Z is CR 2Q In one preferrred embodiment, R16"20 are each independently selected from H and R12 as defined above.
In one particularly preferrred embodiment, R16"20 are each independently selected from H, NO2, NR13R14, halogen, alkoxy and an optionally substituted heteroalicyclic group.
In a more preferrred embodiment, R16"20 are each independently selected from H, NO2, halogen, alkoxy and a heteroalicyclic group.
In one preferrred embodiment, R16"20 are each independently selected from H, NO2, F, OMe, N-morpholinyl, NH2, N-pyrrolidinyl, N-piperazinyl, N-thiomorpholinyl, 2,6- dimethylmorpholin-4-yl, 4-benzylpiperazin-l-yl, 3,5-dimethyl-piperidin-l-yl and 4- acetylpiperazin-1 -yl.
More preferably, R16"20 are each independently selected from H, NO2, F, OMe and N- morpholinyl.
In one especially preferred embodiment, the compound of the invention is selected from the following:
4-(lH-indol-3-yl)-N-(3-nitrophenyl)pyrimidin-2-amine;
N-(4-fluorophenyl)-4-( 1 H-indol-3 -yl)pyrimidin-2-amine;
4-( 1 H-indol-3 -yl)-N-(6-methoxypyridin-3 -y l)pyrimidin-2-amine;
4-(lH-indol-3-yl)-N-(4-morpholin-4-ylphenyl)pyrimidin-2-amine;
4-(lH-indol-3-yl)-N-(4-acetylpiperazin-l-ylphenyl)pyrimidin-2-amine;
4-( 1 H-indol-3 -yl)-N-(4-piperazin- 1 -ylphenyl)pyrimidin-2-amine;
4-(lH-indol-3-yl)-N-(4-benzylpiperazin-l-ylphenyl)pyrimidin-2-amine;
4-(lH-indol-3-yl)-N-(2,6-dimethylmorpholin-4-ylphenyl)pyrimidin-2-amine;
Nl-[4-(lH-indol-3-yl)pyrimidin-2-yl]-N,N-dimethylbenzene-l,4-diamine;
4-(lH-indol-3-yl)-N-(2-methyl-4-morpholin-4-ylphenyl)pyrimidin-2-amine;
4-(lH-indol-3-yl)-N-(3,4,5-trimethoxyphenyl)pyrimidin-2-amine;
4-(lH-indol-3-yl)-N-(3-methoxyl-4-morpholin-4-ylphenyl)pyrimidin-2-amine;
N-(3,5-dimethoxyphenyl)-4-(lH-indol-3-yl)pyrimidin-2-amine; 4-(l-methyl-lH-indol-3-yl)-N-(4-moφholin-4-ylphenyl)pyrimidin-2-a-nine;
4-( 1 -methyl- 1 H-indol-3 -yl)-N-(4-acetylpiperazine- 1 -ylphenyl)pyrimidin-2-amine;
N- 1 ,3 -benzodioxol-5-yl-4-( 1 H-indol-3 -yl)pyrimidin-2-amine;
4-[l-(cyclopropylcarbonyl)-lH-indol-3-yl]-N-(4-morpholin-4ylphenyl)pyrimidin-2-amine;
4-(l-acetyl-lH4ndol-3-yl)-N-(4-morpholin-4ylphenyl)pyrimidin-2-amine;
4-(lH-indol-3-yl)-N-(4-methylpiperazin-l-ylphenyl)pyrimidin-2-amine;
4-(7-methoxy-lH-indol-3-yl)-N-(4-acetylpiperazin-l-ylphenyl)pyrimidin-2-amine;
4-(2-me1hyl-lH-indol-3-yl)-N-(4-acetylpiperazin-l-ylphenyl)]pyrimidin-2-amine;
4-(7-methyl-lH-indol-3-yl)-N-(4-acetylpiperazin-l-ylphenyl)]pyrimidin-2-amine;
4-(6-methoxy-lH-indol-3-yl)-N-(4-acetylpiperazin-l-ylphenyl)]pyriπύdin-2-amine;
4-(7-chloro- 1 H-indol-3 -yl)-N-(4-acetylpiperazin- 1 -ylphenyl)]pyrimidin-2-amine;
4-(6-fluoro-lH-indol-3-yl)-N-(4-acetylpiperazin-l-ylphenyl)]pyrimidin-2-amine;
4-(lH-indol-3-yl)-N-[(4-acetylpiperazin-l-yl)-3-methylphenyl]pyriinidin-2-arιiine;
4-(lH-indol-3-yl)-N-(3-methyl-4-1iiiomorpholin-4-ylphenyl)pyrimidin-2-amine;
4-(lH-indol-3-yl)-N-[(2R,6S)-2,6-dimetiiylmoφholin-4-ylphenyl]pyrimidin-2-amine;
4-(lH-indol-3-yl)-N-[(2S,6S)-2,6-dimethylmorpholin-4-ylphenyl]pyrimidin-2-amine;
4-(lH-indol-3-yl)-N-(3,5-dimethylpiperidin-l-ylphenyl)pyrimidin-2-amine; and
4-( 1 H-indol-3 -yl)pyrimidin-2-amine.
In one particularly preferred embodiment, the compound is selected from the following:
4-(lH-Indol-3-yl)-pyrimidin-2-ylamine; [4-(lH-Indol-3-yl)-pyrimidin-2-yl]-(3-nitro-phenyl)-amine; (4-Fluoro-phenyl)- [4-( 1 H-indol-3 -yl)-pyrimidin-2-yl] -amine; [4-(lH-Indol-3-yl)-pyrimidin-2-yl]-(6-methoxy-pyridin-3-yl)-amine; and [4-(l H-Indol-3 -y l)-pyrimidin-2-yl] -(4-morpholin-4-yl-phenyl)-amine .
In one preferred embodiment, the compound of the invention is capable of inhibiting one or more protein kinases selected from CDKl/cyclin B, CDK2/cyclin A, CDK2/cyclin E, CDK4/cyclin Dl, CDK7/cyclin H, CDK9/cyclin Tl, GSK3β, aurora kinase, FLT3 and PLKl, as measured by the appropriate assay. In one particularly preferred embodiment, the compound of the invention exhibits an IC50 value for kinase inhibition of less than about 10 μM, more preferably less than about 5 μM, more preferably less than about 1 μM, more preferably still less than about 0.5 μM, more preferably less than about 0.1 μM, even more preferably, less than about 0.01 μM. Compounds falling within each of these preferred embodiments can be identified from Tables 2 and 3, which show the IC50 values for selected compounds of the invention. Details of the various kinase assays are disclosed in the accompanying Examples section. Compounds (12) and (13) are especially preferred in this regard.
In one preferred embodiment the compound of the invention is capable of exhibiting an antiproliferative effect in human cell lines, as measured by a standard 72h MTT cytotoxicity assay. Preferably, the compound of the invention exhibits an IC50 value of less than 10 μM, more preferably less than 5 μM, even more preferably less than 1 μM as measured by said MTT assay. More preferably still, the compound exhibits an IC50 value' of less than 0.5 less μM, more preferably still less than 0.2 μM or 0.1 μM. Compounds falling within each of these preferred embodiments can be identified from Table 4, which show the IC50 values for selected compounds of the invention. Details of the standard 72h MTT cytotoxicity assay are set forth in the accompanying Examples section. Compound ( 12) is especially preferred in this regard.
THERAPEUTIC USE
The compounds of the present invention have been found to possess antiproliferative activity and are therefore believed to be of use in the treatment of proliferative disorders such as cancers, leukaemias and other disorders associated with uncontrolled cellular proliferation such as psoriasis and restenosis. As defined herein, an antiproliferative effect within the scope of the present invention may be demonstrated. by the ability to inhibit cell proliferation in an in vitro whole cell assay, for example using any of the cell lines A2780, Mia-PaCa-2, A549, HT29 or Saos-2. Using such assays it may be determined whether a compound is antiproliferative in the context of the present invention. A preferred embodiment of the present invention therefore relates to the use of one or more compounds of formula Ia as defined above, or pharmaceutically acceptable salts thereof, in the preparation of a medicament for treating a proliferative disorder.
One preferred embodiment of the invention relates to the use of a compound of formula Ia, or a pharmaceutically acceptable salt thereof, wherein R3, R4, R5, R6, R7, R8, R9, and R10 are each independently H, R11 or R12;
R1 and R2 are each independently H, R11 or R12; or R1 and R2 are linked to form a cyclic group together with the nitrogen to which they are attached, and wherein said cyclic group is optionally substituted with one or more R11 or R12 groups; each R11 is independently a hydrocarbyl group optionally substituted by one or more R12 substituents; . . each R12 is independently selected from OR13, COR13, COOR13, CN, CONR13R14,
NR13R14, SR13, SOR13, SO2R13, SO2OR13, SO2NR13R14, an alicyclic group, halogen, CF3, and NO2; and :
R13 and R14 are each independently H or (CH2)nR15, where n is 0, 1, 2, or 3; and each R15 is independently selected from alkyl, cycloalkyl, aryl, heteroaryl, aryl and an alicyclic group; with the proviso that the compound is other than [4-(lH-indol-3-yl)-pyrimidin-2-yl]-[3-
(1,1 ,2,2-tetrafluoroethoxyphenyl)]-amine; in the preparation of a medicament for treating a proliferative disorder.
As used herein the phrase "preparation of a medicament" includes the use of a compound of the invention directly as the medicament in addition to its use in a screening programme for further therapeutic agents or in any stage of the manufacture of such a medicament.
Preferably, the proliferative disorder is a cancer or leukaemia.
The term proliferative disorder is used herein in a broad sense to include any disorder that requires control of the cell cycle, for example cardiovascular disorders such as restenosis^ cardiomyopathy and myocardial infarction, auto-immune disorders such as glomerulonephritis and rheumatoid arthritis, dermatological disorders such as psoriasis, anti-inflammatory, anti-fungal, antiparasitic disorders such as malaria, emphysema, alopecia, and chronic obstructive pulmonary disorder. In these disorders, the compounds of the present invention may induce apoptosis or maintain stasis within the desired cells as required.
The compounds of the invention may inhibit any of the steps or stages in the cell cycle, for example, formation of the nuclear envelope, exit from the quiescent phase of the cell cycle (GO), Gl progression, chromosome decondensation, nuclear envelope breakdown, START, initiation of DNA replication, progression of DNA replication, termination of DNA replication, centrosome duplication, G2 progression, activation of mitotic or meiotic functions, chromosome condensation, centrosome separation, microtubule nucleation, spindle formation and function, interactions with microtubule motor proteins, chromatid separation and segregation, inactivation of mitotic functions, formation of contractile ring, and cytokinesis functions. In particular, the compounds of the invention may influence certain gene functions such as chromatin binding, formation of replication complexes, replication licensing, phosphorylation or other secondary modification activity, proteolytic degradation, microtubule binding, actin binding, septin binding, microtubule organising centre nucleation activity and binding to components of cell cycle signalling pathways.
In one embodiment of the invention, the compound of the invention is administered in an amount sufficient to inhibit at least one CDK enzyme. Preferably, the CDK enzyme is CDKl, CDK2, CDK3, CDK4, CDK6, CDK7, CDK8 and/or CDK9.
More preferably, the compound of the invention is administered in an amount sufficient to inhibit at least one of CDK2 and/or CDK4.
Another aspect of the invention relates to the use of a compound of formula Ib as defined above, or a pharmaceutically acceptable salt thereof, in the preparation of a medicament for treating a viral disorder. Preferably, the viral disorder is selected from human cytomegalovirus (HCMV), herpes simplex virus type 1 (HSV-I), human immunodeficiency virus type 1 (HFV-I) and varicella zoster virus (VZV). ■
In one preferred embodiment, the invention relates to the use of a compound of formula Ib, or a pharmaceutically acceptable salt thereof, as defined above, wherein R3, R4, R5, R6, R7, R8, R9, and R10 are each independently H, R1 J or R12;
R1 and R2 are each independently H, R11 or R12; or R1 and R2 are linked to form a cyclic group together with the nitrogen to which they are attached, and wherein said cyclic group is optionally substituted with one or more R11 or R12 groups; each R11 is independently a hydrocarbyl group optionally substituted by one or more R12 substituents; each R12 is independently selected from OR13, COR13, COOR13, CN, CONR13R14,
NR13R14, SR13, SOR13, SO2R13, SO2OR13, SO2NR13R14, an alicyclic group, halogen, CF3,= and NO2; and
R13 and R14 are each independently H or (CH2)nR15, where n is 0, 1, 2, or 3; and each R15 is independently selected from alkyl, cycloalkyl, aryl, heteroaryl, aryl and an alicyclic group; in the preparation of a medicament for treating one or more of the following: a viral disorder; a CNS disorder; a stroke; a microbial infection; a fungal disorder; a parasitic disorder; an inflammatory disorder; a cardiovascular disorder; alopecia; and diabetes.
In a more preferred embodiment of the invention, the compound of the invention is administered in an amount sufficient to inhibit one or more of the host cell CDKs involved in viral replication, i.e. CDK2, CDK7, CDK8, and CDK9 [23].
As defined herein, an anti-viral effect within the scope of the present invention may be demonstrated by the ability to inhibit CDK2, CDK7, CDK8 or CDK9. In a particularly preferred embodiment, the invention relates to the use of one or more compounds of the invention in the treatment of a viral disorder which is CDK dependent or sensitive. CDK dependent disorders are associated with an above normal level of activity of one or more CDK enzymes. Such disorders preferably associated with an abnormal level of activity of CDK2, CDK7, CDK8 and/or CDK9. A CDK sensitive disorder is a disorder in which an aberration in the CDK level is not the primary cause, but is downstream of the primary metabolic aberration. In such scenarios, CDK2, CDK7, CDK8 and/or CDK9 can be said to be part of the sensitive metabolic pathway and CDK inhibitors may therefore be active in treating such disorders.
A further aspect of the invention relates to a method of treating a CDK-dependent disorder, said method comprising administering to a subject in need thereof, a compound of formula Ia or Ib, or a pharmaceutically acceptable salt thereof, as defined above in an amount sufficient to inhibit a cyclin dependent kinase.
Preferably, the CDK-dependent disorder is a viral disorder or a proliferative disorder, more preferably cancer.
In one preferred embodiment, the compound of the invention is administered in an amount sufficient to inhibit FLT3. FLT3 is known to play an important role in the pathogenesis of acute myeloid leukemia [79]. Thus, in one particularly preferred embodiment, the proliferative disorder is acute myeloid leukemia.
Another aspect of the invention relates to a method of treating a FLT3 -dependent disorder, said method comprising administering to a subject in need thereof, a compound of formula Ia or Ib, or a pharmaceutically acceptable salt thereof, in an amount sufficient to inhibit FLT3. Another aspect of the invention relates to the use of a compound of formula Ib as defined above, or a pharmaceutically acceptable salt thereof, in the preparation of a medicament for treating diabetes.
In a particularly preferred embodiment, the diabetes is type II diabetes.
GSK3 is one of several protein kinases that phosphorylate glycogen synthase (GS). The stimulation of glycogen synthesis by insulin in skeletal muscle results from the dephosphorylation and activation of GS. GSK3's action on GS thus results in the latter' s deactivation and thus suppression of the conversion of glucose into glycogen in muscles. Type II diabetes (non-insulin dependent diabetes mellitus) is a multi-factorial disease. Hyperglycaemia is due to insulin resistance in the liver, muscles, and other tissues, coupled with impaired secretion of insulin. Skeletal muscle is the main site for insulin-stimulated glucose uptake, there it is either removed from circulation or converted to glycogen.. Muscle glycogen deposition is the main determinant in glucose homeostasis and type II diabetics have defective muscle glycogen storage. There is evidence that an increase in GSK3 activity is important in type II diabetes [24]. Furthermore, it has been demonstrated that GSK3 is over-expressed in muscle cells of type II diabetics and that an inverse correlation exists between skeletal muscle GSK3 activity and insulin action [25].
GSK3 inhibition is therefore of therapeutic significance in the treatment of diabetes, particularly type II, and diabetic neuropathy.
It is notable that GSK3 is known to phosphorylate many substrates other than GS, and is thus involved in the regulation of multiple biochemical pathways. For example, GSK is highly expressed in the central and peripheral nervous systems.
Preferably, the compound is administered in an amount sufficient to inhibit GSK, more preferably GSK3, more preferably still GSK3β. Another aspect of the invention therefore relates to the use of a compound of formula Ib as defined above, or a pharmaceutically acceptable salt thereof, in the preparation of a medicament for treating a CNS disorder, for example neurodegenerative disorders.
Preferably, the CNS disorder is Alzheimer's disease.
Tau is a GSK-3 substrate which has been implicated in the etiology of Alzheimer's disease, In healthy nerve cells, Tau co-assembles with tubulin into microtubules. However, in Alzheimer's disease, tau forms large tangles of filaments, which disrupt the microtubule structures in the nerve cell, thereby impairing the transport of nutrients as well as the transmission of neuronal messages.
Without wishing to be bound by theory, it is believed that GSK3 inhibitors may be able to prevent and/or reverse the abnormal hyperphosphorylation of the microtubule-associated protein tau that is an invariant feature of Alzheimer's disease and a number of other neurodegenerative diseases, such as progressive supranuclear palsy, corticobasal degeneration and Pick's disease. Mutations in the tau gene cause inherited forms of fronto- temporal dementia, further underscoring the relevance of tau protein dysfunction for the neurodegenerative process [26].
Another aspect of the invention therefore relates to the use of a compound of formula Ib as defined above, or a pharmaceutically acceptable salt thereof, in the preparation of a medicament for treating bipolar disorder.
Yet another aspect of the invention relates the use of a compound of formula Ib as defined above, or a pharmaceutically acceptable salt thereof, in the preparation of a medicament for treating a stroke.
Reducing neuronal apoptosis is an important therapeutic goal in the context of head trauma, stroke, epilepsy, and motor neuron disease [27]. Therefore, GSK3 as a pro- apoptotic factor in neuronal cells makes this protein kinase an attractive ^therapeutic target for the design of inhibitory drugs to treat these diseases. ■ .. ••>
Yet another aspect of the invention relates the use of a compound of formula Ib as defined above, or a pharmaceutically acceptable salt thereof, in the preparation of a medicament for treating alopecia.
Hair growth is controlled by the Wnt signalling pathway, in particular Wnt-3. In tissue- culture model systems of the skin, the expression of non-degradable mutants of β-catenin leads to a dramatic increase in the population of putative stem cells, which have greater proliferative potential [28]. This population of stem cells expresses a higher level of non- cadherin-associated β-catenin [29], which may contribute to their high proliferative potential. Moreover, transgenic mice overexpressing a truncated β-catenin in the skin undergo de novo hair-follicle morphogenesis, which normally is only established during embryogenesis. The ectopic application of GSK3 inhibitors may therefore be therapeutically useful in the treatment of baldness and in restoring hair growth following chemotherapy-induced alopecia.
A further aspect of the invention relates to a method of treating a GSK3 -dependent disorder, said method comprising administering to a subject in need thereof, a compound of formula Ib, or a pharmaceutically acceptable salt thereof, as defined above in an amount sufficient to inhibit GSK3.
Preferably, the GSK3 -dependent disorder is diabetes.
Preferably, the compound of the invention, or pharmaceutically acceptable salt thereof, is administered in an amount sufficient to inhibit GSK3β.
In one embodiment of the invention, the compound of the invention is administered in an amount sufficient to inhibit at least one PLK enzyme. The polo-like kinases (PLKs) constitute a family of serine/threonine protein kinases.
Mitotic Drosophila melanogaster mutants at the polo locus display spindle abnormalities [30] and polo was found to encode a mitotic kinase [31]. In humans, there exist three closely related PLKs [32]. They contain a highly homologous amino-terminal catalytic kinase domain and their carboxyl termini contain two or three conserved regions, the polo boxes. The function of the polo boxes remains incompletely understood but they are implicated in the targeting of PLKs to subcellular compartments [33,34], mediation of interactions with other proteins [35], or may constitute part of an autoregulatory domain [36]. Furthermore, the polo box-dependent PLKl activity is required for proper metaphase/anaphase transition and cytokinesis [37,38].
Studies have shown that human PLKs regulate some fundamental aspects of mitosis [39,40]. In particular, PLKl activity is believed to be necessary for the functional maturation of centrosomes in late G2/early prophase and subsequent establishment of a bipolar spindle. Depletion of cellular PLKl through the small interfering RNA (siRNA) technique has also confirmed that this protein is required for multiple mitotic processes and completion of cytokinesis [41].
In a more preferred embodiment of the invention, the compound of the invention is administered in an amount sufficient to inhibit PLKl.
Of the three human PLKs, PLKl is the best characterized; it regulates a number of cell division cycle effects, including the onset of mitosis [42,43], DNA-damage checkpoint activation [44,45], regulation of the anaphase promoting complex [46-48], phosphorylation of the proteasome [49], and centrosome duplication and maturation [50].
Specifically, initiation of mitosis requires activation of M-phase promoting factor (MPF), the complex between the cyclin dependent kinase CDKl and B-type cyclins [51]. The latter accumulate during the S and G2 phases of the cell cycle and promote the inhibitory phosphorylation of the MPF complex by WEEl, MIKl, and MYTl kinases. At the end of the G2 phase, corresponding dephosphorylation by the dual-specificity phosphatase
CDC25C triggers the activation of MPF [52]. In interphase, cyclin B localizes to the cytoplasm [53], it then becomes phosphorylated during prophase and this event causes nuclear translocation [54,55]. The nuclear accumulation of active MPF during prophase is thought to be important for initiating M-phase events [56]. However, nuclear MPF is kept inactive by WEEl unless counteracted by CDC25C. The phosphatase CDC25C itself, localized to the cytoplasm during interphase, accumulates hi the nucleus in prophase [57- 59]. The nuclear entry of both cyclin B [60] and CDC25C [61] are promoted through phosphorylation by PLKl [43]. This kinase is an important regulator of M-phase initiation.
In one particularly preferred embodiment, the compounds of the invention are ATP- antagonistic inhibitors of PLK 1.
In the present context ATP antagonism refers to the ability of an inhibitor compound to diminish or prevent PLK catalytic activity, i.e. phosphotransfer from ATP to a macromolecular PLK substrate, by virtue of reversibly or irreversibly binding at the enzyme's active site in such a manner as to impair or abolish ATP binding.
In another preferred embodiment, the compound of the invention is administered in an amount sufficient to inhibit PLK2 and/or PLK3.
Mammalian PLK2 (also known as SNK) and PLK3 (also known as PRK and FNK) were originally shown to be immediate early gene products. PLK3 kinase activity appears to peak during late S and G2 phase. It is also activated during DNA damage checkpoint activation and severe oxidative stress. PLK3 also plays an important role in the regulation of microtubule dynamics and centrosome function in the cell and deregulated PLK3 expression results in cell cycle arrest and apoptosis [62]. PLK2 is the least well understood homologue of the three PLKs. Both PLK2 and PLK3 may have additional important postmitotic functions [35], A further aspect of the invention relates to a method of treating a PLK-dependent disorder, said method comprising administering to a subject in need thereof, a compound of formula Ib, or a pharmaceutically acceptable salt thereof, as defined above in an amount sufficient to inhibit PLK.
Preferably, the PLK-dependent disorder is a proliferative disorder, more preferably cancer.
Preferably, the compound of the invention, or pharmaceutically acceptable salt thereof, is administered in an amount sufficient to inhibit aurora kinase.
A further aspect of the invention relates to a method of treating an aurora kinase-dependent disorder, said method comprising administering to a subject in need thereof, a compound of formula Ib, or a pharmaceutically acceptable salt thereof, as defined above in an amount sufficient to inhibit aurora kinase.
Preferably, the aurora kinase dependent disorder is a viral disorder as defined above.
PHARMACEUTICAL COMPOSITIONS
Another aspect of the invention relates to a pharmaceutical composition comprising one or more compounds of the invention as defined above admixed with one or more pharmaceutically acceptable diluents, excipients or carriers. Even though the compounds of the present invention (including their pharmaceutically acceptable salts, esters and pharmaceutically acceptable solvates) can be administered alone, they will generally be administered in admixture with a pharmaceutical carrier, excipient or diluent, particularly for human therapy. The pharmaceutical compositions may be for human or animal usage in human and veterinary medicine.
Examples of such suitable excipients for the various different forms of pharmaceutical compositions described herein may be found in the "Handbook of Pharmaceutical Excipients, 2nd Edition, (1994), Edited by A Wade and PJ Weller. Acceptable carriers or diluents for therapeutic use are well known in the pharmaceutical art, and are described, for example, in Remington's Pharmaceutical Sciences, Mack Publishing Co. (A. R. Gennaro edit. 1985).
Examples of suitable carriers include lactose, starch, glucose, methyl cellulose, magnesium stearate, mannitol, sorbitol and the like. Examples of suitable diluents include ethanol, glycerol and water.
The choice of pharmaceutical carrier, excipient or diluent can be selected with regard to the intended route of administration and standard pharmaceutical practice. The pharmaceutical compositions may comprise as, or in addition to, the carrier, excipient or diluent any suitable binder(s), lubricant(s), suspending agent(s), coating agent(s), solubilising agent(s).
Examples of suitable binders include starch, gelatin, natural sugars such as glucose, anhydrous lactose, free-flow lactose, beta-lactose, corn sweeteners, natural and synthetic gums, such as acacia, tragacanth or sodium alginate, carboxymethyl cellulose and polyethylene glycol.
Examples of suitable lubricants include sodium oleate, sodium stearate, magnesium stearate, sodium benzoate, sodium acetate, sodium chloride and the like.
Preservatives, stabilizers, dyes and even flavoring agents may be provided in the pharmaceutical composition. Examples of preservatives include sodium benzoate, sorbic acid and esters of p-hydroxybenzoic acid. Antioxidants and suspending agents may be also used.
SALTS/ESTERS
The compounds of the invention can be present as salts or esters, in particular pharmaceutically acceptable salts or esters. Pharmaceutically acceptable salts of the compounds of the invention include suitable acid addition or base salts thereof. A review of suitable pharmaceutical salts may be found in Berge et al, J Pharm Sci, 66, 1-19 (1977). Salts are formed, for example with strong inorganic acids such as mineral acids, e.g. sulphuric acid, phosphoric acid or hydrohalic acids; with strong organic carboxylic acids, such as alkanecarboxylic acids of 1 to 4 carbon atoms which are unsubstituted or substituted (e.g., by halogen), such as acetic acid; with saturated or unsaturated dicarboxylic acids, for example oxalic, malonic, succinic, maleic, fumaric, phthalic or tetraphthalic; with hydroxycarboxylic acids, for example ascorbic, glycolic, lactic, malic, tartaric or citric acid; with aminoacids, for example aspartic or glutamic acid; with benzoic acid; or with organic sulfonic acids, such as
Figure imgf000029_0001
or aryl-sulfonic acids which are unsubstituted or substituted (for example, by a halogen) such as methane- or p-toluene sulfonic acid.
Esters are formed either using organic acids or alcohols/hydroxides, depending on the functional group being esterified. Organic acids include carboxylic acids, such as alkanecarboxylic acids of 1 to 12 carbon atoms which are unsubstituted or substituted (e.g;-; by halogen), such as acetic acid; with saturated or unsaturated dicarboxylic acid, for example oxalic, malonic, succinic, maleic, fumaric, phthalic or tetraphthalic; with hydroxycarboxylic acids, for example ascorbic, glycolic, lactic, malic, tartaric or citric acid; with aminoacids, for example aspartic or glutamic acid; with benzoic acid; or with organic sulfonic acids, such as (C1-C4)-alkyl- or aryl-sulfonic acids which are unsubstituted or substituted (for example, by a halogen) such as methane- or p-tolueήe sulfonic acid. Suitable hydroxides include inorganic hydroxides, such as sodium hydroxide, potassium hydroxide, calcium hydroxide, aluminium hydroxide. Alcohols include alkanealcohols of 1-12 carbon atoms which may be unsubstituted or substituted, e.g. by a halogen).
ENANTIOMERS/TAUTOMERS
In all aspects of the present invention previously discussed, the invention includes, where appropriate all enantiomers and tautomers of compounds of the invention. The man skilled in the art will recognise compounds that possess an optical properties (one or more chiral carbon atoms) or tautomeric characteristics. The corresponding enantiomers and/or tautomers may be isolated/prepared by methods known in the art.
STEREO AND GEOMETRIC ISOMERS
Some of the compounds of the invention may exist as stereoisomers and/or geometric isomers - e.g. they may possess one or more asymmetric and/or geometric centres and so may exist in two or more stereoisomeric and/or geometric forms. The present invention contemplates the use of all the individual stereoisomers and geometric isomers of those agents, and mixtures thereof. The terms used in the claims encompass these forms, provided said forms retain the appropriate functional activity (though not necessarily to the same degree).
In particular, the compounds of the invention may exist in cis or trans forms, either in isolated form, or as mixtures thereof in any ratio. By way of example, where the compounds of the invention contain morpholinyl or piperidinyl substituents, the methyl groups on the morpholinyl and piperidinyl rings can be either cis or trans.
The present invention also includes all suitable isotopic variations of the agent of pharmaceutically acceptable salt thereof. An isotopic variation of an agent of the present invention or a pharmaceutically acceptable salt thereof is defined as one in which at least one atom is replaced by an atom having the same atomic number but an atomic mass different from the atomic mass usually found in nature. Examples of isotopes that can be incorporated into the agent and pharmaceutically acceptable salts thereof include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulphur, fluorine and chlorine such as 2H, 3H, 13C, 14C, 15N, 17O, 180, 31P, 32P, 35S, 18F and 36Cl, respectively. Certain isotopic variations of the agent and pharmaceutically acceptable salts thereof, for example, those in which a radioactive isotope such as 3H or 14C is incorporated, are useful in drug and/or substrate tissue distribution studies. Tritiated, i.e., 3H, and carbon-14, i.e., 14C, isotopes are particularly preferred for their ease of preparation and detectability. Further, substitution with isotopes such as deuterium, i.e., 2H5 may afford certain therapeutic advantages resulting from greater metabolic stability, for example, increased in vivo half-life or reduced dosage requirements and hence may be preferred in some circumstances. Isotopic variations of the agent of the present invention and pharmaceutically acceptable salts thereof of this invention can generally be prepared by conventional procedures using appropriate isotopic variations of suitable reagents.
SOLVATES
The present invention also includes the use of solvate forms of the compounds of the present invention. The terms used in the claims encompass these forms.
POLYMORPHS
The invention furthermore relates to the compounds of the present invention in their various crystalline forms, polymorphic forms and (an)hydrous forms. It is well established within the pharmaceutical industry that chemical compounds may be isolated in any of such forms by slightly varying the method of purification and or isolation form the solvents used in the synthetic preparation of such compounds.
PRODRUGS
The invention further includes the compounds of the present invention in prodrug form. Such prodrugs are generally compounds of the invention wherein one or more appropriate groups have been modified such that the modification may be reversed upon administration to a human or mammalian subject. Such reversion is usually performed by an enzyme naturally present in such subject, though it is possible for a second agent to be administered together with such a prodrug in order to perform the reversion in vivo. Examples of such modifications include ester (for example, any of those described above)', wherein the reversion may be carried out be an esterase etc. Other such systems will be well known to those skilled in the art. ADMINISTRATION
The pharmaceutical compositions of the present invention may be adapted for oral, rectal, vaginal, parenteral, intramuscular, intraperitoneal, intraarterial, intrathecal, intrabronchial, subcutaneous, intradermal, intravenous, nasal, buccal or sublingual routes of administration.
For oral administration, particular use is made of compressed tablets, pills, tablets, gellules, drops, and capsules. Preferably, these compositions contain from 1 to 250 mg and more preferably from 10-100 mg, of active ingredient per dose.
Other forms of administration comprise solutions or emulsions which may be injected intravenously, intraarterially, intrathecally, subcutaneously, intradermally, intraperitoneally or intramuscularly, and which are prepared from sterile or sterilisable solutions. The pharmaceutical compositions of the present invention may also be in form of suppositories, pessaries, suspensions, emulsions, lotions, ointments, creams, gels, sprays, solutions or dusting powders.
An alternative means of transdermal administration is by use of a skin patch. For example, the active ingredient can be incorporated into a cream consisting of an aqueous emulsion of polyethylene glycols or liquid paraffin. The active ingredient can also be incorporated, at a concentration of between 1 and 10% by weight, into an ointment consisting of a white wax or white soft paraffin base together with such stabilisers and preservatives as may be required.
Injectable forms may contain between 10-1000 mg, preferably between 10-250 mg, of active ingredient per dose.
Compositions may be formulated in unit dosage form, i.e., in the form of discrete portions containing a unit dose, or a multiple or sub-unit of a unit dose. DOSAGE
A person of ordinary skill in the art can easily determine an appropriate dose of one of the instant compositions to administer to a subject without undue experimentation. Typically, a physician will determine the actual dosage which will be most suitable for an individual patient and it will depend on a variety of factors including the activity of the specific compound employed, the metabolic stability and length of action of that compound, the age, body weight, general health, sex, diet, mode and time of administration, rate of excretion, drug combination, the severity of the particular condition, and the individual undergoing therapy. The dosages disclosed herein are exemplary of the average case. There can of course be individual instances where higher or lower dosage ranges are merited, and such are within the scope of this invention.
Depending upon the need, the agent may be administered at a dose of from 0.01 to 30 mg/kg body weight, such as from 0.1 to 10 mg/kg, more preferably from 0.1 to 1 mg/kg body weight.
In an exemplary embodiment, one or more doses of 10 to 150 mg/day will be administered to the patient.
COMBINATIONS
In a particularly preferred embodiment, the one or more compounds of the invention are administered in combination with one or more other therapeutically active agents, for example, existing drugs available on the market. In such cases, the compounds of the invention may be administered consecutively, simultaneously or sequentially with the one or more other active agents.
By way of example, it is known that anticancer drugs in general are more effective when used in combination. In particular, combination therapy is desirable in order to avoid an overlap of major toxicities, mechanism of action and resistance mechanism(s). Furthermore, it is also desirable to administer most drugs at their maximum tolerated doses with minimum time intervals between such doses. The major advantages of combining chemotherapeutic drugs are that it may promote additive or possible synergistic effects through biochemical interactions and also may decrease the emergence of resistance in early tumor cells which would have been otherwise responsive to initial chemotherapy with a single agent. An example of the use of biochemical interactions in selecting drug combinations is demonstrated by the administration of leucovorin to increase the binding of an active intracellular metabolite of 5-fluorouracil to its target, thymidylate synthase, thus increasing its cytotoxic effects.
Numerous combinations are used in current treatments of cancer and leukemia. A more extensive review of medical practices may be found in "Oncologic Therapies" edited by E. E. Vokes and H. M. Golomb, published by Springer.
Beneficial combinations may be suggested by studying the growth inhibitory activity of the test compounds with agents known or suspected of being valuable in the treatment of a particular cancer initially or cell lines derived from that cancer. This procedure can also be used to determine the order of administration of the agents, i.e. before, simultaneously; or after delivery. Such scheduling may be a feature of all the cycle acting agents identified herein.
ASSAYS
Another aspect of the invention relates to the use of a compound of formula Ib as defined above, or a pharmaceutically acceptable salt thereof, in an assay for identifying further candidate compounds capable of inhibiting one or more protein kinases.
Another aspect of the invention relates to the use of a compound of the invention, or a pharmaceutically acceptable salt thereof, in an assay for identifying further candidate compounds capable of inhibiting one or more cyclin dependent kinases, aurora kinase, GSK, FLT3 and PLK. Preferably, the assay is a competitive binding assay.
More preferably, the competitive binding assay comprises contacting a compound of the invention with a protein kinase and a candidate compound and detecting any change in the interaction between the compound of the invention and the protein kinase.
One aspect of the invention relates to a process comprising the steps of:
(a) performing an assay method described hereinabove;
(b) identifying one or more ligands capable of binding to a ligand binding domain; and
(c) preparing a quantity of said one or more ligands.
Another aspect of the invention provides a process comprising the steps of:
(a) performing an assay method described hereinabove;
(b) identifying one or more ligands capable of binding to a ligand binding domain; and
(c) preparing a pharmaceutical composition comprising said one or more ligands.
Another aspect of the invention provides a process comprising the steps of:
(a) performing an assay method described hereinabove;
(b) identifying one or more ligands capable of binding to a ligand binding domain;
(c) modifying said one or more ligands capable of binding to a ligand binding domain; .
(d) performing the assay method described hereinabove;
(e) optionally preparing a pharmaceutical composition comprising said one or more ligands.
The invention also relates to a ligand identified by the method described hereinabove.
Yet another aspect of the invention relates to a pharmaceutical composition comprising a ligand identified by the method described hereinabove. Another aspect of the invention relates to the use of a ligand identified by the method described hereinabove in the preparation of a pharmaceutical composition for use in the treatment of proliferative disorders, viral disorders, a CNS disorder, stroke, alopecia and diabetes.
Preferably, said candidate compound is generated by conventional SAR modification of a compound of the invention.
As used herein, the term "conventional SAR modification" refers to standard methods known in the art for varying a given compound by way of chemical derivatisation.
The above methods may be used to screen for a ligand useful as an inhibitor of one or more protein kinases.
SYNTHESIS
Compounds of general formula I may be prepared by any suitable method known in the art. A convenient synthetic route is shown below:
Figure imgf000037_0001
IV
Figure imgf000037_0002
V
iV-Unsubstituted 1-H-indoles II may be acylated with acid anhydride or acid halide derivatives of R4CH2COOH at C-3 to afford the 3-acyl-lH-indole products III ([69], pp. 262-263). If R6 is other than H, this substituent is next introduced, followed by acylation with appropriate carbonyl compounds containing the group R3, to provide the intermediate 1,3-dicarbonyl compounds IV. These can be condensed directly with guanidines VI; alternatively they are first converted to the enaminones V5 from which 4-(lH-Indol-3-yl)- pyrimidin-2-ylamines I can be obtained [70].
A further aspect of the invention therefore relates to a process for preparing a compound of formula Ib as defined above, said process comprising the steps of:
Figure imgf000038_0001
(a) condensing a compound of formula IV with a guanidine of formula VI to form a compound of formula I; or
(b) (i) converting a compound of formula IV to a compound of formula V; and (ii) condensing said compound of formula V with a guanidine of formula VI to form a compound of formula I.
Preferably, the compound of formula IV is prepared by acylating a compound of formula III
Figure imgf000038_0002
Preferably, the compound of formula III is prepared by acylating a compound of formula II with an acid anhydride or acid halide derivative OfR4CH2COOH
Figure imgf000039_0001
II
In one preferred embodiment, said compound of formula III is prepared by a process which comprises treating a compound of formula II as defined above with (i) zinc chloride and ethylmagnesium bromide, and (ii) acetyl chloride.
The present invention is further described by way of the following non-limiting examples.
EXAMPLES
Example 1 General
NMR spectra were recorded using a Varian INOVA-500 instrument. Chemical shifts are reported in parts per million relative to internal tetramethylsilane standard. Mass spectra were obtained using a Waters ZQ2000 single quadrupole mass spectrometer with electrospray ionization (ESI). Analytical and preparative RP-HPLC was performed using Vydac 218TP54 (250 x 4.6 mm) and 218TP1022 (250 x 22 mm) columns, respectively. Linear gradient elution using H2OMeCN systems (containing 0.1 % CF3COOH) at flow rates of 1 mL/min (analytical) and 9 mL/min (preparative) was performed. Purity was assessed by integration of chromatograms (λ = 254 nm). Silica gel (EM Kieselgel 60, 0.040-0.063 mm, Merck) or ISOLUTE pre-packed columns (Jones Chromatography Ltd. UK) were used for flash chromatography.
The structures of selected compounds of the invention are shown in Table 1.
Example 2 4-(lH-Indol-3-yl)-pyrimidin-2-ylamine (1)
Figure imgf000040_0001
A mixture of l-(lH-indol-3-yl)-ethanone (2 mmol, 3.18 g) in dimethoxymethyl-dimethyl- amine (60 mmol, 7.18 g, 8 mL) was heated under reflux for 16 h. Excess dimethoxymethyl-dimethyl-amine was evaporated in vacuo to leave an orange residue of 3-dimethylamino-l-(lH-indol-3-yl)-propenone, which was used in the next reaction without further purification. A mixture of this material (5 mmol, 1.07 g) and guanidine carbonate (5 mmol, 0.94 g) in 2-methoxylethanol (20 mL) was heated at 125 0C for 22 h.
The solvent was evaporated and the residue was purified by silica gel column chromatography (elution with 5:1 EtOAc/PE and then EtOAc). The fractions containing the desired product were combined and evaporated. The residue was recrystallised from MeOH to afford the pure title compound (0.80 g, 76 %) as colourless crystals. 1H-NMR (300 MHz, DMSO-J6) δ: 6 A3 (s, IH, NH), 6.94 (d, IH, J= 5.3 Hz, pyrimidinyl-H), 7.14- 7.27 (m, 2H, Ar-H)5 7.49 (d, IH, J= 8.0 Hz, Ar-H), 8.10 (d, IH, J= 5.4 Hz, pyrhnidinyl- H), 8.18 (s, IH, indole C2-H), 8.59 (d, IH, J= 7.6 Hz, Ar-H).
Example 3 [4-(lH-Indol-3-yl)-pyrimidin-2-yl]-(3-nitro-phenyl)-amine (2)
Figure imgf000041_0001
A mixture of l-(lH-indol-3-yl)-ethanone (1.00 g, 6.28 mmol) and tert-butoxy-bis- (dimethyl-amino)-methane (1.5 mL, 1.16 mmol) was heated at 100 0C for 15 h. After cooling, and concentration under vacuum, the residue was treated with cold diethyl ether. The resulting yellow precipitate was filtered and dried to afford 3-dimethylamino-l-(lH- indol-3-yl)-propenone (0.40 g, 1.86 mmol. 29.7 %). 1H-NMR (500 MHz, DMSO-^6) δ: 2.95 (6H, br. s, N(CHa)2), 5.74 (IH, d, C=CH, J= 12.6 Hz), 7.08 (IH, dd, ArH, J = 8.3, 8.3 Hz), 7.10 (IH, dd, ArH, J= 7.8, 7.8 Hz), 7.38 (IH, d, ArH, J= 7.8 Hz), 7.49 (IH, d, C=CH, J= 12.6 Hz), 8.12 (IH, s, ArH), 8.26 (IH, d, ArH, J=7.8 Hz), 11.56 (IH, s, NH). ESI-MS: m/z = 214.98 [M+H]+, C13H14N2O requires 214.3; Anal. RP-HPLC (0-60 % MeCN gradient) tR = 13.2 min (> 98 % purity).
3-Dimethylamino-l-(lH-indol-3-yl)-propenone (0.173 g, 0.8 mmol), iV-(3-nitro-phenyl)- guanidine nitrate (0.197 g, 0.8 mmol) and potassium carbonate (0.139 g, 1.0 mmol) were combined in 2-methoxyethanol (4 mL) and heated at 115 0C for 22 h. After cooling, the inorganics were filtered off and the filtrate was concentrated to dryness. The crude product was purified by silica gel column chromatography. Pooling of the desired fractions, evaporation, and drying afforded the pure title compound (0.083 g, 0.25 mmol, 31 %). 1H- NMR (500 MHz, DMS(W6) S: 7.13 (IH, dd, ArH, J= 8.3, 8.3 Hz), 7.20 (IH, dd, ArH, J= 8.3, 8.3 Hz), 7.39 (IH, d, ArH, J= 5.8 Hz), 7.48 (IH, d, ArH, J= 8.3 Hz), 7.59 (IH, dd, ArH, J= 8.3, 8.3 Hz), 7.80 (IH, dd, ArH, J= 8.3, 2.3 Hz), 8.14 (IH, dd, ArH, J= 8.3, 2.3 Hz), 8.40 IH, d, ArH, J= 2.3 Hz), 8.42 (IH, d, ArH, J= 5.8 Hz), 8.53 (IH, d, ArH, J= 8.3 Hz), 8.97(1H, s, ArH), 9.98 (IH, s, NH), 11.89 (IH, s, NH). ESI-MS m/z = 331.94 [M+H]+, C18H13N5O2 requires 331.33. Anal. RP-HPLC (0-60 % MeCN gradient) tR = 18.53 min (> 98 % purity).
Example 4
The following compounds were prepared using procedures analogous to those described in
Example 3 above:
(4-Fluoro-phenyl)-[4-(lH-indol-3-yl)-pyrimidin-2-yl]-amine (3)
Figure imgf000042_0001
1H-NMR (500 MHz, DMSO-J6) δ: 7.13 (IH, dd, ArH, J= 8.3, 8.3 Hz), 7.19 (IH, dd, ArH, J= 8.3, 8.3 Hz), 7.19 (2H, d, ArH, J= 8.3 Hz), 7.32 (IH, d, ArH, J= 5.8 Hz), 7.46 (IH, d, ArH, J= 8.3 Hz), 7.75 (2H, d, ArH, J= 8.3 Hz), 8.27 (IH, d, ArH, J= 5.8 Hz), 8.40 (IH, s, ArH), 8.47 (IH, d, ArH, J= 8.3 Hz), 9.68 (IH, s, NH), 11.94 (IH, s, NH). ESI-MS m/z = 305.05 [M+H]+, C18H13FN4 requires 304.32. Anal. RP-HPLC (0-60 % MeCN gradient) tR = 18.24 min (> 98 % purity). [4-(lH-Indol-3-yl)-pyrimidin-2-yl]-(6-methoxy-pyridin-3-yl)-amine (4)
Figure imgf000043_0001
1H-NMR (500 MHz, CD3OD) δ: 3.96 (3H, s, CH5), 6.90 (IH5 d, J = 9.0 Hz, ArH), 7.16 (IH, t, J= 8.5 Hz, ArA), 7.23 (IH, t, J= 8.5 Hz, ArH)5 7.31 (IH, d, J= 7.5 Hz, ArH), 7.46 (IH, d, J = 5.5 Hz, pyrimidinyl-H), 7.98 (IH, m, ArH), 8.12 (IH, d, J = 5.5 Hz, pyrimidinyl-H), 8.28 (2H, m, ArH), 8.39 (IH, s, ArH). ESI-MS m/z = 317.99 [M+H]+, C18Hi5N5O requires 317.34. Anal. RP-HPLC (10-70 % MeCN gradient) tR = 12.88 min (purity > 98 %).
[4-(lH-Indol-3-yl)-pyrimidin-2-yl]-(4-morpholin-4-yl-phenyl)-amine (5)
Figure imgf000043_0002
3-Dimethylamino-l-(lH-indol-3-yl)-propenone (0.10 g, 0.46 mmol), iV-(4-morpholin-4-yl- phenyl)-guanidine nitrate (0.11 g, 0.46 mmol) and potassium carbonate (64 mg, 0.46 mmol) were combined in 2-methoxyethanol (4 mL) and the mixture was heated under microwave irradiation at 180 0C for 20 min. After cooling, the reaction product was precipitated by addition of water (25 mL), and was collected by filtration. The product was purified by silica gel chromatography. Pooling of the desired fractions afforded the pure target compound (40 mg, 23 %). 1H-NMR (500 MHz, CD3OD) δ: 11.73 (IH, br. s, NH); 9.12 (IH, s, ArH); 8.57 (IH, d, ArH, J= 7.8 Hz); 8.26 (2H, d, ArH, J= 5.4 Hz); 7.65 (2H, d, ArH, J= 8.8 Hz); 7.45 (IH, d, ArH, J= 7.8 Hz); 7.18 (2H, m, ArH); 7.12 (IH, t, ArH, J
= 7.8 Hz); 6.92 (2H, d, ArH, J= 9.3 Hz); 3.75 (4H, t, ArH5 J= 4.39 Hz); 3.05 (IH, d, ArH, J= 5.15 Hz). ESI-MS m/z =371.98 [M+H]+, C22H21N5O requires 371.44. Anal. RP-HPLC (10-70 % MeCN gradient) tR = 12.02 min (purity > 98 %).
Example 5
Compounds (6) to (31) were synthesised by analogous methods and in accordance with the protocols set forth below.
3-Acetylation of substituted indoles
[Concise Synthesis and Structure- Activity Relationships of Combrestatin A-4 Analogues: 1- and 3-Aroylindoles as Novel Classes of Potent Antitubulin Agents. J. Med. Chem., 2004, Vol. 47, No. 17, 4247-4257]
To a mixture of substituted indole (2.03 mmol) and anhydrous zinc chloride (560mg, 4.07 mmol) in dry dichloromethane (15ml), was added ethylmagnesium bromide (2.7ml, 1.0M solution in THF) over 10 min at room temperature. After stirring for Ih, acetyl chloride (239mg, 217μl, 3.05 mmol) was added dropwise over 5 min. After stirring for another Ih, aluminium chloride (270mg, 2.03 mmol) was added and the resultant mixture stirred for 5h. Water was added (15ml) and extracted with dichloromethane (20ml). The organic layer was dried (MgSO4) and evaporated to give a red solid. Chromatography (2:1 petroleum ether-ethyl acetate) gave the desired 3 -acetyl indole as a colourless solid.
Formation of 1-acylindole pyrimidines: Compounds (18) and (19)
Compound (5) (50mg, 0.135 mmol) was dissolved in dry dimethylformamide (10ml) and sodium hydride (4mg, 1.2eq) added. When evolution of hydrogen had ceased, acid chloride (1.2eq) was added and the mixture stirred at room temperature for a further 4h. Solvent removed in Genevac and the crude product suspended in MeOH (2ml) and absorbed on SPE column. Chromatography (95:5 ethyl acetate-MeOH) gave the desired product as a yellow solid. Preparation of indoles modified at the 2-position (Compound 22), the 6-position
(Compounds 24 and 26), and the 7-position (Compounds 21, 23 and 25), and other indoles modified at the 1 -position (Compound 16). These indoles were obtained as starting materials.
4-(lH-indol-3-yl)-N-(4-acetylpiperazin-l-ylphenyl)pyrimidin-2-amine (6) Anal. RP-HPLC: tκ 13.01 min. (0-60% MeCN).
1H NMR (DMSOd6, 500 MHz): δH 2.05 (3H, s, CH3), 3.03 (2H, dd, J 5.0, 2 x CHH), 3.09 (2H, dd, J 5.0, 2 x CHH), 3.58 (4H, m, incl. J 5.0, 2 x CH2), 6.95 (2H, d, J 9.0, 2 x Ar-H), 7.13 (IH, dd, J 7.5, Ar-H), 7.14-7.20 (2H, m, pyrim-H and Ar-H), 7.46 (IH, dd, J 7.5, Ar- H), 7.67 (2H, d, J 9.0, 2 x Ar-H), 8.26 (2H, m, incl. J 5.0, pyrim-H and Ar-H), 8.57 (IH, dd, J7.5, Ar-H), 9.14 (IH, s, NH) and 11.74 (IH, s, NH). MS (ESf) m/z 412.97 [M+H]+ (C24H24N6O requires 412.49).
4~(lH-indol-3-yl)-N-(4-piperazin-l-ylphenyl)pyrimidin-2-amine (7)
Anal. RP-HPLC: fe 9.48 min. (10-70% MeCN). .
1H NMR (DMSOd6, 500 MHz): δH 2.85 (4H, dd, J 5.0, 2 x CH2), 2.99 (4H, dd, J 5.0, 2 x
CH2), 6.89 (2H, d, J8.5, 2 x Ar-H), 7.12 (IH, dd, J7.5, Ar-H), 7.16-7.20 (2H, m, pyrim-H and Ar-H), 7.45 (IH, dd, J 7.5, Ar-H), 7.63 (2H, d, J 8.5, 2 x Ar-H), 8.26 (2H, m, incl. J
5.0, pyrim-H and Ar-H), 8.57 (IH, dd, J 7.5, Ar-H), 9.09 (IH, s, NH) and 11.73 (IH, s,
NH).
MS (ESI+) m/z 371.02 [M+H]+ (C22H22N6 requires 370.45). :
4-(lH-indol-3-yl)-N-(4-benzylpiperazin-l-ylphenyl)pyrimidin-2-amine (8)
Anal. RP-HPLC: tR 12.31 min. (10-70% MeCN).
1H NMR (DMSOd6, 500 MHz): δH 2.51 (4H, dd, J 5.0, 2 x CH2), 3.09 (4H, dd, J 5.0, 2 x
CH2), 3.53 (2H, s, CH2), 6.91 (2H, d, J 9.0, 2 x Ar-H), 7.12 (IH, dd, J 8.0, Ar-H), 7.16-
7.20 (2H, m, pyrim-H and Ar-H), 7.26 (IH, dd, J 9.0, 4.0, Ar-H), 7.33-7.35 (4H, m, 4 x
Ar-H), 7.45 (IH, d, J 8.0, Ar-H), 7.63 (2H, d, J 9.0, 2 x Ar-H), 8.25 (2H, m, incl. J 5.0, pyrim-H and Ar-H)5 8.56 (IH, dd, J 8.0, 1.0, Ar-H), 9.09 (IH, s, NH) and 11.73 (IH, s,
NH).
MS (ESI+) m/z 460.93 [M]+ (C29H28N6 requires 460.57).
4-(lH-indol-3-yl)-N-(2,6-dimethylmorpholin-4-ylphenyl)pyrimidin-2-amine (9)
Two batches were made. Batch Ol is 20:1 cis:trans. Batch 02 is 1:1 cis:trans.
This compound was the result of a cyclisation with a guanidine containing a 4:1 cis:trans ratio of diastereoisomers. The early fractions from Prep-HPLC contained a 20:1 cisitrans mixture (Batch 01) and later fractions had a 1 :1 mixture (Batch 02). The assay data refers to Batch 01.
Anal. RP-HPLC: tκ 14.05 min. (10-70% MeCN).
1H NMR (DMSOd6, 500 MHz): δH 1.16 (6H, d, J 6.5, 2 x CH3), 2.22 (2H, dd, J 11.0, 2 x
CHH), 3.50 (2H, dd, J 11.0, 2 x CHH), 3.71 (2H, m, incl. J 11.0, 6.5, 2 x CHCH3), 6.92
(2H, dd, J8.0, 2 x Ar-H), 7.12-7.15 (IH, m, Ar-H), 7.17-7.19 (2H, m, pyrim-H and Ar-H),
7.46 (IH, d, J 8.0, Ar-H), 7.65 (2H, d, J 8.0, 2 x Ar-H), 8.26 (2H, m, incl. J 5.0, pyrim-H and Ar-H), 8.56 (IH, d, J 8.0, Ar-H), 9.10 (IH, s, NH) and 11.73 (IH, s, NH).
MS (ESI+) m/z 399.98 [M+H]+ (C24H25N5O requires 399.49).
N'-[4-(lH-indol-3-yl)pyrimidin-2-yl]-N,N-dimethylbenzene-l, 4-diamine (10)
Anal. RP-HPLC: tR 9.45 min. (10-70% MeCN).
1H NMR (DMSOd6, 500 MHz): δH 2.86 (6H, s, 2 x CH3), 6.74 (2H, d, J 7.0, 2 x Ar-H),
7.11 (IH, dd, J 8.0, Ar-H), 7.14-7.20 (2H, m, pyrim-H and Ar-H), 7.45 (IH, d, J 8.0, Ar-
H), 7.58 (2H, d, J 7.0, 2 x Ar-H), 8.23-8.26 (2H, m, incl. J 5.0, pyrim-H and Ar-H), 8.56
(IH, d, J 8.0, Ar-H), 8.99 (IH, s, NH) and 11.71 (IH, s, NH).
MS (ESI+) m/z 330.05 [MH-H]+ (C20H19N5 requires 329.40).
4-(lH-indol-3-yl)-N-(2-methyl-4-morpholin-4-ylphenyl)pyήmidin-2-amine (11) Anal. RP-HPLC: /R 12.48 min. (10-70% MeCN). 1H NMR (DMSOd6, 500 MHz): δH 2.19 (3H5 s, CH3), 3.11 (4H5 dd, J 5.0, 2 x CH2), 3.77 (4H, dd, J 5.0, 2 x CH2), 6.81 (IH, dd, J 9.0, 2.5, Ar-H), 6.86 (IH5 d, J 2.5, Ar-H)5 6.95 (IH5 dd, J 8.0, Ar-H)5 7.09-7.14 (2H, m, pyrim-H and Ar-H), 7.27 (IH, d, J 9.0, Ar-H)5 7.39 (IH5 d, J8.0, Ar-H), 8.16 (IH, d, J 5.5, pyrim-H), 8.20 (2H, br s, 2 x Ar-H), 8.41 (IH, s, NH) and 11.65 (IH, s, NH). MS (ESt) m/z 385.98 [M+H]+ (C23H23N5O requires 385.46).
4-(lH-indol-3-yl)-N-(3, 4, 5-trimethoxyphenyl)pyrimidin-2-amine (12)
Anal. RP-HPLC: tκ 13.60 min. (10-70% MeCN).
1H NMR (DMSOd6, 500 MHz): δH 3.64 (3H, s, CH3), 3.75 (6H5 s5 2 x CH3), 7.13 (IH5 dd,
J 7.5, Ar-H), 7.19 (IH5 dd, J 7.5, Ar-H), 7.23-7.26 (3H5 m, pyrim-H and 2 x Ar-H)5 7.46
(IH, d, J 7.5, Ar-H), 8.30 (IH, s, Ar-H), 8.32 (IH, d, J 5.O5 pyrim-H), 8.58 (IH, d, J 7.5,
Ar-H), 9.24 (IH5 s, NH) and 11.76 (IH, s, NH).
MS (ESl+) m/z 376.97 [M+H]+ (C21H20N4O3 requires 376.41).
4-(lH-indol-3-yl)-N-(3-methoxyl-4~morpholin-4-ylphenyl)pyrimidin-2-amine (13)
Anal. RP-HPLC: tR 10.67 min. (10-70% MeCN).
1H NMR (DMSO-d65 500 MHz): δH 2.93 (4H5 dd, J 5.0, 2 x CH2), 3.72 (4H, dd, J 5.0, 2 x
CH2), 3.76 (3H, S5 CH3), 6.85 (IH5 d, J 9.0, Ar-H)5 7.12 (IH5 dd, J 7.5, Ar-H), 7.17-7.23
(2H5 m, pyrim-H and Ar-H), 7.37 (IH, dd, J 9.0, 2.0, Ar-H), 7.45-7.49 (2H, m, 2 x Ar-H),
8.28 (IH, s, Ar-H)5 8.30 (IH5 d, J 5.O5 pyrim-H)5 8.58 (IH5 d, J 7.5, Ar-H), 9.20 (IH, s,
NH) and 11.76 (IH, s, NH).
MS (ESf) m/z 402.03 [M]+ (C23H23N5O2 requires 401.46).
N-(3,5-dimethoxyphenyl)-4-(lH-indol-3~yl)pyrimidin-2-amine (14)
Anal. RP-HPLC: tR 15.06 min. (10-70% MeCN).
1H NMR (DMSO-d6, 500 MHz): δH 3.73 (6H5 s, 2 x CH3), 6.13 (IH5 s, Ar-H), 7.14 (3H, s,
3 x Ar-H), 7.19 (IH5 dd, J 8.0, Ar-H), 7.28 (IH, d, J 5.5, pyrim-H), 7.46 (IH, d, J 8.0, Ar-
H), 8.30 (IH, s, Ar-H), 8.34 (IH, d, J5.5, pyrim-H), 8.62 (IH, dd, J 8.0, Ar-H)5 9.33 (IH,
S5 NH) and 11.79 (IH, s, NH). MS (ESI+) m/z 344.90 [M]" (C20H18N4O2 requires 346.38).
4-(l-methyl-lH-indol-3-yl)-N-(4-morpholin-4-ylphenyl)pyrimidin-2-amine (15)
Anal. RP-HPLC: tR 13.64 min. (10-70% MeCN).
1H NMR (DMSO-d6j 500 MHz): δH 3.06 (4H5 dd, J 5.0, 2 x CH2), 3.75 (4H, dd, J 5.0, 2 x
CH2), 3.88 (3H, s, CH3), 6.93 (2H, d, J 8.0, 2 x Ar-H), 7.12 (IH, d, J 5.5, pyrim-H), 7.18
(IH, dd, J 7.5, Ar-H), 7.26 (IH, dd, J 7.5, Ar-H), 7.52 (IH, d, J 7.5, Ar-H), 7.65 (2H, d, J
8.0, 2 x Ar-H), 8.26-8.28 (2H, m, pyrim-H and Ar-H), 8.58 (IH, d, J 7.5, Ar-H) and 9.13
(IH5 S5 NH).
MS (ESl+) m/z 385.98 [M+H]+ (C23H23N5O requires 385.46).
4-(l-methyl-lH-indol-3~yl)-N-(4-acetylpiperazine~l-ylphenyl)pyrimidin-2-amine (16) Anal. RP-HPLC: tR 12.95 min. (10-70% MeCN).
1H NMR (DMSO-d6, 500 MHz): δH 2.04 (3H, s, CH3), 3.02 (2H, dd, J5.0, 2 x CHH), 3.09 (2H, dd, J 5.0, 2 x CHH), 3.59 (4H, q, J 5.0, 2 x CH2), 3.88 (3H, s, CH3), 6.95 (2H, d, J 9.5, 2 x Ar-H), 7.12 (IH, d, J 5.5, pyrim-H), 7.18 (IH, dd, J 7.5, Ar-H), 7.26 (IH, dd, J 7.5, Ar-H)5 7.52 (IH, d, J 7.5, Ar-H), 7.66 (2H, d, J 9.5, 2 x Ar-H), 8.26-8.28 (2H, m, incl. J5.5, pyrim-H and Ar-H)5 8.58 (IH, d, J7.5, Ar-H) and 9.15 (IH, s, NH). MS (ESI+) m/z 426.91 [M+H]+ (C25H26N6O requires 426.51).
N-1, 3-benzodioxol-5-yl-4-(lH-indol-3-yl)pyrimidin-2-amine (17)
Anal. RP-HPLC: tκ 12.95 min. (10-70% MeCN).
1H NMR (DMSO-d6, 500 MHz): δH 5.99 (2H, s, CH2), 6.86 (IH5 d, J 8.5, Ar-H), 7.13 (IH, dd, J 7.5, Ar-H)5 7.17-7.20 (2H, m, 2 x Ar-H), 7.22 (IH, d, J 5.5, pyrim-H), 7.46 (IH, d,
7.5, Ar-H), 7.55 (IH, s, Ar-H), 8.28 (IH5 s, Ar-H)5 8.29 (IH, d, J 5.5, pyrim-H), 8.57 (IH5 d, J7.5, Ar-H), 9.25 (IH, s, NH) and 11.76 (IH5 s, NH).
MS (ESI+) m/z 331.01 [M+H]+ (Ci9H14N4O2 requires 330.34). 4-[l-(cyclopropylcarbonyl)-lH-indol-3-yl]-N-(4-morpholin-4ylphenyl)pyrimidin-2-amine
(18)
Anal. RP-HPLC: tκ 14.35 min. (10-70% MeCN).
1H NMR (DMSOd6, 500 MHz): δH 1.14-1.20 (4H5 m, 2 x cycloprop-CH2), 2.87-2.91 (IH, m, cycloprop-CH), 3.04-3.08 (4H5 m5 2 x CH2), 3.74-3.77 (4H, m, 2 x CH2), 6.92-6.95
(2H5 m5 2 x Ar-H), 7.34 (IH5 dd, J 7.5, Ar-H)5 7.40 (IH5 dd, J 7.5, Ar-H)5 7.44 (IH, d, J
5.5, pyrim-H), 7.61-7.65 (2H5 m, 2 x Ar-H), 8.40 (IH, d, J 8.5, Ar-H)5 8.43 (IH5 d, J 5.5, pyrim-H), 8.72 (IH5 dd, J7.5, Ar-H), 9.10 (IH, S5 Ar-H) and 9.35 (IH, s, NH).
MS (ESI+) m/z 439.89 [M]+ (C26H25N5O2 requires 439.51).
4-(l -acetyl- lH-indol-3-yl)-N-(4-morpholin-4ylphenyl)pyrimidin-2-amine (19)
Anal. RP-HPLC: tR 14.44 min. (10-70% MeCN).
1H NMR (DMSOd6, 500 MHz): δH 2.76 (3H, s, CH3), 3.06 (4H, dd, J 5.0, 2 x CH2), 3.75
(4H5 dd, J 5.0, 2 x CH2), 6.94 (2H, d, J 8.5, 2 x Ar-H)5 7.34 (IH, dd, J 8.5, Ar-H), 7.38-
7.42 (2H, m, incl. J 5.0, pyrim-H and Ar-H), 7.63 (2H, d5 J 8.5, 2 x Ar-H)5 8.40 (IH, d5 J
8.5, Ar-H), 8.43 (IH, d, J 5.0, pyrim-H), 8.65-8.67 (IH, m, Ar-H)5 8.73 (IH5 s, Ar-H) and
9.35 (1H5 S5 NH).
MS (ESI+) m/z 414.95 [M+H]+ (C24H23N5O2 requires 413.47).
4-(lH-indol-3-yl)-N-(4-methylpiperazin-l-ylphenyl)pyrimidin-2-amine (20)
Anal. RP-HPLC: tR 9.89 min. (10-70% MeCN).
1H NMR (DMSO-d6, 500 MHz): δH 2.23 (3H5 s, CH3), 2.47 (4H, dd, J 5.0, 2 x CH2), 3.08
(4H, dd, J 5.0, 2 x CH2), 6.91 (2H, d, J 9.0, 2 x Ar-H), 7.12 (IH5 dd, J 8.0, Ar-H), 7.16-
7.20 (2H, m, incl. J 5.0, pyrim-H and Ar-H), 7.45 (IH, d, J 8.0, Ar-H)5 7.63 (2H, d, J 9.0, 2 x Ar-H)5 8.26 (IH, d, J 5.0, pyrim-H), 8.27 (IH5 s, Ar-H), 8.75 (IH, d, J 8.0, Ar-H), 9.10
(IH, s, Ar-H) and 11.73 (IH, s, NH).
MS (ESI+) m/z 385.02 [M+H]+ (C23H24N6 requires 384.48).
4-(7-methoxy-lH-indol-3-yl)-N-(4-acetylpiperazin-l-ylphenyl)pyrimidin-2-amine (21) ; Anal. RP-HPLC: tκ 9.89 min. (10-70% MeCN). 1H NMR (DMSOd6, 500 MHz): δH 2.05 (3H, s, CH3), 3.02 (2H, dd, J 5.0, 2 x CHH), 3.09
(2H, dd, J 5.0, 2 x CHH), 3.59 (4H, dd, J 5.0, 2 x CH2), 3.94 (3H, s, CH3), 6.76 (IH, d, J 7.5, Ar-H), 6.94 (2H, d, J 9.0, 2 x Ar-H), 7.05 (IH, dd, J 7.5, Ar-H), 7.20 (IH, d, J 5.0, pyrim-H), 7.66 (2H, d, J 9.0, 2 x Ar-H), 8.14-8.16 (2H, m, 2 x Ar-H), 8.26 (IH, d, J 5.0, pyrim-H), 9.12 (IH, s, Ar-H) and 11.87 (IH, s, NH). MS (ESI+) m/z 443.39 [M+H]+ (C25H26N6O2 requires 442.51).
4-(2-methyl-lH-indol-3~yl)-N-(4-acetylpipemzin~l-ylphenyl)]pyrimidin-2-amine (22) Anal. RP-HPLC: fe. 12.09 min. (10-70% MeCN).
1H NMR (DMSOd6, 500 MHz): δH 2.04 (3H, s, CH3), 2.99 (3H, s, CH3), 3.00 (2H, dd, J 5.0, 2 x CHH), 3.07 (2H, dd, J 5.0, 2 x CHH), 3.58 (4H, dd, J 5.0, 2 x CH2), 6.91 (2H, d, J 9.0, 2 x Ar-H), 6.98 (IH, d, J 5.0, pyrim-H), 7.06 (IH, dd, J 8.0, Ar-H), 7.10 (IH, dd, J 8.0, Ar-H), 7.36 (IH, d, J8.0, Ar-H), 7.67 (2H, d, J9.0, 2 x Ar-H), 8.15 (IH, d, J8.0, Ar- H), 8.34 (IH, d, J5.0, pyrim-H), 9.14 (IH, s, Ar-H) and 11.56 (IH, s, NH). MS (ESI+) m/z 427. ,39 [M+H]+ (C25H26N6O requires 426.51).
4-(7-methyl-lH-indol-3-yl)-N-(4-acetylpiperazin-l-ylphenyl)]pyrimidin-2-amine (23) Anal. RP-HPLC: fe 12.00 min. (10-70% MeCN).
1H NMR (DMSO-d6, 500 MHz): δH 1.99 (3H, s, CH3), 2.48 (3H, s, CH3), 2.98 (2H, dd, J 5.0, 2 x CHH), 3.05 (2H, dd, J 5.0, 2 x CHH), 3.55 (4H, dd, J 5.0, 2 x CH2), 6.91 (2H, d, J 9.0, 2 x Ar-H), 6.94 (IH, d, J 7.5, Ar-H), 7.00 (IH, dd, J 7.5, Ar-H), 7.18 (IH, d, J 5.0, pyrim-H), 7.62 (2H, d, J 9.0, 2 x Ar-H), 8.21-8.23 (2H, m, incl. J 5.0, pyrim-H and Ar-H), 8.37 (IH, d, J7.5, Ar-H), 9.09 (IH, s, Ar-H) and 11.67 (IH, s, NH). MS (ESI+) m/z 427.38 [M+H]+ (C25H26N6O requires 426.51).
4-(6-methoxy-lH4ndol-3-yl)-N-(4-acetylpiperazin-l-ylphenyl)]pyrimidin-2-amine (24) Anal. RP-HPLC: tR 11.27 min. (10-70% MeCN).
1H NMR (DMSO-d6, 500 MHz): δH 2.01 (3H5 s, CH3), 2.99 (2H, dd, J 5.0, 2 x CHH), 3.06 (2H, dd, J5.0, 2 x CHH), 3.55 (4H, dd, J 5.0, 2 x CH2), 3.76 (3H, s, CH3), 6.73 (IH, dd, J 9.0, 2.0, Ar-H), 6.90-6.92 (3H, m, incl. J 9.0, 3 x Ar-H), 7.12 (IH, d, J 5.0, pyrim-H), 7.62 (2H5 d, J 9.O5 2 x Ar-H), 8.10 (IH, d, J 2.0 Ar-H), 8.21 (IH, d, J 5.0, pyrim-H), 8.40 (IH, d, J9.0, Ar-H), 9.08 (IH, s, Ar-H) and 11.50 (IH, s, NH). MS (ESf) m/z 443.28 [M+H]+ (C25H26N6O2 requires 442.51).
4-(7-chloro-lH-indol-3-yl)-N-(4-acetylpiperazin-l-ylphenyl)]pyrimidin-2-amine (25) Anal. RP-HPLC: fe. 11.27 min. (10-70% MeCN).
1H NMR (DMSOd6, 500 MHz): δH 2.05 (3H, s, CH3), 3.03 (2H, dd, J 5.0, 2 x CHH), 3.10 (2H, dd, J5.0, 2 x CHH), 3.60 (4H5 dd, J5.0, 2 x CH2), 6.95 (2H, d, J9.5, 2 x Ar-H)5 7.13 (IH5 dd, J 8.0, Ar-H)5 7.25-7.29 (2H5 m5 incl. J 5.O5 pyrim-H and Ar-H), 7.65 (2H, d, J 9.5,
2 x Ar-H), 8.29 (IH, d, J 5.0, pyrim-H), 8.36 (IH5 s, Ar-H)5 8.59 (IH5 d5 J 8.O5 Ar-H), 9.19 (IH, s, Ar-H) and 12.11 (IH, s, NH).
MS (ESI+) m/z 447.37 [M+H]+ (C24H23N6OCl requires 446.93).
4-(6-fluoro-lH-indol-3-yl)-N-(4-acetylpiperazin-l-ylphenyl)]pyrimidin-2-amine (26) Anal. RP-HPLC: fe 11.27 min. (10-70% MeCN).
1HNMR (DMSO-d6, 500 MHz): δH 2.05 (3H, s, CH3), 3.03 (2H, dd, J 5.0, 2 x CHH)5 3.10 (2H, dd, J 5.0, 2 x CHH), 3.59 (4H, dd, J 5.0, 2 x CH2), 6.94-7.00 (3H5 m, incl. J 10.0, 9.5,
3 x Ar-H), 7.19 (IH, d, J 5.5, pyrim-H), 7.24 (IH, dd, J 10.0, 1.0, Ar-H)5 7.64 (2H5 dd, J 9.5, 3.0, 2 x Ar-H), 8.27 (IH, d, J 5.5, pyrim-H), 8.29 (IH, d, J 3.0, Ar-H)5 8.58-8.62 (IH5 m, Ar-H), 9.17 (IH5 S5 Ar-H) and 11.79 (IH, s, NH).
MS (ESI+) m/z 431.34 [MH-H]+ (C24H23N6OF requires 430.48).
4-(lH4ndol-3-yl)-N-[(4-acetylpiperazin-l-yl)-3-methylphenyl]pyrimidin-2-amine (27) Anal. RP-HPLC: fe 13.44 min. (10-70% MeCN).
1H NMR (DMSO-d65 500 MHz): δH 2.05 (3H5 s, CH3), 2.31 (3H, s, CH3), 2.76 (2H, dd, J 5.0, 2 x CHH), 2.83 (2H, dd, J 5.0, 2 x CHH), 3.58 (4H, dd, J 5.0, 2 x CH2), 6.99 (IH, d, J 8.5, Ar-H), 7.13 (IH5 dd, J7.5, Ar-H), 7.17-7.23 (2H, m, incl. J7.5, 5.5, pyrim-H and Ar- H)5 7.46 (IH5 d, J 7.5, Ar-H), 7.50 (IH, dd, J 8.5, 2.5, Ar-H), 7.73 (IH, d, J 2.5, Ar-H)5 8.28 (2H, m, incl. J 5.5, pyrim-H and Ar-H), 8.59 (IH5 d, J 7.5, Ar-H), 9.19 (IH, s, Ar-H) and 11.76 (IH5 S5 NH). MS (ESI+) m/z All 30 [M+H]+ (C25H26N6O requires 426.51).
4-(lH-indol-3-yl)-N-(3-methyl-4-thiomorpholin-4-ylphenyl)pyrimidin-2-amine (28) Anal. RP-HPLC: tR 16.86 min. (10-70% MeCN).
1H NMR (DMSO-d6, 500 MHz): δH 2.26 (3H, s, CH3), 2.76 (4H, dd, J 5.0, 2 x CH2), 3.06 (4H, dd, J 5.0, 2 x CH2), 7.00 (IH, d, J 8.5, Ar-H), 7.12 (IH, dd, J 7.5, Ar-H), 7.20 (IH, dd, J 7.5, Ar-H), 7.22 (IH, d, J 5.0, pyrim-H), 7.46 (IH, d, J 7.5, Ar-H), 7.50 (IH, dd, J 8.5, 2.0, Ar-H), 7.72 (IH, d, J 2.0, Ar-H), 8.28 (2H, m, incl. J 5.0, pyrim-H and Ar-H), 8.59 (IH, d,J7.5, Ar-H), 9.18 (IH, s, Ar-H) and 11.76 (IH, s, NH). MS (ESI+) m/z 402.31 [M+H]+ (C23H23N5S requires 401.53).
4-(lH-indol-3-yl)~N-[(2R, 6SJ-2, 6-dimethylmorpholin-4-ylphenyl]pyrimidin-2-amine (29) The cis isomer was prepared from guanidine synthesised using cis-2,6- dimethylmorpholine. Characterisation data for Compound (29) was essentially identical to Compound (9) (Compound (9) contains ~5% of Compound (30) below)
4-(lH-indol~3-yl)-N-[(2S, 6S)-2, 6-dimethylmorpholin-4-ylphenyl]pyrimidin-2-amine (30)
The tram isomer was obtained from Prep-HPLC (0.1% TFA) on batch 02 of Compound
(9).
Anal. RP-HPLC: tR 14.76 min. (10-70% MeCN).
1H NMR (DMSOd6, 500 MHz): δH 1.27 (6H, d, J 6.5, 2 x CH3), 2.81-2.86 (2H, m, 2 x
CHH), 3.16-3.21 (2H, m, 2 x CHH), 4.06-4.10 (2H, m, 2 x CHCH3), 6.98 (2H, dd, J6.5, 2 x Ar-H), 7.13 (IH, dd, J 7.5, Ar-H), 7.21 (IH, dd, J 7.5, Ar-H), 7.30 (IH, br s, pyrim-H),
7.52-7.56 (2H, m, 2 x Ar-H), 7.48 (IH, d, J7.5, Ar-H), 8.19 (IH, br s, pyrim-H), 8.46 (2H, dd, J6.5, 2 x Ar-H), 9.65 (IH, br s, NH) and 12.03 (IH, s, NH).
MS (ESt) m/z 400.35 [M+H]+ (C24H25N5O requires 399.49).
4-(lH-indol-3-yl)-N-(3,5-dimethylpiperidin-l-ylphenyl)pyrimidin-2-amine (31)
The 4:1 cis:trans mixture obtained was the result of a cyclisation with a guanidine containing a 4:1 cis:trans ratio of diastereoisomers. Anal. RP-HPLC: tR 11.95 min. (cis; 90%), 12.54 min. (trans; 10%) (10-70% MeCN).
1H NMR (DMSO-d6, 500 MHz): δH (cis) 0.91 (6H, d, J 6.5, 2 x CH3), 1.69-1.77 (2H, m, CH2), 2.11 (2H, dd, J 11.5, 2 x CHH), 3.55 (2H, d, J 11.5, 2 x CHH), 3.56-4.17 (2H, m, 2 x CHCH3), 6.90 (2H5 d, J 8.5, 2 x Ar-H), 7.11 (IH, dd, J 8.5, 2 x Ar-H), 7.17-7.20 (2H, m, incl. J 8.5, Ar-H and pyrim-H), 7.45 (IH, d, J 8.5, Ar-H), 7.61 (2H, d, J 8.5, 2 x Ar-H), 7.66-7.73 (2H, m, 2 x Ar-H), 8.26 (2H, d, incl. J 5.5, pyrim-H and Ar-H), 8.57 (IH, d, J 7.5, Ar-H), 8.46 (2H, dd, J 6.5, 2 x Ar-H), 9.07 (IH, s, NH) and 11.73 (IH, s, NH); δH (trans - observable signals) 1.00 (6H, d, J 6.5, 2 x CH3), 1.60-1.64 (2H, m, CH2), 1.95-2.05 (2H, m, 2 x CHH), 2.72 (2H, dd, J 11.5, 6.5, 2 x CHH), 3.08 (2H, dd, J 11.5, 4.0, 2 x CHH). MS (ESI+) m/z 398.26 [M+H]+ (C25H27N5 requires 397.52).
Example 6 Kinase assays
The compounds from the examples above were investigated for their ability to inhibit the enzymatic activity of various protein kinases. This was achieved by measurement of incorporation of radioactive phosphate from ATP into appropriate polypeptide substrates. Recombinant protein kinases and kinase complexes were produced or obtained commercially. Assays were performed using 96-well plates and appropriate assay buffers (typically 25 mM β-glycerophosphate, 20 mM MOPS, 5 mM EGTA, 1 mM DTT, 1 mM Na3VO3, pH 7.4), into which were added 2 - 4 μg of active enzyme with appropriate substrates. The reactions were initiated by addition of Mg/ATP mix (15 mM MgCl2 + 100 μM ATP with 30-50 kBq per well of [γ-32P]-ATP) and mixtures incubated as required at 30 0C. Reactions were stopped on ice, followed by filtration through p81 filterplates or GF/C filterplates (Whatman Polyfiltronics, Kent, UK). After washing 3 times with 75 mM aq orthophosphoric acid, plates were dried, scintillant added and incorporated radioactivity measured in a scintillation counter (TopCount, Packard Instruments, Pangbourne, Berks, UK). Compounds for kinase assay were made up as 10 mM stocks in DMSO and diluted into 10 % DMSO in assay buffer. Data was analysed using curve-fitting software (GraphPad Prism version 3.00 for Windows, GraphPad Software, San Diego California USA) to determine ICs0 values (concentration of test compound which inhibits kinase activity by 50 %.). Results for representative example compounds are summarised in Tables 2 and 3.
CDK 7 and 9 assays
CTD peptide substrate (biotinyl-Ahx-(Tyr-Ser-Pro-Thr-Ser-Pro-Ser)4-NH2; 1 - 2 mg/mL) and recombinant human CDK7/cyclin H, CDK9/cyclin Tl, or CDK9/cyclin K (0.5 - 2 μg) were incubated for 45 min at 30 °C in the presence of varying amounts of test compound in 20 mM MOPS pH 7.2, 25mM β-glycerophosphate, 5 roM EGTA, 1 mM DTT, ImM sodium vanadate, 15 mM MgCl2, and 100 μM ATP (containing a trace amount of 32PyATP) in a total volume of 25 μL in a 96-well microtiter plate. The reaction was stopped by placing the plate on ice for 2 min. Avidin (50 μg) was added to each well, and the plate was incubated at room temp for 30 min. The samples were transferred to a 96- well P81 filter plate, and washed (4 x 200 μL per well) with 75 mM phosphoric acid. Microscint 40 scintillation liquid (50 μL) was added to each well, and the amount of 32P incorporation for each sample was measured using a Packard Topcount microplate scintillation counter. Results for representative example compounds are summarised in Tables 2 and 3.
Aurora- A (human) kinase assay
This was achieved by measurement of incorporation of radioactive phosphate froπTATP into Kemptide substrate (LRRASLG), upon phosphorylation by commercially obtained aurora-A (human, Upstate, Dundee, UK). Assays were performed using 96-well plates and appropriate assay buffers (2OmM Tris, 25mM β-glycerophosphate, 5mM EGTA, ImM DTT, ImM sodium vanadate, pH 7.5), into which were added 2-5ng of active enzyme with 500 μM substrate (Kemptide). The reactions were initiated by addition of MgATP mix (15mM MgCl2 + lOOμM ATP with 15-25 kBq per well of [7-32P]-ATP) and mixtures incubated for 30 min at 30°C. Reactions were stopped by addition of an equal volume of 75 mM aq orthophosphoric acid, followed by filtration through p81 fϊlterplates (Whatman Polyfiltronics, Kent, UK). After washing 4 times with 75 mM aq orthophosphoric acid, plates were dried, scintillant added and incorporated radioactivity measured in a scintillation counter (TopCount, Packard Instruments, Pangbourne, Berks, UK). Compounds for kinase assay were made up as 10 mM stocks in DMSO and diluted into 10 % DMSO in assay buffer. Data was analysed using curve-fitting software (XLfit version 4.0.2, IDBS, Guildford, Surrey, UK) to determine IC5O values (concentration of test compound which inhibits kinase activity by 50 %).
Aurora-B (human) kinase assay
This was achieved by measurement of incorporation of radioactive phosphate from ATP into Kemptide substrate (LRRASLG), upon phosphorylation by commercially obtained aurora-B (human, Upstate, Dundee, UK). Assays were performed using 96-well plates and appropriate assay buffers (2OmM Tris, 25mM β-glycerophosphate, 5mM EGTA, ImM DTT, ImM sodium vanadate, pH 7.5), into which were added 75ng of pre-activated enzyme with 500 μM substrate (Kemptide). The reactions were initiated by addition of MgATP mix (15mM MgCl2 + lOOμM ATP with 15-25 kBq per well of [γ-32P]-ATP) and mixtures incubated for 60 min at 3O0C. Reactions were stopped by addition of an equal volume of 75 mM aq orthophosphoric acid, followed by filtration through p81 filterplates (Whatman Polyfiltronics, Kent, UK). After washing 4 times with 75 mM aq orthophosphoric acid, plates were dried, scintillant added and incorporated radioactivity measured in a scintillation counter (TopCount, Packard Instruments, Pangbourne, Berks, UK). Compounds for kinase assay were made up as 10 mM stocks in DMSO and diluted into 10 % DMSO in assay buffer. Data was analysed using curve-fitting software (XLfit version 4.0.2, IDBS, Guildford, Surrey, UK) to determine IC50 values (concentration of test compound which inhibits kinase activity by 50 %).
Pre-Activation of Aurora-B (human)
Aurora-B (human, Upstate, Dundee, UK) was pre-activated immediately prior to kinase assay in appropriate buffers (2OmM Tris, 25mM β-glycerophosphate, 5mM EGTA, ImM DTT, ImM sodium vanadate, pH 7.5) by incubating 15 μg of enzyme with 4 μg INCENP (Upstate, Dundee, UK) in the presence of MgATP mix (15mM MgCl2 + lOOμM ATP) for
15 min at 30°C.
FU3 kinase assay
This was achieved by measurement of incorporation of radioactive phosphate from ATP into myelin basic protein (MBP) substrate, upon phosphorylation by commercially obtained Flt-3 (Upstate, Dundee, UK). Assays were performed using 96-well plates and appropriate assay buffers (2OmM Tris, 25mM β-glycerophosphate, 5mM EGTA, ImM DTT, ImM sodium vanadate, pH 7.5), into which were added 5ng of active enzyme with 0.4 mg/ml substrate (MBP). The reactions were initiated by addition of MgATP mix (15mM MgCl2 + lOOμM ATP with 15-25 kBq per well of [γ-32P]-ATP) and mixtures incubated for 30 min at 30°C. Reactions were stopped by addition of an equal volume of 75 mM aq orthophosphoric acid, followed by filtration through p81 filterplates (Whatman Polyfiltronics, Kent, UK). After washing 4 times with 75 mM aq orthophosphoric acid, plates were dried, scintillant added and incorporated radioactivity measured in a scintillation counter (TopCount, Packard Instruments, Pangbourne, Berks, UK). Compounds for kinase assay were made up as 10 mM stocks in DMSO and diluted into 10 % DMSO in assay buffer. Data was analysed using curve-fitting software (XLfit version 4.0.2, IDBS, Guildford, Surrey, UK) to determine IC50 values (concentration of test compound which inhibits kinase activity by 50 %).
GSK-3β kinase assay
GSK-3 was obtained from New England Biolabs (UK) Ltd., Hitchin, Herts. The recombinant enzyme was isolated from a strain of E. coli that carries a clone expressing GSK-3β derived from a rabbit skeletal muscle cDNA library [Wang, Q.M.; Fiol, CJ.; DePaoli-Roach, A. A.; Roach, PJ. J. Biol. Chem., 1994, 269, 14566]. Inhibition of GSK-3 function was assessed by measurement of phosphorylation of CREB phosphopeptide KRREILSRRPphosphoSYR in the presence of test compounds. Using a 96-well assay format, GSK3 (7.5U) was incubated for 30 min at 30 0C in a total volume of 25 μL in 20 mM MOPS pH 7.2, 25 mM β-glycerophosphate, 5 mM EGTA, 1 mM DTT, 1 mM Na3VO3, 40 μM CREB peptide, 15 mM MgCl2 and 100 μM ATP (containing 0.25 μCi [γ- 32P]-ATP) in the presence of varying concentrations of test compound. The samples were transferred to 96-well p81 filter plates (Whatman Polyfiltronics, Kent, UK), and the plates were washed 4 times with 200 μL/well of 75 mM aq orthophosphoric acid. Scintillation liquid (50 μL) was added to each well, and incorporated radioactivity for each sample was determined using a scintillation counter (TopCount, Packard Instruments, Pangbourne, Berks, UK). Results for representative example compounds are summarised in Tables 2 and 3.
Example 7
MTT cytotoxicity assay
The compounds of the invention were subjected to a standard cellular proliferation assay using human tumour cell lines obtained from the ATCC (American Type Culture Collection, 10801 University Boulevard, Manessas, VA 20110-2209, USA). Standard 72-h MTT (thiazolyl blue; 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide) assays were performed (Haselsberger, K.; Peterson, D. C; Thomas, D. G.; Darling, J. L. Anti Cancer Drugs 1996, 7, 331-8; Loveland, B. E.; Johns, T. G.; Mackay, I. R.; Vaillant, F.; Wang, Z. X.; Hertzog, P. J. Biochemistry International 1992, 27, 501-10). In short: cells were seeded into 96-well plates according to doubling time and incubated overnight at 37 0C. Test compounds were made up in DMSO and a 1/3 dilution series prepared in 100 μL cell media, added to cells (in triplicates) and incubated for 72 ho at 37 0C. MTT was made up as a stock of 5 mg/mL in cell media and filter-sterilised. Media was removed from cells followed by a wash with 200 μL PBS. MTT solution was then added at 20 μL per well and incubated in the dark at 37 0C for 4 h. MTT solution was removed and cells again washed with 200 μL PBS. MTT dye was solubilised with 200 μL per well of DMSO with agitation. Absorbance was read at 540 nm and data analysed using curve-fitting software (GraphPad Prism version 3.00 for Windows, GraphPad Software, San Diego California USA) to determine IC50 values (concentration of test compound which inhibits cell growth by 50 %). Results for representative example compounds are summarised in Table 4. Various modifications and variations of the described aspects of the invention will be apparent to those skilled in the art without departing from the scope and spirit of the invention. Although the invention has been described in connection with specific preferred embodiments, it should be understood that the invention as claimed should not be unduly limited to such specific embodiments. Indeed, various modifications of the described modes of carrying out the invention which are obvious to those skilled in the relevant fields are intended to be within the scope of the following claims.
Table 1: Structures of exemplified compounds
Figure imgf000059_0001
Figure imgf000060_0001
Figure imgf000061_0001
Figure imgf000062_0001
Figure imgf000063_0001
Figure imgf000064_0001
64 Table 2: Kinase inhibition of selected compounds
Figure imgf000065_0001
Table 3: Kinase inhibition of selected compounds
Figure imgf000065_0002
Figure imgf000066_0001
* Drosophila Aurora A
Table 4: MTT Cytoxicity data for cell lines Mia-PaCa-2 and A2780 (IC50 values in μM after 96 hours incubation) for selected compounds of the invention
Figure imgf000066_0002
REFERENCES
1. Manning, G.; Whyte, D. B.; Martinez, R.; Hunter, T.; Sudarsanam, S. The protein kinase complement of the human genome. Science 2002, 298, 1912-1934.
2. Kostich, M.; English, J.; Madison, V.; Gheyas, F.; Wang, L. et al. Human members of the eukaryotic protein kinase family. Genome Biology 2002, 3, research0043.0041-0043.0012.
3. Dancey, J.; Sausville, E. A. Issues and progress with protein kinase inhibitors for cancer treatment. Nat. Rev. Drug Disc. 2003, 2, 296-313.
4. Cockerill, G. S.; Lackey, K. E. Small molecule inhibitors of the class 1 receptor tyrosine kinase family. Current Topics in Medicinal Chemistry 2002, 2, 1001-1010.
5. Fabbro, D.; Ruetz, S.; Buchdunger, E.; Cowan-Jacob, S. W.; Fendrich, G. et al. Protein kinases as targets for anticancer agents: from inhibitors to useful drugs. PharmacolTher. 2002, 93, 79-98.
6. Cohen, P. Protein kinases — the major drug targets of the twenty-first century? Nat. Rev. Drug Disc. 2002, 1, 309-315.
7. Bridges, A. J. Chemical inhibitors of protein kinases. Chem.Rev. 2001, 101(8), 2541-2571.
8. Wang, S.; Meades, C; Wood, G.; Osnowski, A.; Fischer, P. M. N-(4-(4^ methylthiazol-5-yl) pyrimidin-2-yl)-N-phenylamines as antiproliferative compounds. PCT Intl. Patent Appl. Publ. WO 2003029248; Cyclacel Limited, UK.
9. Wu, S. Y.; McNae, L; Kontopidis, G.; McClue, S. J.; Mclnnes, C. et al. Discovery of a Novel Family of CDK Inhibitors with the Program LIDAEUS: Structural Basis for Ligand-Induced Disordering of the Activation Loop. Structure 2003, 11, 399- 410.
10. Fischer, P. M.; Wang, S.; Wood, G. Inhibitors of cyclin dependent kinases as anticancer agent. PCT Intl. Patent Appl. Publ. WO 02/079193; Cyclacel Limited, UK,.
11. Wang, S.; Fischer, P. M. Anti-cancer compounds. US Patent Appl. Publ. 2002/0019404. 12. Fischer, P. M.; Wang, S. 2-substituted 4-heteroaryl-pyrimidines and their use in the treatmetn of proliferative disorders. PCT Intl. Patent Appl. PuU. WO 2001072745; Cyclacel Limited, UK.
13. Knockaert, M.; Greengard, P.; Meijer, L. Pharmacological inhibitors of cyclin- dependent kinases. Trends Pharmacol. Set 2002, 23, 417 -425.
14. Fischer, P. M.; Endicott, J.; Meijer, L. Cyclin-dependent kinase inhibitors. Progress in Cell Cycle Research; Editions de Ia Station Biologique de Roscoff: Roscoff, France, 2003; pp 235-248.
15. Fravolini, A.; Grandolini, G.; Martani, A. New heterocyclic ring systems from α- hydroxymethylene ketones. V. Reaction of 2-methyl-6-hydroxymethylene-4,5,6,7- tetrahydrobenzothiazol-7-one with amines and amidines. Gazz. CMm. Ital. 1973, 103, 1063-1071.
16. Cleaver, L.; Croft, J. A.; Ritchie, E.; Taylor, W. C. Chemical studies of the Proteaceae. IX. Synthesis of 5-alkylresorcinols from aliphatic precursors. Aust. J. Chem. 1976, 29, 1989-2001.
17. Fadda, A. A.; El-Houssini, M. S. Synthesis of cyclic ketones by activated nitriles. J. Ind. Chem. Soc. 1990, 67, 915-917.
18. Kost, A. N.; Ovseneva, L. G. Synthesis of 4-substituted dihydroresorcinols. Zh. Obshch. KUm 1962, 32, 3983-3986.
19. Lehmann, G.; Luecke, B.; Schick, H.; Hilgetag, G. 2-Substituted 7-oxo-4,5,6,7- tetrahydrobenzothiazoles. Z. Chem. 1967, 7, 422.
20. Bell, R. P.; Davis, G. G. Kinetics of the bromination of some enols and their anions. J. Chem. Soc 1965, 353-361.
21. Fravolini, A.; Grandolini, G.; Martani, A. New heterocyclic ring systems from α- hydroxymethylene ketones. III. Pyrazolobenzothiazoles and thiazolo- benzoisoxazoles. Gazz. Chim. Ital. 1973, 103, 755-769.
22. Bredereck, H.; Effenberger, F.; Botsch, H. Acid amide reactions. XLV. Reactivity of formamidines, dimethylformamide diethyl acetal (amide acetal), and bis(dimethylamino)methoxymethane (aminal ester). Chem. Ber. 1964, 97, 3397- 3406. 23. Wang D, De Ia Fuente C, Deng L, Wang L, Zilberman I5 Eadie C, Healey M, Stein D, Denny T, Harrison LE, Meijer L, Kashanchi F. Inhibition of human immunodeficiency virus type 1 transcription by chemical cyclin-dependent kinase inhibitors. J. Virol. 2001; 75: 7266-7279.
24. Chen, Y.H.; Hansen, L.; Chen, M.X.; Bjorbaek, C; Vestergaard, H.; Hansen, T.; Cohen, P.T.; Pedersen, O. Diabetes, 1994, 43, 1234.
25. Nikoulina, S.E.; Ciaraldi, T.P.; Mudaliar, S.; Mohideen, P.; Carter, L.; Henry, R.R. Diabetes, 2000, 49, 263.
26. Goedert, M. Curr. Opin. Gen. Dev., 2001, 11, 343.
27. Mattson, M.P. Nat. Rev. MoI Cell. Biol, 2000, 1, 120.
28. Zhu, A. J.; Watt, F.M. Development, 1999, 126, 2285.
29. DasGupta, R.; Fuchs, E. Development, 1999, 126, 4557.
30. Sunkel et al., J. Cell ScL, 1988, 89, 25.
31. Llamazares et al., Genes Dev., 1991, 5, 2153.
32. Glover et al., Genes Dev., 1998, 12, 3777.
33. Lee et al., Proc. Natl. Acad. Set USA, 1998, 95, 9301.
34. Leung et al., Nat. Struct. Biol, 2002, 9, 719.
35. Kauselmann et al., EMBO J., 1999, 18, 5528.
36. Nigg, Curr. Opin. Cell Biol, 1998, 10, 776.
37. Yuan et al., Cancer Res., 2002, 62, 4186.
38. Seong et al., J. Biol. Chem., 2002, 277, 32282.
39. Lane et al., J. Cell. Biol, 1996, 135, 1701.
40. Cogswell et al., Cell Growth Differ., 2000, 11, 615.
41. Liu et al., Proc. Natl. Acad ScI USA, 2002, 99, 8672.
42. Toyoshima-Morimoto et al., Nature, 2001 , 410, 215.
43. Roshak et al., Cell. Signalling, 2000, 12, 405.
44. Smits et al., Nat. Cell Biol, 2000, 2, 672.
45. van Vugt et al., J. Biol. Chem., 2001, 276, 41656.
46. Sumara et al., MoI. Cell, 2002, 9, 515.
47. Golan et al., J. Biol Chem., 2002, 277, 15552. 48. Kotani et al., MoI. Cell, 1998, 1, 371.
49. Feng et al., Cell Growth Differ., 2001, 12, 29.
50. Dai et al., Oncogene, 2002, 21, 6195.
51. Nurse, iVαrwre, 1990, 344, 503.
52. Nigg, Nat Rev. MoI. Cell Biol, 2001, 2, 21.
53. Hagting et al., EMBO J, 1998, 17, 4127.
54. Hagting et al., Curr. Biol, 1999, 9, 680.
55. Yang et al., J 5to/. Chem., 2001 , 276, 3604.
56. Takizawa et al., Cwrr. Opin. Cell Biol, 2000, 12, 658.
57. Seki et al., MoI. Biol. Cell, 1992, 3, 1373.
58. Heald et al., Cell, 1993, 74, 463.
59. Dalai et al., MoI. Cell. Biol, 1999, 19, 4465.
60. Toyoshima-Morimoto et al., Nature, 2001, 410, 215.
61. Toyoshima-Morimoto et al., EMBO Rep., 2002, 3, 341.
62. Wang et al., MoI. Cell. Biol, 2002, 22, 3450.
63. Tyrrell, E.; Brookes, P. Synthesis 2003, 469-483.
64. Molander, G. A.; Biolatto, B. J. Org. Chem. 2003, 68, 4302-4314.
65. Bredereck, H.; Effenberger, F.; Botsch, H. Chem. Ber. 1964, 97, 3397-3406.
66. Zimmermann, J.; Caravatti, G.; Mett, H.; Meyer, T.; Muller, M. et al. Arch. Pharm. Pharm. Med. Chem. 1996, 329, 371-376.
67. Haselsberger, K.; Peterson, D. C; Thomas, D. G.; Darling, J. L. Anti Cancer Drugs 1996, 7, 331-8.
68. Loveland, B. E.; Johns, T. G.; Mackay, I. R.; Vaillant, F.; Wang, Z. X.; Hertzog, P. J. Biochemistry International 1992, 27, 501-10.
69. Joule, J.A.; Smith, G.F. Heterocyclic Chemistry, Van Nostrand Reinhold (UK) Co. Ltd.: Wokingham, 1983.
70. Bredereck, H.; Effenberger, F.; Botsch, H. Chem. Ber., 1964, 97, 3397.
71. Cohen, P. Nat. Rev. Drug Disc. , 2002, 1, 309.
72. Fischer, P.M. Curr. Med. Chem., 2004, 11, 1563. 73. Kidwai, M.; Rastogi, S.; Saxena, S. Bulletin of the Korean Chemical Society, 2003,
24, 1575.
74. Zimmermann, J.; Caravatti, G.; Mett, H.; Meyer, T.; Mueller, M.; Lydon, N.B.; Fabbro, D. Arch. Pharm. (Weinheim), 1996, 329, 371.
75. Carmena, M.; Earnshaw, W.C. Nat. Rev. MoI Cell Biol. , 2003, 4, 842.
76. Stirewalt, D.L.; Radich, J.P. Nat. Rev. Cancer, 2003, 3, 650.
77. Fischer, P.M.; Endicott, J.; Meijer, L. Progr. Cell Cycle Res. , 2003, 5, 235.
78. Cohen, P.; Goedert, M. Nat. Rev. Drug Disc. , 2004, 3, 479.
79. Reilly, J.T. Leukemia & Lymphoma, 2003, 44, 1.

Claims

1. A compound of formula I, or a pharmaceutically acceptable salt thereof,
Figure imgf000072_0001
wherein R3, R4, R5, R6, R7, R8, R9, and R10 are each independently H, R11 or R12;
R1 and R2 are each independently H, R11 or R12; or R1 and R2 are linked to form a cyclic group together with the nitrogen to which they are attached, and wherein said cyclic group is optionally substituted with one or more R11 or R12 groups; each R11 is independently a hydrocarbyl group optionally substituted by one or more R12 substituents; each R12 is independently selected from OR13, COR13, COOR13, CN, CONR13R14,
NR13R14, SR13, SOR13, SO2R13, SO2OR13, SO2NR13R14, R13, halogen, CF3, NO2 and an alicyclic group itself optionally substituted by one or more R or R groups; and each R13 and each R14 are independently H or (CH2)nR15, where n is 0, 1, 2, or 3; and each R15 is independently selected from alkyl, cycloalkyl, aryl, heteroaryl, aryl and an alicyclic group; with the proviso that the compound is other than:
^-(lH-indol-S-yO-pyrimidin^-ylJ-tS^ljl^^-tetrafluoroethoxyphenyOJ-amine;
3-[6-(4-bromophenyl)-2-(l-piperazinyl)-4-pyrimidinyl]-lH-indole;
3-[6-(4-bromophenyl)-2-(l-pyrrolidinyl)-4-pyrimidinyl]-lH-indole; or
3-[6-(4-bromophenyl)-2-(4-morpholinyl)-4-pyrimidinyl]-lH-indole.
2. A compound according to claim 1 wherein R1 and R2 are each independently H, R11 or R ; or R and R are linked to form a cyclic group together with the nitrogen to which they are attached, wherein said cyclic group contains from two to nine carbon atoms and one or two heteroatoms selected from N, O, and S, and wherein said cyclic group is optionally substituted with one or two substituents selected from R11 and R12.
3. A compound according to claim 1 or claim 2 wherein R1 and R2 are each independently H, R11 or R12.
4. A compound according to any preceding claim wherein R1 and R2 are each independently H or R11.
5. A compound according to any preceding claim wherein one of R1 and R2 is H and the other is R11.
6. A compound according to any preceding claim wherein R11 is a hydrocarbyl group containing from 1 to 24 carbon atoms, optionally containing up to six heteroatoms selected from N, O, and S.
7. A compound according to claim 6 wherein the hydrocarbyl group is optionally substituted by up to six R12 substituents.
8. A compound according to any one of claims 1 to 6 wherein R11 is an aryl group, a heteroaryl group, an aryl-alicyclic group or an alicyclic group, each of which may be optionally substituted by one or more R12 substituents.
9. A compound according to any preceding claim wherein R11 is selected from phenyl, pyridinyl and
Figure imgf000073_0001
each of which may be optionally substituted by one or more R12 substituents.
10. A compound according to any preceding claim wherein R11 is a phenyl or pyridinyl group, each of which may be optionally substituted by one or more R12 substituents.
11. A compound according to any preceding claim wherein R3, R4, R5, R6, R7, R8, R9, and R10 are each independently H or R12.
12. A compound according to any preceding claim wherein R3 and R4 are both H.
13. A compound according to any preceding claim wherein: R9, and R10 are both H;
R5 is H or alkyl;
R6 is H, alkyl, CO-alkyl or CO-cycloalkyl;
R7 is H5 alkyl, alkoxy or halo; and
R8 is H, alkoxy or halo. >
14. A compound according to any preceding claim wherein each R15 is independently selected from ethyl, ethyl, isopropyl, n-butyl, isobutyl, t-butyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, phenyl, pyridinyl, pyrrolidinyl, pyrrolyl, morpholinyl, piperazinyl, piperidinyl, triazolyl, tetrazolyl and thiazolyl.
15. A compound according to any preceding claim wherein the alicyclic group contains one or more heteroatoms.
16. A compound according to any preceding claim wherein R12 is an alicyclic group optionally substituted by one or more R13 or COR13 groups.
17. A compound according to claim 16 wherein R12 is a morpholinyl, piperazinyl, thiomorpholinyl or piperidinyl group optionally substituted by one or more R13 or COR13 groups.
18. A compound according to claim 17 wherein R12 is a morpholinyl, piperazinyl, thiomorpholinyl or piperidinyl group optionally substituted by one or more alkyl, aralkyl or CO-alkyl groups.
19. A compound according to claim 18 wherein R12 is a morpholinyl, piperazinyl, thiomorpholinyl or piperidinyl group optionally substituted by one or more methyl, benzyl or COMe groups.
20. A compound according to claim 19 wherein R12 is selected from the following:
Figure imgf000075_0001
21. A compound according to any one of claims 1 to 15 wherein each R12 is independently selected from OH, OMe, COMe, CHO, CO2Me5 COOH5 CN, CONH2, NHMe, NH2, NMe2, SH, SMe, SOMe, SO2Me, SO2NHMe, SO2NH2, Cl, Br, F, I, CF3, NO2, N-morpholinyl, N-pyrrolidinyl, N-piperazinyl, N-thiomorpholinyl, 2,6- dimethylmorpholin-4-yl, 4-benzylpiperazin-l-yl, 3,5-dimethylpiperidin-l-yl and 4- acetylpiperazin- 1 -yl.
22. A compound according to claim 1 of formula Ic, or a pharmaceutically acceptable salt thereof,
Figure imgf000076_0001
Ic wherein
R " are as defined in claim 1 ;
Z is N or CR 2z0υ;. and
R , 1160-"2^0 are each independently H, R l1lOΛ«r r R» 1 I2A
23. A compound according to claim 22 wherein Z is N.
24. A compound according to claim 22 wherein Z is CR 20.
25. A compound according to claim 22 wherein R16"20 are each independently selected from H, NO2, NR13R14, halogen, alkoxy and an optionally substituted heteroalicyclic group.
26. A compound according to claim 25 wherein R16'20 are each independently selected from H, NO2, F, OMe, N-morpholinyl, NH2, N-pyrrolidinyl, N-piperazinyl, N- thiomorpholinyl, 2,6-dimethylmorpholin-4-yl, 4-benzylpiperazin-l-yl, 3,5-dimethyl- piperidin- 1 -yl and 4-acetylpiperazin- 1 -yl .
27. A compound according to claim 1 which is selected from the following:
4-(lH-indol-3-yl)-N-(3-nitrophenyl)pyrimidin-2-amine;
N-(4-fluorophenyl)-4-(lH-indol-3-yl)pyrimidin-2-amine;
4-(lH-indol-3-yl)-N-(6-methoxypyridin-3-yl)pyrimidin-2-amine;
4-(l H-indol-3 -yl)-N-(4-morpholin-4-ylphenyl)pyrimidin-2-amine;
4-(lH-indol-3-yl)-N-(4-acetylpiperazin-l-ylphenyl)pyrimidin-2-amine;
4-(lH-indol-3-yl)-N-(4-piperazin-l-ylphenyl)pyrimidin-2-amine;
4-(lH-indol-3-yl)-N-(4-benzylpiperazin-l-ylphenyl)pyrimidin-2-amine;
4-(lH-indol-3-yl)-N-(2,6-dimethylmorpholin-4-ylphenyl)pyrimidin-2-amine;
N'-[4-(lH-indol-3-yl)pyrimidin-2-yl]-N,N-dimethylbenzene-l,4-diamine;
4-(lH-indol-3-yl)-N-(2-memyl-4-moφholm-4-ylphenyl)pyrimidin-2-amine;
4-(lH-indol-3-yl)-N-(3,4,5-trimethoxyphenyl)pyrimidin-2-amine;
4-(lH-indol-3-yl)-N-(3-methoxyl-4-morpholin-4-ylphenyl)pyrimidin-2-amine;
N-(3 , 5 -dimethoxyphenyl)-4-( 1 H-indol-3 -yl)pyrimidin-2-amine;
4-(l-methyl-lH-indol-3-yl)-N-(4-morpholin-4-ylphenyl)pyrimidin-2-amine;
4-(l-methyl-lH-indol-3-yl)-N-(4-acetylpiperazine-l-ylρhenyl)pyrimidin-2-amine;
N-l,3-benzodioxol-5-yl-4-(lH-indol-3-yl)pyrimidin-2-amine;
4- [ 1 -(cyclopropylcarbonyl)- 1 H-indol-3 -yl] -N-(4-morpholin-4ylphenyl)pyrimidin-2-amine;
4-(l -acetyl- 1 H-indol-3 -yl)-N-(4-morpholin-4ylphenyl)pyrimidin-2-amine;
4-( 1 H-indol-3 -yl)-N-(4-methylpiperazin- 1 -ylphenyl)pyrimidin-2-amine;
4-(7-methoxy-lH-indol-3-yl)-N-(4-acetylpiperazin-l-ylphenyl)pyrimidin-2-amine;
4-(2-methyl-lH-indol-3-yl)-N-(4-acetylpiperazin-l-ylphenyl)]pyrimidin-2-amine;
4-(7-memyl-lH-mdol-3-yl)-N-(4-ace1ylpiperazm-l-ylphenyl)]pyrimidin-2-amine;
4-(6-methoxy- lH-indol-3-yl)-N-(4-acetylpiperazin- 1 -ylphenyl)]pyrimidin-2-amine;
4-(7-chloro- 1 H-indol-3 -yl)-N-(4-acetylpiperazin- 1 -ylphenyl)]pyrimidin-2-amine;
4-(6-fluoro-lH-indol-3-yl)-N-(4-acetylpiperazin-l-ylphenyl)]pyrimidin-2-amine;
4-( 1 H-indol-3 -yl)-N- [(4-acetylpiperazin- 1 -yl)-3 -methylphenyl]pyrimidin-2-amine;
4-(lH-indol-3-yl)-N-(3-methyl-4-thiomorpholin-4-ylphenyl)pyrimidin-2-amine;
4-(lH-indol-3-yl)-N-[(2R56S)-2,6-dimethylmorpholin-4-ylphenyl]pyrimidin-2-amine;
4-(lH-indol-3-yl)-N-[(2S,6S)-2,6-dimethylmorpholin-4-ylphenyl]pyrimidin-2-amine;
4-(lH-indol-3-yl)-N-(3,5-dimethylpiperidin-l-ylphenyl)pyrimidin-2 -amine; and
4-(lH-indol-3-yl)pyrimidin-2-amine.
28. A compound according to any preceding claim which exhibits an IC50 value for kinase inhibition of less than 10 μM.
29. A compound according to any preceding claim which exhibits an IC50 value for kinase inhibition of less than 1 μM.
30. A compound according to any preceding claim which exhibits an ICs0 value for kinase inhibition of less than 0.1 μM.
31. A pharmaceutical composition comprising a compound according to any preceding claim admixed with a pharmaceutically acceptable diluent, excipient or carrier.
32. A compound of formula I, or pharmaceutically acceptable salt thereof, as defined in any one of claims 1 to 30, for use in medicine.
33. Use of a compound of formula Ia, or a pharmaceutically acceptable salt thereof,
Figure imgf000078_0001
Ia
wherein R3, R4, R5, R6, R7, R8, R9, and R10 are each independently H, R11 or R12; R and R are each independently H, R or R , or R and R are linked to form a cyclic group together with the nitrogen to which they are attached, and wherein said cyclic group is optionally substituted with one or more R11 or R12 groups; each R11 is independently a hydrocarbyl group optionally substituted by one or more R12 substituents; each R12 is independently selected from OR13, COR13, COOR13, CN5 CONR13R14;
NR13R14, SR13, SOR13, SO2R13, SO2OR13, SO2NR13R14, R13, halogen, CF3, NO2 and an alicyclic group itself optionally substituted by one or more R12 or R13 groups; and each R13 and each R14 are independently H or (CH2)nR15, where n is 0, 1, 2, or 3; and each R15 is independently selected from alkyl, cycloalkyl, aryl, heteroaryl, aryl and an alicyclic group; with the proviso that the compound is other than [4-(lH-indol-3-yl)-pyrimidin-2-yl]-[3-
(l,l,2,2-tetrafluoroethoxyphenyl)]-amine; in the preparation of a medicament for treating a proliferative disorder.
34. Use according to claim 33 wherein the proliferative disorder is cancer or leukemia.
35. Use according to claim 33 wherein the proliferative disorder is glomerulonephritis, rheumatoid arthritis, psoriasis or chronic obstructive pulmonary disorder.
36. Use of a compound of formula Ib, or a pharmaceutically acceptable salt thereof,
Figure imgf000079_0001
Ib
wherein R3, R4, R5, R6, R7, R8, R9, and R10 are each independently H, R11 or R12; R1 and R2 are each independently H, R11 or R12; or R1 and R2 are linked to form a cyclic group together with the nitrogen to which they are attached, and wherein said cyclic group is optionally substituted with one or more R11 or R12 groups; each R11 is independently a hydrocarbyl group optionally substituted by one or more R12 substituents; each R12 is independently selected from OR13, COR13, COOR13, CN, CONR13R14,
NR13R14, SR13, SOR13, SO2R13, SO2OR13, SO2NR13R14, R13, halogen, CF3, NO2 and an alicyclic group itself optionally substituted by one or more R12 or R13 groups; and each R13 and each R14 are independently H or (CH2)nR15, where n is 0, 1, 2, or 3; and each R15 is independently selected from alkyl, cycloalkyl, aryl, heteroaryl, aryl and an alicyclic group; in the preparation of a medicament for treating a viral disorder.
37. Use according to claim 36 wherein the viral disorder is selected from human cytomegalovirus (HCMV), herpes simplex virus type 1 (HSV-I), human immunodeficiency virus type 1 (HIV-I), and varicella zoster virus (VZV).
38. Use of a compound of formula Ib, or a pharmaceutically acceptable salt thereof, as defined in claim 36, in the preparation of a medicament for treating a CNS disorder.
39. Use according to claim 38 wherein the CNS disorder is Alzheimer's disease or bipolar disorder.
40. Use of a compound of formula Ib, or a pharmaceutically acceptable salt thereof, as defined in claim 36, in the preparation of a medicament for treating alopecia.
41. Use of a compound of formula Ib, or a pharmaceutically acceptable salt thereof, as defined in claim 36, in the preparation of a medicament for treating a stroke.
42. Use according to any one of claims 33 to 41 wherein the compound is administered in an amount sufficient to inhibit at least one CDK enzyme.
43. Use according to claim 42 wherein the CDK enzyme is CDKl, CDK2, CDK3, CDK4, CDK6, CDK7, CDK8 and/or CDK9.
44. Use according to any one of claims 33 to 41 wherein the compound is administered in an amount sufficient to inhibit aurora kinase.
45. Use according to any one of claims 33 to 41 wherein the compound is administered in an amount sufficient to inhibit FLT3.
46. Use of a compound of formula Ib, or a pharmaceutically acceptable salt thereof, as defined in claim 36, in the preparation of a medicament for treating diabetes or diabetic neuropathy.
47. Use according to claim 46 wherein the diabetes is Type II diabetes.
48. Use according to any one of claims 46 or 47 wherein the compound is administered in an amount sufficient to inhibit GSK. ;
49. Use according to claim 48 wherein the compound is administered in an amount sufficient to inhibit GSK3β.
50. Use of a compound of formula Ib, or a pharmaceutically acceptable salt thereof, as defined in claim 36, in the preparation of a medicament for treating one or more of a microbial infection, a fungal disorder, a parasitic disorder, an inflammatory disorder, and a cardiovascular disorder.
51. Use of a compound of formula Ib5 or a pharmaceutically acceptable salt thereof, as defined in claim 36, in an assay for identifying further candidate compounds capable of inhibiting one or more of a cyclin dependent kinase, an aurora kinase, FLT3, and a glycogen synthase kinase.
52. Use according to claim 51 wherein said assay is a competitive binding assay.
53. Use according to any one of claims 33 to 53 wherein the compound is as defined in any one of claims 1 to 30.
54. A process for preparing a compound of formula Ib as defined in claim 36, said process comprising the steps of:
Figure imgf000082_0001
(a) condensing a compound of formula IV with a guanidine of formula VI to form a compound of formula I; or
(b) (i) converting a compound of formula IV to a compound of formula V; and (ii) condensing said compound of formula V with a guanidine of formula VI to form a compound of formula I.
55. A process according to claim 54 wherein said compound of formula IV is prepared by acylating a compound of formula III
Figure imgf000083_0001
III
56. A process according to claim 55 wherein said compound of formula III is prepared by acylating a compound of formula II with an acid anhydride or acid halide derivative of R4CH2COOH
Figure imgf000083_0002
II
57. A process according to claim 55 wherein said compound of formula III is prepared by a process which comprises treating a compound of formula II as defined in claim 56 with (i) zinc chloride and ethylmagnesium bromide, and (ii) acetyl chloride.
58. A method of treating an aurora kinase-dependent disorder, said method comprising administering to a subject in need thereof, a compound of formula Ib as defined in claim 36, or a pharmaceutically acceptable salt thereof, in an amount sufficient to inhibit aurora kinase.
59. A method of treating a FLT3 -dependent disorder, said method comprising administering to a subject in need thereof, a compound of formula Ib as defined in claim 36, or a pharmaceutically acceptable salt thereof, in an amount sufficient to inhibit FLT3.
60. A method of treating an CDK-dependent disorder, said method comprising administering to a subject in need thereof, a compound of formula Ib as defined in claim 36, or a pharmaceutically acceptable salt thereof, in an amount sufficient to inhibit a cyclin dependent kinase.
61. A method of treating a GSK-dependent disorder, said method comprising administering to a subject in need thereof, a compound of formula Ib as defined in claim 36, or a pharmaceutically acceptable salt thereof, in an amount sufficient to inhibit GSK.
PCT/GB2006/000087 2005-01-11 2006-01-11 4- (1h-indol-3-yl) -pyrimidin-2-ylamine derivates and their use in therapy WO2006075152A1 (en)

Priority Applications (8)

Application Number Priority Date Filing Date Title
EP06701760A EP1836194A1 (en) 2005-01-11 2006-01-11 4- (1h-indol-3-yl) -pyrimidin-2-ylamine derivates and their use in therapy
US11/794,449 US20090318446A1 (en) 2005-01-11 2006-01-11 4-(1H-Indol-3-yl)-Pyrimidin-2-Ylamine Derivatives and Their Use in Therapy
MX2007008373A MX2007008373A (en) 2005-01-11 2006-01-11 4- (1h-indol-3-yl) -pyrimidin-2-ylamine derivates and their use in therapy.
AU2006205710A AU2006205710A1 (en) 2005-01-11 2006-01-11 4- (1H-indol-3-yl) -pyrimidin-2-ylamine derivates and their use in therapy
CA002592723A CA2592723A1 (en) 2005-01-11 2006-01-11 4- (1h-indol-3-yl) -pyrimidin-2-ylamine derivates and their use in therapy
JP2007549955A JP2008526824A (en) 2005-01-11 2006-01-11 4- (1H-Indol-3-yl) -pyrimidin-2-ylamine derivatives and their use in therapy
BRPI0606313-6A BRPI0606313A2 (en) 2005-01-11 2006-01-11 4- (1h-indol-3-yl) -pyrimidin-2-ylamine derivatives, process for their preparation and use in therapy
IL184313A IL184313A0 (en) 2005-01-11 2007-07-01 4-(1h-indol-3-yl)-pyrimidin-2-ylamine derivates and their use in therapy

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
GBGB0500492.4A GB0500492D0 (en) 2005-01-11 2005-01-11 Compound
GB0500492.4 2005-01-11

Publications (1)

Publication Number Publication Date
WO2006075152A1 true WO2006075152A1 (en) 2006-07-20

Family

ID=34203901

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/GB2006/000087 WO2006075152A1 (en) 2005-01-11 2006-01-11 4- (1h-indol-3-yl) -pyrimidin-2-ylamine derivates and their use in therapy

Country Status (11)

Country Link
US (1) US20090318446A1 (en)
EP (1) EP1836194A1 (en)
JP (1) JP2008526824A (en)
CN (1) CN101111490A (en)
AU (1) AU2006205710A1 (en)
BR (1) BRPI0606313A2 (en)
CA (1) CA2592723A1 (en)
GB (1) GB0500492D0 (en)
IL (1) IL184313A0 (en)
MX (1) MX2007008373A (en)
WO (1) WO2006075152A1 (en)

Cited By (50)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008028860A1 (en) * 2006-09-08 2008-03-13 F. Hoffmann-La Roche Ag Benzotriazole kinase modulators
EP2040546A2 (en) * 2006-06-22 2009-04-01 Merck & Co., Inc. Tyrosine kinase inhibitors
US7566736B2 (en) 2004-12-06 2009-07-28 Aventis Pharma S.A. Substituted indoles, compositions containing them, method for the production thereof and their use
JP2011520824A (en) * 2008-05-16 2011-07-21 エフ.ホフマン−ラ ロシュ アーゲー Inhibitor of JNK
US20110263541A1 (en) * 2008-10-27 2011-10-27 Shanghai Genomics, Inc Kinase inhibitors and their use as pharmaceutical agents
US8119655B2 (en) 2005-10-07 2012-02-21 Takeda Pharmaceutical Company Limited Kinase inhibitors
US8278450B2 (en) 2007-04-18 2012-10-02 Takeda Pharmaceutical Company Limited Kinase inhibitors
EP2563774A1 (en) * 2010-04-27 2013-03-06 Hutchison Medipharma Limited Pyrimidinyl indole compounds
US8410112B2 (en) 2008-11-10 2013-04-02 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US8586751B2 (en) 2009-06-12 2013-11-19 Bristol-Myers Squibb Company Nicotinamide compounds useful as kinase modulators
US8598156B2 (en) 2010-03-25 2013-12-03 Glaxosmithkline Llc Chemical compounds
US8623869B2 (en) 2010-06-23 2014-01-07 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US8765751B2 (en) 2011-09-30 2014-07-01 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US8822469B2 (en) 2011-06-22 2014-09-02 Vertex Pharmaceuticals Incorporated Pyrrolo[2,3-B]pyrazines useful as inhibitors of ATR kinase
US8841449B2 (en) 2011-11-09 2014-09-23 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US8841308B2 (en) 2008-12-19 2014-09-23 Vertex Pharmaceuticals Incorporated Pyrazin-2-amines useful as inhibitors of ATR kinase
US8841337B2 (en) 2011-11-09 2014-09-23 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US8841450B2 (en) 2011-11-09 2014-09-23 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US8846686B2 (en) 2011-09-30 2014-09-30 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US8846918B2 (en) 2011-11-09 2014-09-30 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US8846917B2 (en) 2011-11-09 2014-09-30 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US8853217B2 (en) 2011-09-30 2014-10-07 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US8877759B2 (en) 2011-04-05 2014-11-04 Vertex Pharnaceuticals Incorporated Aminopyrazines as ATR kinase inhibitors
US8912198B2 (en) 2012-10-16 2014-12-16 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US8962631B2 (en) 2010-05-12 2015-02-24 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US8969356B2 (en) 2010-05-12 2015-03-03 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US9035053B2 (en) 2011-09-30 2015-05-19 Vertex Pharmaceuticals Incorporated Processes for making compounds useful as inhibitors of ATR kinase
US9062008B2 (en) 2010-05-12 2015-06-23 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US9096602B2 (en) 2011-06-22 2015-08-04 Vertex Pharmaceuticals Incorporated Substituted pyrrolo[2,3-B]pyrazines as ATR kinase inhibitors
US9096584B2 (en) 2010-05-12 2015-08-04 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
EP2736895B1 (en) 2011-07-27 2016-01-06 Astrazeneca AB 2-(2,4,5-substituted-anilino) pyrimidine derivatives as egfr modulators useful for treating cancer
US9309250B2 (en) 2011-06-22 2016-04-12 Vertex Pharmaceuticals Incorporated Substituted pyrrolo[2,3-b]pyrazines as ATR kinase inhibitors
US9334244B2 (en) 2010-05-12 2016-05-10 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US9340546B2 (en) 2012-12-07 2016-05-17 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US9630956B2 (en) 2010-05-12 2017-04-25 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US9663519B2 (en) 2013-03-15 2017-05-30 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US9670215B2 (en) 2014-06-05 2017-06-06 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US9791456B2 (en) 2012-10-04 2017-10-17 Vertex Pharmaceuticals Incorporated Method for measuring ATR inhibition mediated increases in DNA damage
US10160760B2 (en) 2013-12-06 2018-12-25 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
WO2019143719A1 (en) 2018-01-16 2019-07-25 Syros Pharmaceuticals, Inc. Inhibitors of cyclin-dependent kinase 7 (cdk7)
WO2019143730A1 (en) 2018-01-16 2019-07-25 Syros Pharmaceuticals, Inc. Inhibitors of cyclin-dependent kinase 7 (cdk7)
US10478430B2 (en) 2012-04-05 2019-11-19 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase and combination therapies thereof
US10513509B2 (en) 2016-05-26 2019-12-24 Recurium Ip Holdings, Llc EGFR inhibitor compounds
US10813929B2 (en) 2011-09-30 2020-10-27 Vertex Pharmaceuticals Incorporated Treating cancer with ATR inhibitors
EP3789386A1 (en) * 2017-01-26 2021-03-10 Hanmi Pharm. Co., Ltd. Pyrimidine compound and pharmaceutical use thereof
WO2021066443A1 (en) * 2019-09-30 2021-04-08 한미약품 주식회사 Pharmaceutical composition for treating acute myeloid leukemia containing flt3 inhibitor and mdm2 inhibitor
US11179394B2 (en) 2014-06-17 2021-11-23 Vertex Pharmaceuticals Incorporated Method for treating cancer using a combination of Chk1 and ATR inhibitors
US11464774B2 (en) 2015-09-30 2022-10-11 Vertex Pharmaceuticals Incorporated Method for treating cancer using a combination of DNA damaging agents and ATR inhibitors
US11666888B2 (en) 2018-02-05 2023-06-06 Bio-Rad Laboratories, Inc. Chromatography resin having an anionic exchange-hydrophobic mixed mode ligand
EP3974423A4 (en) * 2019-05-22 2023-06-21 Shanghai Hansoh Biomedical Co., Ltd. Indole derivative-containing inhibitor, preparation method therefor and application thereof

Families Citing this family (25)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20120117905A (en) 2010-01-28 2012-10-24 프레지던트 앤드 펠로우즈 오브 하바드 칼리지 Compositions and methods for enhancing proteasome activity
CA2819373A1 (en) 2010-12-09 2012-06-14 Amgen Inc. Bicyclic compounds as pim inhibitors
CZ305457B6 (en) 2011-02-28 2015-09-30 Ústav organické chemie a biochemie, Akademie věd ČR v. v. i. Pyrimidine compounds inhibiting formation of nitrogen monoxide and prostaglandin E2, process for their preparation and use
US9321756B2 (en) 2011-03-22 2016-04-26 Amgen Inc. Azole compounds as PIM inhibitors
CN103635230B (en) 2011-05-12 2017-10-31 普罗蒂斯特斯治疗公司 Albumen homeostasis conditioning agent
KR101283895B1 (en) * 2011-08-04 2013-07-16 한국해양과학기술원 Composotion comprising 5-hydroxyindole compound or the extract of Scalarispongia sp. for preventing and treating cancer
WO2013059634A1 (en) 2011-10-20 2013-04-25 The Regents Of The University Of California Use of cdk9 inhibitors to reduce cartilage degradation
WO2014116228A1 (en) 2013-01-25 2014-07-31 President And Fellows Of Harvard College Usp14 inhibitors for treating or preventing viral infections
WO2015073528A1 (en) 2013-11-12 2015-05-21 Proteostasis Therapeutics, Inc. Proteasome activity enhancing compounds
CN104876914B (en) * 2014-02-28 2017-04-19 山东轩竹医药科技有限公司 Pyrimidine derivative type anaplastic lymphoma kinase inhibitor
CN108558842B (en) * 2014-03-06 2021-06-04 山东轩竹医药科技有限公司 Pyrimidine derivative anaplastic lymphoma kinase inhibitor
JP6930913B2 (en) 2014-10-14 2021-09-01 ザ リージェンツ オブ ザ ユニバーシティ オブ カリフォルニアThe Regents Of The University Of California Usage of CDK9 and BRD4 inhibitors to inhibit inflammation
CA2995430C (en) * 2015-09-25 2023-07-11 Dizal (Jiangsu) Pharmaceutical Co., Limited Compounds and methods for inhibiting jak
CN107987054B (en) * 2017-11-28 2020-05-19 四川大学 CDK2 inhibitor
WO2019154177A1 (en) * 2018-02-12 2019-08-15 恩瑞生物医药科技(上海)有限公司 Pyrimidine compound, preparation method thereof and medical use thereof
AU2019203034B1 (en) * 2018-07-25 2019-09-26 Hanmi Pharm. Co., Ltd. Pyrimidine compounds and pharmaceutical compositions for preventing or treating cancers including the same
KR101954370B1 (en) * 2018-07-25 2019-03-05 한미약품 주식회사 Pyrimidine compounds and pharmaceutical composition for preventing or treating cancers comprising the same
WO2020071550A1 (en) * 2018-10-04 2020-04-09 京都薬品工業株式会社 Cdk8 inhibitor and use of same
JP2020070270A (en) * 2018-11-01 2020-05-07 御木本製薬株式会社 Fibronectin gene expression promoter
US20210403495A1 (en) * 2018-11-01 2021-12-30 Syros Pharmaceuticals, Inc. Inhibitors of cyclin-dependent kinase 7 (cdk7)
US11633399B2 (en) 2018-12-25 2023-04-25 Sol-Gel Technologies Ltd. Treatment of skin disorders with compositions comprising an EGFR inhibitor
WO2020171649A1 (en) * 2019-02-22 2020-08-27 한미약품 주식회사 Pharmaceutical combination comprising flt3 inhibitor and iap antagonist for treatment of acute myeloid leukemia
WO2020176599A1 (en) 2019-02-27 2020-09-03 The Regents Of The University Of California Azepino-indoles and other heterocycles for treating brain disorders
WO2021084541A1 (en) * 2019-10-31 2021-05-06 Sol-Gel Technologies Ltd. Treatment of hair loss disorders with a topical egfr inhibitor
WO2022098083A1 (en) * 2020-11-05 2022-05-12 한미약품 주식회사 Pharmaceutical composition for treating leukemia comprising flt3 inhibitor

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000078731A1 (en) * 1999-06-18 2000-12-28 Celltech R&D Limited 5-cyano-2-aminopyrimidine derivatives
WO2004089913A1 (en) * 2003-04-11 2004-10-21 Novartis Ag Aminopyrimidine derivatives and their medical use

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE3751742T3 (en) * 1986-01-13 2002-11-21 American Cyanamid Co., Wayne 4,5,6-substituted 2-pyrimidinamines
GB9523675D0 (en) * 1995-11-20 1996-01-24 Celltech Therapeutics Ltd Chemical compounds
TW440563B (en) * 1996-05-23 2001-06-16 Hoffmann La Roche Aryl pyrimidine derivatives and a pharmaceutical composition thereof
US20040171630A1 (en) * 2001-06-19 2004-09-02 Yuntae Kim Tyrosine kinase inhibitors

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000078731A1 (en) * 1999-06-18 2000-12-28 Celltech R&D Limited 5-cyano-2-aminopyrimidine derivatives
WO2004089913A1 (en) * 2003-04-11 2004-10-21 Novartis Ag Aminopyrimidine derivatives and their medical use

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
BIAO JIANG, CAI-GUANG YANG: "Synthesis of indolylpyrimidines via cross-coupling of indolylboronic acid with chloropyrimidines: Facile synthesis of meridianin D", HETEROCYCLES, vol. 53, no. 7, 2000, pages 1489 - 1498, XP008060011 *
FRESNEDA, MOLINA, BLEDA: "Synthesis of the indole alkaloids meridianins from the tunicate Aplidium meridianum", TETRAHEDRON, vol. 57, 2001, pages 2355 - 2363, XP002367722 *
FRESNEDA, MOLINA, DELGADO, BLEDA: "Synthetic studies towards the 2-aminopyrimidine alkaloids variolins and meridianins from marine origin", TETRAHEDRON LETTERS, vol. 41, 2000, pages 4777 - 4780, XP002367723 *
GOMPEL, LEOST, JOFFE, PURICELLI, FRANCO, PALERMO, MEIJER: "Meridianins, a new family of protein kinase inhibitors isolated from the Ascidian Aplidium meridianum", BIOORGANIC & MEDICINAL CHEMISTRY LETTERS, vol. 14, 2004, pages 1703 - 1707, XP002367721 *
HERNANDEZ FRANCO, LAURA ET AL: "Indole alkaloids from the Tunicate Aplidium meridianum", JOURNAL OF NATURAL PRODUCTS , 61(9), 1130-1132 CODEN: JNPRDF; ISSN: 0163-3864, 1998, XP002367720 *

Cited By (86)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7566736B2 (en) 2004-12-06 2009-07-28 Aventis Pharma S.A. Substituted indoles, compositions containing them, method for the production thereof and their use
US8119655B2 (en) 2005-10-07 2012-02-21 Takeda Pharmaceutical Company Limited Kinase inhibitors
EP2040546A2 (en) * 2006-06-22 2009-04-01 Merck & Co., Inc. Tyrosine kinase inhibitors
EP2040546A4 (en) * 2006-06-22 2009-12-23 Merck & Co Inc Tyrosine kinase inhibitors
CN101511359B (en) * 2006-09-08 2012-09-05 霍夫曼-拉罗奇有限公司 Benzotriazole kinase modulators
WO2008028860A1 (en) * 2006-09-08 2008-03-13 F. Hoffmann-La Roche Ag Benzotriazole kinase modulators
JP2010502668A (en) * 2006-09-08 2010-01-28 エフ.ホフマン−ラ ロシュ アーゲー Benzotriazole kinase modulator
KR101177729B1 (en) 2006-09-08 2012-09-07 에프. 호프만-라 로슈 아게 Benzotriazole kinase modulators
US8962622B2 (en) 2006-09-08 2015-02-24 Roche Palo Alto Llc Benzotriazole kinase modulators
US8278450B2 (en) 2007-04-18 2012-10-02 Takeda Pharmaceutical Company Limited Kinase inhibitors
JP2011520824A (en) * 2008-05-16 2011-07-21 エフ.ホフマン−ラ ロシュ アーゲー Inhibitor of JNK
US20110263541A1 (en) * 2008-10-27 2011-10-27 Shanghai Genomics, Inc Kinase inhibitors and their use as pharmaceutical agents
US8309550B2 (en) * 2008-10-27 2012-11-13 Shanghai Genomics, Inc. Kinase inhibitors and their use as pharmaceutical agents
US8410112B2 (en) 2008-11-10 2013-04-02 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US10961232B2 (en) 2008-12-19 2021-03-30 Vertex Pharmaceuticals Incorporated Substituted pyrazines as ATR kinase inhibitors
US9701674B2 (en) 2008-12-19 2017-07-11 Vertex Pharmaceuticals Incorporated Substituted pyrazines as ATR kinase inhibitors
US8841308B2 (en) 2008-12-19 2014-09-23 Vertex Pharmaceuticals Incorporated Pyrazin-2-amines useful as inhibitors of ATR kinase
US10479784B2 (en) 2008-12-19 2019-11-19 Vertex Pharmaceuticals Incorporated Substituted pyrazin-2-amines as inhibitors of ATR kinase
US9365557B2 (en) 2008-12-19 2016-06-14 Vertex Pharmaceuticals Incorporated Substituted pyrazin-2-amines as inhibitors of ATR kinase
US8586751B2 (en) 2009-06-12 2013-11-19 Bristol-Myers Squibb Company Nicotinamide compounds useful as kinase modulators
US8598156B2 (en) 2010-03-25 2013-12-03 Glaxosmithkline Llc Chemical compounds
AU2010351938B2 (en) * 2010-04-27 2014-10-30 Hutchison Medipharma Limited Pyrimidinyl indole compounds
US8501166B2 (en) 2010-04-27 2013-08-06 Hutchison Medipharma Limited Pyrimidinyl indole compounds
EP2563774A4 (en) * 2010-04-27 2013-07-10 Hutchison Medipharma Ltd Pyrimidinyl indole compounds
EP2563774A1 (en) * 2010-04-27 2013-03-06 Hutchison Medipharma Limited Pyrimidinyl indole compounds
US8962631B2 (en) 2010-05-12 2015-02-24 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US9630956B2 (en) 2010-05-12 2017-04-25 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US9334244B2 (en) 2010-05-12 2016-05-10 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US9096584B2 (en) 2010-05-12 2015-08-04 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US9062008B2 (en) 2010-05-12 2015-06-23 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US8969356B2 (en) 2010-05-12 2015-03-03 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US8623869B2 (en) 2010-06-23 2014-01-07 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US8877759B2 (en) 2011-04-05 2014-11-04 Vertex Pharnaceuticals Incorporated Aminopyrazines as ATR kinase inhibitors
US8822469B2 (en) 2011-06-22 2014-09-02 Vertex Pharmaceuticals Incorporated Pyrrolo[2,3-B]pyrazines useful as inhibitors of ATR kinase
US9096602B2 (en) 2011-06-22 2015-08-04 Vertex Pharmaceuticals Incorporated Substituted pyrrolo[2,3-B]pyrazines as ATR kinase inhibitors
US9309250B2 (en) 2011-06-22 2016-04-12 Vertex Pharmaceuticals Incorporated Substituted pyrrolo[2,3-b]pyrazines as ATR kinase inhibitors
US10858336B2 (en) 2011-07-27 2020-12-08 Astazeneca Ab 2-(2,4,5-substituted-anilino)pyrimidine compounds
US10017493B2 (en) 2011-07-27 2018-07-10 Astrazeneca Ab 2-(2,4,5-substituted-anilino)pyrimidine compounds
EP3009431B1 (en) 2011-07-27 2017-10-25 Astrazeneca AB 2-(2,4,5-substituted-anilino)pyrimidine derivatives as egfr modulators useful for treating cancer
US9732058B2 (en) 2011-07-27 2017-08-15 Astrazeneca Ab 2-(2,4,5-substituted-anilino)pyrimidine compounds
EP2736895B1 (en) 2011-07-27 2016-01-06 Astrazeneca AB 2-(2,4,5-substituted-anilino) pyrimidine derivatives as egfr modulators useful for treating cancer
US11524951B2 (en) 2011-07-27 2022-12-13 Astrazeneca Ab 2-(2,4,5-substituted-anilino)pyrimidine compounds
US8765751B2 (en) 2011-09-30 2014-07-01 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US8846686B2 (en) 2011-09-30 2014-09-30 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US10822331B2 (en) 2011-09-30 2020-11-03 Vertex Pharmaceuticals Incorporated Processes for preparing ATR inhibitors
US10208027B2 (en) 2011-09-30 2019-02-19 Vertex Pharmaceuticals Incorporated Processes for preparing ATR inhibitors
US10813929B2 (en) 2011-09-30 2020-10-27 Vertex Pharmaceuticals Incorporated Treating cancer with ATR inhibitors
US8853217B2 (en) 2011-09-30 2014-10-07 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US9862709B2 (en) 2011-09-30 2018-01-09 Vertex Pharmaceuticals Incorporated Processes for making compounds useful as inhibitors of ATR kinase
US9035053B2 (en) 2011-09-30 2015-05-19 Vertex Pharmaceuticals Incorporated Processes for making compounds useful as inhibitors of ATR kinase
US8841450B2 (en) 2011-11-09 2014-09-23 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US8841449B2 (en) 2011-11-09 2014-09-23 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US8846918B2 (en) 2011-11-09 2014-09-30 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US8841337B2 (en) 2011-11-09 2014-09-23 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US8846917B2 (en) 2011-11-09 2014-09-30 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US11110086B2 (en) 2012-04-05 2021-09-07 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase and combination therapies thereof
US10478430B2 (en) 2012-04-05 2019-11-19 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase and combination therapies thereof
US9791456B2 (en) 2012-10-04 2017-10-17 Vertex Pharmaceuticals Incorporated Method for measuring ATR inhibition mediated increases in DNA damage
US8912198B2 (en) 2012-10-16 2014-12-16 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US11117900B2 (en) 2012-12-07 2021-09-14 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US11370798B2 (en) 2012-12-07 2022-06-28 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US9718827B2 (en) 2012-12-07 2017-08-01 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US10392391B2 (en) 2012-12-07 2019-08-27 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US9650381B2 (en) 2012-12-07 2017-05-16 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US10787452B2 (en) 2012-12-07 2020-09-29 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US9340546B2 (en) 2012-12-07 2016-05-17 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US9663519B2 (en) 2013-03-15 2017-05-30 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US10160760B2 (en) 2013-12-06 2018-12-25 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US11485739B2 (en) 2013-12-06 2022-11-01 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US10815239B2 (en) 2013-12-06 2020-10-27 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US10093676B2 (en) 2014-06-05 2018-10-09 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US9670215B2 (en) 2014-06-05 2017-06-06 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US10800781B2 (en) 2014-06-05 2020-10-13 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US11179394B2 (en) 2014-06-17 2021-11-23 Vertex Pharmaceuticals Incorporated Method for treating cancer using a combination of Chk1 and ATR inhibitors
US11464774B2 (en) 2015-09-30 2022-10-11 Vertex Pharmaceuticals Incorporated Method for treating cancer using a combination of DNA damaging agents and ATR inhibitors
US10513509B2 (en) 2016-05-26 2019-12-24 Recurium Ip Holdings, Llc EGFR inhibitor compounds
US12049460B2 (en) 2016-05-26 2024-07-30 Recurium Ip Holdings, Llc EGFR inhibitor compounds
US11098030B2 (en) 2016-05-26 2021-08-24 Recurium Ip Holdings, Llc EGFR inhibitor compounds
EP3789386A1 (en) * 2017-01-26 2021-03-10 Hanmi Pharm. Co., Ltd. Pyrimidine compound and pharmaceutical use thereof
WO2019143719A1 (en) 2018-01-16 2019-07-25 Syros Pharmaceuticals, Inc. Inhibitors of cyclin-dependent kinase 7 (cdk7)
EP3740206A4 (en) * 2018-01-16 2021-10-20 Syros Pharmaceuticals, Inc. Inhibitors of cyclin-dependent kinase 7 (cdk7)
EP3740207A4 (en) * 2018-01-16 2021-10-13 Syros Pharmaceuticals, Inc. Inhibitors of cyclin-dependent kinase 7 (cdk7)
WO2019143730A1 (en) 2018-01-16 2019-07-25 Syros Pharmaceuticals, Inc. Inhibitors of cyclin-dependent kinase 7 (cdk7)
US11666888B2 (en) 2018-02-05 2023-06-06 Bio-Rad Laboratories, Inc. Chromatography resin having an anionic exchange-hydrophobic mixed mode ligand
EP3974423A4 (en) * 2019-05-22 2023-06-21 Shanghai Hansoh Biomedical Co., Ltd. Indole derivative-containing inhibitor, preparation method therefor and application thereof
WO2021066443A1 (en) * 2019-09-30 2021-04-08 한미약품 주식회사 Pharmaceutical composition for treating acute myeloid leukemia containing flt3 inhibitor and mdm2 inhibitor

Also Published As

Publication number Publication date
JP2008526824A (en) 2008-07-24
EP1836194A1 (en) 2007-09-26
BRPI0606313A2 (en) 2009-06-16
CA2592723A1 (en) 2006-07-20
IL184313A0 (en) 2007-10-31
AU2006205710A1 (en) 2006-07-20
US20090318446A1 (en) 2009-12-24
CN101111490A (en) 2008-01-23
GB0500492D0 (en) 2005-02-16
MX2007008373A (en) 2007-09-06

Similar Documents

Publication Publication Date Title
US20090318446A1 (en) 4-(1H-Indol-3-yl)-Pyrimidin-2-Ylamine Derivatives and Their Use in Therapy
US7576091B2 (en) Thiazolo-, oxazalo and imidazolo-quinazoline compounds capable of inhibiting protein kinases
US20060241297A1 (en) Pyridinylamino-pyrimidine derivatives as protein kinase inhibitors
US20090215805A1 (en) 4-Heteroaryl Pyrimidine Derivatives and use thereof as Protein Kinase Inhibitors
US7902361B2 (en) Pyrimidin-4-yl-3, 4-thione compounds and their use in therapy
US20070021419A1 (en) 2-Aminophenyl-4-phenylpyrimidines as kinase inhibitors
EP1940846B1 (en) Pyrimidin-4-yl-3,4-dihydro-2h-pyrrolo[1,2a]pyrazin-1-one compounds
AU2005210254A1 (en) Pyridinyl - or pyrimidinyl thiazoles with protein kinase inhibiting activity

Legal Events

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

Ref document number: 2006205710

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2006701760

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2007549955

Country of ref document: JP

WWE Wipo information: entry into national phase

Ref document number: 2592723

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 184313

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: 556243

Country of ref document: NZ

WWE Wipo information: entry into national phase

Ref document number: 5216/DELNP/2007

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: MX/a/2007/008373

Country of ref document: MX

ENP Entry into the national phase

Ref document number: 2006205710

Country of ref document: AU

Date of ref document: 20060111

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

WWP Wipo information: published in national office

Ref document number: 2006205710

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 200680003768.8

Country of ref document: CN

WWP Wipo information: published in national office

Ref document number: 2006701760

Country of ref document: EP

ENP Entry into the national phase

Ref document number: PI0606313

Country of ref document: BR

Kind code of ref document: A2

WWE Wipo information: entry into national phase

Ref document number: 11794449

Country of ref document: US