WO2009118567A2 - Pyrimidines, triazines and their use as pharmaceutical agents - Google Patents

Pyrimidines, triazines and their use as pharmaceutical agents Download PDF

Info

Publication number
WO2009118567A2
WO2009118567A2 PCT/GB2009/050298 GB2009050298W WO2009118567A2 WO 2009118567 A2 WO2009118567 A2 WO 2009118567A2 GB 2009050298 W GB2009050298 W GB 2009050298W WO 2009118567 A2 WO2009118567 A2 WO 2009118567A2
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
compound
formula
hereinbefore defined
groups
Prior art date
Application number
PCT/GB2009/050298
Other languages
French (fr)
Other versions
WO2009118567A3 (en
Inventor
Peter Martin Fischer
Shudong Wang
Andrey Zaytsev
Original Assignee
The University Of Nottingham
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 The University Of Nottingham filed Critical The University Of Nottingham
Priority to US12/934,798 priority Critical patent/US20110092490A1/en
Priority to CN2009801123665A priority patent/CN102143953B/en
Publication of WO2009118567A2 publication Critical patent/WO2009118567A2/en
Publication of WO2009118567A3 publication Critical patent/WO2009118567A3/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
    • C07D417/04Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • 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
    • A61P3/00Drugs for disorders of the metabolism
    • 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
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D251/00Heterocyclic compounds containing 1,3,5-triazine rings
    • C07D251/02Heterocyclic compounds containing 1,3,5-triazine rings not condensed with other rings
    • C07D251/12Heterocyclic compounds containing 1,3,5-triazine rings not condensed with other rings having three double bonds between ring members or between ring members and non-ring members
    • C07D251/14Heterocyclic compounds containing 1,3,5-triazine rings not condensed with other rings having three double bonds between ring members or between ring members and non-ring members with hydrogen or carbon atoms directly attached to at least one ring carbon atom
    • C07D251/16Heterocyclic compounds containing 1,3,5-triazine rings not condensed with other rings having three double bonds between ring members or between ring members and non-ring members with hydrogen or carbon atoms directly attached to at least one ring carbon atom to only one ring carbon atom
    • C07D251/18Heterocyclic compounds containing 1,3,5-triazine rings not condensed with other rings having three double bonds between ring members or between ring members and non-ring members with hydrogen or carbon atoms directly attached to at least one ring carbon atom to only one ring carbon atom with nitrogen atoms directly attached to the two other ring carbon atoms, e.g. guanamines

Definitions

  • the present invention relates to substituted pyrimidine and [1,3,5]triazine derivatives that have broad therapeutic applications via inhibiting one or more protein kinases.
  • the invention also provides processes for preparing compounds, pharmaceutically acceptable compositions comprising the compounds, and the use of the compounds and methods of using the compounds and compositions in the treatment of various diseases, conditions, or disorders.
  • the protein kinase family is one of the largest in the human genome, comprising 500 genes.
  • 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, whose terminal phosphate group transfers covalently to its macromolecular substrates.
  • the protein kinases may be categorized by the substrates they phosphorylate, e.g. protein-serine/threonine, protein-tyrosine.
  • Protein kinases mediate intracellular signalling by effecting a phosphoryl transfer from a nucleoside triphosphate to a protein acceptor that is involved in a signalling pathway. These phosphorylation events are triggered in response to a variety of extracellular and other stimuli and act as molecular on/off switches that can modulate or regulate the target protein biological function.
  • An extracellular stimulus may affect one or more cellular_responses related to cell growth, migration, differentiation, secretion of, hormones, activation of transcription factors, muscle contraction, giucdse " metabolism, control of proteirr' synthesis, and regulation of the cell cycle.
  • diseases are associated with abnormal cellular responses triggered by protein kinase-mediated ⁇ events. These diseases include, but are not limited to allergies and asthma, Alzheimer's disease, autoimmune diseases, bone diseases, cancer, cardiovascular diseases, inflammatory diseases, hormone- related diseases, metabolic diseases, neurological and neurodegenerative diseases. Accordingly, there has been a substantial effort in medicinal chemistry to find protein kinase inhibitors that are effective as therapeutic agents.
  • CDKs Cyclin-dependent kinases
  • CDKs may be classified into two major groups, reflecting their functions.
  • the cell cycle regulator CDKs composed primarily of CDK1, CDK2, CDK3, CDK4 and CDK6 function with their cyclin partners including cyclin A, B, D1 , D2, D3, E, and F to regulate promotion of the cell cycle.
  • the transcription regulator CDKs 1 which include CDK7, CDK8, CDK9 and CDK11 work together with cyclin C, H, K, L1, L2, T1 and T2, tend to play roles in transcriptional regulation.
  • CDKs have been implicated in cell proliferation disorders, particularly in cancer.
  • Cell proliferation is a result of the direct or indirect deregulation of the cell division cycle and the CDKs play a critical role in the regulation of the various phases of this cycle. Therefore, inhibitors of CDKs and their associated cyclins are useful targets for cancer therapy.
  • CDKs also play a role in apoptosis and T-cell development, which is predominantly due to the CDK functions in regulation of transcription.
  • clear clinical activity has very recently been obtained in chronic lymphocytic leukaemia (CLL) with CDK inhibitor flavopiridol.
  • CLL chronic lymphocytic leukaemia
  • flavopiridol CDK inhibitor flavopiridol.
  • CLL is characterised by cellular resistance to apoptosis through up-regulation of anti-apoptotic proteins.
  • Inhibition of transcription at the level of CDK9 which is necessary for mRNA elongation, selectively reinstates apoptosis in CLL cells.
  • CDK9 which is necessary for mRNA elongation
  • CDK inhibitors that restrain viral replication including human immunodeficiency virus, human cytomegalovirus, herpes virus, and varicella-zoster virus have been reported.
  • CDKs are a novel strategy for potential treatment of cardiovascular diseases including cardiohypertrophy.
  • Cardiohypertrophy is characterised by global increases in mRNA and protein synthesis.
  • CDK7 and CDK9 are closely associated with cardiac hypertrophy as they are the main drivers for transcription. Therefore inhibition of CDK9 and its associated cyclins is a relevant drug target for cardiovascular diseases.
  • Inhibition of CDK is also useful for the treatment of neurodegenerative disorders such as Alzheimer's disease.
  • the appearance of Paired Helical Filaments, associated with Alzheimer's disease, is caused by the hyperphosphorylation of Tau protein by CDK5/p25.
  • Inhibition of one or more other serine/threonine kinases including the Aurora kinases, Glycogen synthesis kinases (GSKs), polo-like kinases (PLKs) and tyrosine kinases including Ableson tyrosine kinase (BCR- ABL), FMS-related tyrosine kinases (FLT), IkB kinases (IKK), Janus kinases (JAK), platelet-derived growth factor (PDGF) receptor tyrosine kinases, vascular endothelial growth factor (VEGF) receptor tyrosine kinases, and Src family are also useful for the treatment of numerous diseases, conditions or disorders
  • GSK3 is known to phosphorylate many substrates and is thus involved in the regulation of multiple biochemical pathways. For example, GSK is highly expressed in the central and peripheral nervous systems. GSK3 inhibition is therefore of therapeutic significance in the treatment of CNS disorders such as Parkinsons and Alzheimers diseases.
  • GSK3 is over-expressed in muscle cells of type Il diabetics and that an inverse correlation exists between skeletal muscle GSK3 activity and insulin action. GSK3 inhibition is therefore of therapeutic significance in the treatment of diabetes, particularly type II, and diabetic neuropathy.
  • Aurora kinases and PLK are also important therapeutic targets for treatment of proliferative disorders. Based on their known functions inhibition of Aurora kinases and PLKs activity should disrupt mitosis leading to cell cycle arrest and therefore slowing tumour growth and induce apoptosis.
  • the present invention provides a novel class of substituted-2-anilino-4-arylpyrimidines and -4-aryl- [1,3,5]triazin-2-ylphenylamines with broad therapeutic application as protein kinase inhibitors, specifically compounds which are substituted at the'2-jj,4-,-and .5- and/or 6- positions of. pyrimidines or at the 2 T , A- and 6-positions of [1,3,6]triazines. These compounds have been synthetically difficult to access. We have found that the invention offers a class of compounds which are effective in protein kinase inhibition, offer important benefits in terms of selective inhibition, and are potentially effective therapeutics.
  • a first aspect of the present invention relates to a compound of formula I and its pharmaceutically acceptable salts or solvates and physiologically hydrolysable, solubilising or immobilisable derivatives:
  • Ar is optionally substituted and is a 5-membered heteroaryl ring wherein X 1 and X 2 are one or two heteroatoms or Ar is a 6-membered aromatic ring, wherein heteroatoms are selected from S, O, N, Se and wherein optional substituents include R 1 and R 2 ;
  • Y is N or CR 3 ;
  • R 5 to R 9 are linked to form a cyclic ether containing one or more oxygen atoms
  • R 3 when present, is selected from alkyl and R 13 as hereinbefore defined, with the proviso that when Y is CR 3 , Ar is a 5-membered heterocycle comprising one or two N heteroatoms and Z is NH, then R 3 is selected from C 3+ alkyl and R 13 as hereinbefore defined;
  • R 4 is selected from H, alkyl and R 13 as hereinbefore defined, with the proviso that when R 3 is absent, R 4 is selected from alkyl and R 13 as hereinbefore defined;
  • R 1 , R 2 , R 4 , R?, R 6 , R-, K 8 and R 9 and R 3 or R 12 where present comprise a group R 10 or R 11 wherein R 10 and R 11 comprise one or more solub ⁇ lising moieties chosen from i) neutral hydrophilic groups, ii) ionisable organic acids, iii) ionisable organic bases and combinations thereof.
  • a further aspect of the invention relates to a compound of formula I wherein at least one of R 10 or R 11 further comprises an immobilising moiety chosen from iv) chemical functions or moieties providing covalent or non-covalent attachment or binding to a solid phase or an immobile receptor.
  • the invention relates to the use of a compound of formula I or a pharmaceutically acceptable salt, solvate or physiologically hydrolysable, solubilising or immobilising derivative thereof, in the manufacture of a medicament for treating a condition mediated by an enzyme selected from one or more CDK, aurora kinase, GSK, PLK, BCR-ABL, FLT, IKK, JAK, PDGF or VEGF and Src family enzymes, particularly from one or more CDK2, CDK7, CDK8, CDK9, CDK11 , GSK-3, aurora kinase, PLK or at least one tyrosine kinase.
  • an enzyme selected from one or more CDK, aurora kinase, GSK, PLK, BCR-ABL, FLT, IKK, JAK, PDGF or VEGF and Src family enzymes, particularly from one or more CDK2, CDK7, CDK8, CDK9, CDK11 , GSK-3, aurora kin
  • a compound of formula I or a pharmaceutically acceptable salt, solvate, or physiologically hydrolysable, solubilising or immobilising derivative thereof in a method for treating a condition mediated by an enzyme selected from one or more CDK, aurora kinase, GSK, PLK, BCR-ABL 1 FLT, IKK, JAK, PDGF or VEGF and Src family enzymes, particularly from one or more CDK2, CDK7, CDK8, CDK9, CDK11, GSK-3, aurora kinase, PLK or tyrosine kinase.
  • an enzyme selected from one or more CDK, aurora kinase, GSK, PLK, BCR-ABL 1 FLT, IKK, JAK, PDGF or VEGF and Src family enzymes, particularly from one or more CDK2, CDK7, CDK8, CDK9, CDK11, GSK-3, aurora kinase, PLK or tyrosine kinas
  • a further aspect of the invention relates to the use of a compound of formula I or a pharmaceutically acceptable salt or solvate or physiologically hydrolysable, solubilising or immobilising derivative thereof, in an assay for identifying candidate compounds capable of treating a condition mediated by an enzyme selected from one or more CDK, aurora kinase, GSK, PLK, BCR-ABL, FLT, IKK, JAK, PDGF or VEGF and Src family enzymes, particularly from one or more CDK2, CDK7, CDK8, CDK9, CDK11, GSK-3, aurora kinase, PLK or tyrosine kinase.
  • an enzyme selected from one or more CDK, aurora kinase, GSK, PLK, BCR-ABL, FLT, IKK, JAK, PDGF or VEGF and Src family enzymes, particularly from one or more CDK2, CDK7, CDK8, CDK9, CDK11, GSK-3, aurora
  • a further -aspect of -the- invention- relates -.to a pharmaceutical composition
  • a pharmaceutical composition comprising a compound : of formula I or a pharmaceutically acceptable salt or solvate, or physiologically hydrolysable, solubilising or immobilising derivative thereof, in association with one or more diluents, carriers or excipients.
  • alkyl includes both straight chain and branched alkyl groups.
  • the alkyl group may be substituted (mono- or poly-) or unsubstituted. Suitable substituents include, for example, halo, CF 3 , OH, CN, NO 2 , SO 3 H, SO 2 NH 2 , SO 2 Me, NH 2 , COOH, CONH 2 and alkoxy.
  • the alkyl group is a Ci -20 alkyl group, more preferably a C 1-15 , more preferably still a C 1-12 alkyl group, more preferably still, a C 1-6 alkyl group, more preferably a C 1-3 alkyl group.
  • Particularly preferred alkyl groups include, for example, methyl, ethyl, propyl, isopropyl, butyl, isobutyl, tert-butyl, pentyl and hexyl.
  • heteroalkyl includes an alkyl group as defined above which comprises one or more heteroatoms.
  • cycloalkyl refers to a cyclic alkyl group which may be substituted (mono- or poly-) or unsubstituted. Suitable substituents include, for example, halo, CF 3 , OH, CN, NO 2 , SO 3 H, SO 2 NH 2 , SO 2 Me, NH 2 , COOH, CONH 2 and alkoxy.
  • cycloheteroalkyl refers to a cyclic heteroalkyl group which may be substituted (mono- or poly-) or unsubstituted. Suitable substituents include, for example, halo, CF 3 , OH, CN, NO 2 , SO 3 H, SO 2 NH 2 , SO 2 Me, NH 2 , COOH, CONH 2 and alkoxy. Preferred cycloheteroalkyl groups include morpholino, piperazinyl and piperidinyl groups.
  • aryl refers to an aromatic, substituted (mono- or poly-) or unsubstituted group, and includes, for example, phenyl, naphthyl etc.
  • suitable substituents include, for example, halo, CF 3 , OH, CN, NO 2 , SO 3 H, SO 2 NH 2 , SO 2 Me, NH 2 , COOH, CONH 2 and alkoxy.
  • heteroaryl refers to an aromatic, substituted (mono- or poly-) or unsubstituted group, which comprises one or more heteroatoms.
  • Preferred heteroatoms include N, S, O.
  • Preferred heteroaryl groups include pyrrole, pyrazole, pyrimidine, pyrazine, pyridine, q ⁇ inoline, triazine, triazole, thiophene, selenazol, thiazole and furan.
  • suitable substituents include, for example, halo, CF 3 , OH, CN, NO 2 , SO 3 H, SO 2 NH 2 , SO 2 Me, NH 2 , COOH, CONH 2 and alkoxy.
  • halo or halogeno refers to F, Cl, Br or I.
  • X 1 and X 2 are each independently selected from NH or N, O, S 1 Se, CH and C R 15 and at least one of X 1 and X 2 is selected from NH or N, O, S and Se, and wherein R 15 is as hereinbefore defined for R 1 , and all other variables are as hereinbefore defined.
  • a compound of formula I' wherein: one of X 1 and X 2 is CH or CR 15 , and the other of X 1 and X 2 is S, O, NH, NR 15 , or Se; or one of X 1 and X 2 is S, O or Se, and the other of X 1 and X 2 is N; or one ofX 1 and X 2 is N, and the other of X 1 and X 2 is NH or NR 15 ; and wherein all other variables are as hereinbefore defined. More preferably X 1 is S and X 2 is N or X 2 is S and X 1 is N.
  • a compound of formula I' or I" comprises a mono- or di-substituted phenyl, thiazol-4-yl, thiazol-5-yl, imidazol-4-yl, imidazol-5-yl, pyrrol-4-yl or pyrrol-5-yl group attached to the pyrimidine or [1,3,5]triazine ring through one of the ring carbon atoms; most preferably a phenyl, thiazol-4-yl or thiazol-5- yl group.
  • R 10 or R 11 comprises a neutral hydrophilic group (i) as hereinbefore defined, this preferably includes groups containing mono-, di- and polyhydroxylated saturated or unsaturated aliphatic, alicyclic or aromatic systems, carbohydrate derivatives, ethers and polyethers optionally containing one or more hydroxyl groups, O- and/or S-containing heterocyclic systems optionally containing one or more hydroxy! groups, aliphatic or aromatic systems containing a carboxamide, sulfoxide, sulfone, or sulfonamide function, and halogenated alkylcarbonyl groups.
  • R 1 ° or R 11 comprises an ⁇ bnisabie'organic acid (i ⁇ ) as hereinbefore defined * this preferably includes groups comprising one or more of the functions COOH, SO 3 H, OSO 3 H, PO 3 H 2 , and OPO 3 H 2 .
  • R 10 and R 11 may consist of natural or unnatural amino acid residues and peptides, or their derivatives.
  • R 24 and R 25 are each independently H, alkyl, aralkyl, CO-alkyl or aryl, with the proviso that at least one of R 24 and R 25 is other than H, or R 24 and R 25 are linked to form a cyclic group optionally containing one or more heteroatoms selected from N, O and S, and wherein said alkyl, aryl or cyclic group is optionally substituted by one or more substituents selected from halogeno, NO 2 , OH, , alkoxy, NH 2 , COOH, CH 2 CO 2 -alkyl, CONH 2 and CF 3 ;
  • N-thiomorpholinyl each of which may be optionally substituted by one or more alkyl, alkoxy, aryl, CHO or
  • each R 10 or R 11 is independently selected from a C 1-30 i hydrocarbyl- group, optionally comprising up. to twelve heteroatoms. selected, from N, S r and O, and optionally bearing up to six substituents each independently selected from a group R 15 as hereinbefore defined or comprising a moiety R 14 as hereinbefore defined, and a group R 15 .
  • a compound of formula I as hereinbefore defined bears up to six substituents selected from R 1 to R 9 , R 12 and R 16 as hereinbefore defined each comprising one or more heteroatoms selected from N, S, and O, and alternatively or additionally each comprising one or more moieties R 14 or groups R 15 as hereinbefore defined, wherein the combined substituents comprise up to ten heteroatoms or atoms N, S and O.
  • Z is NH or NR 16 .
  • Y is N or CR 3 .
  • R 13 is selected from alkyl-R 10 , alkyl-cycloalkyl which may be part unsaturated, alkyl- cycloheteroalkyl, aryl, aryl-R 10 , aralkyl, aralkyl-R 10 , alkyl-heteroaryl, halogeno, NO 2 , CN, OH, O-alkyl, O- cycloalkyl which may be part unsaturated, O-aryl, O-heteroaryl, O-R 10 , NH 2 , NH-alkyl, part unsaturated NH-cycloalkyl, NH-cycloheteroalkyl, NH-aryl, NH-heteroaryl, N-(alkyl) 2 , N-(aryl) 2 , N-(alkyl)(cycloalkyl), N- (alkyl)(cycloheteroalkyl), N-(aIkyl)(aryl),
  • R 1 is selected from NH-alkyl, part unsaturated NH-cycloalkyl, NH-heteroaryl, O-alkyl, O- cycloalkyl which may be part unsaturated, O-heteroaryl, alkyl-heteroaryl, alkyl-cycloalkyl which may be part unsaturated.
  • R 2 is selected from H, alkyl, such as C 1-s -alkyl, aryl, NH-alkyl, part unsaturated NH-cycloalkyl, NH-heteroaryl, O-alkyl, O-cycloalkyl which may be part unsaturated, O-heteroaryl, alkyl-heteroaryl and alkyl-cycloalkyl which may be part unsaturated.
  • alkyl such as C 1-s -alkyl, aryl, NH-alkyl, part unsaturated NH-cycloalkyl, NH-heteroaryl, O-alkyl, O-cycloalkyl which may be part unsaturated, O-heteroaryl, alkyl-heteroaryl and alkyl-cycloalkyl which may be part unsaturated.
  • R 5 is selected from H, O-alkyl, particularly OCH 3 , CF 3 , alkyl or halogeno.
  • R 6 and R 8 are independently selected from a sulphonyl, carbonyl, amide or sulphonamide, or thioether link to an unsubstituted or substituted 6 membered cyclic or heterocyclic, or aromatic or heteroaromatic ring, wherein substituents are as hereinbefore defined. More preferably R 6 and R 8 are , independently selected . from SO 2 -cycloheteroalkyl, SO 2 rcycloalkyl ; SO 2 -heteroaryl, SO-cycloheteroalkyl,..
  • R 6 and R 8 are independently selected from ⁇ /-linked N- (alkyl)(cycloheteroalkyl), SO 2 -cycloheteroalkyl and CO-cycloheteroalkyl most preferably such as N- (alkyl)(morpholino), ⁇ /-(alkyl)(piperazine), ⁇ /-(aIkyl)(piperadine), SO 2 -piperazines, SO 2 -morpholines, CO- piperazines, CO-morpholines, CO-piperadine or the like.
  • R 7 is selected from alkyl, for example C 1-5 alkyl, CONH 2 , CONH-alkyl, CN, OH, CF 3 , O-alkyl, halogeno, NH 2 , NH-alkyl and NHCO-alkyl.
  • R 9 is selected from H, halogeno, O-alkyl, more preferably H, halogeno and 0-Ci -5 alkyl.
  • R 3 is selected from C 1-6 alkyl, more preferably, i-propyl, i-butyl or t-butyl, or R 13 as hereinbefore defined. More preferably R 3 is selected from C 4+ alkyl and R 13 as hereinbefore defined, or from R 13 as hereinbefore defined.
  • R 3 is selected from CN, CF 3 , halogeno, NO 2 , NH 2 , NH-alkyl, N- (alkyl)(R 10 ), NH-cycloheteroalkyl, NHSO 2 R 10 , CONH 2 , CONH-(alkyl), CON-(alkyl)(R 10 ), R 10 , CO- cycloheteroalkyl, CO-heteroaryl, CONH-heteroaryl, CH 2 -cycloheteroalkyl, CH 2 -heteroaryl, cycloheteroalkyl, heteroaryl, and C 2-6 or C 4-6 alkyl, wherein alkyl, cycloheteroalkyl, aryl, aralkyl, heteroaryl groups may be further substituted with one or more groups selected from halogeno, NO 2 , CN OH, O-methyl, NH 2 , COOH, CONH 2 and CF 3 .
  • R 4 is selected from alkyl and R 13 as hereinbefore defined; more preferably R 4 is selected from amino, halogeno, such as Cl, and alkyl.
  • R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , R 8 , R 9 , R 12 and R 16 for example one, two, three or four thereof, shall correspond to or contain one or more of the group R 10 or R 11 .
  • R 3 and R 4 comprise or contain one or more of the group R 10 or R 11 .
  • Two or more groups R 10 and/or R 11 may be the same or different.
  • R 1 , R 2 , R 3 , R 5 or R 7 comprises or contains a solubilising moiety R 10 or R 11 .
  • One preferred embodiment of the invention relates to a compound of formula I' wherein: one of X 1 and X 2 is selected from S, O 1 NH, NR 15 and Se and the other thereof is N.
  • Y is CR 3 or N;
  • each R 6 or R 8 is independently SO 2 -cycloheteroalkyl, SO 2 -heteroaryl, SO- cycloheteroalkyl, SO-heteroaryl, CO-cycloheteroalkyl or CO-heteroaryl; preferably, the cycloheteroalkyl group is a /V-alkyl-morpholino, ⁇ /-alkylpiperazine, ⁇ /-alkylpiperadine; More preferably R 7 is alkyl, CN, OH, CF 3 , O-alkyl, halogeno, NH 2 , CONH-R 10 , NHR 10 , or NHCO-R 10 .
  • R 10 or R 11 corresponds to or is contained within R 1 , R 2 , R 3 or R 5 and R 9 is H.
  • Especially preferred compounds of the invention are those of formula I' wherein one X 1 or X 2 is ⁇ S, another X 1 or X 2 is N 1 Y is CR 3 or N, Z is NH, R 1 and R 2 are amino, alkyl, heteroaryl or aryl, R 3 is C 1-4 alkyl, CN 1 CF 3 , halogeno, NO 2 O-alkyl, NH 2 , NH-alkyl, N(alkyl) 2 , CO 2 alkyl, CO-alkyl, CONH 2 , CONH-alkyl or heteroaryl, R 4 is amino, halogeno or alkyl; R 5 is OMe 1 alkyl, or halogeno, each R 6 or R 8 is independently S0 2 -cycloheteroalkyl, SO-cyclohetero
  • Especially preferred compounds of the invention are those of formula I" wherein: Z is NH;
  • R 1 and R 2 are amino, alkyl, or aryl or R 2 is H; more preferably R 1 and R 2 are selected from NH-alkyl, part unsaturated NH-cycloalkyl, NH-heteroaryl, O-alkyl, O-cycloalkyl which may be part unsaturated, O- heteroaryl, alkyl-heteroaryl, alkyl-cycloalkyl which may be part unsaturated; or R 2 is H, alkyl, such as C 1-5 - alkyl or aryl, such as C 6 aryl;
  • R 3 is CN, CONH-alkyl, CF 3 , halogeno, NO 2 , heteroaryl or is contained with R 13 ;
  • R 4 is amino, halogeno or alkyl;
  • R 5 is OMe, alkyl, or halogeno;
  • each R 6 and R 8 is independently selected from SO 2 -cycloheteroalkyl, SO-cycloheteroalkyl, CO- cycloheteroalkyl and CO-heteroaryl;
  • R 7 is alkyl, OH, CF 3 , O-alkyl, halogeno, or NH 2 ;
  • R 9 is H; and the solubilising moiety corresponds to or is contained within R 1 , R 2 , R 3 or R 5 .
  • ntion include, compounds of formula I': "•• ⁇
  • PzC piperazine-1-carbonyl or piperazin-1-ylmethanone
  • MePzC 4-methylpiperazine-1 -carbonyl or 4-methylpiperazin-1 -ylmethanone I or
  • MC morpholin-4-carbonyl or morpholin-4-yl-methanone
  • MePzS 4-methylpiperazin-1-ylsulfonyl ⁇ - N
  • BPdC 1-benzylpiperidin-4-carbonyl o r 1-benzylpiperidin-4-ylmethanone
  • PdC piperidine-4-carbonyI or piperidin-4-ylmethanone
  • MePdC 1-methylpiperidin-4-ylmethanone or 1-methylpiperidin-4-carbonyl
  • MePz 4 ⁇ methylpiperazin-1-yl ON.
  • MeDz 4-methyl-1 ,4-diazepan-1-yl
  • R 10 or R 11 alternatively or additionally comprise devices for immobilisation thereof.
  • Such devices may be chemical functions that can be used for covalent attachment to solid phases such as functionalised polymers (e.g. agarose, polyacrylamide, polystyrene efc.) as commonly found in matrices (microtitre plate wells, microbeads, membranes, efc.) used for biochemical assays and affinity chromatography.
  • the devices may be small molecules (e.g. biotin) or polypeptides (e.g. antigens), which can be used for non-covalent immobilisation through binding to an immobilised receptor (e.g. avidin or streptavidin in the case of biotin, or a specific antibody in the case of antigens).
  • an immobilised receptor e.g. avidin or streptavidin in the case of biotin, or a specific antibody in the case of antigens.
  • a process for the preparation of a compound of formula I as hereinbefore defined comprises: (1) reacting a compound of formula III (as illustrated hereinbelow), where Ar is a mono- or di-substituted phenyl, thiazol-4-yl, thiazol-5-yl, imidazol-4-yl, imidazol-5-yl, pyrrol-4-yl or pyrrol-5-yl, preferably Ar is a phenyl, thiazol-4-yl or thiazol-5-yl group, Y is N, or CR 3 and L 1 is a leaving group, with a compound of formula IV (as illustrated hereinbelow), where Z and R 5 to R 9 are as hereinbefore defined;
  • the compound of formula I is a compound of formula V as hereinbefore defined, more preferably where Ar is a mono- or di-substituted thiazol-4-yl, thiazol-5-yl, imidazol-4-yl, imidazol-5-yl, pyrrol-4-yl or pyrrol-5-yl attached to the pyrimidine or [1,3,5]triazine ring through one of the ring carbon atoms, most preferably Ar is a thiazol-4-yl or thiazol-5-yl group; or is a compound of formula I" as hereinbefore defined, more preferably wherein the compounds of formula I" bear a mono- or di- substituted phenyl attached to the pyrimidine or [1,3,5]triazine ring through one of the ring carbon atoms.
  • L 1 is any leaving group including N(alkyl) 2 , halogen, ester, thioester, more preferably, NMe 2 , and the process (1) comprises a variety of methods for example as disclosed in Fischer PM, Wang S. WO 2001072745; and Wang, S.; et. al WO 2003029248, Cyclacel Limited, UK and references therein.
  • process (2) is conducted via a variety of methods known in the art, particularly, methods described by Liu, C (Liu, C, et al. 2007, Tetrahedron Lett. 48, 435) and Hodous, B (Hodous, B. L. J Med Chem, 50, 611).
  • the compound of formula XI (as illustrated hereinbelow) is obtained by reacting a compound of formula VIlI (as illustrated hereinbelow) where L 2 is any leaving group, preferably a halogeno group, and Y, L 3 and R 4 are as hereinbefore defined, with a compound of formula X (as illustrated hereinbelow), where Ar is as hereinbefore defined and L4 is any boronic acid or derivatives.
  • amidines Vl' (as illustrated hereinbelow) wherein Z and R 4 to R 9 are as hereinbefore defined in the presence of POCI 3 followed by alkylation reaction with anilines of formula XII (as illustrated hereinabove) to obtain [1 ,3,5]triazine triazines of formula I';
  • the compound of formula P is as hereinbefore defined, more preferably is a mono- or di- substituted thiazol-4-yl, thiazol-5-yl, imidazol-4-yl, imidazol-5-yl, pyrrol-4-yl or pyrrol-5-yl attached to the pyrimidine or [1,3,5]triazine ring through one of the ring carbon atoms; most preferably is a thiazol-4-yl or thiazol-5-yl group.
  • process (4) uses the method described previously (Wang, S. et al.J Med Chem, 2004, 47, 1662-75).
  • guanidines VHI' are obtained by reaction of cyanamide or certain of its derivatives using the method of Katritzky, A. R.; et al. Synthetic Communications 1995, 25, 1173.
  • a compound of formula XIIP is obtained by reacting phenyl isothiocyanate sodium hydrogen cyanamide to provide ⁇ /-cyanothiourea XIP.
  • such medicament is suitable for inhibition of a proliferative disorder mediated by a CDK or PLK, preferably is useful in the treatment of a proliferative disorder, such as cancers, leukaemias and other disorders associated with uncontrolled cellular proliferation such as psoriasis and restenosis, a viral disorder, a cardiovascular disease, a CNS disorder, an autoimmune disease, a bond disease, a hormone-related disease, a metabolic disorder, stroke, alopecia, an inflammatory disease or an infectious disease.
  • a proliferative disorder such as cancers, leukaemias and other disorders associated with uncontrolled cellular proliferation such as psoriasis and restenosis, a viral disorder, a cardiovascular disease, a CNS disorder, an autoimmune disease, a bond disease, a hormone-related disease, a metabolic disorder, stroke, alopecia, an inflammatory disease or an infectious disease.
  • the compound of formula I is capable of inhibiting one or more of the host cell kinases involved in cell proliferation, viral replication, a cardiovascular disorder, neurodegeneration, autoimmunity, a metabolic disorder, stroke, alopecia, an inflammatory disease or an infectious disease.
  • a proliferative disorder requires treatment of a susceptible neoplasm and may be selected from the group consisting of chronic lymphocytic ie ⁇ kaemia, lymphoma, leukaemia, breast cancer, lung cancer, prostate cancer, colon cancer, melanoma, pancreatic cancer, ovarian cancer, squamous carcinoma, carcinoma of head and neck, endometrial cancer, and aesophageal carcinoma.
  • 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 and cardiomyopathy, auto-immune disorders such as glomerulonephritis and rheumatoid arthritis, dermatological disorders such as psoriasis, anti-inflammatory, anti-fungal, antiparasitic disorders such as malaria, emphysema and alopecia.
  • the compounds of the present invention may induce apoptosis or maintain stasis within the desired cells as required.
  • an effect against a proliferative disorder mediated by a kinase 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 including, but not limiting to A549, A2780, HT29, Saos-2, HCT-116, HeLa, MCF-7, NCI-H460 or by showing inhibition of a CDK enzyme such as CDK1, CDK2, CDK4, CDK5, CDK6, CDK7, CDK8, CDK9, CDK11, or other protein kinases in an appropriate assay.
  • CDK enzyme such as CDK1, CDK2, CDK4, CDK5, CDK6, CDK7, CDK8, CDK9, CDK11, or other protein kinases in an appropriate assay.
  • PLKs regulate some fundamental aspects of mitosis. Both PLK1 and PLK2 may have additional post-mitotic functions. Deregulated PLK expressions result in cell cycle arrest and apoptosis. Compounds of the invention are therefore believed to be of use in treating PLK-mediated conditions, particularly proliferative disorders.
  • a further embodiment relates to the use of compounds of the invention, or pharmaceutically acceptable salts thereof, in the manufacture of a medicament capable of treating a viral disorder mediated by one or more of the host cell CDKs involved in viral replication, i.e. CDK1 , CDK2, CDK4, CDK7, CDK8, CDK9 or CDK11 as hereinbefore defined.
  • a medicament capable of treating a viral disorder mediated by one or more of the host cell CDKs involved in viral replication, i.e. CDK1 , CDK2, CDK4, CDK7, CDK8, CDK9 or CDK11 as hereinbefore defined.
  • a medicament capable of treating a viral disorder mediated by one or more of the host cell CDKs involved in viral replication, i.e. CDK1 , CDK2, CDK4, CDK7, CDK8, CDK9 or CDK11 as hereinbefore defined.
  • a viral disorder mediated by one or more of the host cell CDKs involved in viral replication, i.e. CDK1
  • Such medicament is useful in the treatment of viral disorders, such as human cytomegalovirus (HCMV), herpes simplex virus type 1 (HSV-1), human immunodeficiency virus type 1 (HIV-1), and varicella zoster virus (VZV).
  • HCMV human cytomegalovirus
  • HSV-1 herpes simplex virus type 1
  • HV-1 human immunodeficiency virus type 1
  • VZV varicella zoster virus
  • CDK dependent disorders are associated with an above normal level of activity of one or more CDK enzymes. Such disorders are typically associated with an abnormal, level of activity of CDK1, Cp.K2,. CDK4, CDK7j CDK8, CDK9 - and/or CDK11.
  • 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, CDK1 , CDK2, CDK4, CDK7, CDK8 CDK9 and/or CDK11 can be said to be part of the sensitive metabolic pathway and inhibitors of these CDKs may therefore be active in treating such disorders.
  • the medicament of the invention is capable of inhibiting CDK2, CDK7, and/or CDK9.
  • Yet another embodiment relates to the use of compounds of the invention, or pharmaceutically acceptable salts thereof, in the manufacture of a medicament capable of treating cardiovascular diseases mediated by one or more CDKs.
  • a medicament capable of treating cardiovascular diseases mediated by one or more CDKs.
  • Preferably such medicament is useful in treating cardiovascular diseases.
  • a cardiovascular disease may be selected from the group consisting of ischaemic heart disease (also known as myocardial infarction or angina), hypertension, heart failure, restenosis and cardiomyopathy.
  • ischaemic heart disease also known as myocardial infarction or angina
  • hypertension also known as myocardial infarction or angina
  • heart failure also known as restenosis
  • cardiomyopathy cardiomyopathy
  • Cardiac hypertrophy is characterised by global increases in mRNA and protein synthesis.
  • CDK9 activity has been demonstrated to be necessary for hypertrophy in cardiomyocytes.
  • Heart-specific activation of CDK9 by cyclin T1 was found to provoke hypertrophy.
  • Compounds of the invention are believed to inhibit
  • CDK9 and are therefore believed to be of use in the prevention and treatment of cardiac hypertrophy.
  • Yet another embodiment relates to the use of a compound of the invention in the manufacture of a medicament capable of treating neurodegenerative disorders mediated by one or more GSKs or CDKs.
  • a medicament capable of treating neurodegenerative disorders mediated by one or more GSKs or CDKs.
  • Preferably such medicament is useful in the treatment of neurodegenerative disorders such as Alzheimer's disease.
  • Tau is a GSK-3 substrate which has been implicated in the etiology of Alzheimer's disease.
  • Tau co-assembles with tubulin into microtubules.
  • 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.
  • Another, embodiment - relates to the use, of. compounds of the invention, or pharmaceutically , acceptable- salts thereof, in the manufacture of a medicament for treating a metabolic disorder mediated by one or more GSKs.
  • the medicament is useful in treating metabolic disorders.
  • Metabolic disorders include Type Il diabetes (non insulin dependent diabetes mellitus) and diabetic neuropathy. Compounds of the invention are believed to inhibit GSK-3, which is implicated in Type Il diabetes.
  • GSK3 is one of several protein kinases that phosphorylate glycogen synthase (GS) and is involved in the stimulation of glycogen synthesis by insulin in skeletal muscle. 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 Il 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 Il diabetes.
  • Another embodiment relates to the use of compounds of the invention, or pharmaceutically acceptable salts thereof, in the manufacture of a medicament for treating bipolar disorder mediated by one or more kinases. Preferably such medicament is useful in treating bipolar disorder.
  • Yet another embodiment relates to the use of compounds of the invention, or pharmaceutically acceptable salts thereof, in the manufacture of a medicament for treating a stroke mediated by one or more GSKs.
  • a medicament for treating a stroke mediated by one or more GSKs.
  • Preferably such medicament is useful in 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 embodiment relates to the use of compounds of the invention, or pharmaceutically acceptable salts thereof, in the manufacture of a medicament for treating alopecia mediated by one or more GSKs.
  • a medicament for treating alopecia mediated by one or more GSKs Preferably such medicament is useful in treating alopecia.
  • GSK3 inhibitors may 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 condition mediated by one or more enzymes selected from a CDK, aurora kinase, GSK, PLK or tyrosine kinase enzyme as hereinbefore defined.
  • Ia one, preferred embodiment such -condition is a GSK3-dependent. disorder, said ⁇ method comprising . administering to a subject in need thereof, a compound of the invention or a pharmaceutically acceptable salt thereof, as defined above in an amount sufficient to inhibit GSK3.
  • the compound of the invention, or pharmaceutically acceptable salt thereof is administered in an amount sufficient to inhibit GSK3 ⁇ .
  • the invention in another preferred embodiment, relates to a method of treating a PLK-dependent disorder, said method comprising administering to a subject in need thereof, a compound of the invention or a pharmaceutically acceptable salt thereof, as defined above in an amount sufficient to inhibit a PLK.
  • the compound of the invention is administered in an amount sufficient to inhibit PLK1 , PLK2 and/or PLK3.
  • the invention in another preferred embodiment, relates to a method of treating an aurora kinase-dependent disorder, said method comprising administering to a subject in need thereof, a compound of the invention or a pharmaceutically acceptable salt thereof, as defined above in an amount sufficient to inhibit an aurora kinase.
  • a compound of the invention Preferably is administered in an amount sufficient to inhibit aurora kinase A, aurora kinase B or aurora kinase C.
  • the invention in another preferred embodiment, relates to a method of treating a tyrosine kinase-dependent disorder, said method comprising administering to a subject in need thereof, a compound of the invention or a pharmaceutically acceptable salt thereof, as defined above in an amount sufficient to inhibit a tyrosine kinase.
  • the compound of the invention is administered in an amount sufficient to inhibit at least one of BCR-ABL, IKK, FLT3, JAK, LCK, PDGF, Src, or VEGF.
  • the invention in another preferred embodiment, relates to a method of selectively treating a protein kinase-dependent disorder, said method comprising administering to a subject in need thereof, a compound of the invention or a pharmaceutically acceptable salt thereof, as defined above in an amount sufficient to inhibit a selected protein kinase.
  • a method of selectively treating a protein kinase-dependent disorder comprising administering to a subject in need thereof, a compound of the invention or a pharmaceutically acceptable salt thereof, as defined above in an amount sufficient to inhibit a selected protein kinase.
  • said method comprising contacting said protein kinase with a compound of the invention.
  • the compound of the invention is administered in an amount sufficient to inhibit at least one of a CDK, GSK, aurora kinase, or PLK 1 or a tyrosine kinase including, but not limiting to BCR-ABL, IKK, FLT3, JAK, LCK, PDGF, Src, or VEGF.
  • a CDK CDK
  • GSK aurora kinase
  • PLK 1 a tyrosine kinase including, but not limiting to BCR-ABL, IKK, FLT3, JAK, LCK, PDGF, Src, or VEGF.
  • the protein kinase is a CDK.
  • CDK inhibitors under development suffer from a number of problems including a promiscuous kinase inhibitor profile which, apart from multiple CDK inhibition, also potently inhibits other kinases, resulting in observations of toxicity.
  • Other CDK inhibitors under clinical and late-clinical predevelopment are either pan-specific, belonging to the oligo-specific CDK2-CDK7-CDK9 class or are CDK4/6 specific.
  • discovery-stage compounds with modest CDK9 selectivity > 10 fold with respect to CDK2 and /or CDK7 have been reported, the determinants for CDK9 selectivity are not currently understood in the published art.
  • CDK7 has an additional role as a general CDK-activating kinase (CAK), while CDK9 appears to function exclusively in the regulation of transcription.
  • CAK general CDK-activating kinase
  • CDK9 also has functions in pre-mRNA splicing.
  • CDK9 is necessary and sufficient for effective reversal of apoptotic resistance in CLL.
  • CCD C-terminal domain
  • CDK9 is unique in apparently lacking cell-cycle related roles.
  • studies on the effect of depletion of CDK1, CDK2, CDK7 and CDK9 on cellular apoptosis suggest that inhibition of cell cycle CDK functions may not contribute to the elimination of CLL cells and may in fact be undesirable because of antiproliferative effects on nontransformed cells in general, which may manifest as toxicity.
  • the compound of formula I is capable of inhibiting at least one CDK enzyme, preferably at least one of CDK2, CDK7 and CDK9.
  • a compound of formula I is capable of inhibiting a CDK, more particularly CDK2, CDK7 or CDK9 at sub-micromolar IC 50 values, more preferably at IC 50 of less than 0.5 micromolar, more preferably less than 0.25 micromolar.
  • Such compounds of formula I include compounds of formula I':
  • compounds of formula I are capable of exhibiting an antiproliferative effect in human cell lines, as measured by a standard 72h MTT cytotoxicity assay.
  • the compound of formula I exhibits an IC 50 value of less than 1 micromolar.
  • Such compounds of formula I include compounds of formula I':
  • a method of treating a proliferative disease or disorder, a viral disorder, a cardiovascular disease, a CNS disorder, an autoimmune disease, a metabolic disorder, stroke, alopecia, an inflammatory disease or an infectious disease comprising administering to a subject in need thereof, a compound of formula I as hereinbefore defined in an effective amount.
  • a compound of the invention in the manufacture of a medicament as hereinbefore defined includes the use of the compound directly, or in any stage of the manufacture of such a medicament, or in vitro in a screening programme to identify further agents for the prevention or treatment of the hereinbefore defined diseases or conditions.
  • a further aspect of the invention relates to the use of a compound of formula I or a pharmaceutically acceptable salt or solvate or physiologically hydrolysable, solubilising or immobilising derivative thereof, in an assay for identifying candidate compounds capable of treating one or more disorders or diseases as hereinbefore defined.
  • a compound is of use in identifying candidate compounds capable of inhibiting a protein kinase, more preferably one or more of a CDK 1 aurora kinase, GSK 1 PLK or tyrosine kinase enzyme.
  • a pharmaceutical composition comprising a therapeutically effective amount of a compound of formula I or its physiologically acceptable salt and physiologically hydrolysable derivative as hereinbefore defined in association with one or more pharmaceutical carriers, excipients or diluents.
  • Suitable carriers, excipients or diluents may be selected having regard to the intended mode of administration and standard practice.
  • the pharmaceutical compositions may be for human or animal usage in human and veterinary medicine, preferably for treatment of a condition, disease or disorder as hereinbefore defined or in inhibiting one or more protein kinase enzyme, more preferably one or more of a CDK, aurora kinase, GSK, PLK or tyrosine kinase enzyme.
  • Suitable carriers include lactose, starch, glucose, methyl cellulose, magnesium stearate, mannitol, sorbitol and the like.
  • a therapeutically effective amount is any amount from 0.1% to 99.9% w/w.
  • a composition of the invention is suitably for any desired mode of administration including oral, rectal, vaginal, parenteral, intramuscular, intraperitoneal, intraarterial, intrathecal, intrabronchial, subcutaneous, intradermal, intravenous, nasal, buccal or sublingual and the like.
  • a composition for oral administration is suitably formulated as a compressed tablet, tablet, capsule, gel capsule, powder, solution, dispersion, suspension, drops or the like.
  • Such forms may be produced according to known methods and may include any suitable binder, lubricant, suspending agent, coating agent or solubilising agent or combinations thereof.
  • a composition for administration by means of injection is suitably formulated as a sterile solution or emulsion from a suitable solution or powder.
  • a composition may be in the form of suppositories, pessaries, suspensions, emulsions, lotions, creams, ointments, skin patches, gels, solgels, sprays, solutions or dusting powders.
  • An indicated daily dosage is from about 1mg to about IOOOmg and compositions generally contain from about 0.25mg to about 250mg of the active ingredient per dose.
  • a composition may include one or more additional active ingredients or may be administered together with compositions comprising other active, ingredients for the treatment of the. same, or different condition.
  • Coadministration may be simultaneously, consecutively or sequentially.
  • An additional active ingredient is suitably selected from other existing anticancer agents. This may be desirable to prevent an overlap of major toxicities, mechanism of action and resistance mechanisms and to enable administration of drugs at their maximum tolerated doses with minimum time intervals between doses. Coadministration is also favoured to promote additive or possible synergistic effects. Selection of other active ingredients and regime of administration may be having regard to a knowledge of agents which are effective in treatment of cell lines derived from the cancer to be treated.
  • Suitable antiproliferative agents that may be used in combination with a compound of the invention include DNA damaging agents, anti-metabolites, anti-tumour antibiotics, dihydrofolate reductase inhibitors, pyrimidine analogues, purine analogues, cyclin-dependant kinase inhibitors, thymidylate synthase inhibitors, DNA intercalators, DNA cleavers, topoisomerase inhibitors, anthracyclines, vinca drugs, mitomycins, bleomycins, cytotoxic nucleosides, pteridine drugs, diynenes, podophyllotoxins, platinum containing drugs, differentiation inducers and taxanes. Suitable examples of these drugs are known in the art.
  • the compounds of the invention display a CDK and cell line selectivity which is not displayed by known anti-proliferative drugs and therefore, co-administration is recommended having regard to desired selectivity.
  • a compound as hereinbefore defined may be in free form, i.e.normally as a base, or in any suitable salt or ester form. Free forms of the compound may be converted into salt or ester form and vice versa, in conventional manner.
  • Suitable salts include hydrochloride, dihydrochloride, hydroformate, amide, succinate, half succinate, maleate, acetate, trifluoroacetate, fumarate, phthalate, tetraphthalate, benzoate, . sulfonate, . sulphate, , phosphate, oxalate,, malonate, hydrogen- malonate, ascorbate, glycolate,.
  • acids for acid addition salt formation include the corresponding acids, i.e. hydrochloric, formic, amino acid, succinic, maleic, acetic, trifluoroacetic, fumaric, phthalic, tetraphthalic, benzoic, sulfonic, sulphuric, phosphoric, oxalic, malonic, ascorbic, glycolic, lactic, malic, tartaric, citric, aspartic or glutamic acids and the like.
  • acids i.e. hydrochloric, formic, amino acid, succinic, maleic, acetic, trifluoroacetic, fumaric, phthalic, tetraphthalic, benzoic, sulfonic, sulphuric, phosphoric, oxalic, malonic, ascorbic, glycolic, lactic, malic, tartaric, citric, aspartic or glutamic acids and the like.
  • Suitable esters include those obtained with the above acids, with hydroxides such as sodium, potassium, calcium or the like, or with alcohols.
  • the compounds of formula I may be present as one or both enantiomeric or tautomeric forms, or stereo or geometric isomeric forms, where relevant. Such forms may be identified and prepared or isolated by methods known in the art. Reference herein to compounds of formula I also encompasses reference to crystalline forms, polymorphs, hydrous and anhydrous forms and prodrugs thereof.
  • The. invention - is -not restricted to. the details of .any. foregoing embodiments.
  • the invention extends to any novel one, or any novel combination, of the features disclosed in this specification (including any accompanying claims, abstract and drawings), or to any novel one, or any novel combination, of the steps of any method or process so disclosed.
  • Cyanuric chloride (4.58 g, 24.81 mmol, 1 eq.) was dissolved in 20 ml of dry THF in an oven-dried three- necked flask equipped with a dropping funnel, a bubbler and a thermometer. The resulting solution was cooled down to -20 0 C and the Grignard reagent was added dropwise via the dropping funnel keeping temperature of the reaction mixture below -15 0 C during the addition. After the addition completed, the content of the flask was stirred for 45 min at -15 0 C. The reaction mixture was quenched with water (100 ml), extracted with ethyl acetate (3 X 50 ml).
  • 2,4-Dichloro-6-(3'-methoxyphenyl)-1,3,5-triazine (0.379 g, 1.48 mmol, 1 eq.) was dissolved in DMF (7 ml) in the presence of NaHCO 3 (0.249 g, 2.96 mmol, 2 eq.) and 4-aminophenol (1.48 mmol, 1 eq.) was introduced into the flask.
  • the reaction mixture was allowed to stir at room temperature till the starting materials disappeared by TLC.
  • the reaction mixture was quenched with water (30 ml). Resulted precipitate was filtered and washed with several times with water.
  • the aqueous solution was extracted with ethyl acetate (3 X 10 ml).
  • Aqueous ammonia (1 ml) was added to a solution of 2-anilino-4-(3'-methoxyphenyI)-6-chloro-1 ,3,5- triazine (0.152 mmol) in 1,4-dioxane (2 ml) and the reaction mixture was slowly heated to 60 0 C over 2 hours.
  • the content of the flask was diluted with water (2 ml), extracted with diethyl ether (3 x 2 ml). Combined organic layers were dried over MgSO 4 , the solvent was evaporated under reduced pressure and the residue was subjected to a flash column chromatography.
  • 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.
  • the reactions were initiated by addition of Mg/ATP mix (15 mM MgCI 2 + 100 ⁇ M ATP with 30-50 kBq per well of [ ⁇ - 32 P]-ATP) and mixtures incubated as required at 30 °C. Reactions were stopped on ice, followed by filtration through p81 filterplates or GF/C filterplates (Whatman Polyfiltronics, Kent, UK).
  • IC 50 values concentration of test compound which inhibits kinase activity by 50 %).
  • MTT cytotoxicity assay The compounds from the examples above were subjected to a standard cellular proliferation assay using the method described previously (Haselsberger, K. et al. Anti Cancer Drugs 1996, 7, (3), 331-8. Loveland, B. E. et al. Biochemistry International 1992, 27, (3), 501-10). Human tumour cell lines were obtained from ECACC (European Collection of Cell Cultures). Standard 72-h MTT (thiazolyl blue; 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyItetrazolium bromide, 2mg/ml in phosphate buffered saline) assays were performed.
  • test compounds were made up in DMSO and a 1/3 dilution series prepared in 100 ⁇ L cell media, added to cells (in triplicates) and incubated for 72 hr at 37 0 C.
  • 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 ⁇ L PBS. MTT solution was then added at 20 ⁇ L per well and incubated in the dark at 37 0 C for 4 h. MTT solution was removed and cells again washed with 200 ⁇ L PBS.
  • MTT dye was solubilised with 200 ⁇ L per well of DMSO with agitation. Absorbance was read at 550 nm on an Anthos Labtec Systems plate reader. The data analysis used program Deltasoft 3TM and Microsoft Excel to determine IC 50 or Gl 50 " values (concentration of test compound which inhibits cell growth by 50 %).
  • CLL apoptosis assay Compounds were thawed on ice and aliquotted to 0.5ml microcentrifuge tubes and stored at -20'C to avoid multiple freeze-thaw cycles. Compound aliquots were thawed on ice and diluted as required in sterile PBS immediately prior to drugging experiment.
  • Primary CLL cells were isolated from ACD whole blood using standard Ficoll (Ficoll-Paque Plus.GE Healthcare) separation and selected for B cells with R ⁇ setteSep B ' cell enrichment cocktail (StemCell tecfh); Cells- were incubated at 1 E6-3E6 cells/ml in RPMI 1640 plus 10% human serum and antibiotics at 37'C in 24 well plates.
  • Inhibitor compounds were added at time 0 and a sample was maintained with media vehicle only. At 24 hours, cells were transferred to a 12x75 tube for Annexin-PI viability assay. Cells were centrifuged at 1500rpm for 5 minutes then incubated at RT in dark for 30 minutes with appropriate reagent plus binding buffer with calcium. After incubation, 80OuI of binding buffer was added for flow cytometry analysis on the EPICS-XL (Beckman-Coulter).
  • Mia-Paca-2 0.468 mean 0.433

Abstract

A compound of formula (I) and its pharmaceutically acceptable salts or solvates and physiologically hydrolysable, solubilising or immobilisable derivatives wherein: Ar is a 5-membered heteroaryl ring wherein X1 and X2 are one or two heteroatoms or Ar is a 6-membered aromatic ring, wherein heteroatoms are selected from S, O, N, Se; Z is NH, NHCO, NHSO2, N-alkyl, CH2NH, CH2N-alkyl, CH2, CH2CH2, CH=CH, CH2CONH, SO2, or SO; Y is N or CR3; R1, R2, R5, R6, R7, R8 and R9 are each independently H, or a substituent; R3, when present, is selected from alkyl and a substituent, with the proviso that when Y is CR3, Ar is a 5- membered heterocycle comprising one or two N heteroatoms and Z is NH, then R3 is selected from C3+ alkyl and a substituent; R4 is selected from H, alkyl and R13 as hereinbefore defined, with the proviso that when R3 is absent, R4 is selected from alkyl and a substituent; processes for the preparation thereof, intermediates and precursors therefore and the use thereof as a medicament, and therapeutic compositions comprising the compound.

Description

PYRIMIDINES, TRIAZINES AND THEIR USE AS PHARMACEUTICAL AGENTS
The present invention relates to substituted pyrimidine and [1,3,5]triazine derivatives that have broad therapeutic applications via inhibiting one or more protein kinases. The invention also provides processes for preparing compounds, pharmaceutically acceptable compositions comprising the compounds, and the use of the compounds and methods of using the compounds and compositions in the treatment of various diseases, conditions, or disorders.
BACKGROUND
The search for new therapeutic agents has been greatly aided in recent years by a better understanding of the structure of enzymes and their biomolecules associated with diseases. One important class of enzymes that has been the subject of extensive study is the proteiη kinase family.
The protein kinase family is one of the largest in the human genome, comprising 500 genes. 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, whose terminal phosphate group transfers covalently to its macromolecular substrates. The protein kinases may be categorized by the substrates they phosphorylate, e.g. protein-serine/threonine, protein-tyrosine.
Protein kinases mediate intracellular signalling by effecting a phosphoryl transfer from a nucleoside triphosphate to a protein acceptor that is involved in a signalling pathway. These phosphorylation events are triggered in response to a variety of extracellular and other stimuli and act as molecular on/off switches that can modulate or regulate the target protein biological function. An extracellular stimulus may affect one or more cellular_responses related to cell growth, migration, differentiation, secretion of, hormones, activation of transcription factors, muscle contraction, giucdse "metabolism, control of proteirr' synthesis, and regulation of the cell cycle.
Many diseases are associated with abnormal cellular responses triggered by protein kinase-mediated events. These diseases include, but are not limited to allergies and asthma, Alzheimer's disease, autoimmune diseases, bone diseases, cancer, cardiovascular diseases, inflammatory diseases, hormone- related diseases, metabolic diseases, neurological and neurodegenerative diseases. Accordingly, there has been a substantial effort in medicinal chemistry to find protein kinase inhibitors that are effective as therapeutic agents.
A wide variety of molecules capable of inhibiting protein kinase function through antagonising ATP binding are known in the art. We have previously disclosed 2-anilino-4-heteroaryl-pyrimidine compounds with kinase inhibitory properties, particularly against cyclin-dependent kinases (CDKs) (Wang, S.; et al. WO 2003029248, Cyclacel Limited, UK. Fischer, P.M., WO2002079193, Cyclacel Limited, UK. Wang, S.; Fischer, P.M. US2002019404, Cyclacel Limited, UK.; Fischer, P.M.; Wang, S. WO2001072745, Cyclacel Limited, UK) and.2-anilino-4-phenyl-pyrimidine (Wang S., et al. WO2005012262, Cyclacel Limited, UK). Also known are [1,3,5]Triazines with kinase inhibitory properties (Liu C, WO2004032875, Squibb Bristol Myers Co. US; Armistead, D M, et al. WO200125220, Kinetix Pharmaceuticals Inc. US). Cyclin-dependent kinases (CDKs) are serine/threonine protein kinases that associate with various cyclin subunits, playing pivotal roles in the regulation of cell cycle progression and transcriptional cycle. Ten distinct CDKs (CDK1-9 and 11) are involved in a variety of important regulatory pathways in eukaryotic cells, including cell-cycle control, apoptosis, neuronal physiology, differentiation and transcription.
CDKs may be classified into two major groups, reflecting their functions. The cell cycle regulator CDKs composed primarily of CDK1, CDK2, CDK3, CDK4 and CDK6 function with their cyclin partners including cyclin A, B, D1 , D2, D3, E, and F to regulate promotion of the cell cycle. The transcription regulator CDKs1 which include CDK7, CDK8, CDK9 and CDK11 work together with cyclin C, H, K, L1, L2, T1 and T2, tend to play roles in transcriptional regulation.
The CDKs have been implicated in cell proliferation disorders, particularly in cancer. Cell proliferation is a result of the direct or indirect deregulation of the cell division cycle and the CDKs play a critical role in the regulation of the various phases of this cycle. Therefore, inhibitors of CDKs and their associated cyclins are useful targets for cancer therapy.
CDKs also play a role in apoptosis and T-cell development, which is predominantly due to the CDK functions in regulation of transcription. For example, clear clinical activity has very recently been obtained in chronic lymphocytic leukaemia (CLL) with CDK inhibitor flavopiridol. CLL is characterised by cellular resistance to apoptosis through up-regulation of anti-apoptotic proteins. Inhibition of transcription at the level of CDK9, which is necessary for mRNA elongation, selectively reinstates apoptosis in CLL cells. There is however a need for pharmacologically and pharmaceutically superior CDK inhibitors with a well- defined kinase selectivity and cellular specificity profile. and,;anti-CLL efficacy, as well as efficacy against other CDK mediated disorders.
Furthermore, numerous viruses require CDKs, particular CDK2, CDK7, and CDK9, for their replication process. CDK inhibitors that restrain viral replication including human immunodeficiency virus, human cytomegalovirus, herpes virus, and varicella-zoster virus have been reported.
Inhibition of CDKs, particular CDK9, is a novel strategy for potential treatment of cardiovascular diseases including cardiohypertrophy. Cardiohypertrophy is characterised by global increases in mRNA and protein synthesis. CDK7 and CDK9 are closely associated with cardiac hypertrophy as they are the main drivers for transcription. Therefore inhibition of CDK9 and its associated cyclins is a relevant drug target for cardiovascular diseases.
Inhibition of CDK is also useful for the treatment of neurodegenerative disorders such as Alzheimer's disease. The appearance of Paired Helical Filaments, associated with Alzheimer's disease, is caused by the hyperphosphorylation of Tau protein by CDK5/p25. Inhibition of one or more other serine/threonine kinases including the Aurora kinases, Glycogen synthesis kinases (GSKs), polo-like kinases (PLKs) and tyrosine kinases including Ableson tyrosine kinase (BCR- ABL), FMS-related tyrosine kinases (FLT), IkB kinases (IKK), Janus kinases (JAK), platelet-derived growth factor (PDGF) receptor tyrosine kinases, vascular endothelial growth factor (VEGF) receptor tyrosine kinases, and Src family are also useful for the treatment of numerous diseases, conditions or disorders mediated by these kinases.
GSK3 is known to phosphorylate many substrates and is thus involved in the regulation of multiple biochemical pathways. For example, GSK is highly expressed in the central and peripheral nervous systems. GSK3 inhibition is therefore of therapeutic significance in the treatment of CNS disorders such as Parkinsons and Alzheimers diseases.
Furthermore, it has been demonstrated that GSK3 is over-expressed in muscle cells of type Il diabetics and that an inverse correlation exists between skeletal muscle GSK3 activity and insulin action. GSK3 inhibition is therefore of therapeutic significance in the treatment of diabetes, particularly type II, and diabetic neuropathy.
Aurora kinases and PLK are also important therapeutic targets for treatment of proliferative disorders. Based on their known functions inhibition of Aurora kinases and PLKs activity should disrupt mitosis leading to cell cycle arrest and therefore slowing tumour growth and induce apoptosis.
The present invention provides a novel class of substituted-2-anilino-4-arylpyrimidines and -4-aryl- [1,3,5]triazin-2-ylphenylamines with broad therapeutic application as protein kinase inhibitors, specifically compounds which are substituted at the'2-jj,4-,-and .5- and/or 6- positions of. pyrimidines or at the 2T, A- and 6-positions of [1,3,6]triazines. These compounds have been synthetically difficult to access. We have found that the invention offers a class of compounds which are effective in protein kinase inhibition, offer important benefits in terms of selective inhibition, and are potentially effective therapeutics.
BRIEF SUMMARY OF THE DISCLOSURE
A first aspect of the present invention relates to a compound of formula I and its pharmaceutically acceptable salts or solvates and physiologically hydrolysable, solubilising or immobilisable derivatives:
Figure imgf000004_0001
Wherein:
Ar is optionally substituted and is a 5-membered heteroaryl ring wherein X1 and X2 are one or two heteroatoms or Ar is a 6-membered aromatic ring, wherein heteroatoms are selected from S, O, N, Se and wherein optional substituents include R1 and R2; Z is NH, NHCO, NHSO2, N-alkyl, CH2NH, CH2N-alkyl, CH2, CH2CH2, CH=CH, CH2CONH, SO2, or SO;
Y is N or CR3;
R1, R2, R5, R6, R7, R8 and R9 are each independently H, alkyl, or R13 wherein R13 is selected from R10, alkyl-R10, aryl, heteroaryl and combinations of two or more thereof and combinations with one or more alkyl and R11, or R13 is one or more moieties R14 selected from O-, N-, NH-, CO-, COO-, CON-, CONH-, SO2-, SO2N-, SO2NH- linking one or more alkyl, aryl, heteroaryl or R10 or R11 groups or combinations thereof, directly or via a moiety selected from alkylene, arylene, heteroarylene or combination thereof, wherein alkyl, aryl, heteroaryl groups or moieties thereof may be substituted with one or more groups R15 selected from halogeno, NH2, NO2, CN, OH1 COOH, CONH2, C(=NH)NH2, SO3H, SO2NH2, SO2CH3, OCH3, CF3 or R13 is selected from a group R15;
or two of R5 to R9 are linked to form a cyclic ether containing one or more oxygen atoms;
R3, when present, is selected from alkyl and R13 as hereinbefore defined, with the proviso that when Y is CR3, Ar is a 5-membered heterocycle comprising one or two N heteroatoms and Z is NH, then R3 is selected from C3+ alkyl and R13 as hereinbefore defined;
R4 is selected from H, alkyl and R13 as hereinbefore defined, with the proviso that when R3 is absent, R4 is selected from alkyl and R13 as hereinbefore defined;
wherein, at least one of R1, R2, R4, R?, R6, R-, K8 and R9 and R3 or R12 where present, comprise a group R10 or R11 wherein R10 and R11 comprise one or more solubϊlising moieties chosen from i) neutral hydrophilic groups, ii) ionisable organic acids, iii) ionisable organic bases and combinations thereof.
A further aspect of the invention relates to a compound of formula I wherein at least one of R10 or R11 further comprises an immobilising moiety chosen from iv) chemical functions or moieties providing covalent or non-covalent attachment or binding to a solid phase or an immobile receptor.
Further aspects of the invention relate to a process for the preparation of a compound of formula I as hereinbefore defined, to a process for the preparation of precursors or intermediates, and to novel precursors or intermediates.
In a further aspect the invention relates to the use of a compound of formula I or a pharmaceutically acceptable salt, solvate or physiologically hydrolysable, solubilising or immobilising derivative thereof, in the manufacture of a medicament for treating a condition mediated by an enzyme selected from one or more CDK, aurora kinase, GSK, PLK, BCR-ABL, FLT, IKK, JAK, PDGF or VEGF and Src family enzymes, particularly from one or more CDK2, CDK7, CDK8, CDK9, CDK11 , GSK-3, aurora kinase, PLK or at least one tyrosine kinase. In a further aspect of the invention, there is provided a method for treating a condition mediated by one or more enzymes selected from CDK, aurora kinase, GSK, PLK, BCR-ABL, FLT, IKK, JAK, PDGF or VEGF and Src family enzymes, particularly from one or more CDK2, CDK7, CDK8, CDK9, CDK11 , GSK-3, aurora kinase, PLK or tyrosine kinase enzyme, in a human or animal subject, the method comprising administering to a human or animal in need thereof a therapeutically effective amount of a compound of formula I or a pharmaceutically acceptable salt, solvate, or physiologically hydrolysable, solubilising or immobilising derivative thereof.
In a further aspect of the invention, there is provided the use of a compound of formula I or a pharmaceutically acceptable salt, solvate, or physiologically hydrolysable, solubilising or immobilising derivative thereof in a method for treating a condition mediated by an enzyme selected from one or more CDK, aurora kinase, GSK, PLK, BCR-ABL1 FLT, IKK, JAK, PDGF or VEGF and Src family enzymes, particularly from one or more CDK2, CDK7, CDK8, CDK9, CDK11, GSK-3, aurora kinase, PLK or tyrosine kinase.
A further aspect of the invention relates to the use of a compound of formula I or a pharmaceutically acceptable salt or solvate or physiologically hydrolysable, solubilising or immobilising derivative thereof, in an assay for identifying candidate compounds capable of treating a condition mediated by an enzyme selected from one or more CDK, aurora kinase, GSK, PLK, BCR-ABL, FLT, IKK, JAK, PDGF or VEGF and Src family enzymes, particularly from one or more CDK2, CDK7, CDK8, CDK9, CDK11, GSK-3, aurora kinase, PLK or tyrosine kinase.
A further -aspect of -the- invention- relates -.to a pharmaceutical composition comprising a compound : of formula I or a pharmaceutically acceptable salt or solvate, or physiologically hydrolysable, solubilising or immobilising derivative thereof, in association with one or more diluents, carriers or excipients.
BRIEF DESCRIPTION OF THE DRAWINGS
[0001] Embodiments of the invention are further described hereinafter with reference to the accompanying drawings, in which Schemes 1 and 2 illustrate processes for preparing compounds of the invention.
DETAILED DESCRIPTION
As used herein the term "alkyl" includes both straight chain and branched alkyl groups. The alkyl group may be substituted (mono- or poly-) or unsubstituted. Suitable substituents include, for example, halo, CF3, OH, CN, NO2, SO3H, SO2NH2, SO2Me, NH2, COOH, CONH2 and alkoxy. Preferably, the alkyl group is a Ci-20 alkyl group, more preferably a C1-15, more preferably still a C1-12 alkyl group, more preferably still, a C1-6 alkyl group, more preferably a C1-3 alkyl group. Particularly preferred alkyl groups include, for example, methyl, ethyl, propyl, isopropyl, butyl, isobutyl, tert-butyl, pentyl and hexyl.
As used herein, the term "heteroalkyl" includes an alkyl group as defined above which comprises one or more heteroatoms.
As used herein, the term "cycloalkyl" refers to a cyclic alkyl group which may be substituted (mono- or poly-) or unsubstituted. Suitable substituents include, for example, halo, CF3, OH, CN, NO2, SO3H, SO2NH2, SO2Me, NH2, COOH, CONH2 and alkoxy.
Likewise, the term "cycloheteroalkyl" refers to a cyclic heteroalkyl group which may be substituted (mono- or poly-) or unsubstituted. Suitable substituents include, for example, halo, CF3, OH, CN, NO2, SO3H, SO2NH2, SO2Me, NH2, COOH, CONH2 and alkoxy. Preferred cycloheteroalkyl groups include morpholino, piperazinyl and piperidinyl groups.
As used herein, the term "aryl" refers to an aromatic, substituted (mono- or poly-) or unsubstituted group, and includes, for example, phenyl, naphthyl etc. Again, suitable substituents include, for example, halo, CF3, OH, CN, NO2, SO3H, SO2NH2, SO2Me, NH2, COOH, CONH2 and alkoxy.
As used herein, the term "heteroaryl" refers to an aromatic, substituted (mono- or poly-) or unsubstituted group, which comprises one or more heteroatoms. Preferred heteroatoms include N, S, O. Preferred heteroaryl groups include pyrrole, pyrazole, pyrimidine, pyrazine, pyridine, qύinoline, triazine, triazole, thiophene, selenazol, thiazole and furan. Again, suitable substituents include, for example, halo, CF3, OH, CN, NO2, SO3H, SO2NH2, SO2Me, NH2, COOH, CONH2 and alkoxy.
As used herein the term "halo" or "halogeno" refers to F, Cl, Br or I.
lri a first embodiment of the invention there- is- provided a compound' of formula V - . ■''. • ■.. . .- -. -. . ■.;■. /■'- .?■
Figure imgf000007_0001
wherein X1 and X2 are each independently selected from NH or N, O, S1 Se, CH and C R15 and at least one of X1 and X2 is selected from NH or N, O, S and Se, and wherein R15 is as hereinbefore defined for R1, and all other variables are as hereinbefore defined.
Preferably there is provided a compound of formula I' wherein: one of X1 and X2 is CH or CR15, and the other of X1 and X2 is S, O, NH, NR15, or Se; or one of X1 and X2 is S, O or Se, and the other of X1 and X2 is N; or one ofX1 and X2 is N, and the other of X1 and X2 is NH or NR15; and wherein all other variables are as hereinbefore defined. More preferably X1 is S and X2 is N or X2 is S and X1 is N.
In an alternative embodiment of the invention there is provided a compound of formula I"
Figure imgf000008_0001
wherein all variables are as hereinbefore defined.
More preferably, a compound of formula I' or I" comprises a mono- or di-substituted phenyl, thiazol-4-yl, thiazol-5-yl, imidazol-4-yl, imidazol-5-yl, pyrrol-4-yl or pyrrol-5-yl group attached to the pyrimidine or [1,3,5]triazine ring through one of the ring carbon atoms; most preferably a phenyl, thiazol-4-yl or thiazol-5- yl group.
.Where R10 or R11 comprises a neutral hydrophilic group (i) as hereinbefore defined, this preferably includes groups containing mono-, di- and polyhydroxylated saturated or unsaturated aliphatic, alicyclic or aromatic systems, carbohydrate derivatives, ethers and polyethers optionally containing one or more hydroxyl groups, O- and/or S-containing heterocyclic systems optionally containing one or more hydroxy! groups, aliphatic or aromatic systems containing a carboxamide, sulfoxide, sulfone, or sulfonamide function, and halogenated alkylcarbonyl groups.
Where R1 ° or R11 comprises an ϊbnisabie'organic acid (iϊ) as hereinbefore defined* this preferably includes groups comprising one or more of the functions COOH, SO3H, OSO3H, PO3H2, and OPO3H2.
Where R10 or R11 comprises an ionisable basic group (iii) as hereinbefore defined, this preferably includes aliphatic, alicyclic, aromatic, or heterocyclic groups comprising one or more of the functions -0-, -NH2, - NH-, =N-, quarternary amine salts, guanidine, and amidine, optionally substituted by one or more substituents selected from halogen, S02alkyl, alkyl optionally substituted by one or more OH or halogen groups, CHO, COalkyl, aralkyl, COOalkyl and an ether group substituted by one or more OH groups.
In one embodiment R10 and R11 may consist of natural or unnatural amino acid residues and peptides, or their derivatives.
Preferably R10 or R11 is selected from v) OSO3H, PO3H2, OPO3H2; vi) Y where Y1 is selected from aliphatic, alicyclic, aromatic, or heterocyclic groups comprising one or more of the functions -0-, -NH2, -NH-, =N-, amidine, optionally substituted by one or more substituents selected from halogen, S02alkyl, alkyl optionally substituted by one or more OH or halogen groups,
COalkyl, aralkyl, COOalkyl and an ether group substituted by one or more OH groups; (vii) NHCO(CH2)m[NHCO(CH2)m.]p[NHCO(CH2)m..]qY' or NHCO(CHZ)1NH(CH2VY1 where p and q are each 0 or 1, and m, m';m", t and t' are each independently an integer from 1 to 10; and
(viii) (CHz)nNR19COR17, (CH2)n.NR20SO2R18, or SO2R21, where R17, R18 and R21 are each alkyl groups optionally comprising one or more heteroatoms, and which are optionally substituted by one or more substituents selected from OH, NH2, halogen and NO2, R19 and R20 are each independently H or alkyl, and n and n' are each independently 0, 1, 2, or 3;
(ix) an ether or polyether optionally substituted by one or more hydroxyl groups or one or more Y' groups;
(X) (CH2)rNH2; where r is 0, 1 , 2, or 3;
(xi) (CH2VOH; where r" is O, 1 , 2, or 3;
(xii) (CH2VNR22COR23 where R22 is H or alkyl, n" is O, 1 , 2 or 3 and R23 is an aryl or heteroaryl group, each of which may be optionally substituted by one or more substituents selected from halogeno, NO2,
OH, alkoxy, NH2, COOH, CONH2 and CF3;
(xiii) SO2NR24R25 where R24 and R25 are each independently H, alkyl, aralkyl, CO-alkyl or aryl, with the proviso that at least one of R24 and R25 is other than H, or R24 and R25 are linked to form a cyclic group optionally containing one or more heteroatoms selected from N, O and S, and wherein said alkyl, aryl or cyclic group is optionally substituted by one or more substituents selected from halogeno, NO2, OH, , alkoxy, NH2, COOH, CH2CO2-alkyl, CONH2 and CF3;
(xiv) N-piperidinyl, piperidinyl, N-piperazinyl, N-diazepanyl, N-pyridinyl, N-pyrrolidinyl, N-morpholinyl or
N-thiomorpholinyl, each of which may be optionally substituted by one or more alkyl, alkoxy, aryl, CHO or
CO-alkyl groups.
In one preferred embodiment of the invention, each R10 or R 11 is independently selected from a C1-30 i hydrocarbyl- group, optionally comprising up. to twelve heteroatoms. selected, from N, Sr and O, and optionally bearing up to six substituents each independently selected from a group R15 as hereinbefore defined or comprising a moiety R14 as hereinbefore defined, and a group R15.
Preferably a compound of formula I as hereinbefore defined bears up to six substituents selected from R1 to R9, R12 and R16 as hereinbefore defined each comprising one or more heteroatoms selected from N, S, and O, and alternatively or additionally each comprising one or more moieties R14 or groups R15 as hereinbefore defined, wherein the combined substituents comprise up to ten heteroatoms or atoms N, S and O.
Preferably Z is NH or NR16 .
Preferably Y is N or CR3.
Preferably R13 is selected from alkyl-R10, alkyl-cycloalkyl which may be part unsaturated, alkyl- cycloheteroalkyl, aryl, aryl-R10, aralkyl, aralkyl-R10, alkyl-heteroaryl, halogeno, NO2, CN, OH, O-alkyl, O- cycloalkyl which may be part unsaturated, O-aryl, O-heteroaryl, O-R10, NH2, NH-alkyl, part unsaturated NH-cycloalkyl, NH-cycloheteroalkyl, NH-aryl, NH-heteroaryl, N-(alkyl)2, N-(aryl)2, N-(alkyl)(cycloalkyl), N- (alkyl)(cycloheteroalkyl), N-(aIkyl)(aryl), N-(alkyl)(heteroaryl), NH-R10, N-(R10)(R11), N-(alkyl)(R10), N-
(aryI)(R10), COOH, COO-R10, CONH2, CONH-alkyl, CONH-aryl, CONH-heteroaryl, CON-(alkyl)(R10), CON(aryl)(R10), CON(heteroaryl)(R10), CONH-R10, CON-(R10)(R11), NHCO-alkyl, NHCO-aryl, NHCO- heteroaryl, NHCO-R10, SO3H, SO2-alky[, SO2-alky[-R10, SO2-aryl, SO2-aryl-R10, SO2-heteroaryl, SO2- heteroaryl-R10, SO2NH2, SO2NH-R10, SO2N-(R10)(R11), NHSO2R10, CF3, CO-R10, CO-alkyl, CO-alkyl-R10, CO-cycloheteroalkyl, CO-aryl, CO-aryl-R10 CO-heteroaryl, CO-heteroarylalkyl or R10, wherein alkyl, aryl, aralkyl, heteroaryl groups may be further substituted with one or more groups selected from halogeno, NO2, CN, OH, O-methyl, NH2, COOH, CONH2 and CF3. Preferably a cycloheteroalkyl is a morpholino, piperazinyl or piperadinyl.
Preferably R1 is selected from NH-alkyl, part unsaturated NH-cycloalkyl, NH-heteroaryl, O-alkyl, O- cycloalkyl which may be part unsaturated, O-heteroaryl, alkyl-heteroaryl, alkyl-cycloalkyl which may be part unsaturated.
Preferably R2 is selected from H, alkyl, such as C1-s-alkyl, aryl, NH-alkyl, part unsaturated NH-cycloalkyl, NH-heteroaryl, O-alkyl, O-cycloalkyl which may be part unsaturated, O-heteroaryl, alkyl-heteroaryl and alkyl-cycloalkyl which may be part unsaturated.
Preferably R5 is selected from H, O-alkyl, particularly OCH3, CF3, alkyl or halogeno.
Preferably R6 and R8 are independently selected from a sulphonyl, carbonyl, amide or sulphonamide, or thioether link to an unsubstituted or substituted 6 membered cyclic or heterocyclic, or aromatic or heteroaromatic ring, wherein substituents are as hereinbefore defined. More preferably R6 and R8 are , independently selected . from SO2-cycloheteroalkyl, SO2rcycloalkyl; SO2-heteroaryl, SO-cycloheteroalkyl,.. SO-cycloalkyl, SO-heteroaryl, CO-cycloheteroalkyl, CO-cycloalkyl, CO-heteroaryl, N-(alkyl)(cycloalkyl), N- (alkyl)(cycloheteroalkyl), or N-(alkyl)(heteroaryl) more preferably wherein the cycloheteroalkyl is heteroatom linked and may be unsubstituted or substituted comprising one, two or three heteroatoms selected from N, O, S. More preferably a cycloheteroalkyl is a Λ/-alkyl-morpholino, Λ/-alkyl-piperazine or N- alkyl-piperadine. Most preferably R6 and R8 are independently selected from Λ/-linked N- (alkyl)(cycloheteroalkyl), SO2-cycloheteroalkyl and CO-cycloheteroalkyl most preferably such as N- (alkyl)(morpholino), Λ/-(alkyl)(piperazine), Λ/-(aIkyl)(piperadine), SO2-piperazines, SO2-morpholines, CO- piperazines, CO-morpholines, CO-piperadine or the like.
Preferably R7 is selected from alkyl, for example C1-5 alkyl, CONH2, CONH-alkyl, CN, OH, CF3, O-alkyl, halogeno, NH2, NH-alkyl and NHCO-alkyl.
Preferably R9 is selected from H, halogeno, O-alkyl, more preferably H, halogeno and 0-Ci-5 alkyl.
Preferably R3 is selected from C1-6 alkyl, more preferably, i-propyl, i-butyl or t-butyl, or R13 as hereinbefore defined. More preferably R3 is selected from C4+ alkyl and R13 as hereinbefore defined, or from R13 as hereinbefore defined. More preferably R3 is selected from CN, CF3, halogeno, NO2, NH2, NH-alkyl, N- (alkyl)(R10), NH-cycloheteroalkyl, NHSO2R10, CONH2, CONH-(alkyl), CON-(alkyl)(R10), R10, CO- cycloheteroalkyl, CO-heteroaryl, CONH-heteroaryl, CH2-cycloheteroalkyl, CH2-heteroaryl, cycloheteroalkyl, heteroaryl, and C2-6 or C4-6 alkyl, wherein alkyl, cycloheteroalkyl, aryl, aralkyl, heteroaryl groups may be further substituted with one or more groups selected from halogeno, NO2, CN OH, O-methyl, NH2, COOH, CONH2 and CF3.
Preferably R4 is selected from alkyl and R13 as hereinbefore defined; more preferably R4 is selected from amino, halogeno, such as Cl, and alkyl.
Preferably up to six of R1, R2, R3, R4, R5, R6, R7, R8, R9, R12 and R16, for example one, two, three or four thereof, shall correspond to or contain one or more of the group R10 or R11. Preferably either or both of R3 and R4 comprise or contain one or more of the group R10 or R11. Two or more groups R10 and/or R11 may be the same or different.
Preferably R1, R2, R3, R5 or R7 comprises or contains a solubilising moiety R10 or R11.
One preferred embodiment of the invention relates to a compound of formula I' wherein: one of X1 and X2 is selected from S, O1 NH, NR15 and Se and the other thereof is N. Y is CR3 or N; Z is NH; each R13 is alkyl-R10, alkyl-cycloalkyl which may be part unsaturated, alkyl-cycloheteroalkyl, aryl, aryl-R10, aralkyl, aralkyl-R10, alkyl-heteroaryl, halogeno, NO2, CN, OH, O-alkyl, O-cycloalkyl which may be part unsaturated, O-aryl, O-heteroaryl, O-R10, NH2, NH-alkyl, part unsaturated NH-cycloalkyl, NH-Gyoloheteroalkyli- 1. NH-arylr •. N
Figure imgf000011_0001
H-heteroaryl,.^-- Nτ(alkyl)2-, > N-(alkyl)(cyclόalkyl), N- (alkyl)(cycloheteroalkyl), N-(alkyl)(aryl), N-(alkyl)(heteroaryl), NH-R10, N-(R10)(R11), N-(alkyl)(R10), N- (aryl)(R10), COOH, COO-R10, CONH2, CONH-alkyl, CONH-aryl, CONH-heteroaryl, CON-(alkyl)(R10), CON(aryl)(R10), CON(heteroaryl)(R10), CONH-R10, CON-(R10)(R11), NHCO-alkyl, NHCO-aryl, NHCO- heteroaryl, NHCO-R10, SO3H, SO2-alkyl, SO2-alkyl-R10, SO2-aryl, SO2-aryl-R10, SO2-heteroaryl, SO2- heteroaryl-R10, SO2NH2, SO2NH-R10, SO2N-(R10)(R11), NHSO2R10, CF3, CO-R10, CO-alkyl, CO-alkyl-R10 CO-cycloheteroalkyl, CO-aryl, CO-aryl-R10, CO-heteroaryl, CO-heteroarylalkyl or R10, wherein alkyl, aryl, aralkyl, heteroaryl groups may be further substituted with one or more groups selected from halogeno, NO2, CN, OH, O-methyl, NH2, COOH, CONH2 and CF3. More preferably R4 is amino, halogeno, or alkyl; More preferably Rs is O-alkyl, CF3, alkyl, or halogeno;
More preferably each R6 or R8 is independently SO2-cycloheteroalkyl, SO2-heteroaryl, SO- cycloheteroalkyl, SO-heteroaryl, CO-cycloheteroalkyl or CO-heteroaryl; preferably, the cycloheteroalkyl group is a /V-alkyl-morpholino, Λ/-alkylpiperazine, Λ/-alkylpiperadine; More preferably R7 is alkyl, CN, OH, CF3, O-alkyl, halogeno, NH2, CONH-R10, NHR10, or NHCO-R10.
More preferably a solubilising moiety R10 or R11 corresponds to or is contained within R1, R2, R3 or R5 and R9 is H. Especially preferred compounds of the invention are those of formula I' wherein one X1 or X2 is S, another X1 or X2 is N1 Y is CR3 or N, Z is NH, R1 and R2 are amino, alkyl, heteroaryl or aryl, R3 is C1-4 alkyl, CN1 CF3, halogeno, NO2 O-alkyl, NH2, NH-alkyl, N(alkyl)2, CO2alkyl, CO-alkyl, CONH2, CONH-alkyl or heteroaryl, R4 is amino, halogeno or alkyl; R5 is OMe1 alkyl, or halogeno, each R6 or R8 is independently S02-cycloheteroalkyl, SO-cycloheteroalkyl, SO2-heteroaryl, SO-heteroaryl, CO-cycloheteroalkyl or CO- heteroarylalkyl, R7 is alkyl, OH, CF3, O-alkyl, halogeno, or NH2 and the solubilising moiety corresponds to or is contained within R1, R2, R3 or R5; and wherein R9 is H.
Especially preferred compounds of the invention are those of formula I" wherein: Z is NH;
R1 and R2 are amino, alkyl, or aryl or R2 is H; more preferably R1 and R2 are selected from NH-alkyl, part unsaturated NH-cycloalkyl, NH-heteroaryl, O-alkyl, O-cycloalkyl which may be part unsaturated, O- heteroaryl, alkyl-heteroaryl, alkyl-cycloalkyl which may be part unsaturated; or R2 is H, alkyl, such as C1-5- alkyl or aryl, such as C6 aryl;
R3 is CN, CONH-alkyl, CF3, halogeno, NO2, heteroaryl or is contained with R13; R4 is amino, halogeno or alkyl; R5 is OMe, alkyl, or halogeno; each R6 and R8 is independently selected from SO2-cycloheteroalkyl, SO-cycloheteroalkyl, CO- cycloheteroalkyl and CO-heteroaryl; R7 is alkyl, OH, CF3, O-alkyl, halogeno, or NH2; R9 is H; and the solubilising moiety corresponds to or is contained within R1, R2, R3 or R5. ntion include, compounds of formula I': "•• ■
Figure imgf000012_0001
Figure imgf000012_0002
Figure imgf000013_0001
and compounds of formula I":
Figure imgf000014_0001
as shown in Table 3
Figure imgf000014_0003
and compounds of formula I':
Figure imgf000014_0002
Figure imgf000014_0004
Figure imgf000015_0007
Wherein
MS = morpholine-4-suIfonyl °' "b
PzC = piperazine-1-carbonyl or piperazin-1-ylmethanone
Figure imgf000015_0001
Figure imgf000015_0002
AcPzC = 4-Acetylpiperazine-1-carbonyl
MePzC = 4-methylpiperazine-1 -carbonyl or 4-methylpiperazin-1 -ylmethanone I or
"O
M = morpholino '^
MC = morpholin-4-carbonyl or morpholin-4-yl-methanone
Figure imgf000015_0003
PzS = piperazin-1-ylsulfonyl \^NH
Figure imgf000015_0004
MePzS = 4-methylpiperazin-1-ylsulfonyl ^-N
BPzS = 4-benzylpiperazin-1-ylsulfonyI
BPzC - benzylpiperazine-1 -carbonyl
BPdC = 1-benzylpiperidin-4-carbonyl o
Figure imgf000015_0005
r 1-benzylpiperidin-4-ylmethanone
Figure imgf000015_0006
PdC = piperidine-4-carbonyI or piperidin-4-ylmethanone
Figure imgf000016_0001
MePdC = 1-methylpiperidin-4-ylmethanone or 1-methylpiperidin-4-carbonyl
°JXXrO'
MePdCB - 4-(1-methylpiperidine-4carbonyl)benzoyl o
Figure imgf000016_0002
Pz = piperazin-1-yl
MePz = 4~methylpiperazin-1-yl ON.
Py = pyridine-3-yl Λ^N
PyEtA = 2-(pyιϊdin-3-yl)ethylamino N 3
Figure imgf000016_0003
PyMeA = pyridin-3-ylmethylamino
■ ' ■ ΓΛ
MeDz = 4-methyl-1 ,4-diazepan-1-yl
and their pharmaceutically acceptable salts, solvates and physiologically hydrolysable, solubilising or immobilisable derivatives.
In a further aspect of the inventioh there is provided a compound of formula I as hereinbefore defined wherein one or more R10 or R11 alternatively or additionally comprise devices for immobilisation thereof. Such devices may be chemical functions that can be used for covalent attachment to solid phases such as functionalised polymers (e.g. agarose, polyacrylamide, polystyrene efc.) as commonly found in matrices (microtitre plate wells, microbeads, membranes, efc.) used for biochemical assays and affinity chromatography. Alternatively, the devices may be small molecules (e.g. biotin) or polypeptides (e.g. antigens), which can be used for non-covalent immobilisation through binding to an immobilised receptor (e.g. avidin or streptavidin in the case of biotin, or a specific antibody in the case of antigens).
In a further aspect of the invention there is provided a precursor to a compound of formula I as hereinbefore defined wherein one or more R10 or R11 is a solubilising moiety comprising a natural or unnatural amino acid residue, peptide or derivative as hereinbefore defined.
In a further aspect of the invention there is provided a process for the preparation of a compound of formula 1 as hereinbefore defined. Compounds of formula I may be prepared by any methods known in the art.
Suitably a process for the preparation of a compound of formula I as hereinbefore defined comprises: (1) reacting a compound of formula III (as illustrated hereinbelow), where Ar is a mono- or di-substituted phenyl, thiazol-4-yl, thiazol-5-yl, imidazol-4-yl, imidazol-5-yl, pyrrol-4-yl or pyrrol-5-yl, preferably Ar is a phenyl, thiazol-4-yl or thiazol-5-yl group, Y is N, or CR3 and L1 is a leaving group, with a compound of formula IV (as illustrated hereinbelow), where Z and R5 to R9 are as hereinbefore defined;
or (2) reacting a compound of formula Vl (as illustrated hereinbelow) where Z and R5 to R9 are as hereinbefore defined with a compound of formula VII (as illustrated hereinbelow), where Ar is as hereinbefore defined and Y is N;
or (3) reacting a compound of formula Xl (as illustrated hereinbelow) where Y is N or CR3, L3 is any leaving group, preferable halogeno group and Ar and R4 are as hereinbefore defined, with a compound of formula XII (as illustrated hereinbelow) where Z and R5 to R9 are as hereinbefore defined.
Preferably the compound of formula I is a compound of formula V as hereinbefore defined, more preferably where Ar is a mono- or di-substituted thiazol-4-yl, thiazol-5-yl, imidazol-4-yl, imidazol-5-yl, pyrrol-4-yl or pyrrol-5-yl attached to the pyrimidine or [1,3,5]triazine ring through one of the ring carbon atoms, most preferably Ar is a thiazol-4-yl or thiazol-5-yl group; or is a compound of formula I" as hereinbefore defined, more preferably wherein the compounds of formula I" bear a mono- or di- substituted phenyl attached to the pyrimidine or [1,3,5]triazine ring through one of the ring carbon atoms.
Preferably in process (1) L1 is any leaving group including N(alkyl)2, halogen, ester, thioester, more preferably, NMe2, and the process (1) comprises a variety of methods for example as disclosed in Fischer PM, Wang S. WO 2001072745; and Wang, S.; et. al WO 2003029248, Cyclacel Limited, UK and references therein.
Preferably process (2) is conducted via a variety of methods known in the art, particularly, methods described by Liu, C (Liu, C, et al. 2007, Tetrahedron Lett. 48, 435) and Hodous, B (Hodous, B. L. J Med Chem, 50, 611).
Preferably in process (3), the compound of formula XI (as illustrated hereinbelow) is obtained by reacting a compound of formula VIlI (as illustrated hereinbelow) where L2 is any leaving group, preferably a halogeno group, and Y, L3 and R4 are as hereinbefore defined, with a compound of formula X (as illustrated hereinbelow), where Ar is as hereinbefore defined and L4 is any boronic acid or derivatives. Palladium-catalysed cross-coupling of VIII (Y is N or CR3) with X or derivatives affords 4-arylated 2- halogeno-pyrimidines or [1.3.6]triazines Xl, which are preferably aminated with anilines XII as hereinbefore defined. Alternatively, treatment of VIII with X, and a Grignard reagent such as alkyl magnesium bromide, forms Xl which reacts as hereinbefore defined with XII to provide the compounds of formula I.
More preferably the process is as illustrated in Scheme 1 below: Suitably an alternative process for the preparation of a compound of formula P as hereinbefore defined comprises: -
(4) condensation reaction between a compound of formula VH' (as illustrated hereinbelow) wherein R1, R2, R4, X1, X2, Y and L2 are as hereinbefore defined with a phenylguanidine of formula VIII' (as illustrated hereinbelow) wherein Z and R5 to R9 are as hereinbefore defined to obtain pyrimidines or [1,3,5]tria2ines of formula I';
or (5) treatment of amidines Vl' (as illustrated hereinbelow) wherein Z and R4 to R9 are as hereinbefore defined in the presence of POCI3 followed by alkylation reaction with anilines of formula XII (as illustrated hereinabove) to obtain [1 ,3,5]triazine triazines of formula I';
or (6) condensation reaction of a compound of formula XII' (as illustrated hereinbelow) wherein Z and R5 to R9 are as hereinbefore defined with an amidine of formula XIIP (as illustrated hereinbelow) wherein R1, R2, X1 and X2 are as hereinbefore defined, in the presence of base to yield a [1,3,5]triazine of formula r.
Preferably the compound of formula P is as hereinbefore defined, more preferably is a mono- or di- substituted thiazol-4-yl, thiazol-5-yl, imidazol-4-yl, imidazol-5-yl, pyrrol-4-yl or pyrrol-5-yl attached to the pyrimidine or [1,3,5]triazine ring through one of the ring carbon atoms; most preferably is a thiazol-4-yl or thiazol-5-yl group.
Preferably process (4) uses the method described previously (Wang, S. et al.J Med Chem, 2004, 47, 1662-75).
Preferably in process (4) or (5), VH' may be obtained from the corresponding Vl' by reaction with N1N'- dimethylformamide dimethylacetal (where R4 = H, L = NMe2) or ferf-butoxy-bis(dimethylamino)methane (Bredereck, H.;.et al. Chemische Berichte 1964, 97, (12), 3397).
Preferably ketones Vl' (Y = CH3 ) or amides (Y = NH2) are obtained by cyclisation reaction between II' (L1 = Cl, Br) with IIP (amides while X1 = O; thioamides while X1 = S, X2 = N, R1 = alkyl, NH-alkyl as hereinbefore defined). Compounds Vl' may also be prepared by treatment of ketones X' with HI' followed by the Friedel-Crafts acylation (Y = CH3), or amination (Y = NH2).
Preferably guanidines VHI' are obtained by reaction of cyanamide or certain of its derivatives using the method of Katritzky, A. R.; et al. Synthetic Communications 1995, 25, 1173.
Preferably in process (6) a compound of formula XIIP is obtained by reacting phenyl isothiocyanate sodium hydrogen cyanamide to provide Λ/-cyanothiourea XIP.
More preferably the process is as illustrated in Scheme 2 below: In a further aspect of the invention there are provided novel chemical intermediates of formula I, F and F',
IV, Vl, VII, Xl, XII, VF, VIF, VIIF, XF or XIIF, as hereinbefore defined.
THERAPEUTIC USE
In a further aspect of the invention there is provided the use of one or more compounds of formula I or salts, solvates or derivatives as hereinbefore defined in the manufacture of a medicament for treating a condition mediated by one or more of a CDK, aurora kinase, GSK, PLK and one of Tyrosine kinases as hereinbefore defined, preferably such medicament is capable of inhibiting such enzymes. The compounds of the invention may inhibit any of the steps or stages in the cell cycle.
In one embodiment such medicament is suitable for inhibition of a proliferative disorder mediated by a CDK or PLK, preferably is useful in the treatment of a proliferative disorder, such as cancers, leukaemias and other disorders associated with uncontrolled cellular proliferation such as psoriasis and restenosis, a viral disorder, a cardiovascular disease, a CNS disorder, an autoimmune disease, a bond disease, a hormone-related disease, a metabolic disorder, stroke, alopecia, an inflammatory disease or an infectious disease.
Preferably the compound of formula I is capable of inhibiting one or more of the host cell kinases involved in cell proliferation, viral replication, a cardiovascular disorder, neurodegeneration, autoimmunity, a metabolic disorder, stroke, alopecia, an inflammatory disease or an infectious disease.
A proliferative disorder requires treatment of a susceptible neoplasm and may be selected from the group consisting of chronic lymphocytic ieύkaemia, lymphoma, leukaemia, breast cancer, lung cancer, prostate cancer, colon cancer, melanoma, pancreatic cancer, ovarian cancer, squamous carcinoma, carcinoma of head and neck, endometrial cancer, and aesophageal carcinoma.
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 and cardiomyopathy, auto-immune disorders such as glomerulonephritis and rheumatoid arthritis, dermatological disorders such as psoriasis, anti-inflammatory, anti-fungal, antiparasitic disorders such as malaria, emphysema and alopecia. In these disorders, the compounds of the present invention may induce apoptosis or maintain stasis within the desired cells as required.
As defined herein an effect against a proliferative disorder mediated by a kinase 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 including, but not limiting to A549, A2780, HT29, Saos-2, HCT-116, HeLa, MCF-7, NCI-H460 or by showing inhibition of a CDK enzyme such as CDK1, CDK2, CDK4, CDK5, CDK6, CDK7, CDK8, CDK9, CDK11, or other protein kinases in an appropriate assay. These assays including methods for their performance are described in more detail under Biological
Activity.
Studies have shown that human PLKs regulate some fundamental aspects of mitosis. Both PLK1 and PLK2 may have additional post-mitotic functions. Deregulated PLK expressions result in cell cycle arrest and apoptosis. Compounds of the invention are therefore believed to be of use in treating PLK-mediated conditions, particularly proliferative disorders.
A further embodiment relates to the use of compounds of the invention, or pharmaceutically acceptable salts thereof, in the manufacture of a medicament capable of treating a viral disorder mediated by one or more of the host cell CDKs involved in viral replication, i.e. CDK1 , CDK2, CDK4, CDK7, CDK8, CDK9 or CDK11 as hereinbefore defined. Preferably such medicament is useful in treating a viral disorder.
Assays for determining CDK activity are described in more detail in the accompanying examples. Using such enzymes assays it may be determined whether a compound is anti-viral in the context of the present invention.
Preferably such medicament is useful in the treatment of viral disorders, such as human cytomegalovirus (HCMV), herpes simplex virus type 1 (HSV-1), human immunodeficiency virus type 1 (HIV-1), and varicella zoster virus (VZV).
Typically such disorder 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 are typically associated with an abnormal, level of activity of CDK1, Cp.K2,. CDK4, CDK7j CDK8, CDK9 - and/or CDK11. 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, CDK1 , CDK2, CDK4, CDK7, CDK8 CDK9 and/or CDK11 can be said to be part of the sensitive metabolic pathway and inhibitors of these CDKs may therefore be active in treating such disorders.
For use in the treatment of viral disorders, preferably the medicament of the invention is capable of inhibiting CDK2, CDK7, and/or CDK9.
Yet another embodiment relates to the use of compounds of the invention, or pharmaceutically acceptable salts thereof, in the manufacture of a medicament capable of treating cardiovascular diseases mediated by one or more CDKs. Preferably such medicament is useful in treating cardiovascular diseases.
A cardiovascular disease may be selected from the group consisting of ischaemic heart disease (also known as myocardial infarction or angina), hypertension, heart failure, restenosis and cardiomyopathy.
Cardiac hypertrophy is characterised by global increases in mRNA and protein synthesis. CDK9 activity has been demonstrated to be necessary for hypertrophy in cardiomyocytes. Heart-specific activation of CDK9 by cyclin T1 was found to provoke hypertrophy. Compounds of the invention are believed to inhibit
CDK9 and are therefore believed to be of use in the prevention and treatment of cardiac hypertrophy.
Yet another embodiment relates to the use of a compound of the invention in the manufacture of a medicament capable of treating neurodegenerative disorders mediated by one or more GSKs or CDKs. Preferably such medicament is useful in the treatment of neurodegenerative disorders such as 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. 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.
The appearances of Paired Helical Filaments, associated with Alzeimer's disease, are caused by the hyperphosphorylation of Tau protein by CDK5-p25. Compounds of the invention are believed to inhibit CDK5 and are therefore believed to be of use in the prevention and treatment of neurodegenerative disorders.
Another, embodiment -relates to the use, of. compounds of the invention, or pharmaceutically, acceptable- salts thereof, in the manufacture of a medicament for treating a metabolic disorder mediated by one or more GSKs. Preferably the medicament is useful in treating metabolic disorders.
Metabolic disorders include Type Il diabetes (non insulin dependent diabetes mellitus) and diabetic neuropathy. Compounds of the invention are believed to inhibit GSK-3, which is implicated in Type Il diabetes.
GSK3 is one of several protein kinases that phosphorylate glycogen synthase (GS) and is involved in the stimulation of glycogen synthesis by insulin in skeletal muscle. 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 Il 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 Il diabetes. Another embodiment relates to the use of compounds of the invention, or pharmaceutically acceptable salts thereof, in the manufacture of a medicament for treating bipolar disorder mediated by one or more kinases. Preferably such medicament is useful in treating bipolar disorder.
Yet another embodiment relates to the use of compounds of the invention, or pharmaceutically acceptable salts thereof, in the manufacture of a medicament for treating a stroke mediated by one or more GSKs. Preferably such medicament is useful in treating a stroke.
Reducing neuronal apoptosis is an important therapeutic goal in the context of head trauma, stroke, epilepsy, and motor neuron disease. 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 embodiment relates to the use of compounds of the invention, or pharmaceutically acceptable salts thereof, in the manufacture of a medicament for treating alopecia mediated by one or more GSKs. Preferably such medicament is useful in treating alopecia.
The ectopic application of GSK3 inhibitors may 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 condition mediated by one or more enzymes selected from a CDK, aurora kinase, GSK, PLK or tyrosine kinase enzyme as hereinbefore defined.
Ia one, preferred embodiment such -condition, is a GSK3-dependent. disorder,, said ^method comprising . administering to a subject in need thereof, a compound of the invention or a pharmaceutically acceptable salt thereof, as defined above in an amount sufficient to inhibit GSK3.
Preferably, the compound of the invention, or pharmaceutically acceptable salt thereof, is administered in an amount sufficient to inhibit GSK3β.
In another preferred embodiment, 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 the invention or a pharmaceutically acceptable salt thereof, as defined above in an amount sufficient to inhibit a PLK.
Preferably the compound of the invention is administered in an amount sufficient to inhibit PLK1 , PLK2 and/or PLK3.
In another preferred embodiment, 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 the invention or a pharmaceutically acceptable salt thereof, as defined above in an amount sufficient to inhibit an aurora kinase. Preferably the compound of the invention is administered in an amount sufficient to inhibit aurora kinase A, aurora kinase B or aurora kinase C.
In another preferred embodiment, the invention relates to a method of treating a tyrosine kinase- dependent disorder, said method comprising administering to a subject in need thereof, a compound of the invention or a pharmaceutically acceptable salt thereof, as defined above in an amount sufficient to inhibit a tyrosine kinase.
Preferably the compound of the invention is administered in an amount sufficient to inhibit at least one of BCR-ABL, IKK, FLT3, JAK, LCK, PDGF, Src, or VEGF.
In another preferred embodiment, the invention relates to a method of selectively treating a protein kinase- dependent disorder, said method comprising administering to a subject in need thereof, a compound of the invention or a pharmaceutically acceptable salt thereof, as defined above in an amount sufficient to inhibit a selected protein kinase. Preferably said method comprising contacting said protein kinase with a compound of the invention.
Preferably the compound of the invention is administered in an amount sufficient to inhibit at least one of a CDK, GSK, aurora kinase, or PLK1 or a tyrosine kinase including, but not limiting to BCR-ABL, IKK, FLT3, JAK, LCK, PDGF, Src, or VEGF.
In a preferred embodiment of this aspect, the protein kinase is a CDK. Preferably, the protein kinase is ,:CDK1 , CDK2r.:CDK3v.C-DK4, CDK55, GDK6; CDK7,, GDK8, CDK9= and. CDK11, more preferably,CDK2,,, CDK7 or CDK9.
Known CDK inhibitors under development suffer from a number of problems including a promiscuous kinase inhibitor profile which, apart from multiple CDK inhibition, also potently inhibits other kinases, resulting in observations of toxicity. Other CDK inhibitors under clinical and late-clinical predevelopment are either pan-specific, belonging to the oligo-specific CDK2-CDK7-CDK9 class or are CDK4/6 specific. Although discovery-stage compounds with modest CDK9 selectivity (> 10 fold with respect to CDK2 and /or CDK7) have been reported, the determinants for CDK9 selectivity are not currently understood in the published art.
Our research derives from the consideration that apoptotic ability in CLL and other tumour cells can be reinstated by interference with the expression of anti-apoptotic proteins at the transcriptional level via RNAPII, and should provide a therapeutic margin for the elimination of CLL cells while sparing non- transformed quiescent and proliferative cells. Although other CDKs - including CDK1, CDK2, CDK8 and CDK11 - have been implicated in the regulation of transcription, the roles of CDK7 and CDK9 appear to be most important in this respect. An important difference between CDK7 and CDK9 is the fact that CDK7 has an additional role as a general CDK-activating kinase (CAK), while CDK9 appears to function exclusively in the regulation of transcription. Apart from regulating transcriptional initiation and elongation,
CDK9 also has functions in pre-mRNA splicing.
Results to date strongly suggest that inhibition of CDK9 is necessary and sufficient for effective reversal of apoptotic resistance in CLL. Of all the CDKs involved in RNAPII C-terminal domain (CTD) phosphorylation, CDK9 is unique in apparently lacking cell-cycle related roles. However studies on the effect of depletion of CDK1, CDK2, CDK7 and CDK9 on cellular apoptosis suggest that inhibition of cell cycle CDK functions may not contribute to the elimination of CLL cells and may in fact be undesirable because of antiproliferative effects on nontransformed cells in general, which may manifest as toxicity.
Our research has enabled us to distinguish, both phenotypically and biochemically, between compounds that inhibit RNAP-II CDKs and those that act predominantly through inhibition of cell cycle CDKs (CDK1, CDK2, CDK4, CDK6) or the closely related mitotic kinases.
In one embodiment of the invention the compound of formula I is capable of inhibiting at least one CDK enzyme, preferably at least one of CDK2, CDK7 and CDK9.
Preferably a compound of formula I is capable of inhibiting a CDK, more particularly CDK2, CDK7 or CDK9 at sub-micromolar IC50 values, more preferably at IC50 of less than 0.5 micromolar, more preferably less than 0.25 micromolar.
Such compounds of formula I include compounds of formula I':
Figure imgf000024_0001
Figure imgf000024_0002
Figure imgf000025_0002
and compounds of formula 1":
Figure imgf000025_0001
as shown in Table 3
Figure imgf000025_0003
Figure imgf000026_0002
and compounds of formula I':
Figure imgf000026_0001
Figure imgf000026_0003
w eren a revaed substituents in the above Tables are as given hereinabove. In a further preferred embodiment compounds of formula I are capable of exhibiting an antiproliferative effect in human cell lines, as measured by a standard 72h MTT cytotoxicity assay. Preferably the compound of formula I exhibits an IC50 value of less than 1 micromolar.
Such compounds of formula I include compounds of formula I':
Figure imgf000027_0001
Figure imgf000027_0002
Figure imgf000028_0002
and compounds of formula I":
Figure imgf000028_0001
as shown in Table 3a:
Figure imgf000028_0003
wherein abbreviated substituents in the above Tables are as given hereinabove.
In a further aspect of the invention there is provided a method of treating a proliferative disease or disorder, a viral disorder, a cardiovascular disease, a CNS disorder, an autoimmune disease, a metabolic disorder, stroke, alopecia, an inflammatory disease or an infectious disease, said method comprising administering to a subject in need thereof, a compound of formula I as hereinbefore defined in an effective amount.
The use of a compound of the invention in the manufacture of a medicament as hereinbefore defined includes the use of the compound directly, or in any stage of the manufacture of such a medicament, or in vitro in a screening programme to identify further agents for the prevention or treatment of the hereinbefore defined diseases or conditions.
A further aspect of the invention relates to the use of a compound of formula I or a pharmaceutically acceptable salt or solvate or physiologically hydrolysable, solubilising or immobilising derivative thereof, in an assay for identifying candidate compounds capable of treating one or more disorders or diseases as hereinbefore defined. Preferably a compound is of use in identifying candidate compounds capable of inhibiting a protein kinase, more preferably one or more of a CDK1 aurora kinase, GSK1 PLK or tyrosine kinase enzyme.
PHARMACEUTICAL COMPOSITIONS
In a further aspect of the invention there is provided a pharmaceutical composition comprising a therapeutically effective amount of a compound of formula I or its physiologically acceptable salt and physiologically hydrolysable derivative as hereinbefore defined in association with one or more pharmaceutical carriers, excipients or diluents. Suitable carriers, excipients or diluents may be selected having regard to the intended mode of administration and standard practice. The pharmaceutical compositions may be for human or animal usage in human and veterinary medicine, preferably for treatment of a condition, disease or disorder as hereinbefore defined or in inhibiting one or more protein kinase enzyme, more preferably one or more of a CDK, aurora kinase, GSK, PLK or tyrosine kinase enzyme.
Examples of suitable carriers include lactose, starch, glucose, methyl cellulose, magnesium stearate, mannitol, sorbitol and the like.
A therapeutically effective amount is any amount from 0.1% to 99.9% w/w.
A composition of the invention is suitably for any desired mode of administration including oral, rectal, vaginal, parenteral, intramuscular, intraperitoneal, intraarterial, intrathecal, intrabronchial, subcutaneous, intradermal, intravenous, nasal, buccal or sublingual and the like.
A composition for oral administration is suitably formulated as a compressed tablet, tablet, capsule, gel capsule, powder, solution, dispersion, suspension, drops or the like. Such forms may be produced according to known methods and may include any suitable binder, lubricant, suspending agent, coating agent or solubilising agent or combinations thereof.
A composition for administration by means of injection is suitably formulated as a sterile solution or emulsion from a suitable solution or powder. Alternatively a composition may be in the form of suppositories, pessaries, suspensions, emulsions, lotions, creams, ointments, skin patches, gels, solgels, sprays, solutions or dusting powders.
An indicated daily dosage is from about 1mg to about IOOOmg and compositions generally contain from about 0.25mg to about 250mg of the active ingredient per dose.
A composition may include one or more additional active ingredients or may be administered together with compositions comprising other active, ingredients for the treatment of the. same, or different condition. Coadministration may be simultaneously, consecutively or sequentially. An additional active ingredient is suitably selected from other existing anticancer agents. This may be desirable to prevent an overlap of major toxicities, mechanism of action and resistance mechanisms and to enable administration of drugs at their maximum tolerated doses with minimum time intervals between doses. Coadministration is also favoured to promote additive or possible synergistic effects. Selection of other active ingredients and regime of administration may be having regard to a knowledge of agents which are effective in treatment of cell lines derived from the cancer to be treated.
Suitable antiproliferative agents that may be used in combination with a compound of the invention include DNA damaging agents, anti-metabolites, anti-tumour antibiotics, dihydrofolate reductase inhibitors, pyrimidine analogues, purine analogues, cyclin-dependant kinase inhibitors, thymidylate synthase inhibitors, DNA intercalators, DNA cleavers, topoisomerase inhibitors, anthracyclines, vinca drugs, mitomycins, bleomycins, cytotoxic nucleosides, pteridine drugs, diynenes, podophyllotoxins, platinum containing drugs, differentiation inducers and taxanes. Suitable examples of these drugs are known in the art.
In a particular advantage the compounds of the invention display a CDK and cell line selectivity which is not displayed by known anti-proliferative drugs and therefore, co-administration is recommended having regard to desired selectivity.
A compound as hereinbefore defined may be in free form, i.e.normally as a base, or in any suitable salt or ester form. Free forms of the compound may be converted into salt or ester form and vice versa, in conventional manner. Suitable salts include hydrochloride, dihydrochloride, hydroformate, amide, succinate, half succinate, maleate, acetate, trifluoroacetate, fumarate, phthalate, tetraphthalate, benzoate, . sulfonate, . sulphate,, phosphate, oxalate,, malonate, hydrogen- malonate, ascorbate, glycolate,. lactate, malate, tartarate, citrate, aspartate or glutamate and variants thereof. Suitable acids for acid addition salt formation include the corresponding acids, i.e. hydrochloric, formic, amino acid, succinic, maleic, acetic, trifluoroacetic, fumaric, phthalic, tetraphthalic, benzoic, sulfonic, sulphuric, phosphoric, oxalic, malonic, ascorbic, glycolic, lactic, malic, tartaric, citric, aspartic or glutamic acids and the like.
Suitable esters include those obtained with the above acids, with hydroxides such as sodium, potassium, calcium or the like, or with alcohols.
The compounds of formula I may be present as one or both enantiomeric or tautomeric forms, or stereo or geometric isomeric forms, where relevant. Such forms may be identified and prepared or isolated by methods known in the art. Reference herein to compounds of formula I also encompasses reference to crystalline forms, polymorphs, hydrous and anhydrous forms and prodrugs thereof.
Throughout the description and claims of this specification, the words "comprise" and "contain" and variations of the words, for example "comprising" and "comprises", means "including but not limited to", and is not intended to (and does not) exclude other moieties, additives, components, integers or steps. Throughout the description and claims of this specification, the singular encompasses the plural unless the context otherwise requires. In particular, where the indefinite article is used, the specification is- to be understood as contemplating plurality as well as singularity, unless the context requires otherwise.
Features, integers, characteristics, compounds, chemical moieties or groups described in conjunction with a particular aspect, embodiment or example of the invention are to be understood to be applicable to any other aspect, embodiment or example described herein unless incompatible therewith.
The reader's attention is directed to all papers and documents which are filed concurrently with or previous to this specification in connection with this application and which are open to public inspection with this specification, and the contents of all such papers and documents are incorporated herein by reference.
All of the features disclosed in this specification (including any accompanying claims, abstract and drawings), and/or all of the steps of any method or process so disclosed, may be combined in any combination, except combinations where at least some of such features and/or steps are mutually exclusive. .
Each feature disclosed in this specification (including any accompanying claims, abstract and drawings), may be replaced by alternative features serving the same, equivalent or similar purpose, unless expressly stated otherwise. Thus, unless expressly stated otherwise, each feature disclosed is one example only of a generic series of equivalent or similar features.
The. invention -is -not restricted to. the details of .any. foregoing embodiments. The invention extends to any novel one, or any novel combination, of the features disclosed in this specification (including any accompanying claims, abstract and drawings), or to any novel one, or any novel combination, of the steps of any method or process so disclosed.
EXAMPLES
SYNTHESIS OF COMPOUNDS
General. 1H-NMR spectra were obtained using a Bruker-400 spectrometer. Chemical shifts are reported in parts per million relative to internal tetramethylsilane standard. Coupling constants (J) are quoted to the nearest 0.1 Hz. The following abbreviations are used: s, singlet; d, doublet; t, triplet; q, quartet; qu, quintuplet; m, mult'iplet and br, broad. Mass spectra were obtained using a Waters 2795 single quadrupole mass spectrometer with electrospray ionization (ESI). Microwave assisted chemistry was carried out using CEM Discovery model (Biotage Ltd. UK). TLC (thin-layer chromatography) was performed using alumina plates coated with silica gel G60. Developed plates were air dried and analysed under a UV lamp (254 / 365 nm). Silica gel (EM Kieselgel 60, 0.040-0.063 mm, Merck) or ISOLUTE pre-packed columns was used for.flash chromatography. Melting points (mp) were determined with an Electrothermal melting point apparatus and are uncorrected. Example 1 - Preparation of a compound of formula I
1.1 4-(4-Methyl-2-methylamino4hiazol-5-yl)-2-[4-methyl-3-(morpholine-4-sulfonyl)φhen pyrimidine-5-carbonitrile:
To a solution of 1~(4-methyI-2-methylamino~thiazo!~5-yl)-ethanone (14.5 mmol) in 3mL acetic acid and 10ml dichloromethane cooling on an ice bath bromine (14.5mmol) was added dropwise. The reaction mixture was stirred for 1.5 hours. If the reaction mixture turns to cake further 3mL of AcOH can be added. The solvent was evaporated in vacuo. The residue was partitioned between CH2CI2 and saturated aq. NaHCO3. The organic layer was washed with brine, dried over Na2SO4, and evaporated to give the crude 2-bromo-1-(4-methyl-2-methylamino-thiazoI-5-yl)-ethanone. Buff cream solid (95% yield): mp 149-1500C. 1H-NMR (DMSO-d6) δ: 2.35 (s, 3H, CH3), 2.45 (s, 3H, CH3), 2.85 (s, 3H, CH3), 8.48 (s, 1H, NH). HRMS (ESI) 171.0577 (M+H)+.
To a solution of 2-bromo-1-(4-methyl-2-methylamino-thiazol-5-yl)-ethanone (10 mmol) in ethanol (8ml_) was added a solution of NaCN (20mmol) in 4mL H2O. The reaction mixture was stirred at r.t. for 1 hour. The mixture was filled and the filtrate was concentrated in vacuo. The residue was poured into 3OmL ice water and stirred for 3h. The precipitates were collected and dried to give 3~(4-methyl-2-methylamino- thiazol-5-yl)-3-oxo-propionitrile. Light yellow solid: 1H-NMR (DMSO-c/6) δ: 2.45 (s, 3H, CH3), 2.86 (s, 3H, CH3), 4.38 (s, 2H, CH2), 8.65 (s, 1H1 NH). HRMS (ESI) 194.0365 (M-H)'.
3-(4-Methyl-2-methylamino-thiazol-5-yl)-3-oxo-prbpionitrire (δmifiόl) was treated "with 24"mmol of" N,N- dimethylformamide dimethylacetal in reflux for 3 hrs. The reaction mixture was concentrated in vacuo and ..purified-: by- column, chromatography ■•: to-= give 3-dimethylamino-2-(4-methyl"2÷methyIamino-thϊazoIe-5- carbonyO-acrylonitrile. Orange solid: 1H-NMR (DMSO-d6) δ 2.33 (s, 3H, CH3), 2.82 (d, J = 4.8 Hz, 3H, CH3), 3.26 (s, 3H, CH3), 3.32 (s, 3H, CH3), 7.80 (s, 1 H1 CH), 8.09 (t, J = 4.8 Hz1 1 H1 NH). HRMS (ESI) 250.9323 (M+H)+.
A mixture of 3-dimethylamino-2-(4-methyl-2-methylamino-thiazole-5-carbonyl)-acrylonitrile and an equimolar amount of Λ/-[4-methyl-3-(morpholine-4-sulfonyl)-phenyl]-guanidine in 2-methoxyethanol was microwaved at 1400C for 30min. The mixture was evaporate in vacuo and purified by column chromatography using EtOAC to elute the desired 4-(4-methyl-2-methylamino-thiazol-5-yl)-2-[4-methyl-3- (morpholine-4-sulfonyl)-phenylamino]-pyrimidine-5-carbonitrile. Yellow solid, m.p. 245-2460C. 1H-NMR (DMSO-Cf6) δ: 2.46 (s, 3H1 CH3), 2.89 (d, 3H, J = 4.4 Hz1 CH3), 3.05 (t, 4H1 J = 4.4 Hz1 CH2*2), 3.63 (t, 4H1. J = 4.4 Hz1 CH2*2), 7.43 (d, 1H1 J = 8.4 Hz1 Ph-H)1 7.95 (dd, 1H, J = 8.4, 1.6 Hz, Ph-H), 8.18 (d, 1H, J = 2.0 Hz, Ph-H)1 8.29 (q, 1H, J = 4.8 Hz, NH), 8.82 (s, 1H1 Pyimidinyl-H), 10.46 (bs, 1H1 NH). HRMS (ESI) 486.1421 (M + H+. C21H23N7O3S2 requires 486.1304).
The following compounds were synthesised by an analogous route. " ■' "' '
^.2 2-(4-Hydroxyφhenylamino)-4-(4-methyl-2-methylamino-thiazol-5-yl)φyrimidiπe-5-carbonitrile Prepared by treatment of 3-dimethylamino-2-(4-methyl-2-methylamino-thiazole-5-carbonyl)-acrylonitrile and Λ/-(4~hydroxy-phenyl)-guanidine hydrochloride. HRMS (ESI) 339.1089 (M + H+. C16Hi4N6OS requires 339.0950).
1.3 2-(3Ηydroxy-phenylamino)-4-(4-methyl-2-methylamino-thiazol-5-yI)φyrimidine-5-caώ^
Prepared by treatment of 3-dimethylamino-2-(4-methyl-2-methylamino-thiazole-5-carbonyl)-acrylonitrile and /V-(3-hydroxy-phenyI)-guanidine hydrochloride. HRMS (ESI) 339.1078 (M + H+. C16H14N6OS requires 339.0950).
1.4 4-(4-Methyl-2-methylamino-thiazol-5-yl)-2-[3-(morpholine-4-carbonyl)ψhenylamm^^ carbonitrile
Prepared by treatment of 3-dimethylamino-2-(4-methyl-2-methylamino-thiaEoie-5-carbonyl)-acrylonitrile and Λ/~[3-(morpholine-4-carbonyl)-phenyl]-guanidine hydrochloride. HRMS (ESI) 436.1616 (M + H+. C21H21 N7O2S requires 436.1477).
1.5 2-(3-(4~Acetylpiperazine-1-carbonyl)phenylamino)-4-(4-methyl-2-(methylamino)- thiazol-5- yl)pyrimidine-5-carbonitrile
Prepared" by treatment of 3-dimethylarhrno-2-(4-methyI-2-methyramino-thiazole-5-carbonyI)-acryrdhitrile and 1-(3-(4-acetyl -piperazine-1-carbonyl)phenyl)guanidine. Yellow solid. HRMS (ESI) 477.1973 (M + H+. C23H24N8O2S requires 477.1743).
1.6 3~(5-cyano-4-(4-methyl-2-(methylamino)thiazol-5-yl)pyrimidin-2-ylamino) benzoic acid
Prepared from 2-(3-(4-acetylpiperazine-1-carbonyl)phenyIamino)-4-(4-methyl-2-(methyIamino)- thiazol-5- yl)pyrimidine-5-carbonitrile. Yellow solidHRMS (ESI) 367.1093 (M + H+. Ci7H14N6O2S requires 367.0899).
1.7 4-(4-methyl-2-(methylamino)thiazol-5-yl)-2-(3-nitrophenylamino)pyrimidine-5-carbonitrile
Prepared by treatment of 3-dimethylamino-2-(4-methyl-2-methylamino-thia2ole-5-carbonyl)-acrylonitrile and 1-(3-nitrophenyl)guanidine hydrochloride. HRMS (ESI) 366.0768 (M - H". C16H13N7O2S requires 366.0851).
1.8 4-(5-cyano-4-(4-methyl~2-(methylamino)thiazol-5-yl)pyrimidin-2-ylamino) benzenesulfonamide
Prepared by treatment of 3-dimethylamino-2-(4-methyl-2-methylamino-thiazole-5-carbonyl)-acrylonitrile and 4-guanidinobenzenesulfonamide. HRMS (ESI) 400.0634 (M - H". C16H15N7O2S2 requires 400.0729). 1.9 3-(5-cyano-4-(4-methyl~2-(methyIamino)thiazol-5-yl)pyrimidin-2-ylamino)benzenesulfonamidθ
Prepared by treatment of 3-dimethylamino-2-(4-methyl-2-methylamino-thiazole-5-carbonyl)-acrylonitrile and 3-guanidinobenzenesulfonamide. HRMS (ESI) 401.8300 (M + H+. C16H15N7O2S2 requires 401.0729).
1.10 4-(4-methyl-2-(methylamino) thiazol-5-yl)-2-(3-(4-methylpiperazine- 1- carbonyl)phenylamino)pyrimidine-5-carbonitrile
Prepared by treatment of 3-dimethylamino-2-(4-methyl-2-methylamino-thiazole-5-carbonyl)-acrylonitriIe and 1-(3-(4-methylpiperazine-1-carbonyl)phenyl)guanidine. Yellow solid. HRMS (ESI) 448.8561 (M + H+. C22H24N8OS requires 448.1794).
1.11 4-(4-methyl-2-(methylamino)thiazol-5-yl)-2-(4-morpholinophenylamino) pyrimidine-5-carbonitrile
Prepared by treatment of 3-dimethylamino-2-(4-methyl-2-methylamino-thiazole-5-carbonyl)-acrylonitrile and 1-(4-morpholinophenyl)guanidine. Yellow solid. HRMS (ESI) 407.8810 (M + H+. C20H21N7OS requires 407.1528).
1.12 4-(4-methyl-2-(methylamino) thiazol-5-yl)-2-(3-(morpholinosulfonyl)phenyl-amino)pyrimidine-5- carbonitrile
Prepared by treatment of 3-dimethylamino-2-(4-methyl-2-methylamino-thiazole-5-carbonyl)-acrylonitrile and 1-(3-(morpholinosulfonyl)phenyl)guanidine. Yellow solid. HRMS (ESI) 471.7335 (M + H+. C20H21N7O3S2 requires 471.1147:
1.13 4-(4-methyl-2-(methylamino)thiazol-5-yl)-2-(4-(morpholinosulfonyl)phenyl-amino)pyrimidine-^ carbonitrile
Prepared by treatment of 3-dimethylamino-2-(4-methyl-2-methylamino-thiazole-5-carbonyl)-acrylonitrile and 1-(4-(morpholinosulfonyl)phenyl)guanidine. Yellow solid. HRMS (ESI) 471.7248 (M + H+. C20H21N7O3S2 requires 471.1147.
1.144-(4^methyl-2-(methylamino)thiazol-5-yl)-2-(3-(niethylsulfonyl)phenylamino)pyrimidine-5-ca
Prepared by treatment of 3-dimethy!amino-2-(4-methyl-2-methylamino-trιiazole-5-carbonyl)-acrylonitrile and 1-(3-(methylsulfonyI)phenyl)guanidine. Yellow solid. HRMS (ESI) 400.8207 (M + H+. C17H16N6O2S2 requires 400.0776.
2.0 4-[4-Chloro-6-(3-methoxy-phenyl)-[1Λ 5]triazin-2-ylamino]-phenol
A solution of 2,4,6-trichloro-[1,3,5]-triazine (20 mmol) in toluene cooling on an ice bath was treated with 3- methoxphenyl magnesium bromide (20 mmol) dropwise. The reaction mixture was stirred for 2 h and warm to room temperature. The compound was evaporated to dryness in vacuo to give 2,4-dich!oro-6-(3- methoxy-phenyl)-[1 ,3,5]triazine as white solid. MS (ESI+) m/z 256.00 (M+H)+.
Above compound in MeCN was treated with 4-aminophenol (1 equiv molar) in the presence of diisopropylamine at room temperature for 2hrs. The reaction mixture was purified by flash chromatography using EtOAc-PE (2:1, v/v) to elute 4-[4-chloro-6-(3-methoxy-phenyl)-[1,3,5]triazin~2- ylaminoj-phenol as a yellow solid. MS (ESI+) m/z 328.06
2.1 4-(4-chloro-6-(3-methoxyphenyl)- 1, 3, 5-triazin-2-ylamino)phenol
A small crystal of I2 was placed in an oven-dried flask containing Mg (1.07 g, 44.58 mmol, 1.8 eq.) in dry THF (15 ml) to activate the metal. After the coloration disappeared, a solution of 3-bromoanisole (4.64 g, 24.81 mmol, 1 eq.) in dry THF (15 ml) was added dropwise keeping the reaction mixture warm. After the addition completed, the reaction was stirred for 30 min (the progress of the reaction was monitored by TLC).
Cyanuric chloride (4.58 g, 24.81 mmol, 1 eq.) was dissolved in 20 ml of dry THF in an oven-dried three- necked flask equipped with a dropping funnel, a bubbler and a thermometer. The resulting solution was cooled down to -20 0C and the Grignard reagent was added dropwise via the dropping funnel keeping temperature of the reaction mixture below -150C during the addition. After the addition completed, the content of the flask was stirred for 45 min at -15 0C. The reaction mixture was quenched with water (100 ml), extracted with ethyl acetate (3 X 50 ml). Combined organic extracts were washed with brine (50 ml), dried=over. JVIgSO4. -The solvent was evaporated under reduced pressure and the residue, was subjected to a flash column chromatography eluting with a mixture of petroleum ether (40-60 °C):diethyl ether = 90:10 to yield 2.65 g (42%) of 2,4-dichloro-6-(3'-methoxyphenyl)-1 ,3,5-triazine as a white solid, mp 113.8-114.0 °C( from petroleum ether (40-60 0C)); δH (400 MHz, CDCI3) 8.11 (1H, ddd, J 7.8, 1.4 and 1.0, 6'-H), 7.99 (1H1 dd, J 2.7 and 1.4, 2'-H), 7.43 (1H, app. t, J 8.0, 5'-H), 7.19 (1H, ddd, 8.2, 2.7 and 1.0, 4'-H), 3.91 (3H, s, OCH3)
2,4-Dichloro-6-(3'-methoxyphenyl)-1,3,5-triazine (0.379 g, 1.48 mmol, 1 eq.) was dissolved in DMF (7 ml) in the presence of NaHCO3 (0.249 g, 2.96 mmol, 2 eq.) and 4-aminophenol (1.48 mmol, 1 eq.) was introduced into the flask. The reaction mixture was allowed to stir at room temperature till the starting materials disappeared by TLC. The reaction mixture was quenched with water (30 ml). Resulted precipitate was filtered and washed with several times with water. The aqueous solution was extracted with ethyl acetate (3 X 10 ml). Combined organic layers were washed with brine (10 ml). The combined organic solution was dried over MgSO4. The solvent was evaporated under reduced pressure and the residue was subjected to a flash column chromatography to yield 0.481 g (99%) of yellow solid eluting with petroleum ether (40-60 °C):EtOAc=70:30; mp 164.1-164.3 0C (from toluene); δH (400 MHz, DMSO-αV) 10.50 and 1046 (1H, 2 x s), 9.38 and 9.38 (1H, 2 X s), 7.79-7.93 (2H, m, Ar), 7.39-7.51 (3H, m, Ar), 7.19- 7.21 (1H, m, Ar), 6.76-6.81 (2H1 m, Ar), 3.83 and 3.82 (3H, 2 x s, OCH3); HRMS (ESI) 329.0823 (M + H+.
C16H14N4 35CIO2 requires 329.0805).
The following compounds were synthesised by an analogous route.
2.2 4-chloro-6-(3-methoxyphenyl)-N-(3-nitrophenyl)~ 1, 3, 5-triazin-2-amine
Prepared using 2,4-dichloro-6-(3'-methoxyphenyl)-1,3,5-triazine (0.379 g, 1.48 mmol), NaHCO3 (0.249 g, 2.96 mmol) and 3-nitroaniline (0.204 g, 1.48 mmol) to yield 0.518 g (98%) of a yellow solid eluting with petroleum ether (40-60 °C):AcOEt=80:20; mp 154.7-154.9 0C; HRMS (ESI) 358.0749 (M + H+. C16H13N5O3 35CI requires 358.0707).
2.3 N-(3-bromophenyl)-4-chloro-6-(3-methoxyphenyl)-1,3,5-triazin-2-amine
Prepared using 2,4-dichloro-6-(3'-methoxyphenyl)-1,3,5-triazine (0.379 g, 1.48 mmol), NaHCO3 (0.249 g, 2.96 mmol) and 3-bromoaniline (0.16 ml, 1.48 mmol) to yield 0.429 g (74%) of a white solid; mp 177.5- 177.7 °C(from toluene); HRMS (ESI) 391.0059 (M + H+. C16H13N4O 35CI 79Br requires 390.9961).
2.4 4-chloro-6-(3-methoxyphenyl)-N-(4-methyl-3-(moφholinosulfonyl)phenyl)- 1, 3, 5-triazin-2-amine
Prepared using 2,4-dichloro-6-(3'-methoxyphenyl)-1 ,3,5-triazine (0.379 g, 1.48 mmol), NaHCO3 (0.249 g, 2.96 mmol) and 4-methyl-3-(morpholine-4-sulfonyl)-aniline (0.379 g, 1.48 mmol) to yield 0.348 g (49%) of a white solid; mp 174.1-174.5 0C (from toluene); HRMS (ESI) 476.1213 (M + H+. C21H23N5O4 35CIS requires 476.1159).
General procedure for the preparation of 6-(3-methoxyphenyl)-N-aryl-[1 ,3,5]triazine-2,4-diamines.
35% Aqueous ammonia (1 ml) was added to a solution of 2-anilino-4-(3'-methoxyphenyI)-6-chloro-1 ,3,5- triazine (0.152 mmol) in 1,4-dioxane (2 ml) and the reaction mixture was slowly heated to 60 0C over 2 hours. The content of the flask was diluted with water (2 ml), extracted with diethyl ether (3 x 2 ml). Combined organic layers were dried over MgSO4, the solvent was evaporated under reduced pressure and the residue was subjected to a flash column chromatography.
2.54-(4-amino-6-(3-methoxyphenyI)-1,3,5-triazin-2-ylamino)phenol
Prepared from 4-(4-chloro-6-(3-methoxyphenyl)-1 ,3,5-triazin-2-ylamino)phenoI (50 mg, 0.152 mmol) to yield 33 mg (70%) of a light-yellow solid after column eluting with petroleum ether (40-60 °C):AcOEt = 70:30. mp 93.5-94.0 0C (decomp.); HRMS (ESI) 310.1288 (M + H+. C16H16N5O2 requires 310.1304).
2.6 6-(3-methoxyphenyl)-N2-(3-nitrophenyl)-1,3, 5-triazine-2,4-diamine Prepared from 4-chloro-6-(3-methoxyphenyl)-N-(3-nitrophenyt)-1I3)5-triazin-2-amine (54 mg, 0.152 mmol) to yield 35 mg (68%) of a light-yellow solid after column eluting with petroleum ether (40-60 °C):AcOEt=70:30; mp 196.9-197.2 0C; HRMS (ESI) 339.1213 (M + H+. C16H15N6O3 requires 339.1206).
2.76-(3-methoxyphenyl)-N2-methyl-N4^(3-nitrophenyl)-1,3, 5-triazine-2,4-diamine
Prepared from 4-chloro-6-(3-methoxyphenyl)-N-(3-nitrophenyl)-1,3,5-triazin-2-amine (49 mg, 0.137 mmol), methylamine hydrochloride (14 mg, 0.206 mmol) and Na2CO3 to yield 39 mg (81%) of a light-yellow solid; mp 170.9-171.3 0C (from petroleum ether (40-60 °C)/AcOEt); HRMS (ESI) 353.1328 (M + H+. C17H17N6O3 requires 353.1362).
2.84-(hydroxyamino)-6-(3-methoxyphenyl)-N-(3-nitrophenyl)-1, 3, 5-triazin-2-amine
Prepared from 4-chloro-6-(3-methoxyphenyl)-N-(3-nitrophenyl)-1,3,5-triazin-2-amine (61 mg, 0.171 mmol), hydroxylamine hydrochloride (18 mg, 0.256 mmol), Na2CO3 (27 mg, 0.256 mmol) in 3 ml of DMF to yield 50 mg (83%) of a light-yellow solid; mp 197.1-197.5 0C; HRMS (ESI) 355.1166 (M + H+. C16H15N6O4 requires 355.1155).
2.9 N2-(3-bromophenyl)-6-(3-methoxyphenyl)- 1, 3, 5-triazine-2, 4-diamine
Prepared from N-(3-bromophenyl)-4-chloro-6-(3-methoxyphenyl)-1 ,3,5-triazin-2-amine (73 mg, 0.186 mmol) to yield 45 mg (65%) of a off-white solid after column eluting with petroleum ether (40-60 °C):AcOEt=70:30; mp 141.9-142.1 0C; HRMS (ESI) 372.0458 (M + H+. C16H15N5O79Br requires 372.0460).
2.10 6-(3-methoxyphenyl)-N2-(4-methyl-3~(morpholinosulfonyl)phenyl)- 1, 3, 5-triazine-2, 4-diamine
Prepared from 4-chloro-6-(3-methoxyphenyl)-N-(4-methyl-3-(morpholinosulfonyl)phenyl)-1,3,5-triazin-2- amine (76 mg, 0.160 mmol) to yield 35 mg (48%) of a white solid after column eluting with petroleum ether (40-60 °C):AcOEt=30:70; mp ~ 100 0C (decomp.); HRMS (ESI) 457.1691 (M + H+. C21H25N6O4S requires 457.1658).
3.2 tert-Butyl 5-(4-chloro-6-(3-nitrophenylamino)-1,3,5-triazin-2-yl)-4-phenylthiazol-2-yl(m^
A 1.8 M solution of LDA in THF (1.1 ml, 2.00 mmol, 1.1 eq) was added to a solution of a corresponding thiazole (0.528 g, 1.82 mmol, 1 eq) in dry THF (5 ml) cooled to -78 0C (dry ice - acetone bath). The reaction mixture was stirred for 45 min. In another flask, cyanuric chloride (0.402 g, 2.18 mmol, 1.2 eq) was dissolved in dry THF (5 ml) and the resulting solution was cooled to -78 0C. The solution of formed anion was transferred via cannular to the content of the second flask and the reaction mixture was stirred for another 30 min, quenched with water (20 ml), extracted with ethyl acetate (3 x 30 ml), combined organic layers washed with brine (50 ml), dried over MgSO4, evaporated under reduced pressure and the residue was subjected to a flash column chromatography eluting with petroleum ether (40-60 °C):AcOEt=95:5 to give 0.387 g (49%) of a yellow solid; mp 163 0C (decomp.); HRMS (ESI) 438.0739 (M
+ H+. C18H18N5O2 35CI2 requires 438.0558)
Prepared using the corresponding ferf-butyl 5-(4,6-dichloro-1 ,3,5-triazin-2-yl)-4-phenylthiazol-2- yl(methyl)carbamate (98 mg, 0.224 mmol), NaHCO3 (38 mg, 0.448 mmol) and 3-nitroaniline (31 mg, 0.224 mmol) to yield 60 mg (50%) of the desired compound as yellow solid after column eluting with petroleum ether (40-60 °C):AcOEt=85:15. HRMS (ESI) 540.1460 (M + H+. C24H23N7O4 35CI S requires 540.1221).
3.3 tert-Bufyl5-(4-amino-6-(3-nitrophenylamino)- 1, 3, 5-triazin-2-yl)-4-phenylthiazol-2-yl(methyl)carbamate
δH (400 MHz, CDCI3) 8.34 (1H, br. s), 7.79 (1H, d, J 7.5, Ar), 7.73-7.70 (2H, m, Ar), 7.57 (1H, d, J 7.9, Ar), 7.35-7.25 (4H, m, Ar), 5.29 (2H, br. s, NH2), 3.58 (3H, s, NCH3), 1.61 (9H, s, C(CH3)3); δc (100 MHz, CDCI3) 168.5, 166.4, 163.9, 162.2, 153.0, 148.4, 139.6, 135.7, 130.1, 128.4, 127.6, 125.3, 124.3, 117.3, 114.4, 83.7, 60.4, 33.8, 28.2.
3.4 6-(2-(methylamino)-4-phenylthiazol-5-yl)~N2-(3-nitrophenyl)- 1, 3, 5-triazine-2, 4-diamine
To a suspension of above ferf-butyl5-(4-amino-6-(3-nitrophenylamino)-1,3,5-triazin-2-yI)-4-phenylthiazol-2- yl(methyl)carbamate (37 mg, 0.071 mmol) in 1 ml of DCM, TFA (1 ml) was added and the reaction mixture was stirred at room temperature for 24 hours. The solvents were evaporated under reduced pressure, the residue was neutralised with 5 ml of saturated Na2CO3 and extracted with ethyl acetate (3 x 5 ml). Combined organic layers were dried over MgSO4, evaporated under reduced pressure and the residue was filtered- through ^plug of silica to give 30 mg (100%) Of 'a yellow solid; mp 277 0C; HRMS (ESI) 421.1265 (M + H+. C19H17N8O2S requires 421.1195).
Example 1a - Preparation of a compound of formula IV
1.1a N-[4-Methyl-3-(morpholine-4-sulfonyl)-phenyl]-guanidine was prepared as follows: 2-Methyl-5-nitro-benzenesulfonyl chloride (20mmol) dissolved in 20ml THF was treated with morpholine (40mmol) in the presence of triethylamine (25mmol). After stirring for 2 hours at room temperature the reaction mixture was concentrated in vacuo to give 4-(2-methyl-5-nitro-benzenesulfonyl)-morpholine as a brown solid (95% yield), m.p. 114-1150C. MS (ESl+) m/z 287.82 (M+H)+.
To a mixture of the latter compound (7mmol) in EtOH (10ml) AcOH (5ml) was added. The mixture was heated to 65°C and Fe powder (28mmol) was added portion wise. After refluxing for 1.5 hours the reaction mixture was cooled to room temperature, filtered and washed with a minima amount of EtOAc/EtOH. The filtrate was evaporated to dryness. The resulted precipitates were basified with excess aq NaOH. The aqueous phase was then extracted several times with EtOAc. The organic fractions were combined and evaporated to yield 4-methyl-3-(morpho!ine-4-sulfonyl)-phenylamine. Brown solid (-39% yield), MS (ESI+) m/z 257.09 (M+H)+. A mixture of 4-methyl-3-(morpholine-4-sulfonyl)-phenylamine (15mmoi) in EtOH (20ml) was cooled on an ice bath and treated with HCI (1.3ml, 37% solution in H2O) followed by cyanamide (2.2ml, 50% in H2O, 60mmol) was added dropwise, and the mixture heated at 10O0C for 17 hours. Upon completion of the reaction, the mixture was concentrated. The precipitate was washed with petroleum ether/EtOAc (4:1), filtered and dried to give Λ/-[4-methyl-3-(morpholine-4-sulfonyl)-phenyl]-guanidine as a brown solid. 1H NMR (DMSO-Cf6) δ 2.43 (s, 3H, CH3), 3.05 (s, 4H, CH2 χ2), 3.63 (s, 4H, CH2 χ2), 7.45 (m, 4H, NH2, NHχ2), 7.94 (d, 1 H, J = 8.0Hz, Ph-H), 8.17 (s, 1H, Ph-H), 8.28 (d, 1H, J = 8.0Hz, Ph-H). MS (ESI+) m/z 299.11 (M+H)+.
The following compounds were synthesised by an analogous route.
1-(3-Nitrophenyl)guanidine. 1H NMR (DMSO-d6): δ 7.70 (m, 5H, Ph-Hχ2, NH, NH2), 8.09 (m, 1H, Ph-H),
8.19 (m, 1 H, Ph-H). MS (ESI+) m/z 181.07 (M+H)+.
1-(3-Hydroxyphenyl)guanidine. 1H NMR (DMSO-Cf6) δ 6.82 (m, 2H, Ph-Hχ2), 7.02 (m, 2H, Ph-H*2), 7.30
(s, 4H, NH2 & NH*2), 9.75 (s, 1H, OH). MS (ESI+) m/z 152.08 (M+H)+.
1-(4-Hydroxyphenyl)guanidine. 1H NMR (DMSO-Cf6): δ 6.63 (m, 2H, Ph-H*2), 6.70 (m, 1H, Ph-H), 7.20 (t,
1H, J=8.0Hz, Ph-H), 7.44 (s, 4H, NH2, NH*2), 9.80 (s, 1H, OH). MS (ESI+) m/z 152.31 (M+H)+
1-(3-(Morpholine-4-carbonyl)phenyl)guanidine. 1H NMR (DMSO-d6) δ 3.61 (s, 8H, CH2), 4.28 (m, 1H, NH),
7.24 (t, 1H, J = 1.6Hz, Ph-H), 7.31 (t, 1H, J = 2Hz, Ph-H), 7.33 (t, 1H, J = 2.4Hz, Ph-H), 7.49 (t, 1H, J =
8Hz, Ph-H), 7.57 (s, 2H, NH2), 10.00 (s, 1H, NH). MS (ESI+) m/z 249.10 (M+H)+.
4-Guanidinobenzenesulfonamide. 1H NMR (DMSO-Cf6, 400 MHz): δ 6.80 (d, 1 H, J = 8.4 Hz, NH), 7.24 (s,
1H, NH), 7.39 (d, 2H, J = 8.8 Hz, Ph-H), 7.39 (d, 2H1 J = 8.4 Hz, Ph-H), 7.77 (s, 2H, NH2). MS (ESI+) m/z
215.07 (M + H)+
3-Guanidinobenzenesulfonamide. 1H NMR (DMSO-Cf6, 400 MHz): δ 7.46 (m, 3H, NH & NH2), 7.63 (t, 1 H, J
= 8.0 Hz; Ph-H), -7.65 (m, -1H, Ph-H), .7.69 r(m,< 2H, Pt>Hx2), 7.72 (s, 2H, NH2), 10.36 (s, 1H, NH).- MS
(ESI+) m/z 215.06 (M + H)+
BIOLOGICAL ACTIVITY
Kinase assays. The compounds from the examples above were investigated for their ability to inhibit the enzymatic activity of various protein kinases using the method as disclosed in Wang, S. et al. J Med Chem 2004, 47, 1662. 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 MgCI2 + 100 μM ATP with 30-50 kBq per well of [γ-32P]-ATP) and mixtures incubated as required at 30 °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, Pangboume, 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 IC50 values (concentration of test compound which inhibits kinase activity by 50 %).
MTT cytotoxicity assay. The compounds from the examples above were subjected to a standard cellular proliferation assay using the method described previously (Haselsberger, K. et al. Anti Cancer Drugs 1996, 7, (3), 331-8. Loveland, B. E. et al. Biochemistry International 1992, 27, (3), 501-10). Human tumour cell lines were obtained from ECACC (European Collection of Cell Cultures). Standard 72-h MTT (thiazolyl blue; 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyItetrazolium bromide, 2mg/ml in phosphate buffered saline) assays were performed. 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 hr 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 550 nm on an Anthos Labtec Systems plate reader. The data analysis used program Deltasoft 3™ and Microsoft Excel to determine IC50 or Gl50" values (concentration of test compound which inhibits cell growth by 50 %).
CLL apoptosis assay. Compounds were thawed on ice and aliquotted to 0.5ml microcentrifuge tubes and stored at -20'C to avoid multiple freeze-thaw cycles. Compound aliquots were thawed on ice and diluted as required in sterile PBS immediately prior to drugging experiment. Primary CLL cells were isolated from ACD whole blood using standard Ficoll (Ficoll-Paque Plus.GE Healthcare) separation and selected for B cells with RόsetteSep B' cell enrichment cocktail (StemCell tecfh); Cells- were incubated at 1 E6-3E6 cells/ml in RPMI 1640 plus 10% human serum and antibiotics at 37'C in 24 well plates. Inhibitor compounds were added at time 0 and a sample was maintained with media vehicle only. At 24 hours, cells were transferred to a 12x75 tube for Annexin-PI viability assay. Cells were centrifuged at 1500rpm for 5 minutes then incubated at RT in dark for 30 minutes with appropriate reagent plus binding buffer with calcium. After incubation, 80OuI of binding buffer was added for flow cytometry analysis on the EPICS-XL (Beckman-Coulter).
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 claims.
Example A1
Biological activity of the example compounds is summarized in Tables A1 and A2. Table A1
Kinase inhibition Cytotoxicity
IC50 μM Gl50 μM
CDK2-cyclinE CDK7-cycIinH CDK9-cycIinT1 HCT116 MCF7 MRC5
1.1 0.232 >1 0.041 0.06 0.06 3.14
1.2 0.40 0.23 1.05
1.3 0.045 0.019 0.03 0.07 0.54
1.4 0.147 3.741 0.085 0.59 0.82 4.43
1.5 0.052 0.033 5.88 4.96 79.92
1.6 0.55 1.87 >100
1.8 0.34 1.64 25.99
1.7 0.05 0.08 0.26
2.4 0.53 1.58 0.58
2.6 7.09 7.07 37.60
2.8 3.48 6.42 4.24
2.13 3.32 3.77 4.33
Table A2 - anti-proliferative activity of Example 1.1 human cell line 72-h MTT origin designation IC50 (μM)
MGF-7 0.060
MDA-MB-231 0.923 cervix HeLa 0.661
, . v... . ... .. .. „
HCPl 16"" ' * '• • — " ' *" ' 0.060 lung A549 0.534
NCI-H460 0.681 ovarian carcinoma A2780 0.147
PANC Wl 0.366 pancreatic carcinoma
Mia-Paca-2 0.468 mean 0.433

Claims

1. A compound of formula 1 and its pharmaceutically acceptable salts or solvates and physiologically hydrolysable, solubilising or immobilisable derivatives:
Figure imgf000042_0001
Wherein:
Ar is optionally substituted and is a 5-membered heteroaryl ring wherein X1 and X2 are one or two heteroatoms or Ar is a 6-membered aromatic ring, wherein heteroatoms are selected from S, O, N, Se and wherein optional substituents include R1 and R2;
Z is NH, NHCO, NHSO2, N-alkyl, CH2NH, CH2N-alkyl, CH2, CH2CH2, CH=CH, CH2CONH, SO2, or SO;
Y is N or CR3;
R1, R2, R5, R6, R7, R8 and R9 are each independently H, alkyl, or R13 wherein R13 is selected from R10, alkyl-R10, aryl, heteroaryl and combinations of two or more thereof and combinations with one or more alkyl and R11, or R13 is one or more moieties R14 selected from O-, N-, NH-, CO-, COO-, CON-, CONH-, SO2-, SO2N-, SO2NH- linking one or more alkyl, aryl, heteroaryl or R10 or R11 groups or combinations thereof, directly, or via a moiety. selected from alkylene, arylene, heteroarylene or combinations thereof, wherein alkyt, aryl, heferoaryl groups or moieties thereof may be substituted with one or more groups R15 selected from halogeno, NH2, NO2, CN, OH, COOH, CONH2, C(=NH)NH2, SO3H, SO2NH2, SO2CH3, OCH3, CF3 or R13 is selected from a group R15;
or two of R5 to R9 are linked to form a cyclic ether containing one or more oxygen atoms;
R3, when present, is selected from alkyl and R13 as hereinbefore defined, with the proviso that when Y is CR3, Ar is a 5-membered heterocycle comprising one or two N heteroatoms and Z is NH, then R3 is selected from C3+ alkyl and R13 as hereinbefore defined;
R4 is selected from H, alkyl and R13 as hereinbefore defined, with the proviso that when R3 is absent, R4 is selected from alkyl and R13 as hereinbefore defined
wherein at least one of R1, R2, R4 R5, R6, R7, R8 and R9 and R3 or R12 where present, comprise a group R10 or R11 wherein R10 and R11 comprise one or more solubilising moieties chosen from i) neutral hydrophilic groups, ii) ionisable organic acids, iii) ibnisable organic bases and combinations thereof.
2. A compound as claimed in Claim 1 wherein at least one R10 or R11 further comprises an immobilising moiety chosen from iv) chemical functions or moieties providing covalent or non-covalent attachment or binding to a solid phase or an immobile receptor.
3. A compound as claimed in Claim 1 or 2 wherein X1 is S and X2 is N or X2 is S and X1 is N.
4. A compound as claimed in any of Claims 1 to 3 which is of formula V
Figure imgf000043_0001
wherein X1 and X2 are each independently selected from NH or N, O, S, Se, CH and C R15 and at least one of X1 and X2 is selected from NH or N, O1 S and Se, and wherein R15 is as hereinbefore defined for R1, and all other variables are as hereinbefore defined in Claims 1 to 3.
5. A compound as claimed in Claim 4 wherein: one ofX1 and X2 is CH or CR15, and the other of X1 and X2 is S, O, NH, NR15, or Se; or one of X1 and X2 is S, O or Se, and the other of X1 and X2 is N; or one of X1 and X2 is N, and the other of X1 and X2 is NH or NR15; and wherein all other variables are as hereinbefore defined.
6. A compound as claimed in any of Claims 1 to 3 which is of formula I"
Figure imgf000043_0002
wherein all variables are as hereinbefore defined in any of Claims 1 to 3.
7. A compound as claimed in any of Claims 1 to 3 wherein when R10 or R11 comprises a neutral hydrophilic group (i) as hereinbefore defined, this comprises groups containing mono-, di- and polyhydroxylated saturated or unsaturated aliphatic, alicyclic or aromatic systems, carbohydrate derivatives, ethers and polyethers optionally containing one or more hydroxyl groups, O- and/or S- containing heterocyclic systems optionally containing one or more hydroxyl groups, aliphatic or aromatic systems containing a carboxamide, sulfoxide, sulfone, or sulfonamide function, and halogenated alkylcarbonyl groups; or when R10 or R11 comprises an ionisable organic acid (ii) as hereinbefore defined, this preferably includes groups comprising one or more of the functions COOH1 SO3H, OSO3H, PO3H2, and OPO3H2; or
where R10 or R11 comprises an ionisable basic group (iii) as hereinbefore defined, this preferably includes aliphatic, alicyclic, aromatic, or heterocyclic groups comprising one or more of the functions -0-, -NH2, - NH-, =N-, quarternary amine salts, guanidine, and amidine, optionally substituted by one or more substituents selected from halogen, SO2alkyl, alkyl optionally substituted by one or more OH or halogen groups, CHO, COalkyl, aralkyl, COOalkyl and an ether group substituted by one or more OH groups.
8. A compound as claimed in any of Claims 1 to 7 wherein R10 and R11 consist of natural or unnatural amino acid residues and peptides, or their derivatives; or
R10 or R11 is selected from v) OSO3H, PO3H2, OPO3H2; vi) Y' where Y' is selected from aliphatic, alicyclic, aromatic, or heterocyclic groups comprising one or more of the functions -0-, -NH2, -NH-, =N-, amidine, optionally substituted by one or more substituents selected from halogen, S02alkyl, alkyl optionally substituted by one or more OH or halogen groups,
COalkyl, aralkyl, COOalkyl and an ether group substituted by one or more OH groups;
(vii) NHCO(CH2)JNHCO(CH2X1V]P[NHCO(CH2WIqY1 or NHCO(CH2)tNH(CH2)t, Y1 where p and q are each O or 1 , and m, m',m", t and t1 are each independently an integer from 1 to 10; and
(viii) (CH2)nNR19COR17, (CH2)rfN R20SO2R18, or SO2R21, where R17, R18 and R21 are each alkyl groups optionally comprising one or more heteroatoms, and which are optionally substituted by one or more substituents selected from OH, NH2, halogen and NO2, R19 and R2d are each independently H or alkyl, and.. n and n' are each independently 0, 1 , 2, or 3;
(ix) an ether or polyether optionally substituted by one or more hydroxyl groups or one or more Y" groups;
(x) (CH2)rNH2; where r is 0, 1 , 2, or 3;
(xi) (CH2)^OH; where r' is O, 1 , 2, or 3;
(xii) (CH2)n"NR22COR23 where R22 is H or alkyl, n" is O, 1 , 2 or 3 and R23 is an aryl or heteroaryl group, each of which may be optionally substituted by one or more substituents selected from halogeno, NO2,
OH, alkoxy, NH2, COOH, CONH2 and CF3;
(xiii) SO2NR24R25 where R24 and R25 are each independently H, alkyl, aralkyl, CO-alkyl or aryl, with the proviso that at least one of R24 and R25 is other than H, or R24 and R25 are linked to form a cyclic group optionally containing one or more heteroatoms selected from N, O and S, and wherein said alkyl, aryl or cyclic group is optionally substituted by one or more substituents selected from halogeno, NO2, OH, alkoxy, aryl, NH2, COOH, CH2CO2-alkyl, CONH2 and CF3;
(xiv) N-piperidinyl, piperidinyl, N-piperazinyl, N-diazepanyl, N-pyridinyl, N-pyrrolidinyl, N-morpholinyl or
N-thiomorpholinyl, each of which may be optionally substituted by one or more alkyl, alkoxy, aryl, CHO or
CO-alkyl groups.
9. A compound as claimed in any of Claims 1 to 8 wherein each R10 or R 11 is independently selected from a C1-30 hydrocarbyl group, optionally comprising up to twelve heteroatoms selected from N1 S, and O,
15 and optionally bearing up to six substituents each independently selected from a group R as
,15 hereinbefore defined or comprising a moiety R1 as hereinbefore defined, and a group R
10. A compound selected from compounds of formula I':
Figure imgf000045_0001
Figure imgf000045_0002
Figure imgf000046_0002
anc i compounds of formula 1":
Figure imgf000046_0001
as shown in Table 3
Figure imgf000046_0003
and compounds of formula I':
Figure imgf000047_0001
As shown in Table 4 wherein X 1 =-SC , X -N
Figure imgf000047_0004
Wherein
MS = morpholine-4-sulfonyl °' "o
Figure imgf000047_0002
PzC = piperazine-1-carbonyl or piperazin-1-ylmethanone °
AcPzC = 4-Acetylpiperazine-1-carbonyI
MePzC = 4-methylpiperazine-1 -carbonyl
Figure imgf000047_0003
or 4-methyIpiperazin-1 -ylmethanone
Figure imgf000048_0001
M = morpholino ^
MC
Figure imgf000048_0002
PzS = piperazin-1-ylsulfonyl ^p ^NH
Figure imgf000048_0003
MePzS = 4-methylpiperazin-1-ylsulfonyl ^N^
BPzS = 4-benzylpiperaziπ-1-ylsu!fonyl
BPzC - benzylpiperazine-1-carbonyl
BPdC = i-benzylpiperidin-4-carbonyl o
Figure imgf000048_0004
r 1-benzylpiperidin-4~ylmethanone
^NH
^ PdC = piperidine-4-carbonyl or piperidin-4-ylmethanone δ
Figure imgf000048_0005
MePdC = 1-methylpiperidiπ-4-ylmethanone or i-methylpiperidin-4-carbonyl
MePdCB - 4-(1-methylpiperidine-4carbonyl)benzoyl
Figure imgf000048_0006
Figure imgf000048_0007
Pz = piperazin-1-yl
Figure imgf000048_0008
MePz = 4-methylpiperazin-1-yl
Py = pyridine-3-yI
Figure imgf000048_0009
PyEtA = 2-(pyridin-3-yl)ethylamino u^^^
PyMeA = pyridin-3-ylmethylamino
Figure imgf000048_0010
MeDz = 4-methyl-1,4-diazepan-1-yI
11. A process for the preparation of a compound of formula I as claimed in any of Claims 1 to 10 comprising
(1) reacting a compound of formula III
Figure imgf000049_0001
III where Ar is a mono- or di-substituted phenyl, thiazol-4-yl, thiazol-5-yl, imidazol-4-yl, imidazol-5-yl, pyrrol-4- yl or pyrrol-5-yl, preferably Ar is a phenyl, thiazol-4-yl or thiazol-5-yl group, Y is N, or CR3 and L1 is a leaving group, with a compound of formula IV
Figure imgf000049_0002
where Z and R5 to R9 are as hereinbefore defined;
or (2) reacting a compound of formula Vl
Figure imgf000049_0003
where Z and R5 to R9 are: as hereinbefore defined with a compound of formula VII
Figure imgf000049_0004
where Ar is as hereinbefore defined and Y is N;
or (3) reacting a compound of formula Xl
Figure imgf000049_0005
where Y is N or CR3, L3 is any leaving group, preferable halogeno group and Ar and R4 are as hereinbefore defined, with a compound of formula XII
Figure imgf000049_0006
where Z and R5 to R9 are as hereinbefore defined.
12. A process for the preparation of a compound of formula I' as claimed in any of Claims 4 to 10 comprising
(4) condensation reaction between a compound of formula VII'
Figure imgf000050_0001
where Y is N; or CR3 and wherein R1, R2, R4, X1, X2, Y and L2 are as hereinbefore defined with a phenylguanidine of formula VIII'
Figure imgf000050_0002
wherein Z and R5 to R9 are as hereinbefore defined ;
or (5) treatment of amidines Vl'
Figure imgf000050_0003
where Y is N and wherein Z and R4 to R9 are as hereinbefore defined in the presence of POCI3 followed by alkylation reaction with anilines of formula XII
Figure imgf000050_0004
or (6) condensation reaction of a compound of formula XII'
Figure imgf000050_0005
wherein Z and R5 to R9 are as hereinbefore defined with an amidine of formula Xlip
Figure imgf000051_0001
wherein R1, R2, X1 and X2 are as hereinbefore defined, in the presence of base.
13. A novel precursor or intermediate as claimed in any of Claims 11 and 12.
14. The use of a compound of formula I as claimed in any of Claims 1 to 10 or a pharmaceutically acceptable salt, solvate or physiologically hydrolysable, solubilising or immobilising derivative thereof, in the manufacture of a medicament for treating a condition mediated by an enzyme selected from one or more CDK, aurora kinase, GSK, PLK, BCR-ABL, FLT, IKK, JAK, PDGF or VEGF and Src family enzymes, particularly from one or more CDK2, CDK7, CDK8, CDK9, CDK11 , GSK-3, aurora kinase, PLK or at least one tyrosine kinase.
15. A method for treating a condition mediated by one or more enzymes selected from CDK, aurora kinase, GSK, PLK, BCR-ABL, FLT, IKK, JAK, PDGF or VEGF and Src family enzymes, particularly from one or more CDK2, CDK7, CDK8, CDK9, CDK11 , GSK-3, aurora kinase, PLK or tyrosine kinase enzyme, in a human or animal subject, the method comprising administering to a human or animal in need thereof a therapeutically effective amount of a compound of formula I or a pharmaceutically acceptable salt, solvate, or physiologically hydrolysable, solubilising or immobilising derivative thereof as claimed in any of Claims 1 to 10.
16. The use of a compound of formula I "or a pharmaceutically acceptable salt, solvate, or physiologically hydrolysable, solubilising or immobilising derivative thereof as claimed in any of Claims 1 to 10 in a method for treating a condition mediated by an enzyme selected from one or more CDK, aurora kinase, GSK, PLK, BCR-ABL, FLT, IKK, JAK, PDGF or VEGF and Src family enzymes, particularly from one or more CDK2, CDK7, CDK8, CDK9, CDK11 , GSK-3, aurora kinase, PLK or tyrosine kinase.
17. The use of a compound of formula I or a pharmaceutically acceptable salt or solvate or physiologically hydrolysable, solubilising or immobilising derivative thereof as claimed in any of Claims 1 to 10, in an assay for identifying candidate compounds capable of treating a condition mediated by an enzyme selected from one or more CDK, aurora kinase, GSK, PLK, BCR-ABL, FLT, IKK, JAK, PDGF or VEGF and Src family enzymes, particularly from one or more CDK2, CDK7, CDK8, CDK9, CDK11, GSK- 3, aurora kinase, PLK or tyrosine kinase.
18. A pharmaceutical composition comprising a compound of formula I or a pharmaceutically acceptable salt or solvate, or physiologically hydrolysable, solubilising or immobilising derivative thereof as claimed in any of Claims 1 to 10, in association with one or more diluents, carriers or excipients.
PCT/GB2009/050298 2008-03-26 2009-03-26 Pyrimidines, triazines and their use as pharmaceutical agents WO2009118567A2 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US12/934,798 US20110092490A1 (en) 2008-03-26 2009-03-26 Pyrimidines, triazines and their use as pharmaceutical agents
CN2009801123665A CN102143953B (en) 2008-03-26 2009-03-26 Pyrimidines, triazines and their use as pharmaceutical agents

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
GB0805477.7 2008-03-26
GBGB0805477.7A GB0805477D0 (en) 2008-03-26 2008-03-26 Pyrimidines triazines and their use as pharmaceutical agents

Publications (2)

Publication Number Publication Date
WO2009118567A2 true WO2009118567A2 (en) 2009-10-01
WO2009118567A3 WO2009118567A3 (en) 2010-03-11

Family

ID=39386767

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/GB2009/050298 WO2009118567A2 (en) 2008-03-26 2009-03-26 Pyrimidines, triazines and their use as pharmaceutical agents

Country Status (4)

Country Link
US (1) US20110092490A1 (en)
CN (1) CN102143953B (en)
GB (1) GB0805477D0 (en)
WO (1) WO2009118567A2 (en)

Cited By (51)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2527332A1 (en) * 2011-05-24 2012-11-28 Bayer Intellectual Property GmbH 4-Aryl-N-phenyl-1,3,5-triazin-2-amines containing a sulfoximine group as CDK9 inhibitors
WO2012160034A1 (en) * 2011-05-24 2012-11-29 Bayer Intellectual Property Gmbh 4-aryl-n-phenyl-1,3,5-triazin-2-amines containing a sulfoximine group
WO2013156780A1 (en) * 2012-04-19 2013-10-24 Cancer Research Technology Limited Therapeutic compounds
CN103476759A (en) * 2011-04-19 2013-12-25 拜耳知识产权有限责任公司 Substituted 4-aryl-n-phenyl-1,3,5-triazin-2-amines
US8895733B2 (en) 2010-09-13 2014-11-25 Novartis Ag Triazine-oxadiazoles
WO2014199164A1 (en) * 2013-06-12 2014-12-18 Ampla Pharmaceuticals, Inc. Diaryl substituted heteroaromatic compounds
WO2015006592A1 (en) * 2013-07-11 2015-01-15 Agios Pharmaceuticals, Inc. N,6-bis(aryl or heteroaryl)-1,3,5-triazine-2,4-diamine compounds as idh2 mutants inhibitors for the treatment of cancer
US9227956B2 (en) 2013-04-17 2016-01-05 Pfizer Inc. Substituted amide compounds
WO2016049211A1 (en) * 2014-09-26 2016-03-31 Gilead Sciences, Inc. Aminotriazine derivatives useful as tank-binding kinase inhibitor compounds
WO2016059011A1 (en) 2014-10-16 2016-04-21 Bayer Pharma Aktiengesellschaft Fluorinated benzofuranyl-pyrimidine derivatives containing a sulfone group
US9434979B2 (en) 2009-10-21 2016-09-06 Shin-San Michael Su Methods and compositions for cell-proliferation-related disorders
US9474779B2 (en) 2012-01-19 2016-10-25 Agios Pharmaceuticals, Inc. Therapeutically active compositions and their methods of use
US9512107B2 (en) 2012-01-06 2016-12-06 Agios Pharmaceuticals, Inc. Therapeutically active compositions and their methods of use
US9579324B2 (en) 2013-07-11 2017-02-28 Agios Pharmaceuticals, Inc Therapeutically active compounds and their methods of use
WO2017055196A1 (en) 2015-09-29 2017-04-06 Bayer Pharma Aktiengesellschaft Novel macrocyclic sulfondiimine compounds
WO2017060322A2 (en) 2015-10-10 2017-04-13 Bayer Pharma Aktiengesellschaft Ptefb-inhibitor-adc
WO2017060167A1 (en) 2015-10-08 2017-04-13 Bayer Pharma Aktiengesellschaft Novel modified macrocyclic compounds
US9650340B2 (en) 2012-11-15 2017-05-16 Bayer Pharma Aktiengesellschaft 5-fluoro-N-(pyridin-2-yl)pyridin-2-amine derivatives containing a sulfoximine group
US9650361B2 (en) 2012-11-15 2017-05-16 Bayer Pharam Aktiengesellschaft N-(pyridin-2-yl)pyrimidin-4-amine derivatives containing a sulfoximine group
US9662327B2 (en) 2011-06-17 2017-05-30 Agios Pharmaceuticals, Inc Phenyl and pyridinyl substituted piperidines and piperazines as inhibitors of IDH1 mutants and their use in treating cancer
US9670161B2 (en) 2012-10-18 2017-06-06 Bayer Pharma Aktiengesellschaft 5-fluoro-N-(pyridin-2-yl)pyridin-2-amine derivatives containing a sulfone group
US9708293B2 (en) 2012-10-18 2017-07-18 Bayer Pharma Aktiengesellschaft N-(pyridin-2-yl)pyrimidin-4-amine derivatives containing a sulfone group
US9738625B2 (en) 2013-08-02 2017-08-22 Agios Pharmaceuticals, Inc. Therapeutically active compounds and their methods of use
US9751863B2 (en) 2015-08-05 2017-09-05 Agios Pharmaceuticals, Inc. Therapeutically active compounds and their methods of use
US9770445B2 (en) 2013-07-04 2017-09-26 Bayer Pharma Aktiengesellschaft Sulfoximine substituted 5-fluoro-N-(pyridin-2-yl)pyridin-2-amine derivatives and their use as CDK9 kinase inhibitors
US9790189B2 (en) 2014-04-01 2017-10-17 Bayer Pharma Aktiengesellschaft Disubstituted 5-fluoro pyrimidine derivatives containing a sulfondiimine group
US9856242B2 (en) 2014-03-13 2018-01-02 Bayer Pharma Aktiengesellscaft 5-fluoro-N-(pyridin-2-yl)pyridin-2-amine derivatives containing a sulfone group
US9856279B2 (en) 2011-06-17 2018-01-02 Agios Pharmaceuticals, Inc. Therapeutically active compositions and their methods of use
US9884849B2 (en) 2014-10-16 2018-02-06 Bayer Pharma Aktiengesellschaft Fluorinated benzofuranyl-pyrimidine derivatives containing a sulfoximine group
US9963464B2 (en) 2014-04-11 2018-05-08 Bayer Pharma Aktiengesellschaft Macrocyclic compounds
US9968595B2 (en) 2014-03-14 2018-05-15 Agios Pharmaceuticals, Inc. Pharmaceutical compositions of therapeutically active compounds
US9980961B2 (en) 2011-05-03 2018-05-29 Agios Pharmaceuticals, Inc. Pyruvate kinase activators for use in therapy
US10017495B2 (en) 2013-07-11 2018-07-10 Agios Pharmaceuticals, Inc. Therapeutically active compounds and their methods of use
US10029987B2 (en) 2009-06-29 2018-07-24 Agios Pharmaceuticals, Inc. Therapeutic compounds and compositions
WO2018134213A1 (en) * 2017-01-17 2018-07-26 Astrazeneca Ab Jak1 selective inhibitors
US10072001B2 (en) 2014-06-03 2018-09-11 Gilead Sciences, Inc. Tank-binding kinase inhibitor compounds
WO2018177889A1 (en) 2017-03-28 2018-10-04 Bayer Aktiengesellschaft Novel ptefb inhibiting macrocyclic compounds
WO2018177899A1 (en) 2017-03-28 2018-10-04 Bayer Aktiengesellschaft Novel ptefb inhibiting macrocyclic compounds
US10202339B2 (en) 2012-10-15 2019-02-12 Agios Pharmaceuticals, Inc. Therapeutic compounds and compositions
US10316049B2 (en) 2015-12-17 2019-06-11 Gilead Sciences, Inc. Tank-binding kinase inhibitor compounds
US10376510B2 (en) 2013-07-11 2019-08-13 Agios Pharmaceuticals, Inc. 2,4- or 4,6-diaminopyrimidine compounds as IDH2 mutants inhibitors for the treatment of cancer
US10610125B2 (en) 2009-03-13 2020-04-07 Agios Pharmaceuticals, Inc. Methods and compositions for cell-proliferation-related disorders
US10653710B2 (en) 2015-10-15 2020-05-19 Agios Pharmaceuticals, Inc. Combination therapy for treating malignancies
JP2020517595A (en) * 2017-04-19 2020-06-18 ジェンフリート セラピューティクス(シャンハイ)インコーポレイテッド Novel inhibitor of cyclin-dependent kinase CDK9
US10689414B2 (en) 2013-07-25 2020-06-23 Agios Pharmaceuticals, Inc. Therapeutically active compounds and their methods of use
US10980788B2 (en) 2018-06-08 2021-04-20 Agios Pharmaceuticals, Inc. Therapy for treating malignancies
US11234976B2 (en) 2015-06-11 2022-02-01 Agios Pharmaceuticals, Inc. Methods of using pyruvate kinase activators
US11419859B2 (en) 2015-10-15 2022-08-23 Servier Pharmaceuticals Llc Combination therapy for treating malignancies
US11666888B2 (en) 2018-02-05 2023-06-06 Bio-Rad Laboratories, Inc. Chromatography resin having an anionic exchange-hydrophobic mixed mode ligand
US11701347B2 (en) 2018-02-13 2023-07-18 Bayer Aktiengesellschaft Use of 5-fluoro-4-(4-fluoro-2-methoxyphenyl)-N-{4-[(S-methylsulfonimidoyl)methyl]pyridin-2-yl}pyridin-2-amine for treating diffuse large B-cell lymphoma
US11844758B2 (en) 2013-07-11 2023-12-19 Servier Pharmaceuticals Llc Therapeutically active compounds and their methods of use

Families Citing this family (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9198911B2 (en) 2010-11-02 2015-12-01 The Trustees Of Columbia University In The City Of New York Methods for treating hair loss disorders
WO2013037894A1 (en) * 2011-09-16 2013-03-21 Bayer Intellectual Property Gmbh Disubstituted 5-fluoro pyrimidine derivatives containing a sulfoximine group
JP6769963B2 (en) 2014-08-29 2020-10-14 ティエエッセ ファルマ ソチエタ レスポンサビリタ リミタータ Inhibitor of α-amino-β-carboxymuconic acid semialdehyde decarboxylase
FR3025199B1 (en) * 2014-09-02 2016-09-23 Pf Medicament MACROCYCLIC POLYETHYL N-ARYL-2-AMINO-4-ARYL-PYRIMIDINE DERIVATIVES
CN105237523B (en) * 2015-10-08 2018-06-01 深圳市博圣康生物科技有限公司 Pyrimidine derivatives and preparation method thereof, purposes
CA3172750A1 (en) * 2020-03-23 2021-09-30 Ge Hyeong Lee Novel pyrimidine derivative, and composition for preventing or treating neurodegenerative diseases and cancer, comprising same
WO2022028556A1 (en) * 2020-08-07 2022-02-10 南京药石科技股份有限公司 Cdk9 inhibitor and use thereof
CN112239466B (en) * 2020-11-10 2021-06-08 常州千红生化制药股份有限公司 Succinate salt of a selective CDK4/6 inhibitor and crystalline forms thereof
CN113563275B (en) * 2021-07-27 2024-05-03 中国药科大学 Preparation and application of aminopyrimidine derivative selectively targeting CDK9

Citations (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000025780A1 (en) * 1998-10-29 2000-05-11 Bristol-Myers Squibb Company Compounds derived from an amine nucleus that are inhibitors of impdh enzyme
WO2001072745A1 (en) * 2000-03-29 2001-10-04 Cyclacel Limited 2-substituted 4-heteroaryl-pyrimidines and their use in the treatmetn of proliferative disorders
WO2002079193A1 (en) * 2001-03-29 2002-10-10 Cyclacel Limited Inhibitors of cyclin dependent kinases as anti-cancer agent
WO2003029248A1 (en) * 2001-09-28 2003-04-10 Cyclacel Limited N-(4-(4-methylthiazol-5-yl) pyrimidin-2-yl) -n-phenylamines as antiproliferative compounds
WO2003037346A1 (en) * 2001-10-31 2003-05-08 Cell Therapeutics, Inc. 6-phenyl-n-phenyl-(1,3,5) -triazine-2,4-diamine derivatives and related compounds with lysophphosphatidic acid acyltransferase beta (lpaat-beta) inhibitory activity for use in the treatment of cancer
WO2004041810A1 (en) * 2002-11-05 2004-05-21 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of jak and other protein kinases
WO2004041789A1 (en) * 2002-11-01 2004-05-21 Vertex Pharmaceuticals Incorporated Compositions useful as inhibitors of jak and other protein kinases
WO2004043467A1 (en) * 2002-11-14 2004-05-27 Cyclacel Limited Anti-viral compounds
WO2004043953A1 (en) * 2002-11-14 2004-05-27 Cyclacel Limited Pyrimidine compounds
WO2004056368A1 (en) * 2002-12-19 2004-07-08 Cyclacel Limited Therapeutic applications of 2-substituted 4-heteroarylryrimidines
WO2004089286A2 (en) * 2003-04-04 2004-10-21 Irm Llc Novel compounds and compositions as protein kinase inhibitors
WO2005012262A1 (en) * 2003-07-30 2005-02-10 Cyclacel Limited 2-aminophenyl-4-phenylpyrimidines as kinase inhibitors
WO2005012298A1 (en) * 2003-07-30 2005-02-10 Cyclacel Limited Pyridinylamino-pyrimidine derivatives as protein kinase inhibitors
FR2867188A1 (en) * 2004-03-03 2005-09-09 Oreal New s-triazine derivatives containing p-oxyphenyl and aminobenzalmalonic acids, for use as broad-spectrum ultraviolet (UV) filters, especially in cosmetic or dermatological sunscreen compositions
WO2005116025A2 (en) * 2004-05-26 2005-12-08 Cyclacel Limited 2-substituted-4-heteroaryl-pyrimidines useful for the treatment of proliferative disorders
WO2006021458A2 (en) * 2004-08-27 2006-03-02 Gpc Biotech Ag Pyrimidine derivatives
US20070244140A1 (en) * 2006-04-12 2007-10-18 Wyeth Anilino-pyrimidine phenyl and benzothiophene analogs

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2542880A1 (en) * 2003-10-21 2005-05-12 Cyclacel Limited Pyrimidin-4-yl-3, 4-thione compounds and their use in therapy
US7335770B2 (en) * 2004-03-24 2008-02-26 Reddy U5 Therapeutics, Inc. Triazine compounds and their analogs, compositions, and methods
WO2005111017A1 (en) * 2004-05-07 2005-11-24 Janssen Pharmaceutica, N.V. Triazine compounds as inhibitors of bacterial type iii protein secretion systems
US7709475B2 (en) * 2004-05-21 2010-05-04 Banyu Pharmaceutical Co., Ltd. Selective inhibitors against Cdk4 and Cdk6 having aminothiazole skeleton

Patent Citations (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000025780A1 (en) * 1998-10-29 2000-05-11 Bristol-Myers Squibb Company Compounds derived from an amine nucleus that are inhibitors of impdh enzyme
WO2001072745A1 (en) * 2000-03-29 2001-10-04 Cyclacel Limited 2-substituted 4-heteroaryl-pyrimidines and their use in the treatmetn of proliferative disorders
WO2002079193A1 (en) * 2001-03-29 2002-10-10 Cyclacel Limited Inhibitors of cyclin dependent kinases as anti-cancer agent
WO2003029248A1 (en) * 2001-09-28 2003-04-10 Cyclacel Limited N-(4-(4-methylthiazol-5-yl) pyrimidin-2-yl) -n-phenylamines as antiproliferative compounds
WO2003037346A1 (en) * 2001-10-31 2003-05-08 Cell Therapeutics, Inc. 6-phenyl-n-phenyl-(1,3,5) -triazine-2,4-diamine derivatives and related compounds with lysophphosphatidic acid acyltransferase beta (lpaat-beta) inhibitory activity for use in the treatment of cancer
WO2004041789A1 (en) * 2002-11-01 2004-05-21 Vertex Pharmaceuticals Incorporated Compositions useful as inhibitors of jak and other protein kinases
WO2004041810A1 (en) * 2002-11-05 2004-05-21 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of jak and other protein kinases
WO2004043953A1 (en) * 2002-11-14 2004-05-27 Cyclacel Limited Pyrimidine compounds
WO2004043467A1 (en) * 2002-11-14 2004-05-27 Cyclacel Limited Anti-viral compounds
WO2004056368A1 (en) * 2002-12-19 2004-07-08 Cyclacel Limited Therapeutic applications of 2-substituted 4-heteroarylryrimidines
WO2004089286A2 (en) * 2003-04-04 2004-10-21 Irm Llc Novel compounds and compositions as protein kinase inhibitors
WO2005012262A1 (en) * 2003-07-30 2005-02-10 Cyclacel Limited 2-aminophenyl-4-phenylpyrimidines as kinase inhibitors
WO2005012298A1 (en) * 2003-07-30 2005-02-10 Cyclacel Limited Pyridinylamino-pyrimidine derivatives as protein kinase inhibitors
FR2867188A1 (en) * 2004-03-03 2005-09-09 Oreal New s-triazine derivatives containing p-oxyphenyl and aminobenzalmalonic acids, for use as broad-spectrum ultraviolet (UV) filters, especially in cosmetic or dermatological sunscreen compositions
WO2005116025A2 (en) * 2004-05-26 2005-12-08 Cyclacel Limited 2-substituted-4-heteroaryl-pyrimidines useful for the treatment of proliferative disorders
WO2006021458A2 (en) * 2004-08-27 2006-03-02 Gpc Biotech Ag Pyrimidine derivatives
US20070244140A1 (en) * 2006-04-12 2007-10-18 Wyeth Anilino-pyrimidine phenyl and benzothiophene analogs

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
DESAI VIKAS A ET AL: "2-(2-Amino-4-phenylthiazolyl)-4-(4-methox yanilino)-6-(arylthioureido) s-triazine" JOURNAL OF THE INSTITUTION OF CHEMISTS (INDIA), CALCUTTA, IN, vol. 76, no. 3, 1 January 2004 (2004-01-01), pages 100-102, XP008107533 ISSN: 0020-3254 *

Cited By (118)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10610125B2 (en) 2009-03-13 2020-04-07 Agios Pharmaceuticals, Inc. Methods and compositions for cell-proliferation-related disorders
US10029987B2 (en) 2009-06-29 2018-07-24 Agios Pharmaceuticals, Inc. Therapeutic compounds and compositions
USRE49582E1 (en) 2009-06-29 2023-07-18 Agios Pharmaceuticals, Inc. Therapeutic compounds and compositions
US10988448B2 (en) 2009-06-29 2021-04-27 Agios Pharmaceuticals, Inc. Therapeutic compounds and compositions
US11866411B2 (en) 2009-06-29 2024-01-09 Agios Pharmaceutical, Inc. Therapeutic compounds and compositions
US9434979B2 (en) 2009-10-21 2016-09-06 Shin-San Michael Su Methods and compositions for cell-proliferation-related disorders
US10711314B2 (en) 2009-10-21 2020-07-14 Agios Pharmaceuticals, Inc. Methods for diagnosing IDH-mutant cell proliferation disorders
US9982309B2 (en) 2009-10-21 2018-05-29 Agios Pharmaceuticals, Inc. Method for treating cell proliferation related disorders
US8895733B2 (en) 2010-09-13 2014-11-25 Novartis Ag Triazine-oxadiazoles
CN103476759A (en) * 2011-04-19 2013-12-25 拜耳知识产权有限责任公司 Substituted 4-aryl-n-phenyl-1,3,5-triazin-2-amines
CN103476759B (en) * 2011-04-19 2016-03-16 拜耳知识产权有限责任公司 4-aryl-N-phenyl-1,3,5-triazines-2-the amine replaced
JP2014511889A (en) * 2011-04-19 2014-05-19 バイエル・インテレクチュアル・プロパティ・ゲゼルシャフト・ミット・ベシュレンクテル・ハフツング Substituted 4-aryl-N-phenyl-1,3,5-triazin-2-amine
US10632114B2 (en) 2011-05-03 2020-04-28 Agios Pharmaceuticals, Inc. Pyruvate kinase activators for use in therapy
US9980961B2 (en) 2011-05-03 2018-05-29 Agios Pharmaceuticals, Inc. Pyruvate kinase activators for use in therapy
US11793806B2 (en) 2011-05-03 2023-10-24 Agios Pharmaceuticals, Inc. Pyruvate kinase activators for use in therapy
CN103702979B (en) * 2011-05-24 2016-09-28 拜耳知识产权有限责任公司 4-aryl-N-phenyl-1,3,5-triazine-2-amine containing sulfenimide group
JP2014515363A (en) * 2011-05-24 2014-06-30 バイエル・インテレクチュアル・プロパティ・ゲゼルシャフト・ミット・ベシュレンクテル・ハフツング 4-Aryl-N-phenyl-1,3,5-triazin-2-amine containing a sulfoximine group
EA023418B1 (en) * 2011-05-24 2016-06-30 Байер Интеллектчуал Проперти Гмбх 4-aryl-n-phenyl-1,3,5-triazin-2-amines containing a sulfoximine group
WO2012160034A1 (en) * 2011-05-24 2012-11-29 Bayer Intellectual Property Gmbh 4-aryl-n-phenyl-1,3,5-triazin-2-amines containing a sulfoximine group
US9962389B2 (en) 2011-05-24 2018-05-08 Bayer Intellectual Property Gmbh 4-aryl-N-phenyl-1,3,5-triazin-2-amines containing a sulfoximine group
US9669034B2 (en) 2011-05-24 2017-06-06 Bayer Intellectual Property Gmbh 4-aryl-N-phenyl-1,3,5-triazin-2-amines containing a sulfoximine group
CN103702979A (en) * 2011-05-24 2014-04-02 拜耳知识产权有限责任公司 4-aryl-n-phenyl-1,3,5-triazin-2-amines containing a sulfoximine group
EP2527332A1 (en) * 2011-05-24 2012-11-28 Bayer Intellectual Property GmbH 4-Aryl-N-phenyl-1,3,5-triazin-2-amines containing a sulfoximine group as CDK9 inhibitors
US9662327B2 (en) 2011-06-17 2017-05-30 Agios Pharmaceuticals, Inc Phenyl and pyridinyl substituted piperidines and piperazines as inhibitors of IDH1 mutants and their use in treating cancer
US9856279B2 (en) 2011-06-17 2018-01-02 Agios Pharmaceuticals, Inc. Therapeutically active compositions and their methods of use
US9512107B2 (en) 2012-01-06 2016-12-06 Agios Pharmaceuticals, Inc. Therapeutically active compositions and their methods of use
US9732062B2 (en) 2012-01-06 2017-08-15 Agios Pharmaceuticals, Inc. Therapeutically active compounds and their methods of use
US9656999B2 (en) 2012-01-06 2017-05-23 Agios Pharmaceuticals, Inc. Therapeutically active compounds and their methods of use
US10294215B2 (en) 2012-01-06 2019-05-21 Agios Pharmaceuticals, Inc. Therapeutically active compounds and their methods of use
US11505538B1 (en) 2012-01-06 2022-11-22 Servier Pharmaceuticals Llc Therapeutically active compounds and their methods of use
US10717764B2 (en) 2012-01-19 2020-07-21 Agios Pharmaceuticals, Inc. Therapeutically active compounds and their methods of use
US9850277B2 (en) 2012-01-19 2017-12-26 Agios Pharmaceuticals, Inc. Therapeutically active compositions and their methods of use
US11667673B2 (en) 2012-01-19 2023-06-06 Servier Pharmaceuticals Llc Therapeutically active compounds and their methods of use
US9474779B2 (en) 2012-01-19 2016-10-25 Agios Pharmaceuticals, Inc. Therapeutically active compositions and their methods of use
US10640534B2 (en) 2012-01-19 2020-05-05 Agios Pharmaceuticals, Inc. Therapeutically active compositions and their methods of use
AU2013250912B2 (en) * 2012-04-19 2017-05-25 Changzhou Le Sun Pharmaceuticals Limited Therapeutic compounds
US9062039B2 (en) 2012-04-19 2015-06-23 Changzhou Le Sun Pharmaceuticals Limited Therapeutic compounds
WO2013156780A1 (en) * 2012-04-19 2013-10-24 Cancer Research Technology Limited Therapeutic compounds
US10202339B2 (en) 2012-10-15 2019-02-12 Agios Pharmaceuticals, Inc. Therapeutic compounds and compositions
US9708293B2 (en) 2012-10-18 2017-07-18 Bayer Pharma Aktiengesellschaft N-(pyridin-2-yl)pyrimidin-4-amine derivatives containing a sulfone group
US9670161B2 (en) 2012-10-18 2017-06-06 Bayer Pharma Aktiengesellschaft 5-fluoro-N-(pyridin-2-yl)pyridin-2-amine derivatives containing a sulfone group
US9877954B2 (en) 2012-11-15 2018-01-30 Bayer Pharma Aktiengesellschaft 5-fluoro-N-(pyridin-2-yl)pyridin-2-amine derivatives containing a sulfoximine group
US9650340B2 (en) 2012-11-15 2017-05-16 Bayer Pharma Aktiengesellschaft 5-fluoro-N-(pyridin-2-yl)pyridin-2-amine derivatives containing a sulfoximine group
US9650361B2 (en) 2012-11-15 2017-05-16 Bayer Pharam Aktiengesellschaft N-(pyridin-2-yl)pyrimidin-4-amine derivatives containing a sulfoximine group
US9227956B2 (en) 2013-04-17 2016-01-05 Pfizer Inc. Substituted amide compounds
WO2014199164A1 (en) * 2013-06-12 2014-12-18 Ampla Pharmaceuticals, Inc. Diaryl substituted heteroaromatic compounds
US9770445B2 (en) 2013-07-04 2017-09-26 Bayer Pharma Aktiengesellschaft Sulfoximine substituted 5-fluoro-N-(pyridin-2-yl)pyridin-2-amine derivatives and their use as CDK9 kinase inhibitors
US9724350B2 (en) 2013-07-11 2017-08-08 Agios Pharmaceuticals, Inc. N,6-bis(aryl or heteroaryl)-1,3,5-triazine-2,4-diamine compounds as IDH2 mutants inhibitors for the treatment of cancer
JP2016523977A (en) * 2013-07-11 2016-08-12 アギオス ファーマシューティカルス,インコーポレーテッド N, 6-bis (aryl or heteroaryl) -1,3,5-triazine-2,4-diamine compounds as IDH2 variant inhibitors for the treatment of cancer
US9579324B2 (en) 2013-07-11 2017-02-28 Agios Pharmaceuticals, Inc Therapeutically active compounds and their methods of use
US10946023B2 (en) 2013-07-11 2021-03-16 Agios Pharmaceuticals, Inc. Therapeutically active compounds and their methods of use
US10017495B2 (en) 2013-07-11 2018-07-10 Agios Pharmaceuticals, Inc. Therapeutically active compounds and their methods of use
US10028961B2 (en) 2013-07-11 2018-07-24 Agios Pharmaceuticals, Inc. Therapeutically active compounds and their methods of use
WO2015006592A1 (en) * 2013-07-11 2015-01-15 Agios Pharmaceuticals, Inc. N,6-bis(aryl or heteroaryl)-1,3,5-triazine-2,4-diamine compounds as idh2 mutants inhibitors for the treatment of cancer
US10376510B2 (en) 2013-07-11 2019-08-13 Agios Pharmaceuticals, Inc. 2,4- or 4,6-diaminopyrimidine compounds as IDH2 mutants inhibitors for the treatment of cancer
US10172864B2 (en) 2013-07-11 2019-01-08 Agios Pharmaceuticals, Inc. Therapeutically active compounds and their methods of use
AU2014287122B9 (en) * 2013-07-11 2018-11-01 Les Laboratoires Servier N,6-bis(aryl or heteroaryl)-1,3,5-triazine-2,4-diamine compounds as IDH2 mutants inhibitors for the treatment of cancer
US11844758B2 (en) 2013-07-11 2023-12-19 Servier Pharmaceuticals Llc Therapeutically active compounds and their methods of use
US10111878B2 (en) 2013-07-11 2018-10-30 Agios Pharmaceuticals, Inc. N,6-bis(aryl or heteroaryl)-1,3,5-triazine-2,4-diamine compounds as IDH2 mutants inhibitors for the treatment of cancer
AU2014287122B2 (en) * 2013-07-11 2018-10-25 Les Laboratoires Servier N,6-bis(aryl or heteroaryl)-1,3,5-triazine-2,4-diamine compounds as IDH2 mutants inhibitors for the treatment of cancer
EA032070B1 (en) * 2013-07-11 2019-04-30 Аджиос Фармасьютикалз, Инк. N,6-bis(aryl or heteroaryl)-1,3,5-triazine-2,4-diamine compounds as idh2 mutants inhibitors for the treatment of cancer
US10689414B2 (en) 2013-07-25 2020-06-23 Agios Pharmaceuticals, Inc. Therapeutically active compounds and their methods of use
US11021515B2 (en) 2013-07-25 2021-06-01 Agios Pharmaceuticals, Inc. Therapeutically active compounds and their methods of use
US10093654B2 (en) 2013-08-02 2018-10-09 Agios Pharmaceuticals, Inc. Therapeutically active compounds and their methods of use
US9738625B2 (en) 2013-08-02 2017-08-22 Agios Pharmaceuticals, Inc. Therapeutically active compounds and their methods of use
US10730854B2 (en) 2013-08-02 2020-08-04 Agios Pharmaceuticals, Inc. Therapeutically active compounds and their methods of use
US9856242B2 (en) 2014-03-13 2018-01-02 Bayer Pharma Aktiengesellscaft 5-fluoro-N-(pyridin-2-yl)pyridin-2-amine derivatives containing a sulfone group
US11504361B2 (en) 2014-03-14 2022-11-22 Servier Pharmaceuticals Llc Pharmaceutical compositions of therapeutically active compounds
US10799490B2 (en) 2014-03-14 2020-10-13 Agios Pharmaceuticals, Inc. Pharmaceutical compositions of therapeutically active compounds
US9968595B2 (en) 2014-03-14 2018-05-15 Agios Pharmaceuticals, Inc. Pharmaceutical compositions of therapeutically active compounds
US10449184B2 (en) 2014-03-14 2019-10-22 Agios Pharmaceuticals, Inc. Pharmaceutical compositions of therapeutically active compounds
US9790189B2 (en) 2014-04-01 2017-10-17 Bayer Pharma Aktiengesellschaft Disubstituted 5-fluoro pyrimidine derivatives containing a sulfondiimine group
US9963464B2 (en) 2014-04-11 2018-05-08 Bayer Pharma Aktiengesellschaft Macrocyclic compounds
US10259811B2 (en) 2014-06-03 2019-04-16 Gilead Sciences, Inc. Tank-binding kinase inhibitor compounds
US10072001B2 (en) 2014-06-03 2018-09-11 Gilead Sciences, Inc. Tank-binding kinase inhibitor compounds
EP3517536A1 (en) * 2014-09-26 2019-07-31 Gilead Sciences, Inc. Aminotriazine derivatives useful as tank-binding kinase inhibitor compounds
WO2016049211A1 (en) * 2014-09-26 2016-03-31 Gilead Sciences, Inc. Aminotriazine derivatives useful as tank-binding kinase inhibitor compounds
US10253019B2 (en) 2014-09-26 2019-04-09 Gilead Sciences, Inc. Tank-binding kinase inhibitor compounds
AU2015320675B2 (en) * 2014-09-26 2018-10-18 Gilead Sciences, Inc. Aminotriazine derivatives useful as tank-binding kinase inhibitor compounds
US10040781B2 (en) 2014-09-26 2018-08-07 Gilead Sciences, Inc. Tank-binding kinase inhibitor compounds
CN106715419A (en) * 2014-09-26 2017-05-24 吉利德科学公司 Aminotriazine derivatives useful as TANK-binding kinase inhibitor compounds
US9902716B2 (en) 2014-10-16 2018-02-27 Bayer Pharma Aktiengesellschaft Fluorinated benzofuranyl-pyrimidine derivatives containing a sulfone group
US9884849B2 (en) 2014-10-16 2018-02-06 Bayer Pharma Aktiengesellschaft Fluorinated benzofuranyl-pyrimidine derivatives containing a sulfoximine group
WO2016059011A1 (en) 2014-10-16 2016-04-21 Bayer Pharma Aktiengesellschaft Fluorinated benzofuranyl-pyrimidine derivatives containing a sulfone group
US11234976B2 (en) 2015-06-11 2022-02-01 Agios Pharmaceuticals, Inc. Methods of using pyruvate kinase activators
US9751863B2 (en) 2015-08-05 2017-09-05 Agios Pharmaceuticals, Inc. Therapeutically active compounds and their methods of use
WO2017055196A1 (en) 2015-09-29 2017-04-06 Bayer Pharma Aktiengesellschaft Novel macrocyclic sulfondiimine compounds
WO2017060167A1 (en) 2015-10-08 2017-04-13 Bayer Pharma Aktiengesellschaft Novel modified macrocyclic compounds
WO2017060322A2 (en) 2015-10-10 2017-04-13 Bayer Pharma Aktiengesellschaft Ptefb-inhibitor-adc
US10653710B2 (en) 2015-10-15 2020-05-19 Agios Pharmaceuticals, Inc. Combination therapy for treating malignancies
US11419859B2 (en) 2015-10-15 2022-08-23 Servier Pharmaceuticals Llc Combination therapy for treating malignancies
US10316049B2 (en) 2015-12-17 2019-06-11 Gilead Sciences, Inc. Tank-binding kinase inhibitor compounds
EA037067B1 (en) * 2017-01-17 2021-02-02 Астразенека Аб Jak1 selective inhibitors
KR20230141938A (en) * 2017-01-17 2023-10-10 아스트라제네카 아베 Jak1 selective inhibitors
US10961228B2 (en) 2017-01-17 2021-03-30 Astrazeneca Ab JAK1 selective inhibitors
TWI753089B (en) * 2017-01-17 2022-01-21 瑞典商阿斯特捷利康公司 Jak1 selective inhibitors
KR102659213B1 (en) 2017-01-17 2024-04-18 아스트라제네카 아베 Jak1 selective inhibitors
US11897869B2 (en) 2017-01-17 2024-02-13 Astazeneca Ab JAK1 selective inhibitors
WO2018134213A1 (en) * 2017-01-17 2018-07-26 Astrazeneca Ab Jak1 selective inhibitors
JP2022043059A (en) * 2017-01-17 2022-03-15 アストラゼネカ・アクチエボラーグ Jak1 selective inhibitors
JP7394820B2 (en) 2017-01-17 2023-12-08 アストラゼネカ・アクチエボラーグ JAK1 selective inhibitor
CN110461830B (en) * 2017-01-17 2022-11-01 阿斯利康(瑞典)有限公司 Selective inhibitors of JAK1
KR102585048B1 (en) 2017-01-17 2023-10-05 아스트라제네카 아베 JAK1 selective inhibitor
JP2020506171A (en) * 2017-01-17 2020-02-27 アストラゼネカ・アクチエボラーグAstrazeneca Aktiebolag JAK1 selective inhibitor
CN115925693A (en) * 2017-01-17 2023-04-07 阿斯利康(瑞典)有限公司 Selective inhibitors of JAK1
KR20190104215A (en) * 2017-01-17 2019-09-06 아스트라제네카 아베 JAK1 selective inhibitor
CN110461830A (en) * 2017-01-17 2019-11-15 阿斯利康(瑞典)有限公司 JAK1 selective depressant
US11691986B2 (en) 2017-03-28 2023-07-04 Bayer Aktiengesellschaft PTEFB inhibiting macrocyclic compounds
WO2018177899A1 (en) 2017-03-28 2018-10-04 Bayer Aktiengesellschaft Novel ptefb inhibiting macrocyclic compounds
WO2018177889A1 (en) 2017-03-28 2018-10-04 Bayer Aktiengesellschaft Novel ptefb inhibiting macrocyclic compounds
US11254690B2 (en) 2017-03-28 2022-02-22 Bayer Pharma Aktiengesellschaft PTEFb inhibiting macrocyclic compounds
US11242356B2 (en) 2017-03-28 2022-02-08 Bayer Aktiengesellschaft PTEFb inhibiting macrocyclic compounds
JP2020517595A (en) * 2017-04-19 2020-06-18 ジェンフリート セラピューティクス(シャンハイ)インコーポレイテッド Novel inhibitor of cyclin-dependent kinase CDK9
EP3613737A4 (en) * 2017-04-19 2020-08-26 Genfleet Therapeutics (Shanghai) Inc. Novel inhibitor of cyclin-dependent kinase cdk9
US10952999B2 (en) 2017-04-19 2021-03-23 Genfleet Therapeutics (Shanghai) Inc. Inhibitor of cyclin-dependent kinase CDK9
US11666888B2 (en) 2018-02-05 2023-06-06 Bio-Rad Laboratories, Inc. Chromatography resin having an anionic exchange-hydrophobic mixed mode ligand
US11701347B2 (en) 2018-02-13 2023-07-18 Bayer Aktiengesellschaft Use of 5-fluoro-4-(4-fluoro-2-methoxyphenyl)-N-{4-[(S-methylsulfonimidoyl)methyl]pyridin-2-yl}pyridin-2-amine for treating diffuse large B-cell lymphoma
US10980788B2 (en) 2018-06-08 2021-04-20 Agios Pharmaceuticals, Inc. Therapy for treating malignancies

Also Published As

Publication number Publication date
GB0805477D0 (en) 2008-04-30
US20110092490A1 (en) 2011-04-21
CN102143953B (en) 2013-12-25
CN102143953A (en) 2011-08-03
WO2009118567A3 (en) 2010-03-11

Similar Documents

Publication Publication Date Title
US20110092490A1 (en) Pyrimidines, triazines and their use as pharmaceutical agents
AU2001242629B2 (en) 2-substituted 4-heteroaryl-pyrimidines and their use in the treatment of proliferative disorders
EP1641805B1 (en) Thiazolo-, oxazalo and imidazolo-quinazoline compounds capable of inhibiting protein kinases
EP1678171B1 (en) 2-amino-4-thiazolone-pyrimidine derivatives as protein kinase inhibitors
US20060241297A1 (en) Pyridinylamino-pyrimidine derivatives as protein kinase inhibitors
EP1567522B1 (en) Pyrimidine compounds
AU2004261484A1 (en) 2-aminophenyl-4-phenylpyrimidines as kinase inhibitors
US20090215805A1 (en) 4-Heteroaryl Pyrimidine Derivatives and use thereof as Protein Kinase Inhibitors
AU2001242629A1 (en) 2-substituted 4-heteroaryl-pyrimidines and their use in the treatment of proliferative disorders
CA2592723A1 (en) 4- (1h-indol-3-yl) -pyrimidin-2-ylamine derivates and their use in therapy
CZ2004354A3 (en) Derivatives of N-(4-(4-methylthiazol-5-yl) pyrimidin-2-yl)-N-phenylamine
MXPA06004442A (en) Pyrimidin-4-yl-3, 4-thione compounds and their use in therapy

Legal Events

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

Ref document number: 200980112366.5

Country of ref document: CN

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

Ref document number: 09723706

Country of ref document: EP

Kind code of ref document: A2

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 12934798

Country of ref document: US

122 Ep: pct application non-entry in european phase

Ref document number: 09723706

Country of ref document: EP

Kind code of ref document: A2