WO2006070202A1 - Dérivés de pyrazole ayant une activité de modulation des kinases - Google Patents

Dérivés de pyrazole ayant une activité de modulation des kinases Download PDF

Info

Publication number
WO2006070202A1
WO2006070202A1 PCT/GB2005/005109 GB2005005109W WO2006070202A1 WO 2006070202 A1 WO2006070202 A1 WO 2006070202A1 GB 2005005109 W GB2005005109 W GB 2005005109W WO 2006070202 A1 WO2006070202 A1 WO 2006070202A1
Authority
WO
WIPO (PCT)
Prior art keywords
group
compound
formula
methyl
optionally substituted
Prior art date
Application number
PCT/GB2005/005109
Other languages
English (en)
Inventor
Valerio Berdini
Michael Alistair O'brien
Eva Figueroa Navarro
Paul Graham Wyatt
Original Assignee
Astex Therapeutics Limited
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from GB0428537A external-priority patent/GB0428537D0/en
Application filed by Astex Therapeutics Limited filed Critical Astex Therapeutics Limited
Publication of WO2006070202A1 publication Critical patent/WO2006070202A1/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/14Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing three or more hetero rings

Definitions

  • This invention relates to pyrazole compounds that inhibit or modulate the activity of Cyclin Dependent Kinases (CDK), Glycogen Synthase Kinases (GSK) and Aurora kinases, to the use of the compounds in the treatment or prophylaxis of disease states or conditions mediated by the kinases, and to novel compounds having kinase inhibitory or modulating activity. Also provided are pharmaceutical compositions containing the compounds and novel chemical intermediates.
  • Protein kinases constitute a large family of structurally related enzymes that are responsible for the control of a wide variety of signal transduction processes within the cell (Hardie, G. and Hanks, S. (1995) The Protein Kinase Facts Book. I and II, Academic Press, San Diego, CA).
  • the kinases may be categorized into families by the substrates they phosphorylate (e.g., protein-tyrosine, protein-serine/threonine, lipids, etc.).
  • Protein kinases may be characterized by their regulation mechanisms. These mechanisms include, for example, autophosphorylation, transphosphorylation by other kinases, protein-protein interactions, protein-lipid interactions, and protein- polynucleotide interactions. An individual protein kinase may be regulated by more than one mechanism.
  • Kinases regulate many different cell processes including, but not limited to, proliferation, differentiation, apoptosis, motility, transcription, translation and other signalling processes, by adding phosphate groups to target proteins. These phosphorylation events act as molecular on/off switches that can modulate or regulate the target protein biological function. Phosphorylation of target proteins occurs in response to a variety of extracellular signals (hormones, neurotransmitters, growth and differentiation factors, etc.), cell cycle events, environmental or nutritional stresses, etc. The appropriate protein kinase functions in signalling pathways to activate or inactivate (either directly or indirectly), for example, a metabolic enzyme, regulatory protein, receptor, cytoskeletal protein, ion channel or pump, or transcription factor.
  • Uncontrolled signalling due to defective control of protein phosphorylation has been implicated in a number of diseases, including, for example, inflammation, cancer, allergy/asthma, disease and conditions of the immune system, disease and conditions of the central nervous system, and angiogenesis.
  • Cdks are cdc2 (also known as cdkl) homologous serine-threonine kinase proteins that are able to utilise ATP as a substrate in the phosphorylation of diverse polypeptides in a sequence dependent context.
  • Cyclins are a family of proteins characterised by a homology region, containing approximately 100 amino acids, termed the "cyclin box" which is used in binding to, and defining selectivity for, specific cdk partner proteins.
  • Modulation of the expression levels, degradation rates, and activation levels of various cdks and cyclins throughout the cell cycle leads to the cyclical formation of a series of cdk/cyclin complexes, in which the cdks are enzymatically active.
  • the formation of these complexes controls passage through discrete cell cycle checkpoints and thereby enables the process of cell division to continue.
  • Failure to satisfy the pre-requisite biochemical criteria at a given cell cycle checkpoint, i.e. failure to form a required cdk/cyclin complex can lead to cell cycle arrest and/or cellular apoptosis. Aberrant cellular proliferation, as manifested in cancer, can often be attributed to loss of correct cell cycle control.
  • Inhibition of cdk enzymatic activity therefore provides a means by which abnormally dividing cells can have their division arrested and/or be killed.
  • the diversity of cdks, and cdk complexes, and their critical roles in mediating the cell cycle, provides a broad spectrum of potential therapeutic targets selected on the basis of a defined biochemical rationale.
  • Progression from the Gl phase to the S phase of the cell cycle is primarily regulated by cdk2, cdk3, cdk4 and cdk ⁇ via association with members of the D and E type cyclins.
  • the D-type cyclins appear instrumental in enabling passage beyond the Gl restriction point, where as the cdk2/cyclin E complex is key to the transition from the Gl to S phase. Subsequent progression through S phase and entry into G2 is thought to require the cdk2/cyclin A complex.
  • mitosis, and the G2 to M phase transition which triggers it are regulated by complexes of cdkl and the A and B type cyclins.
  • Gl phase Retinoblastoma protein (Rb), and related pocket proteins such as pl30 are substrates for cdk(2, 4, & 6)/cyclin complexes. Progression through Gl is in part facilitated by hyperphosphorylation, and thus inactivation, of Rb and pl30 by the cdk(4/6)/cyclin-D complexes. Hyperphosphorylation of Rb and pi 30 causes the release of transcription factors, such as E2F, and thus the expression of genes necessary for progression through Gl and for entry into S-phase, such as the gene for cyclin E. Expression of cyclin E facilitates formation of the cdk2/cyclin E complex which amplifies, or maintains, E2F levels via further phosphorylation of Rb.
  • transcription factors such as E2F
  • the cdk2/cyclin E complex also phosphorylates other proteins necessary for DNA replication, such as NPAT, which has been implicated in histone biosynthesis. Gl progression and the Gl/S transition are also regulated via the mitogen stimulated Myc pathway, which feeds into the cdk2/cyclin E pathway. Cdk2 is also connected to the p53 mediated DNA damage response pathway via p53 regulation of p21 levels. p21 is a protein inhibitor of cdk2/cyclin E and is thus capable of blocking, or delaying, the Gl/S transition.
  • the cdk2/cyclin E complex may thus represent a point at which biochemical stimuli from the Rb, Myc and p53 pathways are to some degree integrated. Cdk2 and/or the cdk2/cyclin E complex therefore represent good targets for therapeutics designed at arresting, or recovering control of, the cell cycle in aberrantly dividing cells.
  • cdk5 which is necessary for correct neuronal development and which has also been implicated in the phosphorylation of several neuronal proteins such as Tau, NUDE-I, synapsinl, DARPP32 and the
  • cdk5 is conventionally activated by binding to the p35/p39 proteins.
  • Cdk5 activity can, however, be deregulated by the binding of p25, a truncated version of p35.
  • Conversion of p35 to p25, and subsequent deregulation of cdk5 activity, can be induced by ischemia, excitotoxicity, and ⁇ -amyloid peptide. Consequently p25 has been implicated in the pathogenesis of neurodegenerative diseases, such as Alzheimer's, and is therefore of interest as a target for therapeutics directed against these diseases.
  • Cdk7 is a nuclear protein that has cdc2 CAK activity and binds to cyclin H.
  • Cdk7 has been identified as component of the TFIIH transcriptional complex which has RNA polymerase II C-terminal domain (CTD) activity. This has been associated with the regulation of HIV-I transcription via a Tat-mediated biochemical pathway.
  • Cdk8 binds cyclin C and has been implicated in the phosphorylation of the CTD of RNA polymerase II.
  • the cdk9/cyclin-Tl complex (P-TEFb complex) has been implicated in elongation control of RNA polymerase II.
  • PTEF -b is also required for activation of transcription of the HIV-I genome by the viral transactivator Tat through its interaction with cyclin Tl .
  • Cdk7, cdk8 5 cdk9 and the P-TEFb complex are therefore potential targets for anti-viral therapeutics.
  • Cdk phosphorylation is performed by a group of cdk activating kinases (CAKs) and/or kinases such as weel, Mytl and Mikl .
  • Dephosphorylation is performed by phosphatases such as cdc25(a & c), pp2a, or KAP.
  • Cdk/cyclin complex activity may be further regulated by two families of endogenous cellular proteinaceous inhibitors: the Kip/Cip family, or the INK family.
  • the INK proteins specifically bind cdk4 and cdk ⁇ .
  • pl6 mk4 also known as MTSl is a potential tumour suppressor gene that is mutated, or deleted, in a large number of primary cancers.
  • the Kip/Cip family contains proteins such as p21 c ipi, w afi ; p27 ⁇ ipi and As discussed previously p21 is induced by p53 and is able to inactivate the cdk2/cyclin(E/A) and cdk4/cyclin(Dl/D2/D3) complexes. Atypically low levels of p27 expression have been observed in breast, colon and prostate cancers. Conversely over expression of cyclin E in solid tumours has been shown to correlate with poor patient prognosis. Over expression of cyclin Dl has been associated with oesophageal, breast, squamous, and non-small cell lung carcinomas.
  • Cdk inhibitors could conceivably also be used to treat other conditions such as viral infections, autoimmune diseases and neuro-degenerative diseases, amongst others.
  • Cdk targeted therapeutics may also provide clinical benefits in the treatment of the previously described diseases when used in combination therapy with either existing, or new, therapeutic agents.
  • Cdk targeted anticancer therapies could potentially have advantages over many current antitumour agents as they would not directly interact with DNA and should therefore reduce the risk of secondary tumour development. Diffuse Large B-cell Lymphomas ( " DLBCD
  • p27KIPl is a CDKi key in cell cycle regulation, whose degradation is required for Gl/S transition.
  • p27KIPl expression in proliferating lymphocytes, some aggressive B-cell lymphomas have been reported to show an anomalous p27KIPl staining. An abnormally high expression of p27KIPl was found in lymphomas of this type.
  • CLL chronic lymphocytic leukaemia
  • Flavopiridol and CYC 202 inhibitors of cyclin-dependent kinases induce in vitro apoptosis of malignant cells from B-cell chronic lymphocytic leukemia (B-CLL).
  • Flavopiridol exposure results in the stimulation of caspase 3 activity and in caspase- dependent cleavage of p27(kipl), a negative regulator of the cell cycle, which is overexpressed in B-CLL (Blood. 1998 Nov 15;92(10):3804-16 Flavopiridol induces apoptosis in chronic lymphocytic leukemia cells via activation of caspase-3 without evidence of bcl-2 modulation or dependence on functional p53.
  • Byrd JC Shinn C, Waselenko JK, Fuchs EJ, Lehman TA, Nguyen PL, Flinn IW, Diehl LF, Sausville E, Grever MR). Aurora Kinases
  • Aurora kinases a new family of serine/threonine kinases known as the Aurora kinases has been discovered that are involved in the G2 and M phases of the cell cycle, and which are important regulators of mitosis.
  • Aurora kinases are located at the centrosomes of interphase cells, at the poles of the bipolar spindle and in the mid-body of the mitotic apparatus.
  • Aurora A also referred to in the literature as Aurora 2
  • Aurora B also referred to in the literature as Aurora 1
  • Aurora C also referred to in the literature as Aurora 3
  • the Aurora kinases have highly homologous catalytic domains but differ considerably in their N-terminal portions (Katayama H, Brinkley WR, Sen S.; The Aurora kinases: role in cell transformation and tumorigenesis; Cancer Metastasis Rev. 2003 Dec;22(4):451-64).
  • the substrates of the Aurora kinases A and B have been identified as including a kinesin-like motor protein, spindle apparatus proteins, histone H3 protein, kinetochore protein and the tumour suppressor protein p53.
  • Aurora A kinases are believed to be involved in spindle formation and become localised on the centrosome during the early G2 phase where they phosphorylate spindle-associated proteins (Fxigent et al, Cell, 114: 531-535 (2003). Hirota etal, Cell, 114:585-598, (2003) found that cells depleted of Aurora A protein kinase were unable to enter mitosis. Furthermore, it has been found (Adams, 2001) that mutation or disruption of the Aurora A gene in various species leads to mitotic abnormalities, including centrosome separation and maturation defects, spindle aberrations and chromosome segregation defects.
  • Aurora kinases are generally expressed at a low level in the majority of normal tissues, the exceptions being tissues with a high proportion of dividing cells such as the thymus and testis.
  • elevated levels of Aurora kinases have been found in many human cancers (Giet et ah, J. Cell. ScI 112: 3591-361, (1999) and Katayama (2003).
  • Aurora A kinase maps to the chromosome 20ql 3 region that has frequently been found to be amplified in many human cancers.
  • Aurora- A Amplification and/or over-expression of Aurora- A is observed in human bladder cancers and amplification of Aurora- A is associated with aneuploidy and aggressive clinical behaviour, see Sen et al., J. Natl.Cancer Inst, 94: 1320-1329 (2002).
  • Aurora-B is highly expressed in multiple human tumour cell lines, including leukemic cells [Katayama et al., Gene 244: 1-7) ]. Levels of this enzyme increase as a function of Duke's stage in primary colorectal cancers [Katayama et al., J. Natl Cancer Inst, 91: 1160-1162 (1999)].
  • Royce et al report that the expression of the Aurora 2 gene (known as STKl 5 or BTAK) has been noted in approximately one-fourth of primary breast tumours.
  • STKl 5 or BTAK the expression of the Aurora 2 gene
  • Endometrial carcinoma comprises at least two types of cancer: endometrioid carcinomas (EECs) are estrogen-related tumours, which are frequently euploid and have a good prognosis.
  • EECs endometrioid carcinomas
  • NEECs nonendometrioid carcinomas
  • Cancers which may be particularly amenable to Aurora inhibitors include breast, bladder, colorectal, pancreatic, ovarian, non-Hodgkin's lymphoma, gliomas and nonendometrioid endometrial carcinomas.
  • Leukemias particularly amenable to Aurora inhibitors include Acute Myelogenous Leukemia (AML), chronic myelogenous leukaemia (CML), B-cell lymphoma (Mantle cell), and Acute Lymphoblastic Leukemia (ALL).
  • Glycogen Synthase Kinase-3 is a serine-threonine kinase that occurs as two ubiquitously expressed isoforms in humans (GSK3 ⁇ & beta GSK3 ⁇ ).
  • GSK3 has been implicated as having roles in embryonic development, protein synthesis, cell proliferation, cell differentiation, microtubule dynamics, cell motility and cellular apoptosis. As such GSK3 has been implicated in the progression of disease states such as diabetes, cancer, Alzheimer's disease, stroke, epilepsy, motor neuron disease and/or head trauma.
  • CDKs cyclin dependent kinases
  • the consensus peptide substrate sequence recognised by GSK3 is (Ser/Thr)-X-X- X-(pSer/pThr), where X is any amino acid (at positions (n+1), (n+2), (n+3)) and pSer and pThr are phospho-serine and phospho-threonine respectively (n+4).
  • GSK3 phosphorylates the first serine, or threonine, at position (n). Phospho-serine, or phospho-threonine, at the (n+4) position appear necessary for priming GSK3 to give maximal substrate turnover. Phosphorylation of GSK3 ⁇ at Ser21, or GSK3 ⁇ at Ser9, leads to inhibition of GSK3.
  • GSK3 ⁇ and GSK ⁇ may be subtly regulated by phosphorylation of tyrosines 279 and 216 respectively. Mutation of these residues to a Phe caused a reduction in in vivo kinase activity.
  • the X-ray crystallographic structure of GSK3 ⁇ has helped to shed light on all aspects of GSK3 activation and regulation.
  • GSK3 forms part of the mammalian insulin response pathway and is able to phosphorylate, and thereby inactivate, glycogen synthase. Upregulation of glycogen synthase activity, and thereby glycogen synthesis, through inhibition of GSK3, has thus been considered a potential means of combating type II, or non- insulin-dependent diabetes mellitus (NIDDM): a condition in which body tissues become resistant to insulin stimulation.
  • NIDDM non- insulin-dependent diabetes mellitus
  • PBK phosphoinositide-3 kinase
  • PIP3 second messenger phosphatidylinosityl 3,4,5-trisphos ⁇ hate
  • PDKl 3-phosphoinositide-dedependent protein kinase 1
  • PKB protein kinase B
  • PKB is able to phosphorylate, and thereby inhibit, GSK3 ⁇ and/or GSK ⁇ through phosphorylation of Ser9, or ser21, respectively.
  • the inhibition of GSK3 then triggers upregulation of glycogen synthase activity.
  • Therapeutic agents able to inhibit GSK3 may thus be able to induce cellular responses akin to those seen on insulin stimulation.
  • a further in vivo substrate of GSK3 is the eukaryotic protein synthesis initiation factor 2B (eIF2B).
  • eIF2B is inactivated via phosphorylation and is thus able to suppress protein biosynthesis.
  • Inhibition of GSK3, e.g. by inactivation of the "mammalian target of rapamycin" protein (mTOR), can thus upregulate protein biosynthesis.
  • GSK3 activity via the mitogen activated protein kinase (MAPK) pathway through phosphorylation of GSK3 by kinases such as mitogen activated protein kinase activated protein kinase 1 (MAPKAP-Kl or RSK).
  • MAPK mitogen activated protein kinase
  • RSK mitogen activated protein kinase activated protein kinase 1
  • GSK3 ⁇ is a key component in the vertebrate Wnt signalling pathway. This biochemical pathway has been shown to be critical for normal embryonic development and regulates cell proliferation in normal tissues. GSK3 becomes inhibited in response to Wnt stimulii. This can lead to the de- phosphorylation of GSK3 substrates such as Axin, the adenomatous polyposis coli (APC) gene product and ⁇ -catenin. Aberrant regulation of the Wnt pathway has been associated with many cancers. Mutations in APC, and/or ⁇ -catenin, are common in colorectal cancer and other tumours, ⁇ -catenin has also been shown to be of importance in cell adhesion.
  • APC adenomatous polyposis coli
  • GSK3 may also modulate cellular adhesion processes to some degree.
  • GSK3 may also modulate cellular adhesion processes to some degree.
  • transcription factors such as c-Jun, CCAAT/enhancer binding protein ⁇ (C/EBP ⁇ ), c-Myc and/or other substrates such as Nuclear Factor of Activated T-cells (NFATc), Heat Shock Factor-1 (HSF-I) and the c-AMP response element binding protein (CREB).
  • NFATc Nuclear Factor of Activated T-cells
  • HSF-I Heat Shock Factor-1
  • CREB c-AMP response element binding protein
  • GSK3 The role of GSK3 in modulating cellular apoptosis, via a pro-apoptotic mechanism, may be of particular relevance to medical conditions in which neuronal apoptosis can occur. Examples of these are head trauma, stroke, epilepsy, Alzheimer's and motor neuron diseases, progressive supranuclear palsy, corticobasal degeneration, and Pick's disease.
  • head trauma head trauma
  • stroke epilepsy
  • Alzheimer's and motor neuron diseases progressive supranuclear palsy
  • corticobasal degeneration corticobasal degeneration
  • Pick's disease In vitro it has been shown that GSK3 is able to hyper- phosphorylate the microtubule associated protein Tau. Hyperphosphorylation of Tau disrupts its normal binding to microtubules and may also lead to the formation of intra-cellular Tau filaments. It is believed that the progressive accumulation of these filaments leads to eventual neuronal dysfunction and degeneration. Inhibition of Tau phosphorylation, through inhibition of GSK3, may
  • WO 02/34721 from Du Pont discloses a class of indeno [l,2-c]pyrazol-4-ones as inhibitors of cyclin dependent kinases.
  • WO 01/81348 from Bristol Myers Squibb describes the use of 5-thio-, sulfmyl- and sulfonylpyrazolo [3, 4-b] -pyridines as cyclin dependent kinase inhibitors.
  • WO 00/62778 also from Bristol Myers Squibb discloses a class of protein tyrosine kinase inhibitors.
  • WO 01 /72745 Al from Cyclacel describes 2-substituted 4-heteroaryl-pyrimidines and their preparation, pharmaceutical compositions containing them and their use as inhibitors of cyclin-dependant kinases (cdks) and hence their use in the treatment of proliferative disorders such as cancer, leukaemia, psoriasis and the like.
  • cdks cyclin-dependant kinases
  • WO 99/21845 from Agouron describes 4-aminothiazole derivatives for inhibiting cyclin-dependent kinases (cdks), such as CDKl, CDK2, CDK4, and CDK6.
  • cdks cyclin-dependent kinases
  • the invention is also directed to the therapeutic or prophylactic use of pharmaceutical compositions containing such compounds and to methods of treating malignancies and other disorders by administering effective amounts of such compounds.
  • WO 01/53274 from Agouron discloses as CDK kinase inhibitors a class of compounds which can comprise an amide-substituted benzene ring linked to an N- containing heterocyclic group.
  • indazole compounds are not mentioned genetically, one of the exemplified compounds comprises an indazole 3-carboxylic acid anilide moiety linked via a methylsulfanyl group to a pyrazolopyrimidine.
  • WO 01/98290 discloses a class of 3-aminocarbonyl-2- carboxamido thiophene derivatives as protein kinase inhibitors. The compounds are stated to have multiple protein kinase activity.
  • WO 01/53268 and WO 01/02369 from Agouron disclose compounds that mediate or inhibit cell proliferation through the inhibition of protein kinases such as cyclin dependent kinase or tyrosine kinase.
  • WO 00/39108 and WO 02/00651 both to Du Pont Pharmaceuticals describe broad classes of heterocyclic compounds that are inhibitors of trypsin-like serine protease enzymes, especially factor Xa and thrombin. The compounds are stated to be useful as anticoagulants or for the prevention of thromboembolic disorders.
  • Heterocyclic compounds that have activity against factor Xa are also disclosed in WO 01/1978 Cor Therapeutics) and US 2002/0091116 (Zhu et al).
  • WO 03/035065 discloses a broad class of benzimidazole derivatives as protein kinase inhibitors but does not disclose activity against CDK kinases or GSK kinases.
  • WO 97/36585 and US 5,874,452 disclose biheteroaryl compounds that are inhibitors of farnesyl transferase.
  • WO 03/037274 (Icagen) discloses pyrazole amides as inhibitors of sodium channels.
  • WO 00/43384 (Boehringer Ingelheim) discloses aryl and heteroaryl ureas as antiinflammatory agents.
  • WO 00/07996 discloses pyrazole compounds for use as oestrogen receptor modulators which may be useful in, for example, the treatment of breast and endometrial cancers.
  • WO 2004/000318 discloses amino-subsituted moncyclic compounds as kinase modulators that may be useful in the treatment of cancers.
  • WO 03/062392 discloses aryl imidazole amides as EDG receptor modulators that may be useful in the treatment of cancers.
  • WO 01/68585 discloses a class of amides for use as 5-HT anatagonists.
  • WO 97/40017 discloses a broad class of heterocyclic compounds for use as protein tyrosine phosphatase modulators.
  • the invention provides compounds that have cyclin dependent kinase inhibiting or modulating activity and/or glycogen synthase kinase-3 (GSK3) inhibiting or modulating activity, and/or Aurora kinase inhibiting or modulating activity, and which it is envisaged will be useful in preventing or treating disease states or conditions mediated by the kinases.
  • GSK3 glycogen synthase kinase-3
  • the compounds of the invention will be useful in alleviating or reducing the incidence of cancer.
  • the invention provides a compound of the formula (I):
  • A is selected from a bond, CH 2 and CH(CN);
  • R 1 is selected from:
  • a cycloalkyl group of 3 to 6 ring members optionally substituted by one or more substituents selected from methyl, ethyl, hydroxy, methoxy, ethoxy, fluorine, amino and cyano;
  • a phenyl group optionally substituted by up to three substituents selected from methyl, ethyl, fluorine, chlorine, methoxy, ethoxy and methylsulphonyl, but excluding 2,6-difluorophenyl, 2-fluoro-6-methoxy and 5-chloro-2-methoxyphenyl;
  • a monocyclic heterocyclic group selected from furyl and isoxazolyl, the heterocyclic group being optionally substituted by one or two groups selected from methyl, ethyl, and a group CH 2 R 2 where R 2 is a five or six membered saturated heterocyclic ring containing one or two heteroatom ring members selected from O and N, the heterocyclic ring being optionally substituted by one or two methyl groups;
  • a bicyclic heterocyclic group selected from 2,3-dihydrobenzofuranyl and benzo[c] isoxazolyl, the bicyclic group being optionally substituted by one or two substituents selected from methyl, ethyl, hydroxy, methoxy, ethoxy, fluorine, amino, cyano and chlorine, the bicyclic heterocyclic group being other than a 2,2-dimethyl-2,3-dihydrobenzofuran-7-yl group.
  • R 1 is an optionally substituted cycloalkyl group of 3 to 6 ring members.
  • cycloalkyl groups are cyclopropyl, cyclobutyl, cyclopentyl and cyclohexyl groups.
  • R 1 is an optionally substituted cycloalkyl group of 3 to 5 ring members, for example 3, or 4, or 5 ring members).
  • R 1 can be an optionally substituted cyclopropyl group. More particularly, R 1 can be an unsubstituted cyclopropyl group.
  • R 1 is a cycloalkyl group and in particular a cyclopropyl group
  • the moiety A can be a bond, or a group selected from CH 2 and CH(CN). In one particular subgroup of compounds, A is selected from CH 2 and CH(CN).
  • R 1 can be a phenyl group optionally substituted by up to three substituents selected from methyl, ethyl, fluorine, chlorine, methoxy, ethoxy and methylsulphonyl, but excluding 2,6-difluorophenyl, 2-fluoro-6-methoxy and 5- chloro-2-methoxyphenyl.
  • the phenyl group may be substituted by one, two or three substituents selected from chlorine, fluorine, methoxy, ethoxy and methylsulphonyl.
  • the phenyl group may be selected from 2-ethoxyphenyl, 2- methoxy-5-methylsulphonylphenyl, 2-fluoro-6-chorophenyl, 2,4,6-trifluorophenyl and 5-fiuoro-2-methoxyphenyl.
  • the phenyl group is selected from 2-ethoxyphenyl, 2-methoxy-5-methylsulphonylphenyl and 5-fluoro-2-methoxyphenyl.
  • R 1 is a monocyclic heterocyclic group selected from furyl and isoxazolyl, the heterocyclic group being optionally substituted by one or two groups selected from methyl, ethyl, and a group CH 2 R 2 where R 2 is a five or six membered saturated heterocyclic ring containing one or two heteroatom ring members selected from O and N, the heterocyclic ring being optionally substituted by one or two methyl groups.
  • R 2 is a five or six membered saturated heterocyclic ring, it can be, for example, selected from morpholine, piperidine (e.g. 1-piperidinyl, 2-piperidinyl 3- piperidinyl and 4-piperidinyl), N-methyl piperidine, pyrrolidine (e.g. 1-pyrrolidinyl, 2-pyrrolidinyl and 3-pyrrolidinyl), azetidine, pyran (2H-pyran or 4H-pyran), dioxane, tetrahydropyran (e.g. 4-tetrahydro pyranyl), piperazine, and N-methyl piperazine.
  • piperidine e.g. 1-piperidinyl, 2-piperidinyl 3- piperidinyl and 4-piperidinyl
  • N-methyl piperidine e.g. 1-pyrrolidinyl, 2-pyrrolidinyl and 3-pyrrolidinyl
  • pyran (2H-pyran or 4
  • R 1 is an isoxazole group optionally substituted by one or more methyl groups.
  • R 1 is a furyl group optionally substituted by one or two groups selected from methyl, ethyl, and a group CH 2 R 2 where R 2 is a five or six membered saturated heterocyclic ring containing one or two heteroatom ring members selected from O and N, the heterocyclic ring being optionally substituted by one or two methyl groups.
  • Particular compounds are those in which the furyl group is an optionally substituted 2-furyl group.
  • the furyl group is preferably other than a 3-methyl-2-furyl, 5 -(pyrrolidine- l-ylmethyl)-2-furyl or 5-(4- morpholinylmethyl)-2-furyl group.
  • R 1 is a bicyclic heterocyclic group selected from 2,3-dihydrobenzofuranyl and benzo[c]isoxazolyl optionally substituted by one or two substituents selected from methyl, ethyl, methoxy, ethoxy, fluorine and chlorine, the bicyclic heterocyclic group being other than a 2,2-dimethyl-2,3- dihydrobenzofuran-7-yl group.
  • R 1 is a 2,3-dihydrobenzofuranyl group optionally substituted by one or two substituents selected from methyl, ethyl, methoxy, ethoxy, fluorine and chlorine, but excluding a 2,2-dimethyl-2,3-dihydrobenzofuran- 7-yl group.
  • the 2,3-dihydrobenzofuranyl group may be, for example, an unsubstituted 2,3-dihydrobenzofuranyl group, such as a 2,3-dihydrobenzofuran-6-yl group or 2,3-dihydrobenzofuran-7-yl group.
  • R 1 is a benzo[c]isoxazolyl group optionally substituted by one or two substituents selected from methyl, ethyl, methoxy, ethoxy, fluorine and chlorine. More particularly, the benzo[c]isoxazolyl group may be unsubstituted.
  • the moiety A is selected from a bond, CH 2 and CH(CN).
  • the moiety A is a bond.
  • the moiety A is CH 2 .
  • the moiety A is CH(CN).
  • A is selected from a bond and a CH 2 group.
  • A is selected from CH 2 and CH(CN).
  • the various functional groups and substituents making up the compounds of the formula (I) are typically chosen such that the molecular weight of the compound of the formula (I) does not exceed 1000. More usually, the molecular weight of the compound will be less than 750, for example less than 700, or less than 650, or less than 600, or less than 550. More preferably, the molecular weight is less than 525 and, for example, is 500 or less.
  • the invention provides inter alia:
  • a method for the prophylaxis or treatment of a disease state or condition mediated by a cyclin dependent kinase or glycogen synthase kinase-3 which method comprises administering to a subject in need thereof a compound of the formula (I) as defined herein.
  • a method for alleviating or reducing the incidence of a disease state or condition mediated by a cyclin dependent kinase or glycogen synthase kinase-3 which method comprises administering to a subject in need thereof a compound of the formula (I) as defined herein.
  • a method for treating a disease or condition comprising or arising from abnormal cell growth in a mammal which method comprises administering to the mammal a compound of the formula (I) as defined herein in an amount effective in inhibiting abnormal cell growth.
  • a method for alleviating or reducing the incidence of a disease or condition comprising or arising from abnormal cell growth in a mammal which method comprises administering to the mammal a compound of the formula (I) as defined herein in an amount effective in inhibiting abnormal cell growth.
  • a method for treating a disease or condition comprising or arising from abnormal cell growth in a mammal comprising administering to the mammal a compound of the formula (I) as defined herein in an amount effective to inhibit a cdk kinase (such as cdkl or cdk2) or glycogen synthase kinase-3 activity.
  • a cdk kinase such as cdkl or cdk2
  • a method for alleviating or reducing the incidence of a disease or condition comprising or arising from abnormal cell growth in a mammal comprising administering to the mammal a compound of the formula (I) as defined herein in an amount effective to inhibit a cdk kinase (such as cdkl or cdk2) or glycogen synthase kinase-3 activity.
  • a cdk kinase such as cdkl or cdk2
  • glycogen synthase kinase-3 activity such as cdkl or cdk2
  • a method of inhibiting a cyclin dependent kinase or glycogen synthase kinase-3 which method comprises contacting the kinase with a kinase- inhibiting compound of the formula (I) as defined herein.
  • a method of modulating a cellular process for example cell division
  • a compound of the formula (I) as defined herein for the manufacture of a medicament for the prophylaxis or treatment of a cancer, the cancer being one which is characterised by up-regulation of an Aurora kinase (e.g. Aurora A kinase or Aurora B kinase).
  • an Aurora kinase e.g. Aurora A kinase or Aurora B kinase
  • a method for the prophylaxis or treatment of a disease or condition characterised by up-regulation of an Aurora kinase e.g. Aurora A kinase or Aurora B kinase
  • the method comprising administering a compound of the formula (I) as defined herein.
  • a method for alleviating or reducing the incidence of a disease or condition characterised by up-regulation of an Aurora kinase e.g. Aurora A kinase or
  • Aurora B kinase the method comprising administering a compound of the formula (I) as defined herein.
  • a method for the prophylaxis or treatment of (or alleviating or reducing the incidence of) cancer in a patient suffering from or suspected of suffering from cancer which method comprises (i) subjecting a patient to a diagnostic test to determine whether the patient possesses the IleSl variant of the Aurora A gene; and (ii) where the patient does possess the said variant, thereafter administering to the patient a compound of the formula (I) as defined herein having Aurora kinase inhibiting activity.
  • a method for the prophylaxis or treatment of (or alleviating or reducing the incidence of) a disease state or condition characterised by up-regulation of an Aurora kinase e.g. Aurora A kinase or Aurora B kinase
  • a disease state or condition characterised by up-regulation of an Aurora kinase e.g. Aurora A kinase or Aurora B kinase
  • method comprises (i) subjecting a patient to a diagnostic test to detect a marker characteristic of up-regulation of the Aurora kinase and (ii) where the diagnostic test is indicative of up-regulation of Aurora kinase, thereafter administering to the patient a compound of the formula (I) as defined herein having Aurora kinase inhibiting activity.
  • a pharmaceutical composition comprising a compound of the formula (I) as defined herein and a pharmaceutically acceptable carrier.
  • a pharmaceutical composition for administration in an aqueous solution form comprising a compound of the formula (I), (II) or (III) or any sub-groups or examples thereof as defined herein in the form of a salt having a solubility in water of greater than 25 mg/ml, typically greater than 50 mg/ml and preferably greater than 100 mg/ml.
  • a pharmaceutical composition for parenteral administration comprising a compound of the formula (I) as defined herein and a pharmaceutically acceptable carrier suitable for parenteral administration.
  • compositions for administration by intravenous, intramuscular, intraperitoneal or subcutaneous administration injection or for direct delivery into a target organ or tissue by injection or infusion comprising a compound of the formula (I) as defined herein and a pharmaceutically acceptable carrier suitable for parenteral administration.
  • a pharmaceutical composition comprising a compound of the formula (I) as defined herein in the form of a sterile solution for injection.
  • a compound of formula (I) or a salt e.g. an acid addition salt
  • solvate e.g. tautomer or N-oxide thereof for use in the treatment of B-cell lymphoma.
  • a compound of formula (I) or a salt e.g. an acid addition salt
  • solvate e.g. tautomer or N-oxide thereof for use in the treatment of chronic lymphocytic leukaemia.
  • a compound of formula (I) or a salt e.g. an acid addition salt
  • solvate e.g. tautomer or N-oxide thereof for use in the treatment of diffuse large B cell lymphoma.
  • a method of treatment of B-cell lymphoma, diffuse large B cell lymphoma or chronic lymphocytic leukaemia by administering to a patient in need of such treatment a compound of formula (I) or a salt (e.g. an acid addition salt), solvate, tautomer or N-oxide thereof.
  • a compound of formula (I) or a salt (e.g. an acid addition salt), solvate, tautomer or N-oxide thereof for use in the treatment of leukaemia in particular relapsed or refractory acute myelogenous leukemia, myelodysplastic syndrome, acute lymphocytic leukemia and chronic myelogenous leukemia.
  • a reference to a particular compound also includes ionic, salt, solvate, and protected forms thereof, for example, as discussed below.
  • the salts of the present invention can be synthesized from the parent compound that contains a basic or acidic moiety by conventional chemical methods such as methods described in Pharmaceutical Salts: Properties, Selection, and Use, P. Heinrich Stahl (Editor), Camille G. Wermuth (Editor), ISBN: 3-90639-026-8, Hardcover, 388 pages, August 2002.
  • such salts can be prepared by reacting the free acid or base forms of these compounds with the appropriate base or acid in water or in an organic solvent, or in a mixture of the two; generally, nonaqueous media such as ether, ethyl acetate, ethanol, isopropanol, or acetonitrile are used.
  • Acid addition salts may be formed with a wide variety of acids, both inorganic and organic.
  • acid addition salts include salts formed with an acid selected from the group consisting of acetic, 2,2-dichloroacetic, adipic, alginic, ascorbic (e.g.
  • the acid addition salts salts may also be selected from aspartic (e.g. D-aspartic), carbonic, dodecanoate, isobutyric, laurylsulphonic, mucic, naphthalenesulphonic (e.g. naphthalene-2-sulphonic), toluenesulphonic (e.g. ⁇ -toluenesulphonic), and xinafoic acids.
  • aspartic e.g. D-aspartic
  • carbonic dodecanoate
  • isobutyric laurylsulphonic
  • toluenesulphonic e.g. ⁇ -toluenesulphonic
  • xinafoic acids xinafoic acids.
  • salts consist of salts formed from hydrochloric, hydriodic, phosphoric, nitric, sulphuric, citric, lactic, succinic, maleic, malic, isethionic, fumaric, benzenesulphonic, toluenesulphonic, methanesulphonic, ethanesulphonic, naphthalenesulphonic, valeric, acetic, propanoic, butanoic, malonic, glucuronic and lactobionic acids.
  • One sub-group of salts consists of salts formed from hydrochloric, acetic, adipic, L- aspartic and DL-lactic acids.
  • salts such as acid addition salts have a number of advantages over the corresponding free base. For example, the salts will enjoy one or more of the following advantages over the free base in that they will:
  • Preferred salts for use in the preparation of liquid (e.g. aqueous) compositions of the compounds of formula (I) described herein are salts having a solubility in a given liquid carrier (e.g. water) of greater than 25 mg/ml of the liquid carrier (e.g. water), more typically greater than 50 mg/ml and preferably greater than 100 mg/ml.
  • a liquid carrier e.g. water
  • preferred salts for use in the preparation of liquid (e.g. aqueous) compositions are salts having a solubility in a given liquid carrier (e.g. water) greater than 1 mg/ml, typically greater than 5 mg/ml of the liquid carrier (e.g. water), more typically greater than 15 mg/ml, more typically greater than 20 mg/ml and preferably greater than 25 mg/ml.
  • a liquid carrier e.g. water
  • the preferred acid addition salts are mesylate, ethanesulphonate, D- or L-lactate, and hydrochloride salts.
  • the acid addition salt is the lactate salt, in particular L-lactate or D- lactate, preferably L-lactate.
  • a pharmaceutical composition comprising an aqueous solution containing a compound of the formula (I) as described herein in the form of a salt in a concentration of greater than 25 mg/ml, typically greater than 50 mg/ml and preferably greater than 100 mg/ml.
  • a pharmaceutical composition comprising an aqueous solution containing a compound of the formula (I) as described herein in the form of a salt in a concentration of greater than 1 mg/ml, typically greater than 5 mg/ml of the liquid carrier (e.g. water), more typically greater than 15 mg/ml, more typically greater than 20 mg/ml and preferably greater than 25 mg/ml.
  • the liquid carrier e.g. water
  • a salt may be formed with a suitable cation.
  • suitable inorganic cations include, but are not limited to, alkali metal ions such as Na + and K + , alkaline earth metal cations such as Ca 2+ and Mg 2+ , and other cations such as Al 3+ .
  • suitable organic cations include, but are not limited to, ammonium ion (i.e., NH 4 + ) and substituted ammonium ions (e.g., NH 3 R + , NH 2 R 2 + , NHR 3 + , NR 4 + ).
  • Examples of some suitable substituted ammonium ions are those derived from: ethylamine, diethylamine, dicyclohexylamine, triethylamine, butylamine, ethylenediamine, ethanolamine, diethanolamine, piperazine, benzylamine, phenylbenzylamine, choline, meglumine, and tromethamine, as well as amino acids, such as lysine and arginine.
  • An example of a common quaternary ammonium ion is N(CH 3 ) 4 + .
  • the salt forms of the compounds of the invention are typically pharmaceutically acceptable salts, and examples of pharmaceutically acceptable salts are discussed in Berge et ah, 1977, "Pharmaceutically Acceptable Salts," J. Pharm. ScI, Vol. 66, pp. 1-19.
  • salts that are not pharmaceutically acceptable may also be prepared as intermediate forms which may then be converted into pharmaceutically acceptable salts.
  • Such non-pharmaceutically acceptable salts forms which may be useful, for example, in the purification or separation of the compounds of the invention, also form part of the invention.
  • Compounds of the formula (I) containing an amine function may also form N- oxides.
  • a reference herein to a compound of the formula (I) that contains an amine function also includes the N-oxide.
  • N-oxide may be oxidised to form an N-oxide.
  • N- oxides are the N-oxides of a tertiary amine or a nitrogen atom of a nitrogen- containing heterocycle.
  • N-Oxides can be formed by treatment of the corresponding amine with an oxidizing agent such as hydrogen peroxide or a per-acid (e.g. a peroxycarboxylic acid), see for example Advanced Organic Chemistry, by Jerry March, 4 th Edition, Wiley Interscience, pages. More particularly, N-oxides can be made by the procedure of L. W. Deady (Syn. Comm. 1977, 7, 509-514) in which the amine compound is reacted with m-chloroperoxybenzoic acid (MCPBA), for example, in an inert solvent such as dichloromethane.
  • MCPBA m-chloroperoxybenzoic acid
  • the imidazole group may take either of the following two tautomeric forms A and B.
  • the general formula (I) illustrates form A but the formula is to be taken as embracing both tautomeric forms.
  • the pyrazole ring may also exhibit tautomerism and can exist in the two tautomeric forms C and D below.
  • tautomeric forms include, for example, keto-, enol-, and enolate- forms, as in, for example, the following tautomeric pairs: keto/enol (illustrated below), imine/enamine, amide/imino alcohol, amidine/amidine, nitroso/oxime, thioketone/enethiol, and nitro/aci-nitro.
  • references to compounds of the formula (I) include all optical isomeric forms thereof (e.g. enantiomers, epimers and diastereoisomers), either as individual optical isomers, or mixtures (e.g. racemic mixtures) or two or more optical isomers, unless the context requires otherwise.
  • the group A can include one or more chiral centres.
  • the said carbon atom is typically chiral and hence the compound of the formula (I) will exist as a pair of enantiomers (or more than one pair of enantiomers where more than one chiral centre is present in the compound).
  • optical isomers may be characterised and identified by their optical activity (i.e. as + and - isomers, or d and / isomers) or they may be characterised in terms of their absolute stereochemistry using the "R and S" nomenclature developed by Cahn, Ingold and Prelog, see Advanced Organic Chemistry by Jerry March, 4 th Edition, John Wiley & Sons, New York, 1992, pages 109-114, and see also Cahn, Ingold & Prelog, Angew. Chem. Int. Ed. Engl, 1966, 5, 385-415.
  • Optical isomers can be separated by a number of techniques including chiral chromatography (chromatography on a chiral support) and such techniques are well known to the person skilled in the art.
  • optical isomers can be separated by forming diastereoisomeric salts with chiral acids such as (+)-tartaric acid, (-)- pyroglutamic acid, (-)-di-toluloyl-L-tartaric acid, (+) ⁇ mandelic acid, (-)-malic acid, and (-)-camphorsulphonic, separating the diastereoisomers by preferential crystallisation, and then dissociating the salts to give the individual enantiomer of the free base.
  • chiral acids such as (+)-tartaric acid, (-)- pyroglutamic acid, (-)-di-toluloyl-L-tartaric acid, (+) ⁇ mandelic acid, (-)-malic acid, and (-)-camphorsulphonic
  • compositions containing a compound of the formula (I) having one or more chiral centres wherein at least 55% (e.g. at least 60%, 65%, 70%, 75%, 80%, 85%, 90% or 95%) of the compound of the formula (I) is present as a single optical isomer (e.g.
  • 99% or more (e.g. substantially all) of the total amount of the compound of the formula (I) may be present as a single optical isomer (e.g. enantiomer or diastereoisomer).
  • the compounds of the invention include compounds with one or more isotopic substitutions, and a reference to a particular element includes within its scope all isotopes of the element.
  • a reference to hydrogen includes within its scope 1 H, 2 H (D), and 3 H (T).
  • references to carbon and oxygen include within their scope respectively 12 C, 13 C and 14 C and 16 O and 18 O.
  • the isotopes may be radioactive or non-radioactive.
  • the compounds contain no radioactive isotopes. Such compounds are preferred for therapeutic use.
  • the compound may contain one or more radioisotopes. Compounds containing such radioisotopes may be useful in a diagnostic context.
  • esters such as carboxylic acid esters and acyloxy esters of the compounds of formula (I) bearing a carboxylic acid group or a hydroxyl group are also embraced by Formula (I).
  • formula (I) Also encompassed by formula (I) are any polymorphic forms of the compounds, solvates (e.g. hydrates), complexes (e.g. inclusion complexes or clathrates with compounds such as cyclodextrins, or complexes with metals) of the compounds, and pro-drugs of the compounds.
  • solvates e.g. hydrates
  • complexes e.g. inclusion complexes or clathrates with compounds such as cyclodextrins, or complexes with metals
  • pro-drugs is meant for example any compound that is converted in vivo into a biologically active compound of the formula (I).
  • acyloxymethyl e.g., acyloxymethyl; acyloxyethyl; pivaloyloxymethyl; acetoxymethyl; 1-acetoxy ethyl;
  • prodrugs are activated enzymatically to yield the active compound, or a compound which, upon further chemical reaction, yields the active compound (for example, as in ADEPT, GDEPT, LIDEPT 5 etc.).
  • the prodrug may be a sugar derivative or other glycoside conjugate, or may be an amino acid ester derivative.
  • the compounds of the formulae (I) and sub-groups thereof are inhibitors of cyclin dependent kinases.
  • compounds of the invention have activity against CDKl, CDK2, CDK3, CDK4, CDK5, CDK6 and CDK7 kinases, and in particular cyclin dependent kinases selected from CDKl, CDK2, CDK3, CDK4, CDK5 and CDK6.
  • Preferred compounds are compounds that inhibit one or more CDK kinases selected from CDKl, CDK2, CDK4 and CDK5, for example CDKl and/or CDK2.
  • CDK4, CDK8 and/or CDK9 may be of interest.
  • Compounds of the invention also have activity against glycogen synthase kinase-3 (GSK-3).
  • Compounds of the invention also have activity against Aurora kinases.
  • Preferred compounds of the invention are those having IC 50 values of less than 0.1 ⁇ M.
  • Many of the compounds of the invention exhibit selectivity for the Aurora A kinase compared to CDKl and CDK2 and such compounds represent one preferred embodiment of the ivention.
  • many compounds of the invention have IC 50 values against Aurora A that are between a tenth and a hundredth of the IC 5 O against CDKl and CDK2.
  • the compounds of the invention will be useful in treating conditions such as viral infections, type II or non-insulin dependent diabetes mellitus, autoimmune diseases, head trauma, stroke, epilepsy, neurodegenerative diseases such as Alzheimer's, motor neurone disease, progressive supranuclear palsy, corticobasal degeneration and Pick's disease for example autoimmune diseases and neurodegenerative diseases.
  • One sub-group of disease states and conditions where it is envisaged that the compounds of the invention will be useful consists of viral infections, autoimmune diseases and neurodegenerative diseases.
  • CDKs play a role in the regulation of the cell cycle, apoptosis, transcription, differentiation and CNS function. Therefore, CDK inhibitors could be useful in the treatment of diseases in which there is a disorder of proliferation, apoptosis or differentiation such as cancer.
  • RB+ve tumours may be particularly sensitive to CDK inhibitors.
  • RB-ve tumours may also be sensitive to CDK inhibitors.
  • cancers which may be inhibited include, but are not limited to, a carcinoma, for example a carcinoma of the bladder, breast, colon (e.g. colorectal carcinomas such as colon adenocarcinoma and colon adenoma), kidney, epidermis, liver, lung, for example adenocarcinoma, small cell lung cancer and non-small cell lung carcinomas, oesophagus, gall bladder, ovary, pancreas e.g.
  • a carcinoma for example a carcinoma of the bladder, breast, colon (e.g. colorectal carcinomas such as colon adenocarcinoma and colon adenoma), kidney, epidermis, liver, lung, for example adenocarcinoma, small cell lung cancer and non-small cell lung carcinomas, oesophagus, gall bladder, ovary, pancreas e.g.
  • exocrine pancreatic carcinoma, stomach, cervix, thyroid, prostate, or skin for example squamous cell carcinoma
  • a hematopoietic tumour of lymphoid lineage for example leukemia, acute lymphocytic leukemia, B-cell lymphoma, T-cell lymphoma, Hodgkin's lymphoma, non-Hodgkin's lymphoma, hairy cell lymphoma, or Burkett's lymphoma
  • a hematopoietic tumour of myeloid lineage for example acute and chronic myelogenous leukemias, myelodysplastic syndrome, or promyelocytic leukemia
  • thyroid follicular cancer a tumour of mesenchymal origin, for example fibrosarcoma or habdomyosarcoma
  • a tumour of the central or peripheral nervous system for example astrocytoma, neuroblastoma, glioma or schwannoma
  • the cancers may be cancers which are sensitive to inhibition of any one or more cyclin dependent kinases selected from CDKl, CDK2, CDK3, CDK4, CDK5 and CDK6, for example, one or more CDK kinases selected from CDKl , CDK2, CDK4 and CDK5, e.g. CDKl and/or CDK2.
  • Whether or not a particular cancer is one which is sensitive to inhibition by a cyclin dependent kinase or an aurora kinase may be determined by means of a cell growth assay as set out in Example 57 and 58 below or by a method as set out in the section headed "Methods of Diagnosis".
  • CDKs are also known to play a role in apoptosis, proliferation, differentiation and transcription and therefore CDK inhibitors could also be useful in the treatment of the following diseases other than cancer; viral infections, for example herpes virus, pox virus, Epstein-Barr virus, Sindbis virus, adenovirus, HIV, HPV, HCV and HCMV; prevention of AIDS development in HIV-infected individuals; chronic inflammatory diseases, for example systemic lupus erythematosus, autoimmune mediated glomerulonephritis, rheumatoid arthritis, psoriasis, inflammatory bowel disease, and autoimmune diabetes mellitus; cardiovascular diseases for example cardiac hypertrophy, restenosis, atherosclerosis; neurodegenerative disorders, for example Alzheimer's disease, AIDS-related dementia, Parkinson's disease, amyotropic lateral sclerosis, retinitis pigmentosa, spinal muscular atropy and cerebellar degeneration; glomerulonephritis; myelody
  • cyclin-dependent kinase inhibitors can be used in combination with other anticancer agents.
  • the cyclin-dependent kinase inhibitor flavopiridol has been used with other anticancer agents in combination therapy.
  • the disease or condition comprising abnormal cell growth in one embodiment is a cancer.
  • cancers include human breast cancers (e.g. primary breast tumours, node-negative breast cancer, invasive duct adenocarcinomas of the breast, non- endometrioid breast cancers); and mantle cell lymphomas.
  • human breast cancers e.g. primary breast tumours, node-negative breast cancer, invasive duct adenocarcinomas of the breast, non- endometrioid breast cancers
  • mantle cell lymphomas e.g. primary breast tumours, node-negative breast cancer, invasive duct adenocarcinomas of the breast, non- endometrioid breast cancers
  • other cancers are colorectal and endometrial cancers.
  • Another sub-set of cancers includes breast cancer, ovarian cancer, colon cancer, prostate cancer, oesophageal cancer, squamous cancer and non-small cell lung carcinomas.
  • human breast cancers e.g. primary breast tumours, node-negative breast cancer, invasive duct adenocarcinomas of the breast, non-endometrioid breast cancers
  • ovarian cancers e.g. primary ovarian tumours
  • pancreatic cancers human bladder cancers
  • colorectal cancers e.g.
  • primary colorectal cancers gastric tumours; renal cancers; cervical cancers: neuroblastomas; melanomas; lymphomas; prostate cancers; leukemia; non-endometrioid endometrial carcinomas; gliomas; non-Hodgkin's lymphoma;
  • Cancers which may be particularly amenable to Aurora inhibitors include breast, bladder, colorectal, pancreatic, ovarian, non-Hodgkin's lymphoma, gliomas and nonendometrioid endometrial carcinomas.
  • a particular sub-set of cancers which may be particularly amenable to Aurora inhibitors consist of breast, ovarian, colon, liver, gastric and prostate cancers.
  • the activity of the compounds of the invention as inhibitors of cyclin dependent kinases, Aurora kinases and glycogen synthase kinase-3 can be measured using the assays set forth in the examples below and the level of activity exhibited by a given compound can be defined in terms of the IC 5 0 value.
  • Preferred compounds of the present invention are compounds having an IC 50 value of less than 1 micromolar, more preferably less than 0.1 micromolar.
  • hematological cancers in particular leukemia.
  • the compounds of formula (I) are used to treat hematological cancers, in particular leukemia.
  • leukemias are selected from Acute Myelogenous Leukemia (AML), chronic myelogenous leukaemia (CML), B-cell lymphoma (Mantle cell), and Acute Lymphoblastic Leukemia (ALL).
  • AML Acute Myelogenous Leukemia
  • CML chronic myelogenous leukaemia
  • B-cell lymphoma Mantle cell
  • ALL Acute Lymphoblastic Leukemia
  • the leukemias are selected from relapsed or refractory acute myelogenous leukemia, myelodysplastic syndrome, acute lymphocytic leukemia and chronic myelogenous leukemia.
  • cancers include human breast cancers (e.g. primary breast tumours, node-negative breast cancer, invasive duct adenocarcinomas of the breast, non- endometrioid breast cancers); and mantle cell lymphomas.
  • human breast cancers e.g. primary breast tumours, node-negative breast cancer, invasive duct adenocarcinomas of the breast, non- endometrioid breast cancers
  • mantle cell lymphomas e.g. primary breast tumours, node-negative breast cancer, invasive duct adenocarcinomas of the breast, non- endometrioid breast cancers
  • other cancers are colorectal and endometrial cancers.
  • Another sub-set of cancers includes hematopoietic tumours of lymphoid lineage, for example leukemia, chronic lymphocytic leukaemia, mantle cell lymphoma and B- cell lymphoma (such as diffuse large B cell lymphoma).
  • lymphoid lineage for example leukemia, chronic lymphocytic leukaemia, mantle cell lymphoma and B- cell lymphoma (such as diffuse large B cell lymphoma).
  • One particular cancer is chronic lymphocytic leukaemia.
  • Another particular cancer is mantle cell lymphoma.
  • Another particular cancer is diffuse large B cell lymphoma.
  • the compounds of the invention and in particular those compounds having aurora kinase inhibitory activity, will be particularly useful in the treatment or prevention of cancers of a type associated with or characterised by the presence of elevated levels of aurora kinases, for example the cancers referred to in this context in the introductory section of this application.
  • the starting material (X) is a protected form of 4-aminopyrazole-3- carboxylic acid methyl ester in which protecting group PG 1 can be, for example a tetrahydropyranyl group and protecting group PG 2 can be, for example, a tert- butoxycarbonyl (Boc) group.
  • protecting group PG 1 can be, for example a tetrahydropyranyl group
  • protecting group PG 2 can be, for example, a tert- butoxycarbonyl (Boc) group.
  • the protected 4-aminopyrazole-3-carboxylic acid methyl ester (X) is converted to the aldehyde (XII) by a sequence of steps involving, firstly, the reduction of the ester to the alcohol (XI) and, secondly, the oxidation of the alcohol (XI) to the aldehyde (XII).
  • Reduction of the ester (X) to the alcohol (XI) can be accomplished by a hydride reducing agent such as di-isobutylaluminium hydride (DIBAH).
  • DIBAH di-isobutylaluminium hydride
  • the reduction is typically carried out in a non-aqueous polar solvent such as tetrahydrofuran (THF) at a reduced temperature.
  • Oxidation of the alcohol (XI) to the aldehyde (XII) can be achieved using an oxidising agent such as manganese dioxide in a polar solvent such as acetone.
  • the aldehyde (XII) can then be used to introduce an imidazole group by means of a ring-forming reaction with ammonia and the ⁇ , ⁇ -dibromoketone (XIII) to give the pyrazolylimidazole intermediate (XIV).
  • the ring-forming reaction is typically carried out in a polar solvent such as an alcohol (e.g. methanol) at or near ambient temperature.
  • a polar solvent such as an alcohol (e.g. methanol) at or near ambient temperature.
  • deprotection can be carried out using a mineral acid such as hydrochloric acid.
  • the amine (XV) can be coupled with an acid of the formula R ⁇ -A-CO 2 H or a reactive derivative or activated form thereof to form an amide of the formula (I).
  • Standard amide formation conditions may be used to form the amide (I).
  • the coupling reaction between the carboxylic acid and the amine (XV) can be carried out in the presence of a reagent of the type commonly used in the formation of peptide linkages.
  • reagents include 1,3- dicyclohexylcarbodiimide (DCC) (Sheehan et al, J. Amer. Chem Soc. 1955, 77, 1067), l-ethyl-3-(3'-dimethylaminopropyl)-carbodiimide (EDC) (Sheehan et al, J. Org.
  • uronium-based coupling agents such as O-(7- azabenzotriazol-l-y ⁇ - ⁇ iV ⁇ N'-tetramethyluronium hexafluorophosphate (HATU) (L. A. Carpino, J. Amer. Chem. Soc, 1993, 115, 4397) and phosphonium-based coupling agents such as l-benzo-triazolyloxytris(pyrrolidino)phosphonium hexafluorophosphate (PyBOP) (Castro et al, Tetrahedron Letters, 1990, 3_1, 205).
  • HATU O-(7- azabenzotriazol-l-y ⁇ - ⁇ iV ⁇ N'-tetramethyluronium hexafluorophosphate
  • phosphonium-based coupling agents such as l-benzo-triazolyloxytris(pyrrolidino)phosphonium hexafluorophosphate (PyBOP
  • Carbodiimide-based coupling agents are advantageously used in combination with 1-hydroxyazobenzotriazole (HOAt) or 1-hydroxybenzotriazole (HOBt) (Konig et al, Chem. Ber., 103, 708, 2024-2034).
  • Preferred coupling reagents include EDC and DCC in combination with HOBt.
  • the coupling reaction is typically carried out in a non-aqueous, non-protic solvent such as acetonitrile, dioxan, dimethylsulphoxide (DMSO), dichloromethane, dimethylformamide (DMF) or N-methylpyrrolidine, or in an aqueous solvent optionally together with one or more miscible co-solvents.
  • a non-aqueous, non-protic solvent such as acetonitrile, dioxan, dimethylsulphoxide (DMSO), dichloromethane, dimethylformamide (DMF) or N-methylpyrrolidine
  • a non-interfering base for example a tertiary amine such as triethylamine or N,N- diisopropylethylamine.
  • a reactive derivative of the carboxylic acid e.g. an anhydride or acid chloride
  • Reaction with a reactive derivative such an anhydride is typically accomplished by stirring the amine and anhydride at room temperature in the presence of a base such as pyridine.
  • 4-nitro-3-pyrazole carboxylic acid is esterified by reaction with thionyl chloride to give the acid chloride intermediate followed by reaction with ethanol to form the ethyl ester (XX).
  • the esterification can be carried out by reacting the alcohol and carboxylic acid in the presence of an acidic catalyst, one example of which is thionyl chloride.
  • the reaction is typically carried out at room temperature using the esterifying alcohol (e.g. ethanol) as the solvent.
  • the pyrazole 1 -nitrogen atom is then protected by means of a suitable protecting group PG 1 , for example a tetrahydropyranyl (THP) group or an optionally substituted benzyl group such as a p ⁇ r ⁇ -methoxybenzyl group.
  • THP group can be introduced by reaction with 3,4-dihydropyran in the presence of an acid such as p-toluene sulphonic acid in a non-aqueous solvent such as chloroform.
  • the benzyl group e.g.
  • ;? ⁇ r ⁇ -methoxybenzyl group can be introduced by reacting the nitro ester (XX) with the appropriate benzyl halide in the presence of a base such as sodium carbonate or potassium carbonate.
  • a base such as sodium carbonate or potassium carbonate.
  • the reaction is typically carried out in a polar solvent such as acetonitrile at ambient temperature.
  • the N-protected nitro ester (XXI) can be reduced to the corresponding amino compound (XXII) according to standard methods.
  • the reduction may be effected, for example by catalytic hydrogenation in the presence of a catalyst such as palladium on carbon in a polar solvent such as ethanol or dimethylformamide at a temperature between about room temperature and about 55 0 C.
  • the 4-amino group in the compound (XXII) can then be protected by standard methods to give the protected compound of the formula (X).
  • a Boc protecting group can be introduced by reacting the amine (XXII) with tert-butyl carbonate in the presence of a non-interfering base such as triethylamine.
  • the aminopyrazole (XXII) (see Scheme 2 above) can be acylated by reaction with a compound of the formula R ⁇ -A-CO 2 H under the amide-forming conditions described above to give a compound of the formula (XXIII).
  • the ester (XXIII) can then be subjected to reduction (e.g. with DIBAH) to the corresponding alcohol and subsequent oxidation with an oxidising agent (e.g. MnO 2 ) to give an aldehyde of the formula (XXIV).
  • reduction e.g. with DIBAH
  • an oxidising agent e.g. MnO 2
  • the aldehyde (XXIV) can then be cyclised by reaction with ammonia and the ⁇ , ⁇ - dibromoketone (XVI) under the conditions described above to give the N-protected compound (XV) which can be deprotected under standard conditions to yield a compound of the formula (I).
  • the carboxylic acids of the formula R -A-CO 2 H used in the amide coupling step described in Scheme 1 above are commercially available or can be made by methods well known for the preparation of carboxylic acids.
  • an ether -OR
  • the aldehyde or ketone group is readily regenerated by hydrolysis using a large excess of water in the presence of acid.
  • An amine group may be protected, for example, as an amide (-NRC0-R) or a urethane (-NRC0-0R), for example, as: a methyl amide (-NHCO-CH 3 ); a benzyloxy amide (-NHCO-OCH 2 C 6 H 5 , -NH-Cbz); as a t-butoxy amide (-NHCO-OC(CH 3 ) 3 , -NH-Boc); a 2-biphenyl-2- ⁇ ropoxy amide (-NHCO-OC(CH 3 ) 2 C 6 H 4 C 6 H 5 , -NH- Bpoc), as a 9-fluorenylmethoxy amide (-NH-Fmoc), as a 6-nitroveratryloxy amide (-NH-Nvoc), as a 2-trimethylsilylethyloxy amide (-NH-Teoc), as a 2,2,2- trichloroethyloxy amide (-NH-Troc),
  • protecting groups for amines such as cyclic amines and heterocyclic N-H groups, include toluenesulfonyl (tosyl) and methanesulfonyl (mesyl) groups and benzyl groups such as a. para- methoxybenzyl (PMB) group, or a tetrahydropyranyl (THP) group.
  • tosyl toluenesulfonyl
  • methanesulfonyl methanesulfonyl
  • benzyl groups such as a. para- methoxybenzyl (PMB) group, or a tetrahydropyranyl (THP) group.
  • PMB para- methoxybenzyl
  • THP tetrahydropyranyl
  • a carboxylic acid group may be protected as an ester for example, as: an C 1-7 alkyl ester (e.g., a methyl ester; a t-butyl ester); a C 1-7 haloalkyl ester (e.g., a C 1-7 trihaloalkyl ester); a triC 1-7 alkylsilyl-C 1-7 alkyl ester; or a C 5-20 aryl-C 1-7 alkyl ester (e.g., a benzyl ester; a nitrobenzyl ester); or as an amide, for example, as a methyl amide.
  • an C 1-7 alkyl ester e.g., a methyl ester; a t-butyl ester
  • a C 1-7 haloalkyl ester e.g., a C 1-7 trihaloalkyl ester
  • the compounds may be isolated and purified by a number of methods well known to those skilled in the art and examples of such methods include chromatographic techniques such as column chromatography (e.g. flash chromatography) and HPLC.
  • Preparative LC-MS is a standard and effective method used for the purification of small organic molecules such as the compounds described herein.
  • the methods for the liquid chromatography (LC) and mass spectrometry (MS) can be varied to provide better separation of the crude materials and improved detection of the samples by MS.
  • Optimisation of the preparative gradient LC method will involve varying columns, volatile eluents and modifiers, and gradients. Methods are well known in the art for optimising preparative LC-MS methods and then using them to purify compounds.
  • the solvent should be readily removed from the purified product. This usually means that it has a relatively low boiling point and a person skilled in the art will know recrystallising solvents for a particular substance, or if that information is not available, test several solvents. To get a good yield of purified material, the minimum amount of hot solvent to dissolve all the impure material is used. In practice, 3-5% more solvent than necessary is used so the solution is not saturated. If the impure compound contains an impurity which is insoluble in the solvent it may then be removed by filtration and then allowing the solution to crystallise.
  • the impure compound may be removed by adding a small amount of decolorizing charcoal to the hot solution, filtering it and then allowing it to crystallise.
  • crystallisation spontaneously occurs upon cooling the solution. If it is not, crystallisation may be induced by cooling the solution below room temperature or by adding a single crystal of pure material (a seed crystal). Recrystallisation can also be carried out and/or the yield optimized by the use of an anti-solvent.
  • the compound is dissolved in a suitable solvent at elevated temperature, filtered and then an additional solvent in which the required compound has low solubility is added to aid crystallisation.
  • the crystals are then typically isolated using vacuum filtration, washed and then dried, for example, in an oven or via desiccation.
  • crystallisation from a vapor which includes an evaporation step for example in a sealed tube or an air stream
  • crystallisation from melt crystallisation Technology Handbook 2nd Edition, edited by A. Mersmann, 2001
  • the compound of formula (I) may subjected to recrystallisation (e.g. using 2-propanol or ethanol as the solvent) to increase the purity and to give a crystalline form.
  • the crystals obtained are analysed by an X-ray diffraction method such as X-ray powder diffraction (XRPD) or X-ray crystal diffraction.
  • XRPD X-ray powder diffraction
  • X-ray crystal diffraction X-ray crystal diffraction
  • the active compound While it is possible for the active compound to be administered alone, it is preferable to present it as a pharmaceutical composition (e.g. formulation) comprising at least one active compound of the invention together with one or more pharmaceutically acceptable carriers, adjuvants, excipients, diluents, fillers, buffers, stabilisers, preservatives, lubricants, or other materials well known to those skilled in the art and optionally other therapeutic or prophylactic agents; for example agents that reduce or alleviate some of the side effects associated with chemotherapy.
  • a pharmaceutical composition e.g. formulation
  • a pharmaceutical composition comprising at least one active compound of the invention together with one or more pharmaceutically acceptable carriers, adjuvants, excipients, diluents, fillers, buffers, stabilisers, preservatives, lubricants, or other materials well known to those skilled in the art and optionally other therapeutic or prophylactic agents; for example agents that reduce or alleviate some of the side effects associated with chemotherapy.
  • agents include anti-emetic agents and agents that prevent or decrease the duration of chemotherapy-associated neutropenia and prevent complications that arise from reduced levels of red blood cells or white blood cells, for example erythropoietin (EPO), granulocyte macrophage-colony stimulating factor (GM-CSF), and granulocyte-colony stimulating factor (G-CSF).
  • EPO erythropoietin
  • GM-CSF granulocyte macrophage-colony stimulating factor
  • G-CSF granulocyte-colony stimulating factor
  • the present invention further provides pharmaceutical compositions, as defined above, and methods of making a pharmaceutical composition comprising admixing at least one active compound, as defined above, together with one or more pharmaceutically acceptable carriers, excipients, buffers, adjuvants, stabilizers, or other materials, as described herein.
  • pharmaceutically acceptable refers to compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of a subject (e.g. human) without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
  • a subject e.g. human
  • Each carrier, excipient, etc. must also be “acceptable” in the sense of being compatible with the other ingredients of the formulation.
  • the invention provides compounds of the formula (I) and sub-groups thereof such as formulae (II) and (III) and sub-groups thereof as defined herein in the form of pharmaceutical compositions.
  • compositions can be in any form suitable for oral, parenteral, topical, intranasal, ophthalmic, otic, rectal, intra-vaginal, or transdermal administration.
  • compositions are intended for parenteral administration, they can be formulated for intravenous, intramuscular, intraperitoneal, subcutaneous administration or for direct delivery into a target organ or tissue by injection, infusion or other means of delivery.
  • the delivery can be by bolus injection, short term infusion or longer term infusion and can be via passive delivery or through the utilisation of a suitable infusion pump.
  • compositions adapted for parenteral administration include aqueous and non-aqueous sterile injection solutions which may contain anti-oxidants, buffers, bacteriostats and solutes which render the formulation isotonic with the blood of the intended recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents. Examples of these are described in R. G. Strickly, Solubilizing Excipients in oral and injectable formulations, Pharmaceutical Research, VoI 21(2) 2004, p 201-230.
  • compositions may contain co-solvents, organic solvent mixtures, cyclodextrin complexation agents, emulsifying agents (for forming and stabilizing emulsion formulations), liposome components for forming liposomes, gellable polymers for forming polymeric gels, lyophilisation protectants and combinations of agents for, inter alia, stabilising, the active ingredient in a soluble form and rendering the formulation isotonic with the blood of the intended recipient.
  • the formulations may be presented in unit-dose or multi-dose containers, for example sealed ampoules and vials, and may be stored in a freeze-dried (lyophilized) condition requiring only the addition of the sterile liquid carrier, for example water for injections, immediately prior to use.
  • a drug molecule that is ionizable can be solubilized to the desired concentration by pH adjustment if the drug's pKa is sufficiently away from the formulation pH value.
  • the acceptable range is pH 2-12 for intravenous and intramuscular administration, but subcutaneously the range is pH 2.7-9.0.
  • the solution pH is controlled by either the salt form of the drug, strong acids/bases such as hydrochloric acid or sodium hydroxide, or by solutions of buffers which include but are not limited to buffering solutions formed from glycine, citrate, acetate, maleate, succinate, histidine, phosphate, tris(hydroxymethyl)aminomethane (TRIS), or carbonate.
  • the combination of an aqueous solution and a water-soluble organic solvent/surfactant is often used in injectable formulations.
  • the water-soluble organic solvents and surfactants used in injectable formulations include but are not limited to propylene glycol, ethanol, polyethylene glycol 300, polyethylene glycol 400, glycerin, dimethylacetamide (DMA), N-methyl-2-pyrrolidone (NMP; Pharmasolve), dimethylsulphoxide (DMSO), Solutol HS 15, Cremophor EL, Cremophor RH 60, and polysorbate 80.
  • Such formulations can usually be, but are not always, diluted prior to injection.
  • Propylene glycol, PEG 300, ethanol, Cremophor EL, Cremophor RH 60, and polysorbate 80 are the entirely organic water-miscible solvents and surfactants used in commercially available injectable formulations and can be used in combinations with each other.
  • the resulting organic formulations are usually diluted at least 2-fold prior to rV bolus or FV infusion.
  • Liposomes are closed spherical vesicles composed of outer lipid bilayer membranes and an inner aqueous core and with an overall diameter of ⁇ 100 ⁇ m.
  • moderately hydrophobic drugs can be solubilized by liposomes if the drug becomes encapsulated or intercalated within the liposome.
  • Hydrophobic drugs can also be solubilized by liposomes if the drug molecule becomes an integral part of the lipid bilayer membrane, and in this case, the hydrophobic drag is dissolved in the lipid portion of the lipid bilayer.
  • a typical liposome formulation contains water with phospholipid at -5-20 mg/ml, an isotonicifier, a pH 5-8 buffer, and optionally cholesterol.
  • formulations may be presented in unit-dose or multi-dose containers, for example sealed ampoules and vials, and may be stored in a freeze-dried
  • the pharmaceutical formulation can be prepared by lyophilising a compound of Formula (I) or acid addition salt thereof.
  • Lyophilisation refers to the procedure of freeze-drying a composition. Freeze-drying and lyophilisation are therefore used herein as synonyms.
  • a typical process is to solubilise the compound and the resulting formulation is clarified, sterile filtered and aseptically transferred to containers appropriate for lyophilisation (e.g. vials). In the case of vials, they are partially stoppered with lyo-stoppers.
  • the formulation can be cooled to freezing and subjected to lyophilisation under standard conditions and then hermetically capped forming a stable, dry lyophile formulation.
  • the composition will typically have a low residual water content, e.g. less than 5% e.g. less than 1% by weight based on weight of the lyophile.
  • the lyophilisation formulation may contain other excipients for example, thickening agents, dispersing agents, buffers, antioxidants, preservatives, and tonicity adjusters.
  • Typical buffers include phosphate, acetate, citrate and glycine.
  • antioxidants include ascorbic acid, sodium bisulphite, sodium metabisulphite, monothioglycerol, thiourea, butylated hydroxytoluene, butylated hydroxyl anisole, and ethylenediamietetraacetic acid salts.
  • Preservatives may include benzoic acid and its salts, sorbic acid and its salts, alkyl esters of para- hydroxybenzoic acid, phenol, chlorobutanol, benzyl alcohol, thimerosal, benzalkonium chloride and cetylpyridinium chloride.
  • the buffers mentioned previously, as well as dextrose and sodium chloride, can be used for tonicity adjustment if necessary.
  • Bulking agents are generally used in lyophilisation technology for facilitating the process and/or providing bulk and/or mechanical integrity to the lyophilized cake.
  • Bulking agent means a freely water soluble, solid particulate diluent that when co- lyophilised with the compound or salt thereof, provides a physically stable lyophilized cake, a more optimal freeze-drying process and rapid and complete reconstitution.
  • the bulking agent may also be utilised to make the solution isotonic.
  • the water-soluble bulking agent can be any of the pharmaceutically acceptable inert solid materials typically used for lyophilisation.
  • Such bulking agents include, for example, sugars such as glucose, maltose, sucrose, and lactose; polyalcohols such as sorbitol or mannitol; amino acids such as glycine; polymers such as polyvinylpyrrolidine; and polysaccharides such as dextran.
  • the ratio of the weight of the bulking agent to the weight of active compound is typically within the range from about 1 to about 5, for example of about 1 to about 3, e.g. in the range of about 1 to 2.
  • dosage forms may be via filtration or by autoclaving of the vials and their contents at appropriate stages of the formulation process.
  • the supplied formulation may require further dilution or preparation before delivery for example dilution into suitable sterile infusion packs.
  • Extemporaneous injection solutions and suspensions may be prepared from sterile powders, granules and tablets.
  • the pharmaceutical composition is in a form suitable for i.v. administration, for example by injection or infusion.
  • compositions of the present invention for parenteral injection can also comprise pharmaceutically acceptable sterile aqueous or nonaqueous solutions, dispersions, suspensions or emulsions as well as sterile powders for reconstitution into sterile injectable solutions or dispersions just prior to use.
  • suitable aqueous and nonaqueous carriers, diluents, solvents or vehicles include water, ethanol, polyols (such as glycerol, propylene glycol, polyethylene glycol, and the like), carboxymethylcellulose and suitable mixtures thereof, vegetable oils (such as olive oil), and injectable organic esters such as ethyl oleate.
  • Proper fluidity can be maintained, for example, by the use of coating materials such as lecithin, by the maintenance of the required particle size in the case of dispersions, and by the use of surfactants.
  • compositions of the present invention may also contain adjuvants such as preservatives, wetting agents, emulsifying agents, and dispersing agents. Prevention of the action of microorganisms may be ensured by the inclusion of various antibacterial and antifungal agents, for example, paraben, chlorobutanol, phenol sorbic acid, and the like. It may also be desirable to include isotonic agents such as sugars, sodium chloride, and the like. Prolonged absorption of the injectable pharmaceutical form may be brought about by the inclusion of agents which delay absorption such as aluminum monostearate and gelatin.
  • a compound If a compound is not stable in aqueous media or has low solubility in aqueous media, it can be formulated as a concentrate in organic solvents. The concentrate can then be diluted to a lower concentration in an aqueous system, and can be sufficiently stable for the short period of time during dosing. Therefore in another aspect, there is provided a pharmaceutical composition comprising a non aqueous solution composed entirely of one or more organic solvents, which can be dosed as is or more commonly diluted with a suitable IV excipient (saline, dextrose; buffered or not buffered) before administration (Solubilizing excipients in oral and injectable formulations, Pharmaceutical Research, 21(2), 2004, p201-230).
  • a suitable IV excipient saline, dextrose; buffered or not buffered
  • solvents and surfactants are propylene glycol, PEG300, PEG400, ethanol, dimethylacetamide (DMA), N-methyl-2-pyrrolidone (NMP, Pharmasolve), Glycerin, Cremophor EL, Cremophor RH 60 and polysorbate.
  • Particular non aqueous solutions are composed of 70-80% propylene glycol, and 20-30% ethanol.
  • One particular non aqueous solution is composed of 70% propylene glycol, and 30% ethanol.
  • these solvents are used in combination and usually diluted at least 2-fold before IV bolus or IV infusion.
  • the typical amounts for bolus IV formulations are -50% for Glycerin, propylene glycol, PEG300, PEG400, and -20% for ethanol.
  • the typical amounts for IV infusion formulations are -15% for Glycerin, 3% for DMA 5 and ⁇ 10% for propylene glycol, PEG300, PEG400 and ethanol.
  • the pharmaceutical composition is in a form suitable for i.v. administration, for example by injection or infusion.
  • the solution can be dosed as is, or can be injected into an infusion bag (containing a pharmaceutically acceptable excipient, such as 0.9% saline or 5% dextrose), before administration.
  • the pharmaceutical composition is in a form suitable for sub-cutaneous (s.c.) administration.
  • Pharmaceutical dosage forms suitable for oral administration include tablets, capsules, caplets, pills, lozenges, syrups, solutions, powders, granules, elixirs and suspensions, sublingual tablets, wafers or patches and buccal patches.
  • compositions containing compounds of the formula (I) can be formulated in accordance with known techniques, see for example, Remington's Pharmaceutical Sciences, Mack Publishing Company, Easton, PA, USA.
  • tablet compositions can contain a unit dosage of active compound together with an inert diluent or carrier such as a sugar or sugar alcohol, eg; lactose, sucrose, sorbitol or mannitol; and/or a non-sugar derived diluent such as sodium carbonate, calcium phosphate, calcium carbonate, or a cellulose or derivative thereof such as methyl cellulose, ethyl cellulose, hydroxypropyl methyl cellulose, and starches such as corn starch. Tablets may also contain such standard ingredients as binding and granulating agents such as polyvinylpyrrolidone, disintegrants (e.g.
  • swellable crosslinked polymers such as crosslinked carboxymethylcellulose
  • lubricating agents e.g. stearates
  • preservatives e.g. parabens
  • antioxidants e.g. BHT
  • buffering agents for example phosphate or citrate buffers
  • effervescent agents such as citrate/bicarbonate mixtures.
  • Capsule formulations may be of the hard gelatin or soft gelatin variety and can contain the active component in solid, semi-solid, or liquid form.
  • Gelatin capsules can be formed from animal gelatin or synthetic or plant derived equivalents thereof.
  • the solid dosage forms can be coated or un-coated, but typically have a coating, for example a protective film coating (e.g. a wax or varnish) or a release controlling coating.
  • a protective film coating e.g. a wax or varnish
  • the coating e.g. a Eudragit TM type polymer
  • the coating can be designed to release the active component at a desired location within the gastro-intestinal tract.
  • the coating can be selected so as to degrade under certain pH conditions within the gastrointestinal tract, thereby selectively release the compound in the stomach or in the ileum or duodenum.
  • the drug can be presented in a solid matrix comprising a release controlling agent, for example a release delaying agent which may be adapted to selectively release the compound under conditions of varying acidity or alkalinity in the gastrointestinal tract.
  • a release controlling agent for example a release delaying agent which may be adapted to selectively release the compound under conditions of varying acidity or alkalinity in the gastrointestinal tract.
  • the matrix material or release retarding coating can take the form of an erodible polymer (e.g. a maleic anhydride polymer) which is substantially continuously eroded as the dosage form passes through the gastrointestinal tract.
  • the active compound can be formulated in a delivery system that provides osmotic control of the release of the compound. Osmotic release and other delayed release or sustained release formulations may be prepared in accordance with methods well known to those skilled in the art.
  • compositions comprise from approximately 1% to approximately 95%, preferably from approximately 20% to approximately 90%, active ingredient.
  • Pharmaceutical compositions according to the invention may be, for example, in unit dose form, such as in the form of ampoules, vials, suppositories, dragees, tablets or capsules.
  • Pharmaceutical compositions for oral administration can be obtained by combining the active ingredient with solid carriers, if desired granulating a resulting mixture, and processing the mixture, if desired or necessary, after the addition of appropriate excipients, into tablets, dragee cores or capsules. It is also possible for them to be incorporated into plastics carriers that allow the active ingredients to diffuse or be released in measured amounts.
  • compositions for topical use include ointments, creams, sprays, patches, gels, liquid drops and inserts (for example intraocular inserts). Such compositions can be formulated in accordance with known methods.
  • compositions for parenteral administration are typically presented as sterile aqueous or oily solutions or fine suspensions, or may be provided in finely divided sterile powder form for making up extemporaneously with sterile water for injection.
  • formulations for rectal or intra- vaginal administration include pessaries and suppositories which may be, for example, formed from a shaped moldable or waxy material containing the active compound.
  • compositions for administration by inhalation may take the form of inhalable powder compositions or liquid or powder sprays, and can be administrated in standard form using powder inhaler devices or aerosol dispensing devices. Such devices are well known.
  • the powdered formulations typically comprise the active compound together with an inert solid powdered diluent such as lactose.
  • the pharmaceutical formulations may be presented to a patient in "patient packs" containing an entire course of treatment in a single package, usually a blister pack.
  • Patient packs have an advantage over traditional prescriptions, where a pharmacist divides a patient's supply of a pharmaceutical from a bulk supply, in that the patient always has access to the package insert contained in the patient pack, normally missing in patient prescriptions.
  • the inclusion of a package insert has been shown to improve patient compliance with the physician's instructions.
  • the compounds of the inventions will generally be presented in unit dosage form and, as such, will typically contain sufficient compound to provide a desired level of biological activity.
  • a formulation intended for oral administration may contain from 0.1 milligrams to 2 grams of active ingredient, e.g. from 1 nanogram to 2 milligrams of active ingredient.
  • particular subranges of compound are 0.1 milligrams to 2 grams of active ingredient more usually from 10 milligrams to 1 gram, for example, 50 milligrams to 500 milligrams or 1 microgram to 20 milligrams (for example 1 microgram to 10 milligrams, e.g. 0.1 milligrams to 2 milligrams of active ingredient).
  • a unit dosage form may contain from 1 milligram to 2 grams, more typically 10 milligrams to 1 gram, for example 50 milligrams to 1 gram, e.g. 100 milligrams to 1 gram, of active compound.
  • the active compound will be administered to a patient in need thereof (for example a human or animal patient) in an amount sufficient to achieve the desired therapeutic effect.
  • the compounds of the formula (I) as defined herein will be useful in the prophylaxis or treatment of a range of disease states or conditions mediated by cyclin dependent kinases, glycogen synthase kinase-3 and Aurora kinases. Examples of such disease states and conditions are set out above.
  • the compounds are generally administered to a subject in need of such administration, for example a human or animal patient, preferably a human.
  • the compounds will typically be administered in amounts that are therapeutically or prophylactically useful and which generally are non-toxic. However, in certain situations (for example in the case of life threatening diseases), the benefits of administering a compound of the formula (I) may outweigh the disadvantages of any toxic effects or side effects, in which case it may be considered desirable to administer compounds in amounts that are associated with a degree of toxicity.
  • the compounds may be administered over a prolonged term to maintain beneficial therapeutic effects or may be administered for a short period only. Alternatively they may be administered in a pulsatile or continuous manner.
  • a typical daily dose of the compound can be in the range from 100 picograms to 100 milligrams per kilogram of body weight, more typically 5 nanograms to 25 milligrams per kilogram of bodyweight, and more usually 10 nanograms to 15 milligrams per kilogram (e.g. 10 nanograms to 10 milligrams such as 1 micrograms to 10 milligrams) per kilogram of bodyweight although higher or lower doses may be administered where required.
  • the quantity of compound administered and the type of composition used will be commensurate with the nature of the disease or physiological condition being treated and will be at the discretion of the physician.
  • the compounds of formula (I) as defined herein can be administered as the sole therapeutic agent or they can be administered in combination therapy with one of more other compounds for treatment of a particular disease state, for example a neoplastic disease such as a cancer as hereinbefore defined.
  • a neoplastic disease such as a cancer as hereinbefore defined.
  • other therapeutic agents or therapies that may be administered or used together (whether concurrently or at different time intervals) with the compounds of the invention include but are not limited to topoisomerase inhibitors, alkylating agents, antimetabolites, DNA binders, microtubule inhibitors (tubulin targeting agents), particular examples being cisplatin, cyclophosphamide, doxorubicin, irinotecan, fiudarabine, 5FU, taxanes, mitomycin C and radiotherapy.
  • therapeutic agents that may be administered together (whether concurrently or at different time intervals) with the compounds of the formula (I) as defined herein include monoclonal antibodies and signal transduction inhibitors.
  • the two or more treatments may be given in individually varying dose schedules and via different routes.
  • the compound of the formula (I) is administered in combination therapy with one, two, three, four or more other therapeutic agents (preferably one or two, more preferably one)
  • the compounds can be administered simultaneously or sequentially.
  • they can be administered at closely spaced intervals (for example over a period of 5-10 minutes) or at longer intervals (for example 1, 2, 3, 4 or more hours apart, or even longer periods apart where required), the precise dosage regimen being commensurate with the properties of the therapeutic agent(s).
  • the compounds of the invention may also be administered in conjunction with non- chemotherapeutic treatments such as radiotherapy, photodynamic therapy, gene therapy; surgery and controlled diets.
  • non- chemotherapeutic treatments such as radiotherapy, photodynamic therapy, gene therapy; surgery and controlled diets.
  • the compound of the formula (I) and one, two, three, four or more other therapeutic agents can be, for example, formulated together in a dosage form containing two, three, four or more therapeutic agents.
  • the individual therapeutic agents may be formulated separately and presented together in the form of a kit, optionally with instructions for their use.
  • a patient Prior to administration of a compound of the formula (I), a patient may be screened to determine whether a disease or condition from which the patient is or may be suffering is one which would be susceptible to treatment with a compound having activity against Aurora and/or cyclin dependent kinases.
  • a biological sample taken from a patient may be analysed to determine whether a condition or disease, such as cancer, that the patient is or may be suffering from is one which is characterised by a genetic abnormality or abnormal protein expression which leads to over-activation of CDKs or to sensitisation of a pathway to normal CDK activity.
  • a condition or disease such as cancer
  • Examples of such abnormalities that result in activation or sensitisation of the CDK2 signal include up-regulation of cyclin E, (Harwell RM, Mull BB, Porter DC, Keyomarsi K.; J Biol Chem.
  • Tumours with mutants of CDC4 or up-regulation, in particular over-expression, of cyclin E or loss of p21 or p27 may be particularly sensitive to CDK inhibitors.
  • a biological sample taken from a patient may be analysed to determine whether a condition or disease, such as cancer, that the patient is or may be suffering from is one which is characterised by upregulation of Aurora kinase and thus may be particularly to Aurora inhibitors.
  • up-regulation includes elevated expression or over-expression, including gene amplification (i.e. multiple gene copies) and increased expression by a transcriptional effect, and hyperactivity and activation, including activation by mutations.
  • the patient may be subjected to a diagnostic test to detect a marker characteristic of over-expression, up-regulation or activation of Aurora kinase or the patient may be subjected to a diagnostic test to detect a marker characteristic of up-regulation of cyclin E, or loss of p21 or p27, or presence of CDC4 variants.
  • diagnosis includes screening.
  • marker we include genetic markers including, for example, the measurement of DNA composition to identify mutations of Aurora or CDC4.
  • the term marker also includes markers which are characteristic of up regulation of Aurora or cyclin E, including enzyme activity, enzyme levels, enzyme state (e.g. phosphorylated or not) and mRNA levels of the aforementioned proteins.
  • Tumours with upregulation of cyclin E, or loss of p21 or p27 may be particularly sensitive to CDK inhibitors. Tumours may preferentially be screened for upregulation of cyclin E, or loss of p21 or p27 prior to treatment.
  • the patient may be subjected to a diagnostic- test to detect a marker characteristic of upregulation of cyclin E, or loss of p21 or p27.
  • the diagnostic tests are typically conducted on a biological sample selected from tumour biopsy samples, blood samples (isolation and enrichment of shed tumour cells), stool biopsies, sputum, chromosome analysis, pleural fluid, peritoneal fluid, or urine.
  • CDC4 also known as Fbw7 or Archipelago
  • Identification of individual carrying a mutation in CDC4 may mean that the patient would be particularly suitable for treatment with a CDK inhibitor.
  • Tumours may preferentially be screened for presence of a CDC4 variant prior to treatment. The screening process will typically involve direct sequencing, oligonucleotide microarray analysis, or a mutant specific antibody.
  • Tumours with activating mutants of Aurora or up-regulation of Aurora including any of the isoforms thereof may be particularly sensitive to Aurora inhibitors. Tumours may preferentially be screened for up-regulation of Aurora or for Aurora possessing the Ile31 variant prior to treatment (Ewart-Toland et al., Nat Genet. 2003 Aug;34(4):403-12). Ewart-Toland et al identified a common genetic variant in STKl 5 (resulting in the amino acid substitution F31I) that is preferentially amplified and associated with the degree of aneuploidy in human colon tumors. These results are consistent with an important role for the Ile31 variant of STKl 5 in human cancer susceptibility. In particular, this polymorphism in Aurora A has been suggested to be a genetic modifier fir developing breast carcinoma (Sun et al, Carcinogenesis, 2004, 25(11), 2225-2230).
  • the aurora A gene maps to the chromosome 20ql 3 region that is frequently amplified in many cancers e.g. breast, bladder, colon, ovarian, pancreatic. Patients ⁇ with a tumour that has this gene amplification might be particularly sensitive to treatments targeting aurora kinase inhibition
  • Screening methods could include, but are not limited to, standard methods such as reverse-transcriptase polymerase chain reaction (RT-PCR) or in- situ hybridisation.
  • RT-PCR reverse-transcriptase polymerase chain reaction
  • telomere amplification is assessed by creating a cDNA copy of the mRNA followed by amplification of the cDNA by PCR.
  • Methods of PCR amplification, the selection of primers, and conditions for amplification, are known to a person skilled in the art.
  • Nucleic acid manipulations and PCR are carried out by standard methods, as described for example in Ausubel, F.M. et al., eds. Current Protocols in Molecular Biology, 2004, John Wiley & Sons Inc., or Innis, M.A. et-al., eds. PCR Protocols: a guide to methods and applications, 1990, Academic Press, San Diego.
  • in situ hybridization comprises the following major steps: (1) fixation of tissue to be analyzed; (2) prehybridization treatment of the sample to increase accessibility of target nucleic acid, and to reduce nonspecific binding; (3) hybridization of the mixture of nucleic acids to the nucleic acid in the biological structure or tissue; (4) post-hybridization washes to remove nucleic acid fragments not bound in the hybridization, and (5) detection of the hybridized nucleic acid fragments.
  • the probes used in such applications are typically labeled, for example, with radioisotopes or fluorescent reporters.
  • Preferred probes are sufficiently long, for example, from about 50, 100, or 200 nucleotides to about 1000 or more nucleotides, to enable specific hybridization with the target nucleic acid(s) under stringent conditions.
  • Standard methods for carrying out FISH are described in Ausubel, F.M. et al, eds. Current Protocols in Molecular Biology, 2004, John Wiley & Sons Inc and Fluorescence In Situ Hybridization: Technical Overview by John M. S. Bartlett in Molecular Diagnosis of Cancer, Methods and Protocols, 2nd ed.; ISBN: 1-59259-760-2; March 2004, pps. 077-088; Series: Methods in Molecular Medicine.
  • the protein products expressed from the mRNAs may be assayed by immunohistochemistry of tumour samples, solid phase immunoassay with microtiter plates, Western blotting, 2-dimensional SDS-polyacrylamide gel electrophoresis, ELISA, flow cytometry and other methods known in the art for detection of specific proteins. Detection methods would include the use of site specific antibodies. The skilled person will recognize that all such well-known techniques for detection of upregulation of cyclin E, or loss of p21 or p27, or detection of CDC4 variants, Aurora up-regulation and mutants of Aurora could be applicable in the present case.
  • Tumours with mutants of CDC4 or up-regulation, in particular over-expression, of cyclin E or loss of p21 or p27 may be particularly sensitive to CDK inhibitors. Tumours may preferentially be screened for up-regulation, in particular over- expression, of cyclin E (Harwell RM, Mull BB, Porter DC, Keyomarsi K.; J Biol Chem. 2004 Mar 26;279(13): 12695-705) or loss of p21 or p27 or for CDC4 variants prior to treatment (Rajagopalan H, Jallepalli PV, Rago C, Velculescu VE, Kinzler KW, Vogelstein B, Lengauer C; Nature. 2004 Mar 4;428(6978):77-81).
  • MCL mantle cell lymphoma
  • MCL is a distinct clinicopathologic entity of non-Hodgkin's lymphoma, characterized by proliferation of small to medium-sized lymphocytes with co-expression of CD5 and CD20, an aggressive and incurable clinical course, and frequent t(l I;14)(ql3;q32) translocation.
  • Over-expression of cyclin Dl mRNA, found in mantle cell lymphoma (MCL) is a critical diagnostic marker. Yatabe et al (Blood.
  • the invention provides the use of the compounds of the formula (I) as hereinbefore defined as antifungal agents.
  • the compounds of the formula (I) may be used in animal medicine (for example in the treatment of mammals such as humans), or in the treatment of plants (e.g. in agriculture and horticulture),. or as general antifungal agents, for example as preservatives and disinfectants.
  • the invention provides a compound of the formula (I) as hereinbefore defined for use in the prophylaxis or treatment of a fungal infection in a mammal such as a human.
  • compounds of the invention may be administered to human patients suffering from, or at risk of infection by, topical fungal infections caused by among other organisms, species of Candida, Trichophyton, Microsporum or Epidermophyton, or in mucosal infections caused by Candida albicans (e.g. thrush and vaginal candidiasis).
  • the compounds of the invention can also be administered for the treatment or prophylaxis of systemic fungal infections caused by, for example, Candida albicans, Cryptococcus neoformans, Aspergillus flavus, Aspergillus fumigatus, Coccidiodies, Paracoccidioides, Histoplasma or Blastomyces.
  • the invention provides an antifungal composition for agricultural (including horticultural) use, comprising a compound of the formula (I) together with an agriculturally acceptable diluent or carrier.
  • the invention further provides a method of treating an animal (including a mammal such as a human), plant or seed having a fungal infection, which comprises treating said animal, plant or seed, or the locus of said plant or seed, with an effective amount of a compound of the formula (I).
  • the invention also provides a method of treating a fungal infection in a plant or seed which comprises treating the plant or seed with an antifungally effective amount of a fungicidal composition containing a compound of the formula (I) as hereinbefore defined.
  • Differential screening assays may be used to select for those compounds of the present invention with specificity for non-human CDK enzymes.
  • Compounds which act specifically on the CDK enzymes of eukaryotic pathogens can be used as antifungal or anti-parasitic agents.
  • Inhibitors of the Candida CDK kinase, CKSI can be used in the treatment of candidiasis.
  • Antifungal agents can be used against infections of the type hereinbefore defined, or opportunistic infections that commonly occur in debilitated and immunosuppressed patients such as patients with leukemias and lymphomas, people who are receiving immunosuppressive therapy, and patients with predisposing conditions such as diabetes mellitus or AIDS, as well as for non-immunosuppressed patients.
  • Assays described in the art can be used to screen for agents which may be useful for inhibiting at least one fungus implicated in mycosis such as candidiasis, aspergillosis, mucormycosis, blastomycosis, geotrichosis, cryptococcosis, chromoblastomycosis, coccidiodomycosis, conidiosporosis, histoplasmosis, maduromycosis, rhinosporidosis, nocaidiosis, para-actinomycosis, penicilliosis, monoliasis, or sporotrichosis.
  • the differential screening assays can be used to identify anti-fungal agents which may have therapeutic value in the treatment of aspergillosis by making use of the CDK genes cloned from yeast such as
  • the CDK assay can be derived from yeast such as Rhizopus arrhizus, Rhizopus oryzae, Absidia corymbifera, Absidia ramosa, or Mucorpusillus. Sources of other CDK enzymes include the pathogen Pneumocystis carinii.
  • in vitro evaluation of the antifungal activity of the compounds can be performed by determining the minimum inhibitory concentration (M.I.C.) which is the lowest concentration of the test compounds, in a suitable medium, at which growth of the particular microorganism fails to occur.
  • M.I.C. minimum inhibitory concentration
  • a series of agar plates, each having the test compound incorporated at a particular concentration is inoculated with a standard culture of, for example, Candida albicans and each plate is then incubated for an appropriate period at 37 °C. The plates are then examined for the presence or absence of growth of the fungus and the appropriate M.I.C. value is noted.
  • a turbidity assay in liquid cultures can be performed and a protocol outlining an example of this assay can be found in Example 51.
  • the in vivo evaluation of the compounds can be carried out at a series of dose levels by intraperitoneal or intravenous injection or by oral administration, to mice that have been inoculated with a fungus, e.g. a strain of Candida albicans or Aspergillus flavus.
  • a fungus e.g. a strain of Candida albicans or Aspergillus flavus.
  • the activity of the compounds can be assessed by monitoring the growth of the fungal infection in groups of treated and untreated mice (by histology or by retrieving fungi from the infection). The activity may be measured in terms of the dose level at which the compound provides 50% protection against the lethal effect of the infection (PD 5 o).
  • the compounds of the formula (I) can be administered alone or in admixture with a pharmaceutical carrier selected in accordance with the intended route of administration and standard pharmaceutical practice.
  • a pharmaceutical carrier selected in accordance with the intended route of administration and standard pharmaceutical practice.
  • they may be administered orally, parenterally, intravenously, intramuscularly or subcutaneously by means of the formulations described above in the section headed "Pharmaceutical Formulations".
  • the daily dosage level of the antifungal compounds of the formula (I) can be from 0.01 to 10 mg/kg (in divided doses), depending on inter alia the potency of the compounds when administered by either the oral or parenteral route.
  • Tablets or capsules of the compounds may contain, for example, from 5 mg to 0.5 g of active compound for administration singly or two or more at a time as appropriate. The physician in any event will determine the actual dosage (effective amount) which will be most suitable for an individual patient and it will vary with the age, weight and response of the particular patient.
  • the antifungal compounds of formula (I) can be administered in the form of a suppository or pessary, or they may be applied topically in the form of a lotion, solution, cream, ointment or dusting powder.
  • they can be incorporated into a cream consisting of an aqueous emulsion of polyethylene glycols or liquid paraffin; or they can be incorporated, at a concentration between 1 and 10%, into an ointment consisting of a white wax or white soft paraffin base together with such stabilizers and preservatives as may be required.
  • anti-fungal agents developed with such differential screening assays can be used, for example, as preservatives in foodstuff, feed supplement for promoting weight gain in livestock, or in disinfectant formulations for treatment of non-living matter, e.g., for decontaminating hospital equipment and rooms.
  • side by side comparison of inhibition of a mammalian CDK and an insect CDK such as the Drosophilia CDK5 gene (Heimlich et al. (1994) FEBS Lett 356:317-21)
  • the present invention expressly contemplates the use and formulation of the compounds of the invention in insecticides, such as for use in management of insects like the fruit fly.
  • certain of the subject CDK inhibitors can be selected on the basis of inhibitory specificity for plant CDK's relative to the mammalian enzyme.
  • a plant CDK can be disposed in a differential screen with one or more of the human enzymes to select those compounds of greatest selectivity for inhibiting the plant enzyme.
  • the present invention specifically contemplates formulations of the subject CDK inhibitors for agricultural applications, such as in the form of a defoliant or the like.
  • the compounds of the invention may be used in the form of a composition formulated as appropriate to the particular use and intended purpose.
  • the compounds may be applied in the form of dusting powders, or granules, seed dressings, aqueous solutions, dispersions or emulsions, dips, sprays, aerosols or smokes.
  • Compositions may also be supplied in the form of dispersible powders, granules or grains, or concentrates for dilution prior to use.
  • Such compositions may contain such conventional carriers, diluents or adjuvants as are known and acceptable in agriculture and horticulture and they can be manufactured in accordance with conventional procedures.
  • compositions may also incorporate other active ingredients, for example, compounds having herbicidal or insecticidal activity or a further fungicide.
  • the compounds and compositions can be applied in a number of ways, for example they can be applied directly to the plant foliage, stems, branches, seeds or roots or to the soil or other growing medium, and they may be used not only to eradicate disease, but also prophylactically to protect the plants or seeds from attack.
  • the compositions may contain from 0.01 to 1 wt.% of the active ingredient. For field use, likely application rates of the active ingredient may be from 50 to 5000 g/hectare.
  • the invention also contemplates the use of the compounds of the formula (I) in the control of wood decaying fungi and in the treatment of soil where plants grow, paddy fields for seedlings, or water for perfusion. Also contemplated by the invention is the use of the compounds of the formula (I) to protect stored grain and other non-plant loci from fungal infestation.
  • DMAW90 Solvent mixture DCM: MeOH, AcOH, H 2 O (90: 18:3 :2)
  • DMAW120 Solvent mixture DCM: MeOH, AcOH, H 2 O (120:18:3:2)
  • DMAW240 Solvent mixture DCM: MeOH, AcOH, H 2 O (240:20:3 :2)
  • the compounds prepared were characterised by liquid chromatography and mass spectroscopy using the systems and operating conditions set out below. Where atoms with different isotopes are present, and a single mass quoted, the mass quoted for the compound is the monoisotopic mass (i.e. 35 Cl; 79 Br etc.).
  • the monoisotopic mass i.e. 35 Cl; 79 Br etc.
  • Preparative LC-MS is a standard and effective method used for the purification of small organic molecules such as the compounds described herein.
  • the methods for the liquid chromatography (LC) and mass spectrometry (MS) can be varied to provide better separation of the crude materials and improved detection of the samples by MS.
  • Optimisation of the preparative gradient LC method will involve varying columns, volatile eluents and modifiers, and gradients. Methods are well known in the art for optimising preparative LC-MS methods and then using them to purify compounds. Such methods are described in Rosentreter U, Huber U.;
  • CFO column fluidic organiser
  • RMA Waters reagent manager
  • UV detector 1100 series "MWD” Multi Wavelength Detector
  • Nebuliser Pressure 50 psig
  • Solvent A H 2 O + 10 mM NH 4 HCO 3 + NH4OH, pH-9.2
  • Solvent B CH 3 CN 3. Make up solvent:
  • Example IF By following the procedure set out in Example IF, using the appropriate starting materials, the compounds of Examples 2 to 13 set out in the table below were prepared.
  • Activated CDK2/CyclinA (Brown et al, Nat. Cell Biol, 1, ⁇ p438-443 5 1999; Lowe, E.D., et al Biochemistry, 41, ppl5625-15634, 2002) is diluted to 125pM in 2.5X strength assay buffer (5OmM MOPS pH 7.2, 62.5 mM ⁇ -glycerophosphate, 12.5mM EDTA, 37.5mM MgCl 2 , 112.5 mM ATP, 2.5 mM DTT, 2.5 mM sodium orthovanadate, 0.25 mg/ml bovine serum albumin), and 10 ⁇ l mixed with 10 ⁇ l of histone substrate mix (60 ⁇ l bovine histone Hl (Upstate Biotechnology, 5 mg/ml), 940 ⁇ l H 2 O, 35 ⁇ Ci ⁇ 33 P-ATP) and added to 96 well plates along with 5 ⁇ l of various dilutions of the test compound in DMSO (up to 2.5%).
  • ⁇ 33 P-ATP which remains unincorporated into the histone Hl is separated from phosphorylated histone Hl on a Millipore MAPH filter plate.
  • the wells of the MAPH plate are wetted with 0.5% orthophosphoric acid, and then the results of the reaction are filtered with a Millipore vacuum filtration unit through the wells. Following filtration, the residue is washed twice with 200 ⁇ l of 0.5% orthophosphoric acid. Once the filters have dried, 20 ⁇ l of Microscint 20 scintillant is added, and then counted on a Packard Topcount for 30 seconds.
  • the % inhibition of the CDK2 activity is calculated and plotted in order to determine the concentration of test compound required to inhibit 50% of the CDK2 activity (IC 50 ).
  • the compounds of Examples 1 to 13 all have IC 50 values no greater than 1 ⁇ M.
  • the CDKl/CyclinB assay is identical to the CDK2/CyclinA above except that CDKl/CyclinB (Upstate Discovery) is used and the enzyme is diluted to 6.25 nM.
  • the compounds of Examples 2 to 5, 7, 10 and 12 all have IC 50 values of less than 1 ⁇ M
  • the compounds of Examples 1, 6, 8 and 13 have IC 50 values of less than 10 ⁇ M
  • the compounds of Examples 9 and 11 exhibit greater than 35% inhibition at a concentration of 1 ⁇ M.
  • AuroraA Upstate Discovery
  • GSK3- ⁇ Upstate Discovery
  • 1OnM 1OnM
  • 7.5nM 25mM MOPS, pH 7.00, 25mg/ml BSA, 0.0025% Brij- 35, 1.25% glycerol, 0.5mM EDTA, 25mM MgCl 2 , 0.025% ⁇ -mercaptoethanol, 37.5mM ATP and and 10 ⁇ l mixed with 10 ⁇ l of substrate mix.
  • the substrate mix for Aurora is 500 ⁇ M Kemptide peptide (LRRASLG 5 Upstate Discovery) in ImI of water with 35 ⁇ Ci ⁇ 33 P-ATP.
  • the substrate mix for GSK3- ⁇ is 12.5 ⁇ M phospho- glycogen synthase peptide-2 (Upstate Discovery) in ImI of water with 35 ⁇ Ci ⁇ 33 P- ATP. Enzyme and substrate are added to 96 well plates along with 5 ⁇ l of various dilutions of the test compound in DMSO (up to 2.5%). The reaction is allowed to proceed for 30 minutes (Aurora) or 3 hours (GSK3- ⁇ ) before being stopped with an excess of ortho-phosphoric acid (5 ⁇ l at 2%). The filtration procedure is as for Activated CDK2/CyclinA assay above.
  • Kinases are diluted to a 1Ox working stock in 20 mM MOPS pH 7.0, ImM EDTA, 0.1% ⁇ -mercaptoethanol, 0.01% Brij-35, 5% glycerol, lmg/ml BSA.
  • One unit equals the incorporation of lnmol of phosphate per minute into 0.1 mg/ml histone Hl, or CDK7 substrate peptide at 30 °C with a final ATP concentration of 100 ⁇ M.
  • the substrate for all the CDK assays is histone Hl, diluted to 1OX working stock in 2OmM MOPS pH 7.4 prior to use.
  • the substrate for CDK7 is a specific peptide diluted to 1OX working stock in deionised water.
  • CDK2/cvclinA CDK2/cvclinE.
  • CDK3/cvclinE CDK5/p35, CDK6/cvclinD3:
  • the enzyme (5-10 mU) is incubated with 8 mM MOPS pH 7.0, 0.2 mM EDTA 5 0.1 mg/ml histone Hl, 10 mM MgAcetate and [ ⁇ - 33 P-ATP] (specific activity approx 500 cpm/pmol, concentration as required).
  • the reaction is initiated by the addition of Mg 2+ [ ⁇ - 33 P-ATP].
  • After incubation for 40 minutes at room temperature the reaction is stopped by the addition of 5 ⁇ l of a 3% phosphoric acid solution. 10 ml of the reaction is spotted onto a P30 filter mat and washed 3 times for 5 minutes in 75mM phosphoric acid and once in methanol prior to drying and counting.
  • the enzyme (5-10 mU) is incubated with 8 mM MOPS pH 7.0, 0.2 mM EDTA 5 500 ⁇ M peptide, 10 mM MgAcetate and [ ⁇ - 33 P- ATP] (specific activity approx 500 cpm/pmol, concentration as required).
  • the reaction is initiated by the addition of Mg 2 +[ ⁇ - 33 P-ATP].
  • After incubation for 40 minutes at room temperature the reaction is stopped by the addition of 5 ⁇ l of a 3% phosphoric acid solution. 10 ml of the reaction is spotted onto a P30 filtermat and washed 3 times for 5 minutes in 75 mM phosphoric acid and once in methanol prior to drying and counting.
  • the anti-proliferative activities of compounds of the invention can be determined by measuring the ability of the compounds to inhibition of cell growth in a number of cell lines. Inhibition of cell growth is measured using the Alamar Blue assay (Nociari, M. M, Shalev, A., Benias, P., Russo, C. Journal of Immunological Methods 1998, 213, 157-167). The method is based on the ability of viable cells to reduce resazurin to its fluorescent product resorufm. For each proliferation assay cells are plated onto 96 well plates and allowed to recover for 16 hours prior to the addition of inhibitor compounds for a further 72 hours.
  • the compounds of Examples 2 to 13 had IC 5 O values of less than 15 ⁇ M.
  • GSK3 ⁇ (human) is diluted to a 1Ox working stock in 5OmM Tris pH 7.5, 0.ImM EGTA, O.lmM sodium vanadate, 0.1% ⁇ -mercaptoethanol, lmg/ml BSA.
  • One unit equals the incorporation of lnmol of phosphate per minute phospho-glycogen synthase peptide 2 per minute.
  • GSK3 ⁇ (5-10 mU) is incubated with 8mM MOPS 7.0, 0.2mM EDTA, 20 ⁇ M YRRAAVPPSPSLSRHSSPHQS(p)EDEEE (phospho GS2 peptide) , 1OmM MgAcetate and [ ⁇ - 33 P-ATP] (specific activity approx 500cpm/pmol, concentration as required).
  • the reaction is initiated by the addition of Mg 2 +[ ⁇ - 33 P-ATP] .
  • After incubation for 40 minutes at room temperature the reaction is stopped by the addition of 5 ⁇ l of a 3% phosphoric acid solution. lO ⁇ l of the reaction is spotted onto a P30 filter mat and washed 3 times for 5 minutes in 5OmM phosphoric acid and once in methanol prior to drying and counting.
  • Aurora A kinase activity can be determined using a Dissociative Enhanced Lanthanide Fluoro Immuno Assay (DELFIA) with a GSK3-derived biotinylated peptide.
  • DELFIA Dissociative Enhanced Lanthanide Fluoro Immuno Assay
  • CGPKGPGRRGRRRTSSFAEG 15 ⁇ M ATP and various dilutions of compound in 1OmM MOPS, pH 7.0, O.lmg/ml BSA, 0.001% Brij-35, 0.5% glycerol, 0.2mM EDTA, 1OmM MgCl 2 , 0.01% ⁇ -mercaptoethanol & 2.5% DMSO.
  • the reaction is allowed to proceed for 60 minutes at room temperature before stopping with lOO ⁇ l STOP buffer containing 10OmM EDTA, 0.05% Surfact-Amps20 (Pierce) and Ix BlockerTM BSA in TBS (Pierce).
  • Detection step The reaction mixture is then transferred to a 96-well Neutravidin-coated plate (Pierce) and incubated for 30 minutes to capture the biotinylated peptide. After washing 5 times with 200 ⁇ l TBST buffer per well, a mixture of anti-phospho- (Ser/Thr)-AKT substrate antibody (Cell Signalling Technology) and Eu-N 1 anti- rabbit IgG (Perkin Elmer) is added to all wells and left for lhour. After a further washing step, DELFIA enhancement solution (Perkin Elmer) is added to all wells. After an incubation of 5 minutes, the wells are counted on a Fusion plate reader.
  • a mixture of anti-phospho- (Ser/Thr)-AKT substrate antibody Cell Signalling Technology
  • Eu-N 1 anti- rabbit IgG Perkin Elmer
  • Assay reactions are set up in 96 well plates in a total reaction volume of 25 ⁇ l with 5nM AuroraB (ProQinase), 3 ⁇ M Biotin-CGPKGPGRRGRRRTSSFAEG, 15 ⁇ M ATP and various dilutions of compound in 25mM TRIS pH 8.5, O.lmg/ml BSA, 0.025% Surfact-Amps 20, 5mM MgCl 2 , ImM DTT, & 2.5% DMSO. The reaction is allowed to proceed for 90 minutes at room temperature before stopping with lOO ⁇ l STOP buffer containing 10OmM EDTA, 0.05% Surfact-am ⁇ s20 (Pierce) and Ix BlockerTM BSA in TBS (Pierce).
  • the detection step is carried out as described for AuroraA.
  • the antifungal activity of the compounds of the formula (I) is determined using the following protocol.
  • the compounds are tested against a panel of fungi including Candida parpsilosis, Candida tropicalis, Candida albicans-ATCC 36082 and Cryptococcus neoformans.
  • the test organisms are maintained on Sabourahd Dextrose Agar slants at 4 °C.
  • Singlet suspensions of each organism are prepared by growing the yeast overnight at 27 °C on a rotating drum in yeast-nitrogen base broth (YNB) with amino acids (Difco, Detroit, Mich.), pH 7.0 with 0.05 M morpholine propanesulphonic acid (MOPS). The suspension is then centrifuged and washed twice with 0.85% NaCl before sonicating the washed cell suspension for 4 seconds (Branson Sonifier, model 350, Danbury, Conn.). The singlet blastospores are counted in a haemocytometer and adjusted to the desired concentration in 0.85% NaCl.
  • test compounds The activity of the test compounds is determined using a modification of a broth microdilution technique.
  • Test compounds are diluted in DMSO to a 1.0 mg/ml ratio then diluted to 64 ⁇ g/ml in YNB broth, pH 7.0 with MOPS (Fluconazole is used as the control) to provide a working solution of each compound.
  • MOPS Fluonazole is used as the control
  • wells 1 and 3 through 12 are prepared with YNB broth, ten fold dilutions of the compound solution are made in wells 2 to 11 (concentration ranges are 64 to 0.125 ⁇ g/ml).
  • Well 1 serves as a sterility control and blank for the spectrophotometric assays.
  • Well 12 serves as a growth control.
  • microtitre plates are inoculated with 10 ⁇ l in each of well 2 to 11 (final inoculum size is 10 4 organisms/ml). Inoculated plates are incubated for 48 hours at 35 °C.
  • the IC50 values are determined spectrophotometrically by measuring the absorbance at 420 run (Automatic Microplate Reader, DuP ont Instruments, Wilmington, Del.) after agitation of the plates for 2 minutes with a vortex-mixer (Vorte-Genie 2 Mixer, Scientific Industries, Inc., Bolemia, N. Y.).
  • the IC50 endpoint is defined as the lowest drug concentration exhibiting approximately 50% (or more) reduction of the growth compared with the control well.
  • MCC Minimum Cytolytic Concentrations
  • compositions are then used to test the activity of the compounds of the invention against tomato blight (Phytophthora infestans) using the following protocol.
  • Tomatoes (cultivar Rutgers) are grown from seed in a soil-less peat-based potting mixture until the seedlings are 10-20 cm tall. The plants are then sprayed to run-off with the test compound at a rate of 100 ppm. After 24 hours the test plants are inoculated by spraying with an aqueous sporangia suspension of Phytophthora infestans, and kept in a dew chamber overnight. The plants are then transferred to the greenhouse until disease develops on the untreated control plants.
  • a tablet composition containing a compound of the formula (I) is prepared by mixing 50 mg of the compound with 197 mg of lactose (BP) as diluent, and 3 mg magnesium stearate as a lubricant and compressing to form a tablet in known manner.
  • BP lactose
  • a capsule formulation is prepared by mixing 100 mg of a compound of the formula (I) with 100 mg lactose and filling the resulting mixture into standard opaque hard gelatin capsules.
  • a parenteral composition for administration by injection can be prepared by dissolving a compound of the formula (I) (e.g. in a salt form) in water containing 10% propylene glycol to give a concentration of active compound of 1.5 % by weight. The solution is then sterilised by filtration, filled into an ampoule and sealed.
  • a parenteral composition for injection is prepared by dissolving in water a compound of the formula (I) (e.g. in salt form) (2 mg/ml) and mannitol (50 mg/ml), sterile filtering the solution and filling into sealable 1 ml vials or ampoules.
  • a compound of the formula (I) e.g. in salt form
  • mannitol 50 mg/ml
  • a composition for sub-cutaneous administration is prepared by mixing a compound of the formula (I) with pharmaceutical grade corn oil to give a concentration of 5 mg/ml.
  • the composition is sterilised and filled into a suitable container.
  • a formulation for i.v. delivery by injection or infusion can be prepared by dissolving the compound of formula (I) (e.g. in a salt form) in water at 20 mg/ml. The vial is then sealed and sterilised by autoclaving.
  • a formulation for i.v. delivery by injection or infusion can be prepared by dissolving the compound of formula (I) (e.g. in a salt form) in water containing a buffer (e.g. 0.2 M acetate pH 4.6) at 20mg/ml. The vial is then sealed and sterilised by autoclaving.
  • a buffer e.g. 0.2 M acetate pH 4.6
  • a composition for sub-cutaneous administration is prepared by mixing a compound of the formula (I) with pharmaceutical grade corn oil to give a concentration of 5 mg/ml.
  • the composition is sterilised and filled into a suitable container.
  • Lyophilised formulation Aliquots of formulated compound of formula (I) or a salt thereof as defined herein are put into 50 mL vials and lyophilized. During lyophilisation, the compositions are frozen using a one-step freezing protocol at (-45 0 C). The temperature is raised to -10 0 C for annealing, then lowered to freezing at -45 0 C, followed by primary drying at +25 0 C for approximately 3400 minutes, followed by a secondary drying with increased steps if temperature to 50 0 C. The pressure during primary and secondary drying is set at 80 millitor.

Abstract

L'invention concerne un composé de formule (I) : ou un sel, tautomère, N-oxyde ou solvate de celui-ci ; selon laquelle A est sélectionné entre une liaison, CH2 et CH(CN) ; RI est sélectionné entre (i) un groupe cycloalkyle à 3 à 6 chaînons cycliques facultativement substitué par un ou plusieurs substituants sélectionnés parmi les groupes méthyle, éthyle, hydroxy, méthoxy, éthoxy, fluor, amino et cyano ; (ii) un groupe phényle facultativement substitué par jusqu'à trois substituants sélectionnés parmi les groupes méthyle, éthyle, fluor, chlore, méthoxy, éthoxy et méthylsulfonyle, mais à l'exception du 2,6-difluorophényle, du 2-fluoro-6-méthoxy et du 5-chloro-2-méthoxyphényle ; (iii) un groupe hétérocyclique monocyclique sélectionné entre un furyle et un isoxazolyle, le groupe hétérocyclique étant facultativement substitué par un ou deux groupes sélectionnés parmi les groupes méthyle, éthyle et un groupe CH2R2 où R2 est un hétérocycle saturé à cinq ou six chaînons contenant un ou deux chaînons du cycle qui sont des hétéroatomes sélectionnés parmi O et N, l'hétérocycle étant facultativement substitué par un ou deux groupes méthyle ; et (iv) un groupe hétérocyclique bicyclique sélectionné entre le 2,3-dihydrobenzofuranyle et le benzo[c]isoxazolyle, le groupe bicyclique étant facultativement substitué par un ou deux substituants sélectionnés parmi les groupes méthyle, éthyle, hydroxy, méthoxy, éthoxy, fluor, amino, cyano et chlore, le groupe hétérocyclique bicyclique étant autre qu'un groupe 2,2-diméthyl-2,3-dihydrobenzofuran-7-yle.
PCT/GB2005/005109 2004-12-30 2005-12-30 Dérivés de pyrazole ayant une activité de modulation des kinases WO2006070202A1 (fr)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US64054604P 2004-12-30 2004-12-30
GB0428537.5 2004-12-30
GB0428537A GB0428537D0 (en) 2004-12-30 2004-12-30 Pharmaceutical compounds
US60/640,546 2004-12-30

Publications (1)

Publication Number Publication Date
WO2006070202A1 true WO2006070202A1 (fr) 2006-07-06

Family

ID=35986149

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/GB2005/005109 WO2006070202A1 (fr) 2004-12-30 2005-12-30 Dérivés de pyrazole ayant une activité de modulation des kinases

Country Status (1)

Country Link
WO (1) WO2006070202A1 (fr)

Cited By (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010074724A1 (fr) 2008-12-22 2010-07-01 Millennium Pharmaceuticals, Inc. Inhibiteurs d'aurora kinases associés avec des anticorps anti-cd20
EP2468882A1 (fr) * 2010-12-27 2012-06-27 Bayer CropScience AG Polypeptides des kinases Aurora pour identifier des composés à effet fongicide
WO2015085289A1 (fr) 2013-12-06 2015-06-11 Millennium Pharmaceuticals, Inc. Combinaison d'inhibiteurs de kinase aurora et d'anticorps anti-cd30
US9126969B2 (en) 2006-07-04 2015-09-08 Aventis Pharma S.A. Pyrazolylbenzimidazole derivatives, compositions containing them and use thereof
US20160316988A1 (en) * 2015-05-01 2016-11-03 Ecolab Usa Inc. Mop bucket
CN107438603A (zh) * 2015-04-22 2017-12-05 里格尔药品股份有限公司 吡唑化合物以及制备和使用该化合物的方法
WO2018172852A1 (fr) * 2017-03-21 2018-09-27 Arbutus Biopharma Corporation Dihydroindène-4-carboxamides substitués, leurs analogues et procédés d'utilisation correspondant
JP2020510061A (ja) * 2017-03-14 2020-04-02 エフ.ホフマン−ラ ロシュ アーゲーF. Hoffmann−La Roche Aktiengesellschaft ピラゾロクロロフェニル化合物、組成物及びその使用方法
WO2021041532A1 (fr) 2019-08-26 2021-03-04 Dana-Farber Cancer Institute, Inc. Utilisation d'héparine pour favoriser la signalisation de l'interféron de type 1
US11370787B2 (en) 2019-08-30 2022-06-28 Rigel Pharmaceuticals, Inc. Pyrazole compounds, formulations thereof, and a method for using the compounds and/or formulations
US11874276B2 (en) 2018-04-05 2024-01-16 Dana-Farber Cancer Institute, Inc. STING levels as a biomarker for cancer immunotherapy

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001057022A2 (fr) * 2000-02-05 2001-08-09 Vertex Pharmaceuticals Incorporated Compositions de pyrazole utiles comme inhibiteurs de erk
WO2003077921A1 (fr) * 2002-03-15 2003-09-25 Vertex Pharmaceuticals, Inc. Azinylaminoazoles utilises comme inhibiteurs de proteine kinases
WO2005002576A2 (fr) * 2003-07-03 2005-01-13 Astex Therapeutics Limited Composes pharmaceutiques
WO2005012256A1 (fr) * 2003-07-22 2005-02-10 Astex Therapeutics Limited Composes 1h-pyrazole 3,4-disubstitues et leur utilisation en tant que kinases dependant des cyclines (cdk) et modulateurs de la glycogene synthase kinase-3 (gsk-3)

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001057022A2 (fr) * 2000-02-05 2001-08-09 Vertex Pharmaceuticals Incorporated Compositions de pyrazole utiles comme inhibiteurs de erk
WO2003077921A1 (fr) * 2002-03-15 2003-09-25 Vertex Pharmaceuticals, Inc. Azinylaminoazoles utilises comme inhibiteurs de proteine kinases
WO2005002576A2 (fr) * 2003-07-03 2005-01-13 Astex Therapeutics Limited Composes pharmaceutiques
WO2005012256A1 (fr) * 2003-07-22 2005-02-10 Astex Therapeutics Limited Composes 1h-pyrazole 3,4-disubstitues et leur utilisation en tant que kinases dependant des cyclines (cdk) et modulateurs de la glycogene synthase kinase-3 (gsk-3)

Cited By (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9126969B2 (en) 2006-07-04 2015-09-08 Aventis Pharma S.A. Pyrazolylbenzimidazole derivatives, compositions containing them and use thereof
WO2010074724A1 (fr) 2008-12-22 2010-07-01 Millennium Pharmaceuticals, Inc. Inhibiteurs d'aurora kinases associés avec des anticorps anti-cd20
EP2468882A1 (fr) * 2010-12-27 2012-06-27 Bayer CropScience AG Polypeptides des kinases Aurora pour identifier des composés à effet fongicide
WO2015085289A1 (fr) 2013-12-06 2015-06-11 Millennium Pharmaceuticals, Inc. Combinaison d'inhibiteurs de kinase aurora et d'anticorps anti-cd30
CN113750094A (zh) * 2015-04-22 2021-12-07 里格尔药品股份有限公司 吡唑化合物以及制备和使用该化合物的方法
CN107438603A (zh) * 2015-04-22 2017-12-05 里格尔药品股份有限公司 吡唑化合物以及制备和使用该化合物的方法
JP2018513177A (ja) * 2015-04-22 2018-05-24 ライジェル ファーマシューティカルズ, インコーポレイテッド ピラゾール化合物ならびにこの化合物を作製および使用するための方法
US11370808B2 (en) 2015-04-22 2022-06-28 Rigel Pharmaceuticals, Inc. Pyrazole compounds and methods for making and using the compounds
JP2021020924A (ja) * 2015-04-22 2021-02-18 ライジェル ファーマシューティカルズ, インコーポレイテッド ピラゾール化合物ならびにこの化合物を作製および使用するための方法
JP7050879B2 (ja) 2015-04-22 2022-04-08 ライジェル ファーマシューティカルズ, インコーポレイテッド ピラゾール化合物ならびにこの化合物を作製および使用するための方法
CN107438603B (zh) * 2015-04-22 2021-08-10 里格尔药品股份有限公司 吡唑化合物以及制备和使用该化合物的方法
US20160316988A1 (en) * 2015-05-01 2016-11-03 Ecolab Usa Inc. Mop bucket
JP2020510061A (ja) * 2017-03-14 2020-04-02 エフ.ホフマン−ラ ロシュ アーゲーF. Hoffmann−La Roche Aktiengesellschaft ピラゾロクロロフェニル化合物、組成物及びその使用方法
US11649241B2 (en) 2017-03-14 2023-05-16 Genentech, Inc. Pyrazolochlorophenyl compounds, compositions and methods of use thereof
US11098010B2 (en) 2017-03-21 2021-08-24 Arbutus Biopharma Corporation Substituted dihydroindene-4-carboxamides and analogs thereof, and methods using same
WO2018172852A1 (fr) * 2017-03-21 2018-09-27 Arbutus Biopharma Corporation Dihydroindène-4-carboxamides substitués, leurs analogues et procédés d'utilisation correspondant
US11874276B2 (en) 2018-04-05 2024-01-16 Dana-Farber Cancer Institute, Inc. STING levels as a biomarker for cancer immunotherapy
WO2021041532A1 (fr) 2019-08-26 2021-03-04 Dana-Farber Cancer Institute, Inc. Utilisation d'héparine pour favoriser la signalisation de l'interféron de type 1
US11370787B2 (en) 2019-08-30 2022-06-28 Rigel Pharmaceuticals, Inc. Pyrazole compounds, formulations thereof, and a method for using the compounds and/or formulations

Similar Documents

Publication Publication Date Title
AU2005321091B2 (en) Pyrazole compounds that modulate the activity of CDK, GSK and Aurora kinases
WO2006070202A1 (fr) Dérivés de pyrazole ayant une activité de modulation des kinases
US8293767B2 (en) 4-(2,6-dichloro-benzoylamino)-1H-pyrazole-3-carboxylic acid piperidin-4-ylamide acid addition salts as kinase inhibitors
WO2006077416A1 (fr) Derives de pyrazole servant a inhiber les kinases dependantes des cyclines (cdk) et les glycogene synthases kinases (gsk)
US20100160324A1 (en) Pyrazole derivatives as that modulate the activity of cdk, gsk and aurora kinases
US20080004270A1 (en) 3,4-Disubstituted Pyrazoles as Cyclin Dependent Kinases (Cdk) or Aurora Kinase or Glycogen Synthase 3 (Gsk-3) Inhibitors
EP1836199A1 (fr) Derives de thiazole et isothiazole modulant l'activite des kinases cdk, gsk et aurora
EP1648449A2 (fr) Composes pharmaceutiques
US20080139620A1 (en) Pyrazole Derivatives For The Inhibition Of Cdk's And Gsk's
EP2049516A2 (fr) Composés pharmaceutiques
WO2006008545A2 (fr) Composes pharmaceutiques

Legal Events

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

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 05823118

Country of ref document: EP

Kind code of ref document: A1