WO2013024282A2 - Compounds and their uses - Google Patents

Compounds and their uses Download PDF

Info

Publication number
WO2013024282A2
WO2013024282A2 PCT/GB2012/051978 GB2012051978W WO2013024282A2 WO 2013024282 A2 WO2013024282 A2 WO 2013024282A2 GB 2012051978 W GB2012051978 W GB 2012051978W WO 2013024282 A2 WO2013024282 A2 WO 2013024282A2
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
optionally substituted
compound
cancer
group
Prior art date
Application number
PCT/GB2012/051978
Other languages
French (fr)
Other versions
WO2013024282A3 (en
Inventor
Gary Newton
Trevor Perrior
Alan Ashworth
Christopher James LORD
Original Assignee
Domainex Limited
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Domainex Limited filed Critical Domainex Limited
Publication of WO2013024282A2 publication Critical patent/WO2013024282A2/en
Publication of WO2013024282A3 publication Critical patent/WO2013024282A3/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/403Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil condensed with carbocyclic rings, e.g. carbazole
    • A61K31/404Indoles, e.g. pindolol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41641,3-Diazoles
    • A61K31/41841,3-Diazoles condensed with carbocyclic rings, e.g. benzimidazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/4353Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/437Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems the heterocyclic ring system containing a five-membered ring having nitrogen as a ring hetero atom, e.g. indolizine, beta-carboline
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/4709Non-condensed quinolines and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • A61K31/713Double-stranded nucleic acids or oligonucleotides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Definitions

  • This invention relates to compounds that are inhibitors of one or both of the kinases TBK1 and ⁇ (otherwise referred to as I-kappa-B-kinase-3 (IKK3), or down-regulators of the expression of one or both of TBK1 and ⁇ , and their use in the treatment of cancers that are dependent on the PDkinase-pathway.
  • the invention also relates to methods of diagnosis, methods of determining the susceptibility of a cancer to particular treatments, and methods of determining suitability of particular treatments.
  • the PDkinase signalling pathway is an important cell survival pathway which involves a number of protein kinases, and also lipid kinases of the phosphoinositide 3-kinase (PI3 kinase) family whose primary role is to phosphorylate inositol lipids.
  • PI3 kinase phosphoinositide 3-kinase
  • the phosphorylated inositol lipids subsequently play an important signalling role by membrane recruitment of effectors having phosphoinositide-recognition motifs.
  • PI3 kinases Three different classes of PI3 kinases have been identified. All members of the family share a common function in phosphorylating the 3 ⁇ group of phosphatidylinositols (Ptdlns), however they act preferentially on different Ptdlns substrates.
  • Ptdlns phosphatidylinositols
  • the PI3 kinases that are most commonly implicated in cancer are the class I PI3 kinases (Engleman, 2009).
  • PDkinase signalling by class I PD kinases is driven by the kinase function of the pi 10 catalytic domain which primarily converts PtdIns(4,5)P2 into PtdIns(3,4,5)P 3 .
  • Proteins that contain a pleckstrin homology (PH) domain become associated with the cell membrane by binding to Ptdlns(3, 4,5)P > and when localised here they can become further activated by other proteins.
  • a major pathway for transduction of the downstream signal from PDkinase involves the activation of AKT.
  • AKT becomes associated with the cell membrane via its PH domain, from where it can become activated by phopsphorylation of Threonine 308 and Serine 473 by PDK-1, and the mTOR/Rictor complex (mTorc2), respectively.
  • AKT can then provide a powerful growth/survival signal by phosphorylation of a number of proteins including TSC-2, which indirectly activates the mTOR/Raptor complex (mTorcl), GSK3 and members of the FOXO family of proteins (Engleman, 2009; Stephens et ah, 2005)
  • AKT is the PDkinase pathway downstream effector most widely implicated in cancer
  • AKT independent pathways include the Bruton tyrosine kinase (BTK), the TEC families of non-receptor tyrosine kinases and serum- and glucocorticoid- regulated kinases (SGKs) (Qui et al., Oncogene, 2000, 19, 5651-5661; Vasudevan et al., Cancer Cell, 2009, 16 (1), 21-32).
  • BTK Bruton tyrosine kinase
  • SGKs serum- and glucocorticoid- regulated kinases
  • Receptor tyrosine kinases such as EGFR or Erb-2
  • EGFR or Erb-2 can activate the PI3 kinase pathway by recruiting the regulatory subunit thereby releasing the auto-inhibitory role that this would normally play (Nagata et al., Cancer Cell, 2004, 6(2), 117-27). This mechanism of activation is primarily associated with class IA isoforms (Williams et al., Biochem. Soc. Trans, 2009, 37, 615-626).
  • the pi 10 isoforms can be activated directly by binding to activated Ras proteins. This is the primary mode of activation for pi 10 ⁇ , and may also be required for activation of ⁇ ⁇ (Kang et al, PNAS, 2006, 103 (5) 1289- 1294, Andrews et al, Science Signalling, 2007, 407, cm2, "PDK Class IB Pathway” ).
  • Mutations in the one of the PI3 kinase catalytic subunits can result in constituitive activation of the pathway (Engleman, 2009). Mutations in the regulatory p85 subunit can result in loss of its inhibitory activity and hence constitutive activation of the pathway (Engleman, 2009).
  • PI3 Kinase catalytic subunits may be over-expressed resulting in amplification of signalling through the PI3 kinase pathway (Engleman 2009).
  • Loss or inactivation of key phosphatases such as PTEN which reverse the action of the PI3 kinases by dephophorylating the 3 position of Ptdlns(3, 4,5)P3 can increase flux through the PI3 kinase pathway (Engleman, 2009; Nagata et al , Cancer Cell, 2004, 6(2), 117-27).
  • a downstream kinase such as PDK- 1 or AKT may be over-expressed or possess an activating mutation (Engleman, 2009).
  • Phosphatase and tensin homolog is a key enzyme that inhibits signalling through the PDkinase pathway and it therefore has tumour suppressing effects. Mutation in the PTEN gene (PTEN(-) mutation) inactivates the enzyme and leads to increased flux through the pathway. PTEN is one of the most commonly mutated or lost tumour suppressors in human cancer. PTEN loss has been reported in endometrial cancer (35%) and glioblastoma (54%) as well as in other cancer types such as breast (32%), colo-rectal carcinoma (30%), prostate (27%) and lung cancer (19%) (Frattini et al, Br. J. Cancer, 2007, 97(8), 1139-45; Nagata et al, 2004).
  • Low levels of PTEN are also reported in a number of cancers. This can occur for example as the result of the loss of one functional copy of the PTEN gene, often described as haploinsufficiency. It is believed that low levels of PTEN can contribute significantly to the oncogenic state especially when accompanied by other changes/mutations that are able to further activate the PI3 kinase pathway (Ying et al., Cancer Discovery, 2011, 1 (2), 158-169; Nagata et al., 2004). It has also been shown that receptor tyrosine kinase signalling can deactivate PTEN. It has been shown that part of the mechanism of action of the Erb2 antibody trastuzumab involves inhibition of Src kinase activity.
  • Src kinase is believed to be responsible for deactivating PTEN in breast cancers where Erb2 is over-expressed, by phophorylation of key tyrosine residues on PTEN. Furthermore, loss of PTEN has been shown to be associated with resistance to trastuzumab, and inhibition of PI3 kinase signalling can increase sensitivity to trastuzumab in PTEN deficient cells (Nagata et al., 2004).
  • PBkinases increase flux through the PDkinase pathway, and so PI3 kinase activating mutations are oncogenic, for example mutations classified as PIK3CA (catalytic - alpha polypeptide) leads to enhanced signalling through the pathway.
  • PIK3CA mutation can be found in a range of malignancies such as colon, lung, ovarian, liver, brain, stomach and breast cancers (Samuels et al., Science, 2004, 304(5670), 554; Engleman, 2009). Genetic amplification of PIK3CA is also found in a number of tumour types including head and neck, squamous cell lung carcinoma, cervical, gastric and oesophageal (Engleman, 2009).
  • I-kappa-B-kinase epsilon, ⁇ (also known as ⁇ , I-kappa-B-kinase-3 (IKK3), or inducible I-kappa-B-kinase (IKKi)), and TANK Binding Kinase- 1, TBK1 (also known as T2K or NF-kappa B-activating kinase), are serine-threonine kinases.
  • ⁇ activity has been linked to a number of disease states including cancer and obesity.
  • IKBE gene encoding ⁇
  • has also been shown to phosphorylate the estrogen receptor, and its activity has been linked to tamoxifen resistance in breast cancer tumours (Guo et al., The Journal of Biological Chemistry, 2010, 285, 3676-3684).
  • is also frequently over-expressed in human ovarian cancer lines and primary tumours.
  • ⁇ over-expression renders cells resistant to cis-platin
  • ⁇ knockdown restores cis-platin sensitivity
  • ⁇ inhibitors may show efficacy in the treatment of certain cancers.
  • TBK1 has been linked to various cancer states although the published literature has not defined its exact role, or the appropriate clinical setting for use of a TBK- 1 inhibitor.
  • the levels of TBK1 mRNA and protein are elevated in certain malignant colon and breast cancer cells.
  • TBK1 is also recruited and activated by the RalB/Sec5 effector complex: in cancer cells, constitutive engagement of this pathway via chronic RalB activation, restricts the initiation of apoptotic programmes.
  • the proto-oncogene KRAS (which encodes the protein K-Ras) is mutated in a wide array of human tumours, most of which are aggressive and respond poorly to standard therapies.
  • the knockdown of TBK1 in a number of K-Ras dependant tumour cell lines has been shown to cause cell death.
  • K- Ras can activate the PI3 kinase pathway it is believed that cell lines which have K-Ras mutations are not necessarily addicted to the PI3 kinase pathway. For example it has been shown in pancreatic ductal adenocarcinoma (PDAC) that low levels of PTEN are also required for activation of the PI3 kinase pathway (Ying et ah, 2011).
  • PDAC pancreatic
  • AKT is a major signalling kinase and is generally considered to be one of the enzymes involved in the PDkinase pathway. Despite this it has been shown that not all tumour cell lines that are dependent on the PDkinase pathway have high levels of activated AKT nor do they all respond to AKT knockdown (Vasudevan et al, 2009). Ou et al.
  • the invention provides a compound that is an inhibitor of one or both of TBKl and ⁇ , or a down-regulator of the expression of one or both of TBKl and ⁇ , for use in a method of treating a cancer that is dependent on the PDkinase pathway. It has surprisingly been found by the current inventors that cancers that are deficient in PTEN activity are sensitive to compounds that are inhibitors of TBKl and ⁇ .
  • cancers which are deficient in PTEN activity are sensitive to down-regulation of the expression of TBKl . From the data obtained by the current inventors, it is anticipated that such cancers are also are sensitive to down-regulation of the expression of ⁇ . It has also been found by the current inventors that cancers with overactive PDkinase signalling are sensitive to TBKl and ⁇ inhibitors. From the data obtained by the current inventors, it is anticipated that cancers with overactive PDkinase signalling are sensitive to down-regulation of the expression of one or both of TBKl and ⁇ .
  • the current inventors found that a human cell line in which both alleles of PTEN had been inactivated was lethally susceptible to siRNA inhibition of TBKl. That result demonstrates that TBKl activity is necessary for the survival of cells that are deficient in PTEN.
  • the inventors also found that TBKl inhibitor compounds were selectively toxic to PTEN (-/-) cell lines over PTEN (+/+) cells lines, showing that TBKl activity is necessary for the survival of cells that are deficient in PTEN, but not for cells that have normally functional PTEN.
  • the inventors have further found that breast tumour cell models that have PTEN and/or PIK3CA defects are more sensitive to TBKl inhibitors than tumour cell models with defects in other genes.
  • a compound that is an inhibitor of one or both of TBKl and ⁇ , or a down-regulator of the expression of one or both of TBKl and ⁇ is effective in the treatment of a cancer that is dependent on the PDkinase pathway.
  • a compound that is an inhibitor of one or both of TBKl and ⁇ , or a down-regulator of the expression of one or both of TBKl and ⁇ is effective in the treatment of a cancer that has a defect in one or both of the PTEN and PIK3CA genes.
  • Figure 1 shows the Normalised Percent Inhibition (NPI) scores following the knockdown of 714 kinase genes with a panel of siRNAs in HCT116 human tumour cells in which both alleles of PTEN had been deactivated (PTEN7-) and HCT116 human tumour cells with wildtype PTEN (PTEN +/+).
  • Figures 2 (a) to (c) show the effects of small molecule dual inhibitors of the kinase domain of TBKl and ⁇ on (PTEN-/-)-HCT116 and (PTEN +/+)-HCT116 human tumour cells, (a) shows the results for DMA-A, (b) shows the results for DMX-B and (c) shows the results for DMX-C.
  • Figure 3 shows the effect on survival fraction for escalating concentrations of the ⁇ / ⁇ inhibitor DMX-C on an ovarian tumour cell line which contains deactivating mutations in the PTEN gene.
  • Figures 3 (b) to (d) show the effects on survival fractions for escalating concentrations of the ⁇ / ⁇ inhibitor DMX-C on breast tumour cell lines which contains deactivating mutations in the PTEN gene.
  • Figure 3 (e) shows the effect on survival fraction for escalating concentrations of the ⁇ / ⁇ ⁇ inhibitor DMX-C on a breast tumour cell lines which contains both deactivating mutations in the PTEN gene and activating mutations in the PIK3CA gene.
  • Figures 4(a) to (c) show the effects on survival fractions for escalating concentrations of the ⁇ / ⁇ ⁇ inhibitor DMX-C on breast tumour cell lines which contains activating mutations in the PIK3CA gene.
  • Figures 5 (a) to (e) show the effects on survival fractions for escalating concentrations of the ⁇ / ⁇ ⁇ inhibitor DMX-C on breast tumour cell lines with wildtype PTEN and PIK3CA genes.
  • Figure 6 shows SF80 (concentration at which 80% of cells survive) and (b) shows SF50 (concentration at which 50% of cells survive) results for DMX-C in cell lines with PTEN and/or PIKCA mutations and those with wild type PTEN and PIKCA.
  • Figure 7 shows the results of a study into TBK1 sensitising to BEZ235 (a dual PDkinase, mTOR inhibitor), the graph shows the survival fraction results for T47D breast tumour cells exposed to various concentrations of BEZ235 in the presence or absence of TBK-1 siRNA , with siRNA targeting PIK3CA used as a positive control.
  • BEZ235 a dual PDkinase, mTOR inhibitor
  • TBK1 and ⁇ have overlapping substrate specificities and it is believed that they have similar binding sites. Accordingly, many compounds that are inhibitors of one of TBK1 and ⁇ ⁇ are also inhibitors of the other. Compounds that are strong inhibitors of both TBK1 and ⁇ are particularly advantageous compounds for use in the current invention.
  • the cancer to be treated may be a cancer that is deficient in PTEN activity.
  • the cancer may have reduced amounts of PTEN, or a level of PTEN function that is lower than PTEN function in normal cells in the individual.
  • Expression levels of PTEN protein can be established by western blotting, or in situ hybridization, using commercially available antibodies to PTEN.
  • the cancer may include at least some cells carrying a loss of function mutation in the PTEN gene. The presence or absence of loss of function mutations may be assessed by determining the presence or absence of one or more mutations or polymorphisms in a nucleic acid sequence encoding at least a portion of the PTEN enzyme.
  • the presence of such mutations can be established by Sanger Sequencing or Next Generation (Massively Parallel or Deep) Sequencing, or the use of mutation- specific PCR assays.
  • the copy number of each gene (and thus loss of heterozygosity in the case of PTEN) can be established by Fluorescent In Situ Hybridisation (FISH).
  • FISH Fluorescent In Situ Hybridisation
  • the cancer may be a cancer that has overactive PBkinase signalling.
  • the level of PBkinase signalling can be assessed by a functional assay known to or readily developed by the person skilled in the art.
  • the cancer may include at least some cells carrying an activating PIK3CA mutation.
  • the presence or absence of an activating PIK3CA mutation may be assessed by determining the presence or absence of one or more mutations or polymorphisms in a nucleic acid sequence encoding at least a portion of the PBkinase pi 10 catalytic subunit.
  • Overactive PB kinase signalling may be detected by an increase in phosphorylation of one or more of the downstream proteins, for example PDK-1 or mTOR. This can be measured in standard assay formats such as the use of a western blot, ELISA or in situ hybridization techniques, which all use commercially available phospho-protein specific antibodies.
  • Cancers with overactive PBkinase signalling include endometrial cancer, glioblastoma, colorectal carcinoma, myeloma and leukemia, and cancers of the breast, prostate, lung (including non small cell lung cancer (NSCLC)), ovary, liver, brain and stomach; in particular, endometrial cancer, glioblastoma, breast cancer, including tamoxifen resistant breast cancer, ovarian cancer, including cis-platin resistant ovarian cancer.
  • NSCLC non small cell lung cancer
  • Compounds of the invention are expected to be particularly useful in the treatment of such cancers.
  • a compound for use in the invention may be a compound that is an inhibitor of one or both of TBK1 and ⁇ .
  • a compound is considered to be an inhibitor of TBK1 if it has an IC 50 for inhibition of TBK-1 of ⁇ 300nM.
  • Preferred compounds have an IC 50 for inhibition of TBK-1 of ⁇ 100nM, particularly ⁇ 50nM, more particularly ⁇ 30nM.
  • the IC 50 for inhibition of TBK-1 can be assessed by assays known to the person skilled in the art, for example a luminescent kinase assay, or a time-resolved fluorescence assay.
  • assays known to the person skilled in the art, for example a luminescent kinase assay, or a time-resolved fluorescence assay.
  • a TBK-1 assay is available from Promega (Promega Corporation, 2800 Woods Hollow Road, Madison, WI53711-5399, USA, or see http://www.promega.com/).
  • An example of an assay is described in the Examples section herein.
  • a compound is considered to be an inhibitor of ⁇ if for inhibition of ⁇ of ⁇ 300nM; Preferred compounds have an IC 50 for inhibition of ⁇ of ⁇ 100nM, particularly ⁇ 50nM, more particularly ⁇ 30nM.
  • the IC 50 for inhibition of ⁇ can be assessed by assays known to the person skilled in the art, for example a luminescent kinase assay, or a time-resolved fluorescence assay.
  • an ⁇ assay is available from Invitrogen (see http://www.invitrogen.com). An example of an assay is described in the Examples section herein.
  • a compound for use in the invention may be selective for TBK1 and/or ⁇ over other kinases, or said compound may also inhibit other kinases. Some compounds have been found to be inhibitors of both TBK1 and ⁇ .
  • Compounds that are inhibitors of one or both of TBK1 and ⁇ include compounds selected from the group consisting of: (a) compounds described in PCT/GB2011/001075 (published as WO2012/010826), that is to say compounds of Formula (I):
  • R 2 represents a phenyl or heteroaryl group which is optionally substituted by one or more substituents independently selected from:
  • R' represents one of the following substituents: OH; N0 2 ; CN; COR c ; O.CO.R c ; C0 2 R b ; NR a .COR c ; NR a C0 2 R b ;
  • R a represents a hydrogen atom or a Ci- 4 alkyl group
  • R b represents a hydrogen atom, a Ci_ 4 alkyl group optionally substituted by a group NR a R a , or a cycloalkyl group in which a C3 ⁇ 4 moiety may be replaced by an oxygen atom or an NR a group
  • R c represents a hydrogen atom, a group -NR a R b , or a Ci_ 4 alkyl group optionally substituted by a group NR a R b ;
  • R and R b together may, when attached to the same nitrogen atom, represent a -(CH 2 )m- group in which a C3 ⁇ 4 moiety may be replaced by an oxygen atom or an -NR a - group;
  • n 4 or 5;
  • n 1 or 2;
  • each of R 3 and R 4 independently represents a hydrogen atom or a Ci- 4 alkyl group; or a salt thereof.
  • Rl , R2, R3, and R5 are independently chosen from the following groups: alkyl, alkylene, alkenyl, alkenylene, alkynyl, carbocycle, cycloalkyl, cycloalkenyl, heterocycle, aryl, heteroaryl, halo, hydro, hydroxyl, alkoxy, alkynyloxy, cycloalkyloxy, heterocycloxy, aryloxy, heteroaryloxy, arylalkoxy, heteroarylalkoxy, mercapto, alkylthio, arylthio, cycloalkylthio, arylalkyl, heteroarylalkyl, heteroarylalkenyl, arylalkynyl, haloalkyl, aldehyde, thiocarbonyl, O-carboxy, C-carboxy, carboxylic acid, ester, C-carboxy salt, carboxyalkyl,
  • R4 is independently chosen from hydro, halo, and an optionally-substituted group chosen from lower alkyl, haloalkyl, alkoxy, arylalkoxy, heteroarylalkoxy, and heterocycloalkoxy;
  • R6 and R7 are independently chosen from hydro, halo, and lower alkyl;
  • preferred compounds of group (b) have an ether group at R5 and are without further substitution at R4, R6 and R7, for example compounds of Formula (Ila):
  • Compound (III) was reported as having activity against both TBK1 and ⁇ .
  • Rl is a 5 or 6 membered cyclic alkyl ring or aromatic cyclic ring, wherein optionally one or more carbons in the ring structure is substituted by an O, N or S;
  • Preferred compounds of group (d) are the compounds:
  • BX-795 has also been reported in, for example, J. Biol. Chem. , 2009, 284(21), 14136-14146.
  • the preferred compounds BX-320 and BX-795 were shown to inhibit TBK1 and ⁇ .
  • R 1 is C 3 _8-cycloalkyl
  • X is O, NR 7 or C 3 _6-heterocycloalkyl
  • R 2 is aryl, heteroaryl, fused or unfused aryl-C 3 _6-heterocycloalkyl or fused or unfused heteroaryI-C 3 _6- heterocycloalkyl, each of which is optionally substituted by one or more substitutents selected from aryl, heteroaryl, Ci_6-alkyl, C 3 _7-cycloalkyl and a group A, wherein said Ci-6-alkyl group is optionally substituted by one or more substituents selected from aryl, heteroaryl, R 10 and a group A, said heteroaryl group is optionally substituted by one or more R ⁇ 10> groups; and wherein said C 3 -6- heterocycloalkyl group optionally contains one or more groups selected from oxygen, sulfur, nitrogen and CO;
  • R 3 is Ci-6-alkyl optionally substituted by one or more substituents selected from aryl, heteroaryl, -NR 4 R 5 , -OR 6 , -NR 7 (CO)R 6 , -NR 7 (CO)NR 4 R 5 , -NR 7 S02R 6 , -NR 7 COOR 7 , - CONR 4 R 5 , C 3 _ 6 -heterocycloalkyl and
  • aryl, heteroaryl and C 3 -6 -heterocycloalkyl groups are each optionally substituted by one or more substituents selected from -Ci_6-alkyl and a group A, wherein said -Ci_6-alkyl group is optionally substituted by one or more substituents selected from aryl, heteroaryl and a group A;
  • A is selected from halogen, hydroxyl, cyano, trifluoromethyl, alkoxy, -N0 2 , -NH 2 , -NR 4 R 5 , -OR 6 , -NR 7 (CO)R 6 , -NR 7 (CO)NR 4 R 5 , -NR 7 COOR 7 , -NR 7 (S0 2 )R 6 , - C0 2 H,
  • each R 4 and R 5 is independently selected from hydrogen, C3_7-cycloalkyl, aryl, heteroaryl, Ci_ 6-alkyl and a C 3 -6-heterocycloalkyl ring optionally further containing one or more groups selected from oxygen, sulfur, nitrogen and CO and optionally substituted by one or more R 10 groups, wherein said Ci_6-alkyl is optionally substituted by one or more substituents selected from halogen, cyano, hydroxyl, aryl, heteroaryl, -NR 8 R 9 , -NR 7 (CO)R 6 , NR 7 COOR 6 , -NR 7 (S0 2 )R 6 , -COOR 6 , -CONR 8 R 9 , OR 10 , -S02R 6 and a C
  • R is Ci-6-alkyl, C3_7-cyloalkyl, C4_7-heteroalkyl, heteroaryl, each of which may be optionally substituted by one of more substituents selected from halogen, R 10 and -NR 8 R 9 ; and R 2 is an aryl or heteroaryl group each of which is optionally substituted by one or more substituents selected from Me, CI, F, CN, NHCOMe, CF 3 , COOH, CONH 2 , OH, NH 2 , NHS0 2 Me, OCF 3 , -NHCOO'Bu, -C0 2 Me, -NMe 2 , 4-methylpiperazin-l-yl, N-morpholinyl, (4-methylpiperazin-l-yl)-CO-, (N-morpholinyl)-CH 2 CH 2 0-, (imidazol-l-yl)-CH 2 - and
  • R 1 is -NR 7 (CO)R u ;
  • R 2 is aryl, heteroaryl, fused aryl-C 3 - 6 -heterocycloalkyl or fused heteroaryl-C 3 -6- heterocycloalkyl, each of which is optionally substituted by one or more substitutents selected from aryl, heteroaryl, Ci_ 6 -alkyl, C 3 _ 6 -heterocycloalkyl and a group A, wherein said Ci-6-alkyl group is in turn optionally substituted by one or more substituents selected from aryl, heteroaryl, C3-6 -heterocycloalkyl and a group A, said heteroaryl group is optionally substituted by one or more R 10 groups; and wherein each C 3 _ 6 -heterocycloalkyl group is optionally substituted by one or more groups selected from Ci_6-alkyl, Ci_6-haloalkyl, and A, and which optionally contains one or
  • A is selected from halogen, hydroxyl, cyano, trifluoromethyl, -NO 2 , -NH 2 , -NR 4 R 5 , -OR 6 , - NR 7 (CO)R 6 , -NR 7 (C0)NR 4 R 5 , -NR 7 COOR 7 , -NR 7 (S0 2 )R 6 , -C0 2 H, -NR 7 (S0 2 )NR 4 R 5 , - COOR 7 , -C0NR 4 R 5 , COR 6 and -S0 2 CH 3 ; each R 4 and R 5 is independently selected from hydrogen, C 3 _7-CycloaIkyI, aryl, heteroaryl, Ci-6-alkyl and a C 3 -6 -heterocycloalkyl ring optionally further containing one or more groups selected from oxygen, sulfur, nitrogen and CO, and optionally substituted by one or more R 1( groups, wherein said Ci- 6 -alkyl is
  • R 4 and R 5 together with the N to which they are attached form a C 3 _6-heterocycloalkyl ring optionally further containing one or more groups selected from oxygen, sulfur, nitrogen and CO, wherein said C 3 _6-heterocycloalkyl ring may be saturated or unsaturated and is optionally substituted with one or more groups selected from NR 8 R 9 and R 10 ; each R 6 is independently selected from Ci_6-alkyl, C 3 _7-cycloalkyl, C 4 _7-heterocycloalkyl, aryl and heteroaryl, each of which may be optionally substituted by one or more substituents selected from halogen, R 10 and -NR 8 R 9 ; each R 7 is selected from hydrogen, Ci_6-alkyl and C 3 _6-cycloalkyl, wherein said Ci_6-alkyl is optionally substituted by one or more halogens; each of R 8 and R 9 is independently selected from hydrogen and Ci-6-alky
  • R 8 and R 9 together with the N to which they are attached form a C 4 -6-heterocycloalkyl ring optionally further containing one or more heteroatoms selected from oxygen and sulfur, wherein said C 4 _6-heterocycloalkyl ring is optionally substituted by one or more R 10 groups; and each R 10 is selected from C 3 -7-cycloalkyl and Ci-6-alkyl optionally substituted by one or more halogens, wherein where R 10 is Ci_6-alkyl and two or more R 10 groups are attached to the same carbon atom, the R 10 groups may be linked to form a spiroalkyl group; and each R 11 is independently selected from Ci-6-alkyl, C 3 _7-cycloalkyl, Ci-6-alkyl-C 3 _7-cycloalkyl, C 4-7 - heterocycloalkyl, aryl and heteroaryl, each of which may be optionally substituted by one or more substituents selected from A.
  • Preferred compounds of Formula (VI) include compounds of Formula (Via):
  • R 2 is selected from aryl or heteroaryl, each of which is optionally substituted by one or more substitutents selected from halo, optionally substituted C 3 _7-heterocycloalkyl, optionally substituted Ci-6-alkyl, heteroaryl, Ci_6-alkyl-C 3 -7 -heterocycloalkyl, CN, NHCO- C 3 -7-heterocycloalkyl, CO-C 3 -7- heterocycloalkyl and NHCO-Ci-6-alkyl, wherein said C 3 -7- heterocycloalkyl is optionally substituted by one or more Ci_6-alkyl or A groups, and said Ci_ 6-alkyl is optionally substituted by one or more halo or NR 4 R 5 groups;
  • R is selected from cyclobutyl, thienyl, cyclopentyl, pyrazinyl, CE -cyclopropyl, sec- butyl, tetrahydrofuranyl, thiazolyl, cyclopropyl, isopropyl, cyclohexyl, CH 2 -cyclopentyl and n-propyl.
  • Compound (VII) was reported as having activity against both TBKl and ⁇ .
  • Compound SUl 1652, of Formula (VIII) disclosed in a poster Wilkinson et ah, "High Throughput Compatible Cellular Assays for Interrogating Toll-Like Receptor Signaling Pathways", Invitrogen, Life Technologies Ltd, 3 Fountain Drive, Inchinnan Business Park Paisley PA4 9RF, UK (www.invitrogen.com).
  • n 0, 1,2, 3, or 4 ;
  • each R 1 which may be the same or different, independently represents H, halogen or a group (X) a (Y) b Z;
  • X represents -O- or -CONH-
  • a is 0 or 1 ;
  • Y represents Ci_ 6 alkylene-
  • b is O or l ;
  • Z represents hydroxy, C 1-6 alkyl, Ci_ 6 haloalkyl, C5-7 heterocyclyl, C 1-6 alkoxyalkyl, C 1-6 haloalkoxyalkyl;
  • R 2 represents a group -(X 1 ) c (Y 1 ) d Z 1 ;
  • X 1 represents -Ci-n alkylene-
  • c is 0 or 1 ;
  • Y represents -0-
  • d is 0 or 1
  • Z 1 represents H, aryl or heteroaryl each of which contains 5-14 ring atoms, Cs_7 heterocyclyl, C5-7 cycloalkyl, C5-7 cycloalkenyl, (each of which aryl, heteroaryl, heterocyclyl, cycloalkyl, cycloalkenyl may be optionally substituted by one or more substituents independently selected from C 1-6 alkyl, Ci_ 6 haloalkyl, halogen, Ci_6 alkoxy, Ci_6 haloalkoxy, S0 2 R 3 , Ci_ 6 hydroxyalkyl);
  • R 3 represents H or C 1-6 alkyl
  • Preferred compounds have the general of structure (Xa):
  • Z is aryl or heteroaryl each of which is optionally substituted by one of more substituents independently selected from halogen, CF 3 , CH 2 OH, SO 2 CH 3 , CH 3 , OCH 3 .
  • R 6 is Br, CN, CONH 2 or CI
  • R 1 denotes A, -[C(R 3 )( R 4 )] 3 ⁇ 4 Ar, -[C(R 3 )( R 4 )] wheneverHet or --[C(R 3 )( R 4 )] wheneverCyc,
  • R 2 denotes H, A, Hal, CN, -[C(R ) 2 ] sanction-Ar', -[C(R 3 ) 2 ] admir-Het', -[C(R 3 ) 2 ] ceremoni-Cyc, OR 3 or N(R ) 2 ,
  • R 3 denotes H or A
  • R 4 denotes H, A, -[C(R 3 ) 2 ] friendshipOR 3 , -[C(R 3 ) 2 ] sanctionCOOR 3 , -[C(R 3 ) 2 ] sanctionN(R 3 ) 2 or -[C(R 3 ) 2 ] anythingHet,
  • Cyc denotes cycloalkyl having 3, 4, 5, 6 or 7 C atoms
  • Ar denotes phenyl which is unsubstituted or mono-, di- or trisubstituted by Hal, A, -[C(R 3 ) 2 ] legalOR 3 , N(R ) 2 , N0 2 , CN, COOR 3 , CON(R ) 2 , NR 3 COA, NR 3 S0 2 A, COR 3 , S0 2 N(R ) 2 and/or S(0) friendshipA,
  • Ar' denotes phenyl which is unsubstituted or mono-, di- or trisubstituted by Hal, A, OR 3 , N(R ) 2 , SR 3 , N0 2 , CN, COOR 3 , CON(R 3 ) 2 , NR COA, NR S0 2 A, S0 2 N(R ) 2 , S(0) admirA, CO- Het ! , Heti, [C(R 3 ) 2 ] sanctionN(R 3 ) 2 , 0[C(R 3 ) 2 ] socialN(R 3 ) 2 , 0[C(R 3 ) 2 LHet !
  • Het denotes a mono- or bicyclic saturated, unsaturated or aromatic heterocycle having 1 to 4 N, and/or O and/or S atoms which is unsubstituted or mono- or disubstituted by Hal, A, OR 3 , N(R ) 2 , N0 2 , CN, COOR 3 , CON(R ) 2 , NR 3 COA, NR 3 S0 2 A, COR s , S0 2 NR 3 and/or S(0) confrontA, Het' denotes a mono-, bi- or tricyclic saturated, unsaturated or aromatic heterocycle having 1 to 4 N, O and/or S atoms which is unsubstituted or may be mono- or disubstituted by Hal, A, OR 3 , N(R 3 ) 2 , SR 3 , N0 2 , CN, COOR 3 , CON(R 3 ) 2 , NR 3 COA, NR 3 S0 2 A, S0 2 N(R
  • Hal denotes F, CI, Br or I
  • N denotes 0, 1 or 2
  • Preferred compounds have the following structures:
  • a compound for use in the invention may be a compound that is a down-regulator of the expression of one or both of TBK1 and ⁇ . Expression of one or both of TBK1 and ⁇ may be down-regulated using anti-sense nucleic acid or RNAi technology, amongst other approaches. The use of these approaches to down-regulate gene expression is now well- established in the art. Anti-sense oligonucleotides may be designed to hybridise to the complementary sequence of nucleic acid, pre-mRNA or mature mRNA, interfering with the production of TBK1 or ⁇ so that its expression is reduced or completely or substantially completely prevented.
  • anti-sense techniques may be used to target control sequences of a gene, e. g. in the 5' flanking sequence, whereby the anti- sense oligonucleotides can interfere with expression control sequences.
  • the construction of anti-sense sequences and their use is described for example in Peyman and Ulman, Chemical Reviews, 1990, 90, 543-584, and Crooke, Ann. Rev. Pharmacol. Toxicol. 1992, 32, 329-376.
  • Oligonucleotides may be generated in vitro or ex vivo for administration or anti-sense RNA may be generated in vivo within cells in which down-regulation is desired.
  • double- stranded DNA may be placed under the control of a promoter in a "reverse orientation" such that transcription of the anti- sense strand of the DNA yields RNA which is complementary to normal mRNA transcribed from the sense strand of the target gene.
  • the complementary anti- sense RNA sequence is thought then to bind with mRNA to form a duplex, inhibiting translation of the endogenous mRNA from the target gene into protein. Whether or not this is the actual mode of action is still uncertain. However, it is established fact that the technique works.
  • the complete sequence corresponding to the coding sequence in reverse orientation need not be used. For example fragments of sufficient length may be used. It is a routine matter for the person skilled in the art to screen fragments of various sizes and from various parts of the coding or flanking sequences of a gene to optimise the level of anti-sense inhibition. It may be advantageous to include the initiating methionine ATG codon, and perhaps one or more nucleotides upstream of the initiating codon. A suitable fragment may have about 14-23 nucleotides, e. g. about 15, 16 or 17.
  • An alternative to anti-sense is to use a copy of all or part of the target gene inserted in sense, that is the same, orientation as the target gene, to achieve reduction in expression of the target gene by co-suppression (see Angell & Baulcombe, The EMBO Journal, 1997, 16(12), 3675- 3684; and Voinnet & Baulcombe, Nature , 1997, 389, 553).
  • RNA interference is a two-step process. First, dsRNA is cleaved within the cell to yield short interfering RNAs (siRNAs) of about 21-23nt length with 5' terminal phosphate and 3' short overhangs (-2nt). The siRNAs target the corresponding mRNA sequence specifically for destruction (Zamore, Nature Structural Biology, 2001, 8, 746-750).
  • RNAi may also be efficiently induced using chemically synthesized siRNA duplexes of the same structure with 3'-overhang ends (Zamore et al., Cell, 2000, 101, 25-33. Synthetic siRNA duplexes have been shown to specifically suppress expression of endogenous and heterologeous genes in a wide range of mammalian cell lines (Elbashir, et al. , Nature, 2001, 411, 494-498). Particular siRNA molecules that find use in the invention include RNAi reagents with the target sequence include:
  • a compound that is an inhibitor of one or both of TBKl and ⁇ , or a down- regulator of the expression of one or both of TBKl and ⁇ may be used as the sole active agent, it is also possible for the compound to be used in combination with one or more further active agents.
  • Such further active agents may be further compounds that are inhibitors of one or both of TBKl and ⁇ , or down-regulators of the expression of one or both of TBKl and ⁇ , or they may be different therapeutic agents, for example agents targeting one of the diseases mentioned above, particularly the same disease as that targeted by the compound of the invention.
  • a further therapeutic agent is selected from the group consisting of inhibitors of mTOR, inhibitors of PDkinase, inhibitors of both mTOR and PDkinase, inhibitors of AKT and inhibitors of PDK1.
  • further therapeutic agents include: UCN01, (NVP)- BAG956, Celecoxib, OSU-03012, BX-795, BX-912, BX-320, AR-12, KP372-1, LY294002, PWT-458, PX-866, CAL-101, XL-147, ZSTK474, GDC-0941, (NVP)-BEZ235, AS-252424, TGX-221, XL-765, wortmannin, PI- 103, Perifosine (KRX-0401), Triciribine (API-2), SR13688, AR-67 (DB-67), AR-42, GSK690693, KP372- 1, A-443654, MK-2206, Rapamycin (Sirolimus
  • Deforolimus (Ridaforolimus, AP-23573), AZD-8055, OSI-027, INK- 128, PP-242. These and further therapeutic agents are described in further detail in Chapell et al, Oncotarget, 2011, 2, 135-164.
  • a compound that is an inhibitor of one or both of TBK1 and IKK8, or a down-regulator of the expression of one or both of TBK1 and ⁇ may be used in combination with an inhibitor of the Ras/Raf/Mek/Erk pathway, in particular in tumours that contain activating mutations in this pathway.
  • Examples of therapeutic agents that inhibit the Ras/Raf/Mek Erk pathway are: Tipifarnib (ZarnestaTM, Rl 15777), Sorafenib (Nexavar®, BAY43-9006), AAL-881, LBT-613, RAF265, XL281, SB-590885, PLX-4720, PLX-4032, L-779,450, GW5074, SB-699393, CI-1040 (PD184352), PD0325901, XL518, Selumtinib (AZD6244, ARRY-142886), RDEA119 (BAY 869766), PD098059, U0126, SL-327. These and further therapeutic agents are described in further detail in Chapell, 2011.
  • the compound of the invention may be co-formulated with the additional agent, or it may be formulated separately and administered consecutively, simultaneously or sequentially with the additional agent.
  • the amount of active ingredient which is required to achieve a therapeutic effect will, of course, vary with the particular compound, the route of administration, the subject under treatment, including the type, species, age, weight, sex, and medical condition of the subject and the renal and hepatic function of the subject, and the particular cancer being treated, as well as its severity.
  • An ordinarily skilled physician or veterinarian can readily determine and prescribe the effective amount of the drug required to prevent, counter or arrest the progress of the condition.
  • Oral dosages of the present invention when used for the indicated effects, will range between about 0.01 mg per kg of body weight per day (mg/kg/day) to about 100 mg/kg/day, preferably 0.01 mg per kg of body weight per day (mg/kg/day) to 10 mg/kg/day, and most preferably 0.1 to 5.0 mg/kg/day, for adult humans.
  • the compositions are preferably provided in the form of tablets or other forms of presentation provided in discrete units containing 0.01, 0.05, 0.1, 0.5, 1.0, 2.5, 5.0, 10.0, 15.0, 25.0, 50.0, 100, and 500 milligrams of the active ingredient for the symptomatic adjustment of the dosage to the patient to be treated.
  • a medicament typically contains from about 0.01 mg to about 500 mg of the active ingredient, preferably from about 1 mg to about 100 mg of active ingredient.
  • the most preferred doses will range from about 0.1 to about 10 mg/kg/minute during a constant rate infusion.
  • compounds of the present invention may be administered in a single daily dose, or the total daily dosage may be administered in divided doses of two, three or four times daily.
  • preferred compounds for the present invention can be administered in intranasal form via topical use of suitable intranasal vehicles, or via transdermal routes, using those forms of transdermal skin patches well known to those of ordinary skill in the art.
  • the dosage administration will, of course, be continuous rather than intermittent throughout the dosage regimen.
  • the pharmaceutical formulations according to the invention include those suitable for oral, parenteral (including subcutaneous, intradermal, intramuscular, intravenous (bolus or infusion), and intraarticular), inhalation (including fine particle dusts or mists which may be generated by means of various types of metered doses pressurized aerosols), nebulizers or insufflators, rectal, intraperitoneal and topical (including dermal, buccal, sublingual, and intraocular) administration, although the most suitable route may depend upon, for example, the condition and disorder of the recipient.
  • the formulations may conveniently be presented in unit dosage form and may be prepared by any of the methods well known in the art of pharmacy. All methods include the step of bringing the active ingredient into association with the carrier which constitutes one or more accessory ingredients. In general the formulations are prepared by uniformly and intimately bringing into association the active ingredient with liquid carriers or finely divided solid carriers or both and then, if necessary, shaping the product into the desired formulation.
  • Formulations of the present invention suitable for oral administration may be presented as discrete units such as capsules, cachets, pills or tablets each containing a predetermined amount of the active ingredient; as a powder or granules; as a solution or a suspension in an aqueous liquid or a non-aqueous liquid; or as an oil-in-water liquid emulsion or a water-in-oil liquid emulsion.
  • the active ingredient may also be presented as a bolus, electuary or paste.
  • the present compounds can, for example, be administered in a form suitable for immediate release or extended release. Immediate release or extended release can be achieved by the use of suitable pharmaceutical compositions comprising the present compounds, or, particularly in the case of extended release, by the use of devices such as subcutaneous implants or osmotic pumps.
  • compositions for oral administration include suspensions which can contain, for example, microcrystalline cellulose for imparting bulk, alginic acid or sodium alginate as a suspending agent, methylcellulose as a viscosity enhancer, and sweeteners or flavoring agents such as those known in the art; and immediate release tablets which can contain, for example, microcrystalline cellulose, dicalcium phosphate, starch, magnesium stearate, calcium sulfate, sorbitol, glucose and/or lactose and/or other excipients, binders, extenders, disintegrants, diluents and lubricants such as those known in the art.
  • Suitable binders include starch, gelatin, natural sugars such as glucose or beta-lactose, corn sweeteners, natural and synthetic gums such as acacia, tragacanth or sodium alginate, carboxymethylcellulose, polyethylene glycol, waxes and the like.
  • Disintegrators include without limitation starch, methylcellulose, agar, bentonite, xanthan gum and the like.
  • the compounds of the invention can also be delivered through the oral cavity by sublingual and/or buccal administration. Molded tablets, compressed tablets or freeze-dried tablets are exemplary forms which may be used.
  • compositions include those formulating the present compound(s) with fast dissolving diluents such as mannitol, lactose, sucrose and/or cyclodextrins. Also included in such formulations may be high molecular weight excipients such as celluloses (avicel) or polyethylene glycols (PEG). Such formulations can also include an excipient to aid mucosal adhesion such as hydroxy propyl cellulose (HPC), hydroxy propyl methyl cellulose (HPMC), sodium carboxy methyl cellulose (SCMC), maleic anhydride copolymer (e.g., Gantrez), and agents to control release such as polyacrylic copolymer (e.g. Carbopol 934).
  • fast dissolving diluents such as mannitol, lactose, sucrose and/or cyclodextrins.
  • high molecular weight excipients such as celluloses (avicel) or polyethylene glycols (P
  • Lubricants, glidants, flavors, coloring agents and stabilizers may also be added for ease of fabrication and use.
  • Lubricants used in these dosage forms include sodium oleate, sodium stearate, magnesium stearate, sodium benzoate, sodium acetate, sodium chloride and the like.
  • the oral drug components can be combined with any oral, nontoxic, pharmaceutically acceptable inert carrier such as ethanol, glycerol, water, and the like.
  • Formulations 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.
  • 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 (lyophilised) condition requiring only the addition of the sterile liquid carrier, for example saline or water-for-injection, immediately prior to use.
  • Extemporaneous injection solutions and suspensions may be prepared from sterile powders, granules and tablets of the kind previously described.
  • compositions for parenteral administration include injectable solutions or suspensions which can contain, for example, suitable non-toxic, parenterally acceptable diluents or solvents, such as mannitol, 1,3-butanediol, water, Ringer's solution, an isotonic sodium chloride solution, or other suitable dispersing or wetting and suspending agents, including synthetic mono- or diglycerides, and fatty acids, including oleic acid, or
  • compositions for nasal, aerosol or inhalation administration include solutions in saline, which can contain, for example, benzyl alcohol or other suitable preservatives, absorption promoters to enhance bioavailability, and/or other solubilizing or dispersing agents such as those known in the art.
  • Formulations for rectal administration may be presented as a suppository with the usual carriers such as cocoa butter, synthetic glyceride esters or polyethylene glycol. Such carriers are typically solid at ordinary temperatures, but liquefy and/or dissolve in the rectal cavity to release the drug.
  • Formulations for topical administration in the mouth include lozenges comprising the active ingredient in a flavoured basis such as sucrose and acacia or tragacanth, and pastilles comprising the active ingredient in a basis such as gelatin and glycerine or sucrose and acacia.
  • exemplary compositions for topical administration include a topical carrier such as Plastibase (mineral oil gelled with polyethylene). It should be understood that in addition to the ingredients particularly mentioned above, the formulations of this invention may include other agents conventional in the art having regard to the type of formulation in question, for example those suitable for oral administration may include flavouring agents.
  • si ⁇ RNA agents are well known to those skilled in the art, for example as described in Xie et a ⁇ ., Drug Discovery Today, 2006 11 (1-2), 67-73. "Harnessing in vivo siRNA delivery for drug discovery and therapeutic development".
  • the invention provides a compound that is an inhibitor of one or both of TBK1 and ⁇ , or a down-regulator of the expression of one or both of TBK1 and ⁇ , for use in a method of treating a cancer that is dependent on the PDkinase pathway comprising:
  • the cancer may be a cancer as described above.
  • the compound may be a compound as described above.
  • a method of treating cancer in an individual in whom the cancer is dependent on the PDkinase pathway comprising administering to the individual a compound that is an inhibitor of one or both of TBKl and ⁇ , or a down- regulator of the expression of one or both of TBKl and ⁇ .
  • the cancer may be a cancer as described above.
  • the compound may be a compound described above.
  • a method of treating cancer in an individual comprising:
  • the cancer may be a cancer as described above.
  • the compound may be a compound as described above.
  • the invention further provides a method of determining the susceptibility of a cancer in an individual to treatment with a compound that is an inhibitor of one or both of TBKl and ⁇ , or a down-regulator of the expression of one or both of TBKl and ⁇ , comprising determining whether the cancer is dependent on the PDkinase pathway, whereby a dependence on the PDkinase pathway indicates that the cancer in the individual has an increased susceptibility to a compound that is an inhibitor of one or both of TBKl and ⁇ , or a down-regulator of the expression of one or both of TBKl and ⁇ .
  • the cancer may be a cancer as described above.
  • the compound may be a compound as described above.
  • the invention further provides a method of determining a suitable treatment for an individual with a cancer comprising:
  • a suitable treatment whereby a compound that is an inhibitor of one or both of TBKl and ⁇ is suitable in the treatment of an individual with a cancer that is dependent on the PDkinase pathway.
  • the cancer may be a cancer as described above.
  • the compound may be a compound as described above.
  • a method of determining a suitable treatment may including determining that a particular treatment is not suitable in the subject. For example, if a cancer is found not to be dependent on the PDkinase pathway, then an inhibitor of one or both of TBKl and ⁇ might not be suitable in the treatment of the individual.
  • the invention further provides a method of predicting the response of a cancer condition in an individual to a treatment with a compound that is an inhibitor of one or both of TBKl and ⁇ by determining whether the cancer is dependent on the PDkinase pathway.
  • the cancer may be a cancer as described above.
  • the compound may be a compound as described above.
  • a cancer may be identified as dependent on the PDkinase pathway by determining the presence in cancer cells from the individual of one or more variations, for example, polymorphisms or mutations, in a nucleic acid encoding a polypeptide that is a component of the PDkinase pathway.
  • a cancer may be identified as being deficient in PTEN activity by determining the presence in cancer cells from the individual of one or more variations, for example, polymorphisms or mutations, in a nucleic acid encoding a PTEN polypeptide.
  • a cancer may be identified as having overactive PDkinase signalling by determining the presence in cancer cells from the individual of one or more variations, for example, polymorphisms or P1K3CA mutations, in a nucleic acid encoding an overactive PD kinase pi 10 polypeptide.
  • a cancer may be identified as being deficient in PTEN activity by determining the presence in cancer cells from the individual of one or more variations, for example, polymorphisms or mutations, in a nucleic acid encoding a PTEN polypeptide.
  • a cancer may be identified as having overactive PDkinase signalling by determining the presence in cancer cells from the individual of one or more variations, for example, polymorphisms or mutations, in a nucleic acid encoding an PD kinase pi 10 polypeptide, for example PIK3CA, which polymorphisms or mutations cause the PI3 kinase to be overactive, or one or more variations in the nucleic acid controlling expression of a PI3 kinase pi 10 polypeptide, for example PIK3CA, which cause the PI3 kinase to be over-expressed.
  • Sequence variations such as mutations and polymorphisms may include a deletion, insertion or substitution of one or more nucleotides, relative to the wild-type nucleotide sequence.
  • the variation may be a gene amplification.
  • the one or more variations may be in a coding or non-coding region of the nucleic acid sequence and, may reduce or abolish the expression of a negatively regulating pathway component polypeptide, for example PTEN.
  • the variant nucleic acid may encode a variant polypeptide which has reduced or abolished activity or may encode a wild- type polypeptide which has little or no expression within the cell, for example through the altered activity of a regulatory element.
  • a variant nucleic acid may have one, two, three, four or more mutations or polymorphisms relative to the wild-type sequence. Furthermore, a cancer may be determined to be heterozygous or homozygous in mutant forms of one or more components, leading to a dependency on the PDkinase pathway.
  • a particular nucleic acid sequence for example a nucleic acid sequence which has a mutation or polymorphism that reduces or abrogates the expression or activity of a PDkinase pathway component, or that increases the expression or activity of a PDkinase pathway component, as the case may be depending on the role that the component plays in the pathway or its control.
  • sequence information can be retained and subsequently searched without recourse to the original nucleic acid itself.
  • scanning a database of sequence information using sequence analysis software may identify a sequence alteration or mutation.
  • Methods according to some aspects of the present invention may comprise determining the binding of an oligonucleotide probe to nucleic acid obtained from the sample, for example, genomic DNA, RNA or cDNA.
  • the probe may comprise a nucleotide sequence which binds specifically to a nucleic acid sequence which contains one or more mutations or
  • the oligonucleotide probe may comprise a label and binding of the probe may be determined by detecting the presence of the label.
  • a method may include hybridisation of one or more (e. g. two) oligonucleotide probes or primers to target nucleic acid. Where the nucleic acid is double-stranded DNA, hybridisation will generally be preceded by denaturation to produce single- stranded DNA.
  • hybridisation may be as part of a PCR procedure, or as part of a probing procedure not involving PCR.
  • An example procedure would be a combination of PCR and low stringency hybridisation.
  • Binding of a probe to target nucleic acid may be measured using any of a variety of techniques at the disposal of those skilled in the art.
  • probes may be radioactively, fluorescently or enzymatically labelled.
  • Other methods not employing labelling of probe include examination of restriction fragment length polymorphisms, amplification using PCR, RN'ase cleavage and allele specific oligonucleotide probing.
  • Probing may employ the standard Southern blotting technique. For instance, DNA may be extracted from cells and digested with different restriction enzymes. Restriction fragments may then be separated by electrophoresis on an agarose gel, before denaturation and transfer to a nitrocellulose filter. Labelled probe may be hybridised to the DNA fragments on the filter and binding determined.
  • Suitable selective hybridisation conditions for oligonucleotides of 17 to 30 bases include hybridization overnight at 42 °C in 6X SSC and washing in 6X SSC at a series of increasing temperatures from 42 °C to 65 °C.
  • Other suitable conditions and protocols are described in Molecular Cloning: a Laboratory Manual: 3rd edition, Sambrook & Russell (2001) Cold Spring Harbor Laboratory Press NY and Current Protocols in Molecular Biology, Ausubel et al. eds. John Wiley & Sons (1992).
  • Nucleic acid which may be genomic DNA, RNA or cDNA, or an amplified region thereof, may be sequenced to identify or determine the presence of polymorphism or mutation therein.
  • a polymorphism or mutation may be identified by comparing the sequence obtained with the database sequence of the component, as set out above. In particular, the presence of one or more polymorphisms or mutations that cause abrogation or loss of function of the
  • sequence Navigator software may be determined using any one of a range of standard techniques. Sequencing of an amplified product may, for example, involve precipitation with isopropanol, resuspension and sequencing using a TaqFS+ Dye terminator sequencing kit. Extension products may be electrophoresed on an ABI 377 DNA sequencer and data analysed using Sequence Navigator software.
  • a specific amplification reaction such as PCR using one or more pairs of primers may conveniently be employed to amplify the region of interest within the nucleic acid sequence, for example, the portion of the sequence suspected of containing mutations or
  • the amplified nucleic acid may then be sequenced as above, and/or tested in any other way to determine the presence or absence of a mutation or polymorphism which reduces PTEN function, or enhances PDkinase function, or the overall flux through the PDkinase pathway.
  • the polymerase chain reaction has been reviewed, for instance in "PCR protocols; A Guide to Methods and Applications", Eds. Innis et al, 1990, Academic Press, New York, Mullis et al, Cold Spring Harbor Symp; Ehrlich (ed), PCR technology, Stockton Press, NY, 1989; and Ehrlich et al, Science, 1991, 252, 1643-1650.
  • PIK3CA A number of methods have been described for the detection of mutant forms of PIK3CA.
  • the most common activating PIK3CA mutations found in cancer are missense mutations resulting from a single base pair change in either: exon 20, coding the helical domain; or exon 9, coding the kinase domain.
  • Examples of common mutations in exon 20 include the following nucleotide changes (corresponding amino acid changes shown in brackets) A3140G (H1047R), A3150T (H1047L).
  • Examples of common mutation in exon 9 include the following nucleotide changes (corresponding amino acid changes shown in brackets) G1633A (E545K), G1624A (E542K), G1634A (E545G).
  • SSCP single strand conformational polymorphism
  • PIK3CA mutants is the use of allele specific PCR using primers specifically designed against common mutants (H1047L, H1047R, E542K and E545K) using the Amplification Refractory Mutation System (ARMSTM, AstraZeneca) and ScopionsTM (DxS), a fluorescent signalling system that detects PCR products (Board et al., Clinical Chemistry, 2008, 54 (4), 757-760).
  • Amplification Refractory Mutation System AstraZeneca
  • ScopionsTM DxS
  • Colonies which lack a functional ADE2 protein appear red due to the accumulation of the ADE2 substrate (Zhang et al., Oncogene 2000, 19, 4346-4353).
  • Another method for the detection of PTEN deletions involves the use of FISH probes directed at the PTEN-locus on chromosome region 10q23 (Chang et al., Leukemia Research, 2006, 30 262- 265).
  • the presence of one or more variations in a component of the PDkinase pathway may be determined by detecting, in one or more cells of a test sample, the presence of the variant component polypeptide, or the absence or reduced amount of the non-mutated component polypeptide (e.g. detection of the levels of PTEN) which is encoded by the nucleic acid sequence.
  • a method of identifying a cancer cell in a sample from an individual as dependent on the PDkinase pathway may include contacting a sample with a specific binding member directed against a variant (e. g. a mutant) polypeptide component of the pathway, and determining binding of the specific binding member to the sample.
  • Another method of identifying a cancer cell in a sample from an individual as dependent on the PDkinase pathway may include contacting a sample with a specific binding member directed against a phosphorylated polypeptide component of the pathway, and determining binding of the specific binding member to the sample.
  • a preferred phosphorylated polypeptide for this purpose is phospho-PDKl (Vasudevan et ah, 201 1).
  • Binding of the specific binding member to the sample may be indicative of the presence of the variant polypeptide component, or phosphorylated polypeptide component, of the pathway in a cell within the sample.
  • Preferred specific binding molecules for use in aspects of the present invention include antibodies and fragments or derivatives thereof ('antibody molecules').
  • the reactivities of a binding member such as an antibody on normal and test samples may be determined by any appropriate means. Tagging with individual reporter molecules is one possibility.
  • the reporter molecules may directly or indirectly generate detectable, and preferably measurable, signals.
  • the linkage of reporter molecules may be directly or indirectly, covalently, e. g. via a peptide bond or non-covalently. Linkage via a peptide bond may be as a result of recombinant expression of a gene fusion encoding binding molecule (e. g. antibody) and reporter molecule.
  • Antibodies for PTEN are commercially available and can be used to assess the amounts of PTEN using standard techniques such as western blotting.
  • Reverse-phase protein array (RPPA) analysis a high throughput technique that can be used to assess multiple tissue samples, has also been used to assess levels of PTEN protein (Vasudevan et ah, 2011).
  • Antibodies for phophorylated kinases such as phospho-PDK- 1 or phospho-mTOR are readily available from commercial suppliers. Sotiriou et al. have described a signature for PI3 kinase pathway activation in breast cancer samples using microarray analysis, this or similar approaches could also be used for identifying tumours with activation of the PI3 kinase pathway (US2011/0038862).
  • tumours that contain a combination of mutations or transformations to members that can activate the PI3 kinase pathway may also be considered dependant on the PI3 kinase pathway.
  • this could take the form of the combination of a PIK3CA mutation and the loss or low levels of PTEN.
  • it may be the over-expression of a growth factor receptor which can activate the PI3 kinase pathway.
  • a growth factor receptor which can activate the PI3 kinase pathway.
  • ErbB2 in breast cancer in combination with a reduction in the amount of PTEN, or with an activating mutation in PIK3CA.
  • Another example would be in activating mutation in a Ras protein for example K-Ras and a mutation in PIK3CA or a reduction in the amount of PTEN.
  • Compound IC50 values were determined at the apparent Km for ATP (20 ⁇ ) based on a radiometric assay (Invitrogen) using 8 or 10 point curves in duplicate.
  • the final reaction conditions contained 400 nM fluorescein-IkBa substrate (DRHDSGLDSMKDE), 20 ⁇ ATP, 2 nM or 8 nM ⁇ or TBK1 kinase respectively, and 3% DMSO in kinase assay buffer consisting of 50 mM HEPES (pH 7.5), 10 mM MgCl, 1 mM EGTA, 0.01% Brij-35.
  • Compound dilutions were prepared from 10 mM DMSO stocks by dilution into DMSO. Compound dilution series were further diluted in kinase assay buffer to give a 12% DMSO stock, the final concentration in the assay being 3% DMSO.
  • the kinase phosphorylation assay was initiated by the addition of the kinase and the reaction was allowed to proceed for 1 hr or 2.5 hr for ⁇ and TBK-1 kinase respectively. Both conditions were within the linearity of the phosphorylation.
  • the reaction was stopped by the addition of 10 mM EDTA, and phosphorylation was detected after a lhr incubation with 1.5 nM terbium-labelled antibody against phosphorylation at Serine 32 on the IkBa peptide, both in TR-FRET dilution buffer (Invitrogen).
  • the compound DMX-A had an IC 50 for inhibition of ⁇ of ⁇ 30nM; and an IC 50 for inhibition of TBK-1 of ⁇ 30nM.
  • the compounds DMX-A, B and C were tested against a number of enzymes that are generally considered members of the PI3 kinase pathway. DMX-A, B and C showed high levels of selectivity for TBKl and ⁇ over the other enzymes tested: i.e.
  • PDK-1, mTOR, ⁇ , ⁇ , PDKy and AKT-l The assays were performed against ⁇ and TBK-1 as described above.
  • the counterscreening assays were performed using radiolabeled P 33 ATP using the Kinase Hotspot Technology (Reaction Biology Corp). Screening was carried out at the Km for ATP for ⁇ - ⁇ and TBK-1 and at 10 ⁇ ATP for the other enzymes, with the exception of screening of DMX-A against AKT-1 which was screened at ⁇ ATP.
  • PTEN wild type comparator HCT-116 cells Both cell lines were screened with a siRNA library targeting -720 kinases and kinase related genes and the affects of each siRNA on cell viability were assessed.
  • SMARTpool siRNAs were transfected with SMARTpool siRNAs using Dharmafect 3 (DF3- Dharmacon).
  • the siRNA library siARRAY - targeting 714 known and putative human protein kinase genes
  • Each well in this library contained a SMARTpool of four distinct siRNA species targeting different sequences of the target transcript.
  • Each plate was supplemented with siCONTROL (ten wells, Dharmacon (USA)).
  • Cells were transfected with the SMARTpool library, where each well of the 96 well-plate contained a pool of four different siRNAs (a SMARTpool) targeting one gene. After five population doublings, cell viability in each well was estimated by use of a highly sensitive luminescent assay measuring cellular ATP levels (Cell Titre Glo, Promega). To identify loss of viability/failure to proliferate effects in each cell line, luminescence readings from each well were log transformed and then centred by the plate median, to account for plate-to-plate variation common in high- throughput screens. Well position effects were identified and eliminated and the quality of data from each plate estimated by calculating Z' -factors (Zhang et al., J. Biomol.
  • NPI scores normalised percent inhibition
  • PTEN +/+ and PTEN -/- HCT 116 human tumour cell lines wherein a NPI score ⁇ 1 indicates a loss of viability, identified that PTEN null cells were particularly sensitive to the knockdown of TBK-1.
  • the NPI results of this experiment are shown in Figure 1.
  • TBK- 1 siRNAs show a selective lethality towards (PTEN-/-)-HCTl 16 cells.
  • DMX-A and DMX-B were dosed at 24 and 96 hours. After 7 days, cell viability was measured using CellTiter Glo Luminescent Cell Viability Assay (Promega, USA). Surviving fractions were calculated and drug sensitivity curves plotted as previously described (Farmer et ah, Nature, 2005 434 (7035), 917-921).
  • DMX-C cells were dosed every three days after initial plating. After 14 days, cell viability was measured using CellTiter Glo Luminescent Cell Viability Assay (Promega, USA).
  • Figure 2(a) shows the DMX-A results
  • Figure 2(b) shows the DMX-B results
  • Figure 2(c) shows the DMX-C results.
  • a leftward shift for the (PTEN-/-) curve indicates a greater sensitivity of this cell type towards the inhibitors tested.
  • an increased sensitivity of the PTEN null cells to TBK- 1 inhibition is indicated by a leftwards shift of the curve relative to the PTEN +/+ HCT 116 line.
  • Figure 6(a) and (b) show SF80 (concentration at which 80% of cells survive) and SF50 (concentration at which 50% of cells survive) results for cell lines with PTEN and/or PIKCA mutations and those with wild type PTEN and PIKCA.
  • DMX-C shows enhanced sensitivity towards cell lines containing either a PTEN mutation or a PIK3CA mutation compared to cell lines that contain both PTEN and PIK3CA wild-type.
  • STR profiling short tandem repeat profiling was used to verify the origin of the panel of cell lines. 8 STR loci were simultaneously amplified in a multiplex PCR reaction (Promega PowerPlex 1.2 System) and compared with the ATCC database. Where appropriate PTEN gene mutation status was determined by Sanger sequencing and western blot analysis and PIK3CA mutation status defined by Sanger sequencing. All of the cell lines used in the panel were human breast cancer cell lines with the exception of A2780 which is a human ovarian cancer cell line.
  • F- TBKI/IKKE sensitising to BEZ235 T47D cell line was purchased from ATCC (USA) and maintained according to the suppliers instructions. The authenticity of the cell line was verified as described above for panel of tumour cell lines. The sensitivity of this cell line to the PI3 kinase inhibitor BEZ235 was measured in the presence or absence of TBK- 1 siRNA, shown in table 1 below. Cells were reverse transfected with siRNA and after 48 hours exposed to BEZ235. BEZ235 exposure was maintained for 7 days, with BEZ235 being replenished 3 days after the initial dosing.
  • siRNAs Two out of four TBK-1 siRNAs increased sensitivity to BEZ235, in a similar fashion to the positive control siRNA targeting PIK3CA.
  • the siRNAs used were from Dharmacon siGenome® SMARTpool siRNA human protein kinase library. Results of this experiment are shown in Figure 7.
  • Qneg represents the negative control (i.e. no siRNA)
  • PIK3CA represents a positive control targeting PI 3CA
  • TBK_l-4 represents 4 different siRNAs targeting TBKl.

Abstract

The invention provides a compound that is an inhibitor of one or both of TBK1 and IKKε, or a down-regulator of the expression of one or both of TBK1 and IKKε, for use in a method of treating a cancer that is dependent on the PI3kinase pathway. The invention further provides a method of treating cancer in an individual in whom the cancer is dependent on the PI3kinase pathway, comprising administering to the individual a compound that is an inhibitor of one or both of TBK1 and IKKε, or a down-regulator of the expression of one or both of TBK1 and IKKε.

Description

Compounds and their uses
This invention relates to compounds that are inhibitors of one or both of the kinases TBK1 and ΙΚΚε (otherwise referred to as I-kappa-B-kinase-3 (IKK3), or down-regulators of the expression of one or both of TBK1 and ΙΚΚε, and their use in the treatment of cancers that are dependent on the PDkinase-pathway. The invention also relates to methods of diagnosis, methods of determining the susceptibility of a cancer to particular treatments, and methods of determining suitability of particular treatments. There are approximately 500 different known protein kinases. Protein kinases serve to catalyze the phosphorylation of an amino-acid side-chain in various substrate proteins.
Studies have shown that protein kinases play a key role in many cell functions, including signal transduction, transcriptional regulation, cell motility, and cell division. The PDkinase signalling pathway is an important cell survival pathway which involves a number of protein kinases, and also lipid kinases of the phosphoinositide 3-kinase (PI3 kinase) family whose primary role is to phosphorylate inositol lipids. The phosphorylated inositol lipids subsequently play an important signalling role by membrane recruitment of effectors having phosphoinositide-recognition motifs. Increased signalling through this pathway is seen in many tumours (Holmes, Nature Reviews Drug Discovery, 2011, 10, 563- 564; Engleman, Nature Reviews Cancer, 2009, 9, 550-562; Stephens et al., Current Opinion in Pharmacology, 2005, 5, 357-365).
Three different classes of PI3 kinases have been identified. All members of the family share a common function in phosphorylating the 3ΌΗ group of phosphatidylinositols (Ptdlns), however they act preferentially on different Ptdlns substrates. The PI3 kinases that are most commonly implicated in cancer are the class I PI3 kinases (Engleman, 2009).
PDkinase signalling by class I PD kinases is driven by the kinase function of the pi 10 catalytic domain which primarily converts PtdIns(4,5)P2 into PtdIns(3,4,5)P3. Proteins that contain a pleckstrin homology (PH) domain become associated with the cell membrane by binding to Ptdlns(3, 4,5)P > and when localised here they can become further activated by other proteins. A major pathway for transduction of the downstream signal from PDkinase involves the activation of AKT. AKT becomes associated with the cell membrane via its PH domain, from where it can become activated by phopsphorylation of Threonine 308 and Serine 473 by PDK-1, and the mTOR/Rictor complex (mTorc2), respectively. AKT can then provide a powerful growth/survival signal by phosphorylation of a number of proteins including TSC-2, which indirectly activates the mTOR/Raptor complex (mTorcl), GSK3 and members of the FOXO family of proteins (Engleman, 2009; Stephens et ah, 2005)
Although AKT is the PDkinase pathway downstream effector most widely implicated in cancer, there are also AKT independent pathways, which include the Bruton tyrosine kinase (BTK), the TEC families of non-receptor tyrosine kinases and serum- and glucocorticoid- regulated kinases (SGKs) (Qui et al., Oncogene, 2000, 19, 5651-5661; Vasudevan et al., Cancer Cell, 2009, 16 (1), 21-32).
There are 4 isoforms of the catalytic pi 10 subunit α, β γ, δ all of which contain binding sites for a regulatory subunit, p85-related proteins in the case of the class IA catalytic subunits pi 10a, pi 10β and pi 10δ and plOl or the closely related p84-p87 subunit in the case of the class IB isotype pi 10γ. The class I catalytic subunits also contain a binding site for Ras proteins. There are a number of mechanisms by which the PDkinase pathway can be activated in cancer:
Receptor tyrosine kinases, such as EGFR or Erb-2, can activate the PI3 kinase pathway by recruiting the regulatory subunit thereby releasing the auto-inhibitory role that this would normally play (Nagata et al., Cancer Cell, 2004, 6(2), 117-27). This mechanism of activation is primarily associated with class IA isoforms (Williams et al., Biochem. Soc. Trans, 2009, 37, 615-626).
The pi 10 isoforms can be activated directly by binding to activated Ras proteins. This is the primary mode of activation for pi 10γ, and may also be required for activation of ρΐ ΐθβ (Kang et al, PNAS, 2006, 103 (5) 1289- 1294, Andrews et al, Science Signalling, 2007, 407, cm2, "PDK Class IB Pathway" ).
Mutations in the one of the PI3 kinase catalytic subunits can result in constituitive activation of the pathway (Engleman, 2009). Mutations in the regulatory p85 subunit can result in loss of its inhibitory activity and hence constitutive activation of the pathway (Engleman, 2009).
One of the PI3 Kinase catalytic subunits may be over-expressed resulting in amplification of signalling through the PI3 kinase pathway (Engleman 2009). Loss or inactivation of key phosphatases such as PTEN, which reverse the action of the PI3 kinases by dephophorylating the 3 position of Ptdlns(3, 4,5)P3 can increase flux through the PI3 kinase pathway (Engleman, 2009; Nagata et al , Cancer Cell, 2004, 6(2), 117-27).
A downstream kinase such as PDK- 1 or AKT may be over-expressed or possess an activating mutation (Engleman, 2009).
Phosphatase and tensin homolog (PTEN) is a key enzyme that inhibits signalling through the PDkinase pathway and it therefore has tumour suppressing effects. Mutation in the PTEN gene (PTEN(-) mutation) inactivates the enzyme and leads to increased flux through the pathway. PTEN is one of the most commonly mutated or lost tumour suppressors in human cancer. PTEN loss has been reported in endometrial cancer (35%) and glioblastoma (54%) as well as in other cancer types such as breast (32%), colo-rectal carcinoma (30%), prostate (27%) and lung cancer (19%) (Frattini et al, Br. J. Cancer, 2007, 97(8), 1139-45; Nagata et al, 2004). Low levels of PTEN are also reported in a number of cancers. This can occur for example as the result of the loss of one functional copy of the PTEN gene, often described as haploinsufficiency. It is believed that low levels of PTEN can contribute significantly to the oncogenic state especially when accompanied by other changes/mutations that are able to further activate the PI3 kinase pathway (Ying et al., Cancer Discovery, 2011, 1 (2), 158-169; Nagata et al., 2004). It has also been shown that receptor tyrosine kinase signalling can deactivate PTEN. It has been shown that part of the mechanism of action of the Erb2 antibody trastuzumab involves inhibition of Src kinase activity. Src kinase is believed to be responsible for deactivating PTEN in breast cancers where Erb2 is over-expressed, by phophorylation of key tyrosine residues on PTEN. Furthermore, loss of PTEN has been shown to be associated with resistance to trastuzumab, and inhibition of PI3 kinase signalling can increase sensitivity to trastuzumab in PTEN deficient cells (Nagata et al., 2004). PBkinases increase flux through the PDkinase pathway, and so PI3 kinase activating mutations are oncogenic, for example mutations classified as PIK3CA (catalytic - alpha polypeptide) leads to enhanced signalling through the pathway. PIK3CA mutation can be found in a range of malignancies such as colon, lung, ovarian, liver, brain, stomach and breast cancers (Samuels et al., Science, 2004, 304(5670), 554; Engleman, 2009). Genetic amplification of PIK3CA is also found in a number of tumour types including head and neck, squamous cell lung carcinoma, cervical, gastric and oesophageal (Engleman, 2009).
I-kappa-B-kinase epsilon, ΙΚΚε, (also known as ΙΚΚε, I-kappa-B-kinase-3 (IKK3), or inducible I-kappa-B-kinase (IKKi)), and TANK Binding Kinase- 1, TBK1 (also known as T2K or NF-kappa B-activating kinase), are serine-threonine kinases.
Aberrant ΙΚΚε activity has been linked to a number of disease states including cancer and obesity. Studies have shown that the gene encoding ΙΚΚε (IKBE) is amplified and over- expressed in certain breast cancer cell lines and patient-derived tumours. Furthermore suppression of IKBE gene expression in these cell lines induces cell death (Boehm et al., Cell, 2007, 129, 1065- 1079). ΙΚΚε has also been shown to phosphorylate the estrogen receptor, and its activity has been linked to tamoxifen resistance in breast cancer tumours (Guo et al., The Journal of Biological Chemistry, 2010, 285, 3676-3684). ΙΚΚε is also frequently over-expressed in human ovarian cancer lines and primary tumours. Moreover ΙΚΚε over-expression renders cells resistant to cis-platin, whereas ΙΚΚε knockdown restores cis-platin sensitivity (Guo et al., The American Journal of Pathology, 2009, 175, 324-333). These observations suggest that ΙΚΚε inhibitors may show efficacy in the treatment of certain cancers. TBK1 has been linked to various cancer states although the published literature has not defined its exact role, or the appropriate clinical setting for use of a TBK- 1 inhibitor. The levels of TBK1 mRNA and protein are elevated in certain malignant colon and breast cancer cells. TBK1 is also recruited and activated by the RalB/Sec5 effector complex: in cancer cells, constitutive engagement of this pathway via chronic RalB activation, restricts the initiation of apoptotic programmes. The proto-oncogene KRAS (which encodes the protein K-Ras) is mutated in a wide array of human tumours, most of which are aggressive and respond poorly to standard therapies. The knockdown of TBK1 in a number of K-Ras dependant tumour cell lines has been shown to cause cell death. Although it is known that K- Ras can activate the PI3 kinase pathway it is believed that cell lines which have K-Ras mutations are not necessarily addicted to the PI3 kinase pathway. For example it has been shown in pancreatic ductal adenocarcinoma (PDAC) that low levels of PTEN are also required for activation of the PI3 kinase pathway (Ying et ah, 2011).
Both ΙΚΚε and TBKl have been shown to phosphorylate and activate AKT in a number of cancer cell lines (Ou et al, Molecular Cell, 2011, 41, 458-70; Xie et al, PNAS, 2011, 108, 16, 6474-6479). AKT is a major signalling kinase and is generally considered to be one of the enzymes involved in the PDkinase pathway. Despite this it has been shown that not all tumour cell lines that are dependent on the PDkinase pathway have high levels of activated AKT nor do they all respond to AKT knockdown (Vasudevan et al, 2009). Ou et al. reported that shRNA knockdown of TBKl in a number of NSCLC cell lines inhibited cell survival and that K-Ras mutation status alone was not sufficient to determine sensitivity. They also reported that a small molecule dual inhibitor of TBKl and ΙΚΚε kinase showed greater efficacy in cell lines that were susceptible to TBK-1 shRNA knockdown. Those cell lines which showed enhanced sensitivity to TBK-1 inhibition also showed reduction of AKT phosphorylation following treatment with the small molecule inhibitor, although the authors were unable to determine what factors conferred sensitivity to TBK-1 inhibition.
There remains a need for improved treatments of cancers. It is important for clinicians to know which mechanisms are driving the malignancy in a particular patient, and to use the appropriate treatment. For example a drug that targets PDkinase signalling pathway will be effective in tumours that use the pathway, but not effective in tumours that do not use it. The invention provides a compound that is an inhibitor of one or both of TBKl and ΙΚΚε, or a down-regulator of the expression of one or both of TBKl and ΙΚΚε, for use in a method of treating a cancer that is dependent on the PDkinase pathway. It has surprisingly been found by the current inventors that cancers that are deficient in PTEN activity are sensitive to compounds that are inhibitors of TBKl and ΙΚΚε. It has also been found by the current inventors that cancers which are deficient in PTEN activity are sensitive to down-regulation of the expression of TBKl . From the data obtained by the current inventors, it is anticipated that such cancers are also are sensitive to down-regulation of the expression of ΙΚΚε. It has also been found by the current inventors that cancers with overactive PDkinase signalling are sensitive to TBKl and ΙΚΚε inhibitors. From the data obtained by the current inventors, it is anticipated that cancers with overactive PDkinase signalling are sensitive to down-regulation of the expression of one or both of TBKl and ΙΚΚε. In particular, the current inventors found that a human cell line in which both alleles of PTEN had been inactivated was lethally susceptible to siRNA inhibition of TBKl. That result demonstrates that TBKl activity is necessary for the survival of cells that are deficient in PTEN. The inventors also found that TBKl inhibitor compounds were selectively toxic to PTEN (-/-) cell lines over PTEN (+/+) cells lines, showing that TBKl activity is necessary for the survival of cells that are deficient in PTEN, but not for cells that have normally functional PTEN. The inventors have further found that breast tumour cell models that have PTEN and/or PIK3CA defects are more sensitive to TBKl inhibitors than tumour cell models with defects in other genes.
The findings demonstrate that a compound that is an inhibitor of one or both of TBKl and ΙΚΚε, or a down-regulator of the expression of one or both of TBKl and ΙΚΚε, is effective in the treatment of a cancer that is dependent on the PDkinase pathway. In particular, a compound that is an inhibitor of one or both of TBKl and ΙΚΚε, or a down-regulator of the expression of one or both of TBKl and ΙΚΚε, is effective in the treatment of a cancer that has a defect in one or both of the PTEN and PIK3CA genes.
BRIEF DESCRIPTION OF THE DRAWINGS
Figure 1 shows the Normalised Percent Inhibition (NPI) scores following the knockdown of 714 kinase genes with a panel of siRNAs in HCT116 human tumour cells in which both alleles of PTEN had been deactivated (PTEN7-) and HCT116 human tumour cells with wildtype PTEN (PTEN +/+).. Figures 2 (a) to (c) show the effects of small molecule dual inhibitors of the kinase domain of TBKl and ΙΚΚε on (PTEN-/-)-HCT116 and (PTEN +/+)-HCT116 human tumour cells, (a) shows the results for DMA-A, (b) shows the results for DMX-B and (c) shows the results for DMX-C. Figure 3 (a) shows the effect on survival fraction for escalating concentrations of the ΤΒΚΙ/ΙΚΚε inhibitor DMX-C on an ovarian tumour cell line which contains deactivating mutations in the PTEN gene.
Figures 3 (b) to (d) show the effects on survival fractions for escalating concentrations of the ΤΒΚΙ/ΙΚΚε inhibitor DMX-C on breast tumour cell lines which contains deactivating mutations in the PTEN gene.
Figure 3 (e) shows the effect on survival fraction for escalating concentrations of the ΤΒΚΙ/Ι Κε inhibitor DMX-C on a breast tumour cell lines which contains both deactivating mutations in the PTEN gene and activating mutations in the PIK3CA gene. Figures 4(a) to (c) show the effects on survival fractions for escalating concentrations of the ΤΒΚΙ/Ι Κε inhibitor DMX-C on breast tumour cell lines which contains activating mutations in the PIK3CA gene.
Figures 5 (a) to (e) show the effects on survival fractions for escalating concentrations of the ΤΒΚΙ/Ι Κε inhibitor DMX-C on breast tumour cell lines with wildtype PTEN and PIK3CA genes.
Figure 6 (a) shows SF80 (concentration at which 80% of cells survive) and (b) shows SF50 (concentration at which 50% of cells survive) results for DMX-C in cell lines with PTEN and/or PIKCA mutations and those with wild type PTEN and PIKCA.
Figure 7 shows the results of a study into TBK1 sensitising to BEZ235 (a dual PDkinase, mTOR inhibitor), the graph shows the survival fraction results for T47D breast tumour cells exposed to various concentrations of BEZ235 in the presence or absence of TBK-1 siRNA , with siRNA targeting PIK3CA used as a positive control.
TBK1 and ΙΚΚε have overlapping substrate specificities and it is believed that they have similar binding sites. Accordingly, many compounds that are inhibitors of one of TBK1 and Ι Κε are also inhibitors of the other. Compounds that are strong inhibitors of both TBK1 and ΙΚΚε are particularly advantageous compounds for use in the current invention.
The cancer to be treated may be a cancer that is deficient in PTEN activity. For example, the cancer may have reduced amounts of PTEN, or a level of PTEN function that is lower than PTEN function in normal cells in the individual. Expression levels of PTEN protein can be established by western blotting, or in situ hybridization, using commercially available antibodies to PTEN. For example, the cancer may include at least some cells carrying a loss of function mutation in the PTEN gene. The presence or absence of loss of function mutations may be assessed by determining the presence or absence of one or more mutations or polymorphisms in a nucleic acid sequence encoding at least a portion of the PTEN enzyme. The presence of such mutations can be established by Sanger Sequencing or Next Generation (Massively Parallel or Deep) Sequencing, or the use of mutation- specific PCR assays. The copy number of each gene (and thus loss of heterozygosity in the case of PTEN) can be established by Fluorescent In Situ Hybridisation (FISH). The cancer may be a cancer that has overactive PBkinase signalling. For example, the level of PBkinase signalling can be assessed by a functional assay known to or readily developed by the person skilled in the art.
For example, the cancer may include at least some cells carrying an activating PIK3CA mutation. The presence or absence of an activating PIK3CA mutation may be assessed by determining the presence or absence of one or more mutations or polymorphisms in a nucleic acid sequence encoding at least a portion of the PBkinase pi 10 catalytic subunit.
Overactive PB kinase signalling may be detected by an increase in phosphorylation of one or more of the downstream proteins, for example PDK-1 or mTOR. This can be measured in standard assay formats such as the use of a western blot, ELISA or in situ hybridization techniques, which all use commercially available phospho-protein specific antibodies.
Cancers with overactive PBkinase signalling include endometrial cancer, glioblastoma, colorectal carcinoma, myeloma and leukemia, and cancers of the breast, prostate, lung (including non small cell lung cancer (NSCLC)), ovary, liver, brain and stomach; in particular, endometrial cancer, glioblastoma, breast cancer, including tamoxifen resistant breast cancer, ovarian cancer, including cis-platin resistant ovarian cancer. Compounds of the invention are expected to be particularly useful in the treatment of such cancers. Cancers in the treatment of which the compounds of the invention are especially useful include cancers harbouring Ras mutations and Ras-dependant tumours, and cancers with PTEN loss of function mutatons or PIK3CA gain of function mutations. A compound for use in the invention may be a compound that is an inhibitor of one or both of TBK1 and ΙΚΚε. A compound is considered to be an inhibitor of TBK1 if it has an IC50 for inhibition of TBK-1 of <300nM. Preferred compounds have an IC50 for inhibition of TBK-1 of <100nM, particularly <50nM, more particularly <30nM. The IC50 for inhibition of TBK-1 can be assessed by assays known to the person skilled in the art, for example a luminescent kinase assay, or a time-resolved fluorescence assay. For example, a TBK-1 assay is available from Promega (Promega Corporation, 2800 Woods Hollow Road, Madison, WI53711-5399, USA, or see http://www.promega.com/). An example of an assay is described in the Examples section herein.
A compound is considered to be an inhibitor of ΙΚΚε if for inhibition of ΙΚΚε of <300nM; Preferred compounds have an IC50 for inhibition of ΙΚΚε of <100nM, particularly <50nM, more particularly <30nM. The IC50 for inhibition of ΙΚΚε can be assessed by assays known to the person skilled in the art, for example a luminescent kinase assay, or a time-resolved fluorescence assay. For example, an ΙΚΚε assay is available from Invitrogen (see http://www.invitrogen.com). An example of an assay is described in the Examples section herein.
A compound for use in the invention may be selective for TBK1 and/or ΙΚΚε over other kinases, or said compound may also inhibit other kinases. Some compounds have been found to be inhibitors of both TBK1 and ΙΚΚε.
Compounds that are inhibitors of one or both of TBK1 and ΙΚΚε include compounds selected from the group consisting of: (a) compounds described in PCT/GB2011/001075 (published as WO2012/010826), that is to say compounds of Formula (I):
Figure imgf000011_0001
(I)
in which:
R1 represents an aliphatic heterocyclyl group having 4, 5, 6 or 7 ring atoms, bonded to the phenyl group shown in formula I through a ring nitrogen atom, and optionally substituted by one or more substituents selected from halogen atoms; OH; =0; Ci-4alkyl, Ci-4alkoxy, Ci_4alkoxyCi_4alkyl, C2-4alkenyl and C2^alkynyl groups, each optionally substituted by one or more substituents independently selected from halogen atoms and NRaRb groups; NO2; CN; NRaRb; CORc; O.CO.Rc; C02Ra; NRa.CORc; NRaC02Rb; C(=NH)NH2; S02Rc; NRaS02Rc; and CH(CF3)NH2;
R2 represents a phenyl or heteroaryl group which is optionally substituted by one or more substituents independently selected from:
halogen atoms;
NRaRb;
Ci_4alkyl, C1-4alkoxy, Ci_4alkoxyCi_4alkyl, C2^alkenyl and C2-4alkynyl groups, each optionally substituted by one or more substituents independently selected from halogen atoms and NRaRb groups; and
-(CH2)P-R' in which p is 0, 1, 2, 3 or 4 and R' represents one of the following substituents: OH; N02; CN; CORc; O.CO.Rc; C02Rb; NRa.CORc; NRaC02Rb;
C(=NH)NH2; S02Rc; NRaS02Rc; and CH(CF3)NH2;
and/or which is optionally substituted on adjacent ring atoms by a group
-NRa.CO.(CH2)n- or -(CH2)n.CO.NRa- forming a fused ring;
Ra represents a hydrogen atom or a Ci-4alkyl group;
Rb represents a hydrogen atom, a Ci_4alkyl group optionally substituted by a group NRaRa, or a cycloalkyl group in which a C¾ moiety may be replaced by an oxygen atom or an NRa group; Rc represents a hydrogen atom, a group -NRaRb, or a Ci_4alkyl group optionally substituted by a group NRaRb;
or R and Rb together may, when attached to the same nitrogen atom, represent a -(CH2)m- group in which a C¾ moiety may be replaced by an oxygen atom or an -NRa- group;
m represents 4 or 5;
n represents 1 or 2; and
each of R3 and R4 independently represents a hydrogen atom or a Ci-4alkyl group; or a salt thereof.
(b) Compounds described in WO2011/046970, that is to say compounds of Formula (II):
Figure imgf000012_0001
and pharmaceutically acceptable salts thereof, wherein:
Rl , R2, R3, and R5 are independently chosen from the following groups: alkyl, alkylene, alkenyl, alkenylene, alkynyl, carbocycle, cycloalkyl, cycloalkenyl, heterocycle, aryl, heteroaryl, halo, hydro, hydroxyl, alkoxy, alkynyloxy, cycloalkyloxy, heterocycloxy, aryloxy, heteroaryloxy, arylalkoxy, heteroarylalkoxy, mercapto, alkylthio, arylthio, cycloalkylthio, arylalkyl, heteroarylalkyl, heteroarylalkenyl, arylalkynyl, haloalkyl, aldehyde, thiocarbonyl, O-carboxy, C-carboxy, carboxylic acid, ester, C-carboxy salt, carboxyalkyl,
carboxyalkenylene, carboxyalkyl salt, carboxyalkoxy, carboxyalkoxyalkanoyl, amino, aminoalkyl, nitro, O-carbamyl, N- carbamyl, O-thiocarbamyl, N-thiocarbamyl, C-amido, N- amido, aminothiocarbonyl, hydroxyaminocarbonyl, alkoxyaminocarbonyl, cyano, nitrile, cyanato, isocyanato, thiocyanato, isothiocyanato, sulfinyl, sulfonyl, sulfonamide, aminosulfonyl, aminosulfonyloxy, sulfonamidecarbonyl, alkanoylaminosulfonyl, trihalomethylsulfonyl, or trihalomethylsulfonamide, wherein any of the foregoing groups are optionally substituted at least once with alkyl, alkylene, alkenyl, alkenylene, alkynyl, carbocycle, cycloalkyl, cycloalkenyl, heterocycle, aryl, heteroaryl, halo, hydro, hydroxyl, alkoxy, alkynyloxy, cycloalkyloxy, heterocycloxy, aryloxy, heteroaryloxy, arylalkoxy, heteroarylalkoxy, mercapto, alkylthio, arylthio, cycloalkylthio, arylalkyl, heteroarylalkyl, heteroarylalkenyl, arylalkynyl, haloalkyl, aldehyde, thiocarbonyl, O-carboxy, C-carboxy, carboxylic acid, ester, C-carboxy salt, carboxyalkyl, carboxyalkenylene, carboxyalkyl salt, carboxyalkoxy, carboxyalkoxyalkanoyl, amino, aminoalkyl, nitro, O-carbamyl, N-carbamyl, O-thiocarbamyl, N- thiocarbamyl, C-amido, N- amido, aminothiocarbonyl, hydroxyaminocarbonyl, alkoxyaminocarbonyl, cyano, nitrile, cyanato, isocyanato, thiocyanato, isothiocyanato, sulfinyl, sulfonyl, sulfonamide,
aminosulfonyl, aminosulfonyloxy, sulfonamidecarbonyl, alkanoylaminosulfonyl, trihalomethylsulfonyl, or trihalomethylsulfonamide, or, R2 and either Rl or R3, together with the carbon atoms to which they are bound, form an optionally-substituted cycloalkyl, heterocycle, aryl, or heteroaryl;
R4 is independently chosen from hydro, halo, and an optionally-substituted group chosen from lower alkyl, haloalkyl, alkoxy, arylalkoxy, heteroarylalkoxy, and heterocycloalkoxy; R6 and R7 are independently chosen from hydro, halo, and lower alkyl;
or R6, taken together with R7, form an aryl or heteroaryl ring.
Compounds of Formula (II) were reported to be inhibitors of both TBK1 and/or ΙΚΚε.
In particular, preferred compounds of group (b) have an ether group at R5 and are without further substitution at R4, R6 and R7, for example compounds of Formula (Ila):
Figure imgf000014_0001
(Ha)
wherein R is H, alkyl, substituted alkyl, substituted or un substituted heterocyclyl groups. (c) the inhibitor compound described in Ou et al., Molecular Cell, 2011, 41, 458-70, referred to herein as the compound of Formula (III):
Figure imgf000014_0002
(III)
Compound (III) was reported as having activity against both TBK1 and ΙΚΚε.
(d) Compounds described in WO2009/030890, that is to say compound of Formula (IV):
Figure imgf000014_0003
(IV) wherein n is from 2-6;
Rl is a 5 or 6 membered cyclic alkyl ring or aromatic cyclic ring, wherein optionally one or more carbons in the ring structure is substituted by an O, N or S;
R2 is a 5 or 6 membered cyclic alkyl ring or arromatic cyclic ring, wherein optionally one or more carbons in the ring structure is substituted by an O, N or S, or is a linear or branched substituted or unsubstituted C1-C6 alkyl, Ci-C6 alkoxy or C2-C6 alkenyl, wherein when substituted the substituent group may be a C(=0)NH2;COOH; OH, NH2, N02, and R3 is a halo, H, CH3, CN, N02.
Compounds of Formula (IV) were reported as having activity against TBK1 and/or ΙΚΚε.
Preferred compounds of group (d) are the compounds:
Figure imgf000015_0001
BX-320 BX-795
BX-795 has also been reported in, for example, J. Biol. Chem. , 2009, 284(21), 14136-14146. The preferred compounds BX-320 and BX-795 were shown to inhibit TBK1 and ΙΚΚε.
(e) Compounds described in WO2009/122180, that is to say compound of Formula (V):
Figure imgf000016_0001
(V) wherein: R1 is C3_8-cycloalkyl;
X is O, NR7 or C3_6-heterocycloalkyl;
R2 is aryl, heteroaryl, fused or unfused aryl-C3_6-heterocycloalkyl or fused or unfused heteroaryI-C3_6- heterocycloalkyl, each of which is optionally substituted by one or more substitutents selected from aryl, heteroaryl, Ci_6-alkyl, C3_7-cycloalkyl and a group A, wherein said Ci-6-alkyl group is optionally substituted by one or more substituents selected from aryl, heteroaryl, R10 and a group A, said heteroaryl group is optionally substituted by one or more R<10> groups; and wherein said C3-6- heterocycloalkyl group optionally contains one or more groups selected from oxygen, sulfur, nitrogen and CO;
R3 is Ci-6-alkyl optionally substituted by one or more substituents selected from aryl, heteroaryl, -NR4R5, -OR6, -NR7(CO)R6, -NR7(CO)NR4R5, -NR7S02R6, -NR7COOR7, - CONR4R5, C3_6-heterocycloalkyl and
Figure imgf000016_0002
wherein said aryl, heteroaryl and C3-6 -heterocycloalkyl groups are each optionally substituted by one or more substituents selected from -Ci_6-alkyl and a group A, wherein said -Ci_6-alkyl group is optionally substituted by one or more substituents selected from aryl, heteroaryl and a group A; A is selected from halogen, hydroxyl, cyano, trifluoromethyl, alkoxy, -N02, -NH2, -NR4R5, -OR6, -NR7(CO)R6, -NR7(CO)NR4R5, -NR7COOR7, -NR7 (S02)R6, - C02H,
-(NR7CS02)NR4R5, -COOR7, -CONR4R5, COR6, S02NR4R5 and -S02CH3; each R4 and R5 is independently selected from hydrogen, C3_7-cycloalkyl, aryl, heteroaryl, Ci_ 6-alkyl and a C3-6-heterocycloalkyl ring optionally further containing one or more groups selected from oxygen, sulfur, nitrogen and CO and optionally substituted by one or more R10 groups, wherein said Ci_6-alkyl is optionally substituted by one or more substituents selected from halogen, cyano, hydroxyl, aryl, heteroaryl, -NR8R9, -NR7 (CO)R6, NR7COOR6, -NR7 (S02)R6, -COOR6, -CONR8R9, OR10, -S02R6 and a C3-6-heterocycloalkyl ring optionally further containing one or more groups selected from oxygen, sulfur, nitrogen and CO and optionally substituted by one or more or R10 groups; or R4 and R5 together with the N to which they are attached form a C3_6-heterocycloalkyl ring optionally further containing one or more groups selected from oxygen, sulfur, nitrogen and CO, wherein said C3_6-heterocycloalkyl ring may be saturated or unsaturated and is optionally substituted with one or more groups selected from NR8R9 and R10 groups; each R6 is independently selected from Ci-6-alkyl, C3_7-cycloalkyl, C4_7-heterocycloalkyl, aryl and heteroaryl, each of which may be optionally substituted by one or more substituents selected from halogen, R10 and -NR8R9; each R7 is selected from hydrogen, Ci-6-alkyl and C3-7-cycloalkyl, wherein said Ci-6-alkyl is optionally substituted by one or more halogens; each of R8 and R9 is independently selected from hydrogen and Ci-6-alkyl, wherein said Ci-6- alkyl group is optionally substituted by one or more halogens; or R8 and R9 together with the N to which they are attached form a C4_6-heterocycloalkyl ring optionally further containing one or more heteroatoms selected from oxygen and sulfur, wherein said C4-6-heterocycloalkyl ring is optionally substituted by one or more R10 groups; and each R is selected from halogen, C3_7-cycloalkyl and Ci_6-alkyl optionally substituted by one or more halogens, wherein where R10 is Ci_6-alkyl and two or more R10 groups are attached to the same carbon atom, the R10 groups may be linked to form a spiroalkyl group.
Compounds of Formula (V) were primarily reported as having activity against TBKl. From the disclosure of WO2009/122180, they appear also to have activity against ΙΚΚε
In particular, there are disclosed in WO2009/122180 compounds for Formula (Va):
Figure imgf000018_0001
(Va) wherein R is Ci-6-alkyl, C3_7-cyloalkyl, C4_7-heteroalkyl, heteroaryl, each of which may be optionally substituted by one of more substituents selected from halogen, R10 and -NR8R9; and R2 is an aryl or heteroaryl group each of which is optionally substituted by one or more substituents selected from Me, CI, F, CN, NHCOMe, CF3, COOH, CONH2, OH, NH2, NHS02Me, OCF3, -NHCOO'Bu, -C02Me, -NMe2, 4-methylpiperazin-l-yl, N-morpholinyl, (4-methylpiperazin-l-yl)-CO-, (N-morpholinyl)-CH2CH20-, (imidazol-l-yl)-CH2- and
Figure imgf000018_0002
(f) Compounds described in WO2010/100431, that is to say compound of Formula (VI):
Figure imgf000019_0001
(VI)
wherein: R1 is -NR7(CO)Ru; R2 is aryl, heteroaryl, fused aryl-C3-6 -heterocycloalkyl or fused heteroaryl-C3-6- heterocycloalkyl, each of which is optionally substituted by one or more substitutents selected from aryl, heteroaryl, Ci_6-alkyl, C3_6 -heterocycloalkyl and a group A, wherein said Ci-6-alkyl group is in turn optionally substituted by one or more substituents selected from aryl, heteroaryl, C3-6 -heterocycloalkyl and a group A, said heteroaryl group is optionally substituted by one or more R10 groups; and wherein each C3_6 -heterocycloalkyl group is optionally substituted by one or more groups selected from Ci_6-alkyl, Ci_6-haloalkyl, and A, and which optionally contains one or more groups selected from oxygen, sulphur, nitrogen and CO; R3 is H, halogen, cyano or Ci_6-alkyl;
A is selected from halogen, hydroxyl, cyano, trifluoromethyl, -NO2, -NH2, -NR4R5, -OR6, - NR7 (CO)R6, -NR7(C0)NR4R5, -NR7COOR7, -NR7(S02)R6, -C02H, -NR7 (S02)NR4R5, - COOR7, -C0NR4R5, COR6 and -S02CH3; each R4 and R5 is independently selected from hydrogen, C3_7-CycloaIkyI, aryl, heteroaryl, Ci-6-alkyl and a C3-6 -heterocycloalkyl ring optionally further containing one or more groups selected from oxygen, sulfur, nitrogen and CO, and optionally substituted by one or more R1( groups, wherein said Ci-6-alkyl is optionally substituted by one or more substituents selected from halogen, cyano, hydroxyl, aryl, heteroaryl, -NR8R9, -NR7(CO)R6, - NR7COOR6, -NR7 (S02)R6, -COOR6, -CONR8R9, OR10, -S02R6 and a C3_6-heterocycloalkyl ring optionally further containing one or more groups selected from oxygen, sulfur, nitrogen and CO and optionally substituted by one or more or R10 groups; or
R4 and R5 together with the N to which they are attached form a C3_6-heterocycloalkyl ring optionally further containing one or more groups selected from oxygen, sulfur, nitrogen and CO, wherein said C3_6-heterocycloalkyl ring may be saturated or unsaturated and is optionally substituted with one or more groups selected from NR8R9 and R10; each R6 is independently selected from Ci_6-alkyl, C3_7-cycloalkyl, C4_7-heterocycloalkyl, aryl and heteroaryl, each of which may be optionally substituted by one or more substituents selected from halogen, R10 and -NR8R9; each R7 is selected from hydrogen, Ci_6-alkyl and C3_6-cycloalkyl, wherein said Ci_6-alkyl is optionally substituted by one or more halogens; each of R8 and R9 is independently selected from hydrogen and Ci-6-alkyl, wherein said C1-6- alkyl group is optionally substituted by one or more halogens; or
R8 and R9 together with the N to which they are attached form a C4-6-heterocycloalkyl ring optionally further containing one or more heteroatoms selected from oxygen and sulfur, wherein said C4_6-heterocycloalkyl ring is optionally substituted by one or more R10 groups; and each R10 is selected from C3-7-cycloalkyl and Ci-6-alkyl optionally substituted by one or more halogens, wherein where R10 is Ci_6-alkyl and two or more R10 groups are attached to the same carbon atom, the R10 groups may be linked to form a spiroalkyl group; and each R11 is independently selected from Ci-6-alkyl, C3_7-cycloalkyl, Ci-6-alkyl-C3_7-cycloalkyl, C4-7- heterocycloalkyl, aryl and heteroaryl, each of which may be optionally substituted by one or more substituents selected from A.
Compounds of Formula (VI) were reported as having activity against TBKl and/or ΙΚΚε.
Preferred compounds of Formula (VI) include compounds of Formula (Via):
Figure imgf000021_0001
(Via) wherein: R2 is selected from aryl or heteroaryl, each of which is optionally substituted by one or more substitutents selected from halo, optionally substituted C3_7-heterocycloalkyl, optionally substituted Ci-6-alkyl, heteroaryl, Ci_6-alkyl-C3-7 -heterocycloalkyl, CN, NHCO- C3-7-heterocycloalkyl, CO-C3-7- heterocycloalkyl and NHCO-Ci-6-alkyl, wherein said C3-7- heterocycloalkyl is optionally substituted by one or more Ci_6-alkyl or A groups, and said Ci_ 6-alkyl is optionally substituted by one or more halo or NR4R5 groups;
and R is selected from cyclobutyl, thienyl, cyclopentyl, pyrazinyl, CE -cyclopropyl, sec- butyl, tetrahydrofuranyl, thiazolyl, cyclopropyl, isopropyl, cyclohexyl, CH2-cyclopentyl and n-propyl. (g) Compound GSK2292978, of Formula (VII), disclosed in poster Richter et al. , "Assessing ΙΚΚε as a Novel Oncology Target", presented at AACR-NCI-EORTC 2009
Figure imgf000021_0002
(VII)
Compound (VII) was reported as having activity against both TBKl and ΙΚΚε. (h) Compound SUl 1652, of Formula (VIII), disclosed in a poster Wilkinson et ah, "High Throughput Compatible Cellular Assays for Interrogating Toll-Like Receptor Signaling Pathways", Invitrogen, Life Technologies Ltd, 3 Fountain Drive, Inchinnan Business Park Paisley PA4 9RF, UK (www.invitrogen.com).
Figure imgf000022_0001
Compound (VIII) was reported as being an inhibitor of TBK1.
(i) Compounds described in WO2005/075465 (also US 2007/014951), that is to say compounds of Formula (X):
Figure imgf000022_0002
(X)
wherein: n is 0, 1,2, 3, or 4 ;
each R1 which may be the same or different, independently represents H, halogen or a group (X)a(Y)bZ;
X represents -O- or -CONH-;
a is 0 or 1 ;
Y represents Ci_6alkylene-;
b is O or l ;
Z represents hydroxy, C1-6 alkyl, Ci_6 haloalkyl, C5-7 heterocyclyl, C1-6 alkoxyalkyl, C1-6 haloalkoxyalkyl; R2 represents a group -(X1)c(Y1)dZ1;
Wherein X1 represents -Ci-n alkylene-;
c is 0 or 1 ;
Y represents -0-;
d is 0 or 1;
Z1 represents H, aryl or heteroaryl each of which contains 5-14 ring atoms, Cs_7 heterocyclyl, C5-7 cycloalkyl, C5-7 cycloalkenyl, (each of which aryl, heteroaryl, heterocyclyl, cycloalkyl, cycloalkenyl may be optionally substituted by one or more substituents independently selected from C1-6 alkyl, Ci_6 haloalkyl, halogen, Ci_6 alkoxy, Ci_6 haloalkoxy, S02R3, Ci_6 hydroxyalkyl);
R3 represents H or C1-6 alkyl;
or pharmaceutically acceptable salts, solvates or physiologically functional derivatives thereof.
Preferred compounds have the general of structure (Xa):
Figure imgf000023_0001
(Xa)
where Z is aryl or heteroaryl each of which is optionally substituted by one of more substituents independently selected from halogen, CF3, CH2OH, SO2CH3, CH3, OCH3.
Compounds of this general structure are also described in Bamborough et ah, Bioorg. Med. Chem. Lett. , 2006, 16, 6236-6240.
Compounds of Formula (X) were reported as being inhibitors of ΙΚΚε. (j) Compounds described in Wang et al. Biorg. Med. Chem. Lett, 2012, 22, 2063-2068, that is to say compounds of formula (XI)
Figure imgf000024_0001
wherein R6 is Br, CN, CONH2 or CI;
wherein R7
re resents
Figure imgf000024_0002
and wherein R re resents
Figure imgf000025_0001
or pharmaceutically acceptable salts, solvates or physiologically functional derivatives thereof.
(k) Compounds described in WO2010127754 (also published as US2012/0053178) that is to say:
1. Compounds of the formula (XII)
Figure imgf000025_0002
(XII) in which R1 denotes A, -[C(R3)( R4)]¾Ar, -[C(R3)( R4)]„Het or --[C(R3)( R4)]„Cyc,
R2 denotes H, A, Hal, CN, -[C(R ) 2]„-Ar', -[C(R3) 2]„-Het', -[C(R3) 2]„-Cyc, OR3 or N(R ) 2,
R3 denotes H or A,
R4 denotes H, A, -[C(R3) 2]„OR3, -[C(R3) 2]„COOR3, -[C(R3) 2]„N(R3) 2 or -[C(R3) 2]„Het, A denotes unbranched or branched alkyl having 1-6 C atoms, in which one or two CH2 groups may be replaced by O, N and/or S atoms and/or by— CH=CH— groups and/or, in addition, 1-7 H atoms may be replaced by F,
Cyc denotes cycloalkyl having 3, 4, 5, 6 or 7 C atoms,
Ar denotes phenyl which is unsubstituted or mono-, di- or trisubstituted by Hal, A, -[C(R3) 2]„OR3, N(R )2, N02, CN, COOR3, CON(R )2, NR3COA, NR3S02A, COR3, S02N(R )2 and/or S(0)„A,
Ar' denotes phenyl which is unsubstituted or mono-, di- or trisubstituted by Hal, A, OR3, N(R ) 2, SR3, N02, CN, COOR3, CON(R3)2, NR COA, NR S02A, S02N(R ) 2, S(0)„A, CO- Het!, Heti, [C(R3) 2]„N(R3)2,
Figure imgf000026_0001
0[C(R3)2]„N(R3) 2, 0[C(R3)2LHet!, NHCOOA, NHCON(R3)2, NHCOO[C(R )2]„N(R3) 2, NHCOO[C(R )2]„Heti, NHCONH[C(R )2]„N(R3) 2, NHCONH[C(R3)2]„Heti, OCONH[C(R3)2]„N(R ) 2, OCONH[C(R ) 2]„Heti, CO-Heti, CHO and/or COA,
Het denotes a mono- or bicyclic saturated, unsaturated or aromatic heterocycle having 1 to 4 N, and/or O and/or S atoms which is unsubstituted or mono- or disubstituted by Hal, A, OR3, N(R )2, N02, CN, COOR3, CON(R )2, NR3COA, NR3S02A, CORs, S02NR3 and/or S(0)„A, Het' denotes a mono-, bi- or tricyclic saturated, unsaturated or aromatic heterocycle having 1 to 4 N, O and/or S atoms which is unsubstituted or may be mono- or disubstituted by Hal, A, OR3, N(R3)2, SR3, N02, CN, COOR3, CON(R3)2, NR3COA, NR3S02A, S02N(R3) 2, S(O)„ A, CO-Heti, Heti, [C(R3) 2]„N(R ) 2, [C(R ) 2]„Heti, 0[C(R )2]„N(R3) 2, 0[C(R3)2]„Heti, NHCOOA, NHCON(R3)2, NHCOO[C(R3)2]„N(R3) 2, NHCOO[C(R3)2]¾Het.sup.l,
NHCONH[C(R3)2]„N(R3) 2, NHCONH[C(R3)2]„Heti, OCONH[C(R3)2]„N(R3) 2,
OCONH[C(R3)2]„Heti, CO-Heti, CHO, COA, =S, =NH, =NA and/or =0 (carbonyl oxygen), Heti denotes a monocyclic saturated heterocycle having 1 to 2 N and/or O atoms, which may be mono- or disubstituted by A, OA, OH, Hal and/or =0 (carbonyl oxygen),
Hal denotes F, CI, Br or I,
N denotes 0, 1 or 2,
and pharmaceutically usable salts, tautomers and stereoisomers thereof, including mixtures thereof in all ratios. Preferred compounds have the following structures:
Figure imgf000027_0001
(Xlld)
Figure imgf000028_0001
Figure imgf000028_0002
(Xllf)
Figure imgf000028_0003
Further compounds are also described in WO2012059171. A compound for use in the invention may be a compound that is a down-regulator of the expression of one or both of TBK1 and ΙΚΚε. Expression of one or both of TBK1 and ΙΚΚε may be down-regulated using anti-sense nucleic acid or RNAi technology, amongst other approaches. The use of these approaches to down-regulate gene expression is now well- established in the art. Anti-sense oligonucleotides may be designed to hybridise to the complementary sequence of nucleic acid, pre-mRNA or mature mRNA, interfering with the production of TBK1 or ΙΚΚε so that its expression is reduced or completely or substantially completely prevented. In addition to targeting coding sequence, anti-sense techniques may be used to target control sequences of a gene, e. g. in the 5' flanking sequence, whereby the anti- sense oligonucleotides can interfere with expression control sequences. The construction of anti-sense sequences and their use is described for example in Peyman and Ulman, Chemical Reviews, 1990, 90, 543-584, and Crooke, Ann. Rev. Pharmacol. Toxicol. 1992, 32, 329-376.
Oligonucleotides may be generated in vitro or ex vivo for administration or anti-sense RNA may be generated in vivo within cells in which down-regulation is desired. Thus, double- stranded DNA may be placed under the control of a promoter in a "reverse orientation" such that transcription of the anti- sense strand of the DNA yields RNA which is complementary to normal mRNA transcribed from the sense strand of the target gene. The complementary anti- sense RNA sequence is thought then to bind with mRNA to form a duplex, inhibiting translation of the endogenous mRNA from the target gene into protein. Whether or not this is the actual mode of action is still uncertain. However, it is established fact that the technique works.
The complete sequence corresponding to the coding sequence in reverse orientation need not be used. For example fragments of sufficient length may be used. It is a routine matter for the person skilled in the art to screen fragments of various sizes and from various parts of the coding or flanking sequences of a gene to optimise the level of anti-sense inhibition. It may be advantageous to include the initiating methionine ATG codon, and perhaps one or more nucleotides upstream of the initiating codon. A suitable fragment may have about 14-23 nucleotides, e. g. about 15, 16 or 17.
An alternative to anti-sense is to use a copy of all or part of the target gene inserted in sense, that is the same, orientation as the target gene, to achieve reduction in expression of the target gene by co-suppression (see Angell & Baulcombe, The EMBO Journal, 1997, 16(12), 3675- 3684; and Voinnet & Baulcombe, Nature , 1997, 389, 553).
Double stranded RNA (dsRNA) has been found to be even more effective in gene silencing than both sense or antisense strands alone (Fire et al., Nature, 1998, 391, 806-811) [. dsRNA mediated silencing is gene specific and is often termed RNA interference (RNAi). RNA interference is a two-step process. First, dsRNA is cleaved within the cell to yield short interfering RNAs (siRNAs) of about 21-23nt length with 5' terminal phosphate and 3' short overhangs (-2nt). The siRNAs target the corresponding mRNA sequence specifically for destruction (Zamore, Nature Structural Biology, 2001, 8, 746-750). RNAi may also be efficiently induced using chemically synthesized siRNA duplexes of the same structure with 3'-overhang ends (Zamore et al., Cell, 2000, 101, 25-33. Synthetic siRNA duplexes have been shown to specifically suppress expression of endogenous and heterologeous genes in a wide range of mammalian cell lines (Elbashir, et al. , Nature, 2001, 411, 494-498). Particular siRNA molecules that find use in the invention include RNAi reagents with the target sequence include:
5'-GAACGUAGAUUAGCUUAUA-3' (TBKl) [SEQ ID NO. 1],
5 -UG AC AG AG AUUU ACU AUC A- 3 ' (TBKl) [SEQ ID NO. 2],
5 '-UAAAGUACAUCCACGUUAU-3 ' (TBKl) [SEQ ID NO. 3],
5'-GGAUAUCGACAGCAGAUUA-3' (TBKl) [SEQ ID NO. 4] ;
5'-ACAAACAGUUCAAGAAGUC-3' (ΙΚΚε) [SEQ ID NO. 5],
5 -GGACCUGCUUCUCCACAUG-3 ' (ΙΚΚε) [SEQ ID NO. 6],
5 -GAAGGCGGCUGCAGAACUG-3 ' (ΙΚΚε) [SEQ ID NO. 7],
5'-GGCAGGAGCUAAUGUUUCG-3' (ΙΚΚε) [SEQ ID NO. 8] .
Whilst a compound that is an inhibitor of one or both of TBKl and ΙΚΚε, or a down- regulator of the expression of one or both of TBKl and ΙΚΚε may be used as the sole active agent, it is also possible for the compound to be used in combination with one or more further active agents. Such further active agents may be further compounds that are inhibitors of one or both of TBKl and ΙΚΚε, or down-regulators of the expression of one or both of TBKl and ΙΚΚε, or they may be different therapeutic agents, for example agents targeting one of the diseases mentioned above, particularly the same disease as that targeted by the compound of the invention. A further therapeutic agent is selected from the group consisting of inhibitors of mTOR, inhibitors of PDkinase, inhibitors of both mTOR and PDkinase, inhibitors of AKT and inhibitors of PDK1. Examples of further therapeutic agents include: UCN01, (NVP)- BAG956, Celecoxib, OSU-03012, BX-795, BX-912, BX-320, AR-12, KP372-1, LY294002, PWT-458, PX-866, CAL-101, XL-147, ZSTK474, GDC-0941, (NVP)-BEZ235, AS-252424, TGX-221, XL-765, wortmannin, PI- 103, Perifosine (KRX-0401), Triciribine (API-2), SR13688, AR-67 (DB-67), AR-42, GSK690693, KP372- 1, A-443654, MK-2206, Rapamycin (Sirolimus), Temsirolimus (Torisel®, CCI-779), Everolimus (Afinitor®, RAD001),
Deforolimus (Ridaforolimus, AP-23573), AZD-8055, OSI-027, INK- 128, PP-242. These and further therapeutic agents are described in further detail in Chapell et al, Oncotarget, 2011, 2, 135-164.
In certain cancers in which activation of the Ras/Raf/Mek/Erk pathway is important, for example those tumours containing activating mutations in KRAS or BRAF, inhibition of the Ras/Raf/Mek/Erk pathway in conjunction with inhibition of the PI3 kinase pathway could provide an important therapeutic strategy (Chappell, 2011; Engleman et ah, Nat. Med., 2008, 14, 1351-1356). A compound that is an inhibitor of one or both of TBK1 and IKK8, or a down-regulator of the expression of one or both of TBK1 and ΙΚΚε may be used in combination with an inhibitor of the Ras/Raf/Mek/Erk pathway, in particular in tumours that contain activating mutations in this pathway. Examples of therapeutic agents that inhibit the Ras/Raf/Mek Erk pathway are: Tipifarnib (Zarnesta™, Rl 15777), Sorafenib (Nexavar®, BAY43-9006), AAL-881, LBT-613, RAF265, XL281, SB-590885, PLX-4720, PLX-4032, L-779,450, GW5074, SB-699393, CI-1040 (PD184352), PD0325901, XL518, Selumtinib (AZD6244, ARRY-142886), RDEA119 (BAY 869766), PD098059, U0126, SL-327. These and further therapeutic agents are described in further detail in Chapell, 2011.
The compound of the invention may be co-formulated with the additional agent, or it may be formulated separately and administered consecutively, simultaneously or sequentially with the additional agent. The amount of active ingredient which is required to achieve a therapeutic effect will, of course, vary with the particular compound, the route of administration, the subject under treatment, including the type, species, age, weight, sex, and medical condition of the subject and the renal and hepatic function of the subject, and the particular cancer being treated, as well as its severity. An ordinarily skilled physician or veterinarian can readily determine and prescribe the effective amount of the drug required to prevent, counter or arrest the progress of the condition.
Oral dosages of the present invention, when used for the indicated effects, will range between about 0.01 mg per kg of body weight per day (mg/kg/day) to about 100 mg/kg/day, preferably 0.01 mg per kg of body weight per day (mg/kg/day) to 10 mg/kg/day, and most preferably 0.1 to 5.0 mg/kg/day, for adult humans. For oral administration, the compositions are preferably provided in the form of tablets or other forms of presentation provided in discrete units containing 0.01, 0.05, 0.1, 0.5, 1.0, 2.5, 5.0, 10.0, 15.0, 25.0, 50.0, 100, and 500 milligrams of the active ingredient for the symptomatic adjustment of the dosage to the patient to be treated. A medicament typically contains from about 0.01 mg to about 500 mg of the active ingredient, preferably from about 1 mg to about 100 mg of active ingredient. Intravenously, the most preferred doses will range from about 0.1 to about 10 mg/kg/minute during a constant rate infusion. Advantageously, compounds of the present invention may be administered in a single daily dose, or the total daily dosage may be administered in divided doses of two, three or four times daily. Furthermore, preferred compounds for the present invention can be administered in intranasal form via topical use of suitable intranasal vehicles, or via transdermal routes, using those forms of transdermal skin patches well known to those of ordinary skill in the art. To be administered in the form of a transdermal delivery system, the dosage administration will, of course, be continuous rather than intermittent throughout the dosage regimen.
The pharmaceutical formulations according to the invention include those suitable for oral, parenteral (including subcutaneous, intradermal, intramuscular, intravenous (bolus or infusion), and intraarticular), inhalation (including fine particle dusts or mists which may be generated by means of various types of metered doses pressurized aerosols), nebulizers or insufflators, rectal, intraperitoneal and topical (including dermal, buccal, sublingual, and intraocular) administration, although the most suitable route may depend upon, for example, the condition and disorder of the recipient.
The formulations may conveniently be presented in unit dosage form and may be prepared by any of the methods well known in the art of pharmacy. All methods include the step of bringing the active ingredient into association with the carrier which constitutes one or more accessory ingredients. In general the formulations are prepared by uniformly and intimately bringing into association the active ingredient with liquid carriers or finely divided solid carriers or both and then, if necessary, shaping the product into the desired formulation.
Formulations of the present invention suitable for oral administration may be presented as discrete units such as capsules, cachets, pills or tablets each containing a predetermined amount of the active ingredient; as a powder or granules; as a solution or a suspension in an aqueous liquid or a non-aqueous liquid; or as an oil-in-water liquid emulsion or a water-in-oil liquid emulsion. The active ingredient may also be presented as a bolus, electuary or paste.
The present compounds can, for example, be administered in a form suitable for immediate release or extended release. Immediate release or extended release can be achieved by the use of suitable pharmaceutical compositions comprising the present compounds, or, particularly in the case of extended release, by the use of devices such as subcutaneous implants or osmotic pumps.
Exemplary compositions for oral administration include suspensions which can contain, for example, microcrystalline cellulose for imparting bulk, alginic acid or sodium alginate as a suspending agent, methylcellulose as a viscosity enhancer, and sweeteners or flavoring agents such as those known in the art; and immediate release tablets which can contain, for example, microcrystalline cellulose, dicalcium phosphate, starch, magnesium stearate, calcium sulfate, sorbitol, glucose and/or lactose and/or other excipients, binders, extenders, disintegrants, diluents and lubricants such as those known in the art. Suitable binders include starch, gelatin, natural sugars such as glucose or beta-lactose, corn sweeteners, natural and synthetic gums such as acacia, tragacanth or sodium alginate, carboxymethylcellulose, polyethylene glycol, waxes and the like. Disintegrators include without limitation starch, methylcellulose, agar, bentonite, xanthan gum and the like. The compounds of the invention can also be delivered through the oral cavity by sublingual and/or buccal administration. Molded tablets, compressed tablets or freeze-dried tablets are exemplary forms which may be used.
Exemplary compositions include those formulating the present compound(s) with fast dissolving diluents such as mannitol, lactose, sucrose and/or cyclodextrins. Also included in such formulations may be high molecular weight excipients such as celluloses (avicel) or polyethylene glycols (PEG). Such formulations can also include an excipient to aid mucosal adhesion such as hydroxy propyl cellulose (HPC), hydroxy propyl methyl cellulose (HPMC), sodium carboxy methyl cellulose (SCMC), maleic anhydride copolymer (e.g., Gantrez), and agents to control release such as polyacrylic copolymer (e.g. Carbopol 934). Lubricants, glidants, flavors, coloring agents and stabilizers may also be added for ease of fabrication and use. Lubricants used in these dosage forms include sodium oleate, sodium stearate, magnesium stearate, sodium benzoate, sodium acetate, sodium chloride and the like. For oral administration in liquid form, the oral drug components can be combined with any oral, nontoxic, pharmaceutically acceptable inert carrier such as ethanol, glycerol, water, and the like.
Formulations 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. 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 (lyophilised) condition requiring only the addition of the sterile liquid carrier, for example saline or water-for-injection, immediately prior to use. Extemporaneous injection solutions and suspensions may be prepared from sterile powders, granules and tablets of the kind previously described.
Exemplary compositions for parenteral administration include injectable solutions or suspensions which can contain, for example, suitable non-toxic, parenterally acceptable diluents or solvents, such as mannitol, 1,3-butanediol, water, Ringer's solution, an isotonic sodium chloride solution, or other suitable dispersing or wetting and suspending agents, including synthetic mono- or diglycerides, and fatty acids, including oleic acid, or
Cremaphor.
Exemplary compositions for nasal, aerosol or inhalation administration include solutions in saline, which can contain, for example, benzyl alcohol or other suitable preservatives, absorption promoters to enhance bioavailability, and/or other solubilizing or dispersing agents such as those known in the art.
Formulations for rectal administration may be presented as a suppository with the usual carriers such as cocoa butter, synthetic glyceride esters or polyethylene glycol. Such carriers are typically solid at ordinary temperatures, but liquefy and/or dissolve in the rectal cavity to release the drug.
Formulations for topical administration in the mouth, for example buccally or sublingually, include lozenges comprising the active ingredient in a flavoured basis such as sucrose and acacia or tragacanth, and pastilles comprising the active ingredient in a basis such as gelatin and glycerine or sucrose and acacia. Exemplary compositions for topical administration include a topical carrier such as Plastibase (mineral oil gelled with polyethylene). It should be understood that in addition to the ingredients particularly mentioned above, the formulations of this invention may include other agents conventional in the art having regard to the type of formulation in question, for example those suitable for oral administration may include flavouring agents. Methods for the therapeutic delivery of si~RNA agents are well known to those skilled in the art, for example as described in Xie et a\., Drug Discovery Today, 2006 11 (1-2), 67-73. "Harnessing in vivo siRNA delivery for drug discovery and therapeutic development".
In a further embodiment, the invention provides a compound that is an inhibitor of one or both of TBK1 and ΙΚΚε, or a down-regulator of the expression of one or both of TBK1 and ΙΚΚε, for use in a method of treating a cancer that is dependent on the PDkinase pathway comprising:
- identifying a cancer in the individual as being dependent on the PDkinase pathway, and
- administering the compound to the individual.
The cancer may be a cancer as described above. The compound may be a compound as described above. In a further embodiment, there is provided a method of treating cancer in an individual in whom the cancer is dependent on the PDkinase pathway, comprising administering to the individual a compound that is an inhibitor of one or both of TBKl and ΙΚΚε, or a down- regulator of the expression of one or both of TBKl and ΙΚΚε. The cancer may be a cancer as described above. The compound may be a compound described above.
In a further embodiment, there is provided a method of treating cancer in an individual comprising:
- identifying the cancer as dependent on the PDkinase pathway, and
- administering to the individual a compound that is an inhibitor of one or both of TBKl and ΙΚΚε, or a down-regulator of the expression of one or both of TBKl and ΙΚΚε.
The cancer may be a cancer as described above. The compound may be a compound as described above.
The invention further provides a method of determining the susceptibility of a cancer in an individual to treatment with a compound that is an inhibitor of one or both of TBKl and ΙΚΚε, or a down-regulator of the expression of one or both of TBKl and ΙΚΚε, comprising determining whether the cancer is dependent on the PDkinase pathway, whereby a dependence on the PDkinase pathway indicates that the cancer in the individual has an increased susceptibility to a compound that is an inhibitor of one or both of TBKl and ΙΚΚε, or a down-regulator of the expression of one or both of TBKl and ΙΚΚε.
The cancer may be a cancer as described above. The compound may be a compound as described above.
The invention further provides a method of determining a suitable treatment for an individual with a cancer comprising:
determining whether the cancer is dependent on the PDkinase pathway; and
determining a suitable treatment, whereby a compound that is an inhibitor of one or both of TBKl and ΙΚΚε is suitable in the treatment of an individual with a cancer that is dependent on the PDkinase pathway. The cancer may be a cancer as described above. The compound may be a compound as described above.
A method of determining a suitable treatment may including determining that a particular treatment is not suitable in the subject. For example, if a cancer is found not to be dependent on the PDkinase pathway, then an inhibitor of one or both of TBKl and ΙΚΚε might not be suitable in the treatment of the individual.
The invention further provides a method of predicting the response of a cancer condition in an individual to a treatment with a compound that is an inhibitor of one or both of TBKl and ΙΚΚε by determining whether the cancer is dependent on the PDkinase pathway.
The cancer may be a cancer as described above. The compound may be a compound as described above. In some embodiments, a cancer may be identified as dependent on the PDkinase pathway by determining the presence in cancer cells from the individual of one or more variations, for example, polymorphisms or mutations, in a nucleic acid encoding a polypeptide that is a component of the PDkinase pathway.
In some embodiments, a cancer may be identified as being deficient in PTEN activity by determining the presence in cancer cells from the individual of one or more variations, for example, polymorphisms or mutations, in a nucleic acid encoding a PTEN polypeptide.
In some embodiments, a cancer may be identified as having overactive PDkinase signalling by determining the presence in cancer cells from the individual of one or more variations, for example, polymorphisms or P1K3CA mutations, in a nucleic acid encoding an overactive PD kinase pi 10 polypeptide.
In some embodiments, a cancer may be identified as being deficient in PTEN activity by determining the presence in cancer cells from the individual of one or more variations, for example, polymorphisms or mutations, in a nucleic acid encoding a PTEN polypeptide. A cancer may be identified as having overactive PDkinase signalling by determining the presence in cancer cells from the individual of one or more variations, for example, polymorphisms or mutations, in a nucleic acid encoding an PD kinase pi 10 polypeptide, for example PIK3CA, which polymorphisms or mutations cause the PI3 kinase to be overactive, or one or more variations in the nucleic acid controlling expression of a PI3 kinase pi 10 polypeptide, for example PIK3CA, which cause the PI3 kinase to be over-expressed.
Sequence variations such as mutations and polymorphisms may include a deletion, insertion or substitution of one or more nucleotides, relative to the wild-type nucleotide sequence. In some embodiments, the variation may be a gene amplification. The one or more variations may be in a coding or non-coding region of the nucleic acid sequence and, may reduce or abolish the expression of a negatively regulating pathway component polypeptide, for example PTEN. The variant nucleic acid may encode a variant polypeptide which has reduced or abolished activity or may encode a wild- type polypeptide which has little or no expression within the cell, for example through the altered activity of a regulatory element. A variant nucleic acid may have one, two, three, four or more mutations or polymorphisms relative to the wild-type sequence. Furthermore, a cancer may be determined to be heterozygous or homozygous in mutant forms of one or more components, leading to a dependency on the PDkinase pathway.
Various methods are available for determining, in a sample obtained from an individual, the presence or absence of a particular nucleic acid sequence, for example a nucleic acid sequence which has a mutation or polymorphism that reduces or abrogates the expression or activity of a PDkinase pathway component, or that increases the expression or activity of a PDkinase pathway component, as the case may be depending on the role that the component plays in the pathway or its control.
Furthermore, having sequenced nucleic acid of an individual or sample, the sequence information can be retained and subsequently searched without recourse to the original nucleic acid itself.
Thus, for example, scanning a database of sequence information using sequence analysis software may identify a sequence alteration or mutation.
Methods according to some aspects of the present invention may comprise determining the binding of an oligonucleotide probe to nucleic acid obtained from the sample, for example, genomic DNA, RNA or cDNA. The probe may comprise a nucleotide sequence which binds specifically to a nucleic acid sequence which contains one or more mutations or
polymorphisms and does not bind specifically to the nucleic acid sequence which does not contain the one or more mutations or polymorphisms, or vice versa. The oligonucleotide probe may comprise a label and binding of the probe may be determined by detecting the presence of the label.
A method may include hybridisation of one or more (e. g. two) oligonucleotide probes or primers to target nucleic acid. Where the nucleic acid is double-stranded DNA, hybridisation will generally be preceded by denaturation to produce single- stranded DNA. The
hybridisation may be as part of a PCR procedure, or as part of a probing procedure not involving PCR. An example procedure would be a combination of PCR and low stringency hybridisation.
Binding of a probe to target nucleic acid (e. g. DNA) may be measured using any of a variety of techniques at the disposal of those skilled in the art. For instance, probes may be radioactively, fluorescently or enzymatically labelled. Other methods not employing labelling of probe include examination of restriction fragment length polymorphisms, amplification using PCR, RN'ase cleavage and allele specific oligonucleotide probing. Probing may employ the standard Southern blotting technique. For instance, DNA may be extracted from cells and digested with different restriction enzymes. Restriction fragments may then be separated by electrophoresis on an agarose gel, before denaturation and transfer to a nitrocellulose filter. Labelled probe may be hybridised to the DNA fragments on the filter and binding determined.
Those skilled in the art are well able to employ suitable conditions of the desired stringency for selective hybridisation, taking into account factors such as oligonucleotide length and base composition, temperature and so on. Suitable selective hybridisation conditions for oligonucleotides of 17 to 30 bases include hybridization overnight at 42 °C in 6X SSC and washing in 6X SSC at a series of increasing temperatures from 42 °C to 65 °C. Other suitable conditions and protocols are described in Molecular Cloning: a Laboratory Manual: 3rd edition, Sambrook & Russell (2001) Cold Spring Harbor Laboratory Press NY and Current Protocols in Molecular Biology, Ausubel et al. eds. John Wiley & Sons (1992).
Nucleic acid, which may be genomic DNA, RNA or cDNA, or an amplified region thereof, may be sequenced to identify or determine the presence of polymorphism or mutation therein. A polymorphism or mutation may be identified by comparing the sequence obtained with the database sequence of the component, as set out above. In particular, the presence of one or more polymorphisms or mutations that cause abrogation or loss of function of the
polypeptide component in the case, for example, of PTEN; or over-expression or constitutive (i.e. unregulated) function of the polypeptide component in the case, for example, of PIK3CA mutations; and thus the activation of the PDkinase pathway as a whole, may be determined. Sequencing may be performed using any one of a range of standard techniques. Sequencing of an amplified product may, for example, involve precipitation with isopropanol, resuspension and sequencing using a TaqFS+ Dye terminator sequencing kit. Extension products may be electrophoresed on an ABI 377 DNA sequencer and data analysed using Sequence Navigator software.
A specific amplification reaction such as PCR using one or more pairs of primers may conveniently be employed to amplify the region of interest within the nucleic acid sequence, for example, the portion of the sequence suspected of containing mutations or
polymorphisms. The amplified nucleic acid may then be sequenced as above, and/or tested in any other way to determine the presence or absence of a mutation or polymorphism which reduces PTEN function, or enhances PDkinase function, or the overall flux through the PDkinase pathway. The polymerase chain reaction (PCR) has been reviewed, for instance in "PCR protocols; A Guide to Methods and Applications", Eds. Innis et al, 1990, Academic Press, New York, Mullis et al, Cold Spring Harbor Symp; Ehrlich (ed), PCR technology, Stockton Press, NY, 1989; and Ehrlich et al, Science, 1991, 252, 1643-1650.
A number of methods have been described for the detection of mutant forms of PIK3CA. The most common activating PIK3CA mutations found in cancer are missense mutations resulting from a single base pair change in either: exon 20, coding the helical domain; or exon 9, coding the kinase domain. Examples of common mutations in exon 20 include the following nucleotide changes (corresponding amino acid changes shown in brackets) A3140G (H1047R), A3150T (H1047L). Examples of common mutation in exon 9 include the following nucleotide changes (corresponding amino acid changes shown in brackets) G1633A (E545K), G1624A (E542K), G1634A (E545G). A method for the detection of mutations in exons 9 and 20 using single strand conformational polymorphism (SSCP) is described in US2011/0038862. Another method that has been used for the detection of
PIK3CA mutants is the use of allele specific PCR using primers specifically designed against common mutants (H1047L, H1047R, E542K and E545K) using the Amplification Refractory Mutation System (ARMS™, AstraZeneca) and Scopions™ (DxS), a fluorescent signalling system that detects PCR products (Board et al., Clinical Chemistry, 2008, 54 (4), 757-760).
A number of methods have been described for the detection of mutant forms of PTEN. Both missense and 'nonsense' mutations (i.e. stop codon, frameshift or splicing mutations) are found in human cancers. The most common of these are nonsense mutations which often result in inactive truncated forms of the protein. A method for the detection of truncated PTEN mRNA has been described using a yeast-based assay, which relies on the incorporation of full length PTEN cDNA into a primer which leads to the expression of a PTEN::ADE2 (EC 4.1.1.21) protein. Colonies which lack a functional ADE2 protein appear red due to the accumulation of the ADE2 substrate (Zhang et al., Oncogene 2000, 19, 4346-4353). Another method for the detection of PTEN deletions involves the use of FISH probes directed at the PTEN-locus on chromosome region 10q23 (Chang et al., Leukemia Research, 2006, 30 262- 265).
Alternatively, the presence of one or more variations in a component of the PDkinase pathway may be determined by detecting, in one or more cells of a test sample, the presence of the variant component polypeptide, or the absence or reduced amount of the non-mutated component polypeptide (e.g. detection of the levels of PTEN) which is encoded by the nucleic acid sequence. A method of identifying a cancer cell in a sample from an individual as dependent on the PDkinase pathway may include contacting a sample with a specific binding member directed against a variant (e. g. a mutant) polypeptide component of the pathway, and determining binding of the specific binding member to the sample. Another method of identifying a cancer cell in a sample from an individual as dependent on the PDkinase pathway may include contacting a sample with a specific binding member directed against a phosphorylated polypeptide component of the pathway, and determining binding of the specific binding member to the sample. A preferred phosphorylated polypeptide for this purpose is phospho-PDKl (Vasudevan et ah, 201 1).
Binding of the specific binding member to the sample may be indicative of the presence of the variant polypeptide component, or phosphorylated polypeptide component, of the pathway in a cell within the sample. Preferred specific binding molecules for use in aspects of the present invention include antibodies and fragments or derivatives thereof ('antibody molecules'). The reactivities of a binding member such as an antibody on normal and test samples may be determined by any appropriate means. Tagging with individual reporter molecules is one possibility. The reporter molecules may directly or indirectly generate detectable, and preferably measurable, signals. The linkage of reporter molecules may be directly or indirectly, covalently, e. g. via a peptide bond or non-covalently. Linkage via a peptide bond may be as a result of recombinant expression of a gene fusion encoding binding molecule (e. g. antibody) and reporter molecule.
Antibodies for PTEN are commercially available and can be used to assess the amounts of PTEN using standard techniques such as western blotting. Reverse-phase protein array (RPPA) analysis, a high throughput technique that can be used to assess multiple tissue samples, has also been used to assess levels of PTEN protein (Vasudevan et ah, 2011).
Antibodies for phophorylated kinases such as phospho-PDK- 1 or phospho-mTOR are readily available from commercial suppliers. Sotiriou et al. have described a signature for PI3 kinase pathway activation in breast cancer samples using microarray analysis, this or similar approaches could also be used for identifying tumours with activation of the PI3 kinase pathway (US2011/0038862).
Additionally tumours that contain a combination of mutations or transformations to members that can activate the PI3 kinase pathway may also be considered dependant on the PI3 kinase pathway. For example this could take the form of the combination of a PIK3CA mutation and the loss or low levels of PTEN. Alternatively it may be the over-expression of a growth factor receptor which can activate the PI3 kinase pathway. For example the over-expression of ErbB2 in breast cancer in combination with a reduction in the amount of PTEN, or with an activating mutation in PIK3CA. Another example would be in activating mutation in a Ras protein for example K-Ras and a mutation in PIK3CA or a reduction in the amount of PTEN. Methods for the detecting the over expression of growth factor receptors or the presence of RAS mutations are well known in the art.
Examples: A - Assays for activity against the IKKs and TBK-1 enzyme
Compounds of the invention (synthesised as described below) were tested for activity against the ΙΚΚε and TBK-1 enzyme as follows: Inhibitions studies were performed using a time-resolved fluorescence-based Lanthascreen™ assay. Phosphorylation of a fluorescein-labelled substrate peptide is measured using terbium- labeled phospho specific antibodies. Terbium is excited at 340 nm and the FRET energy transfer to fluorescein is measured at 495 and 520 nm. The emission ratio between 520 and 495 is a measure of the level of phosphorylation of the substrate by the kinase. Kinase inhibition assays (10 \xL) were performed at 20°C in 384-well plate format.
Compound IC50 values were determined at the apparent Km for ATP (20 μΜ) based on a radiometric assay (Invitrogen) using 8 or 10 point curves in duplicate. The final reaction conditions contained 400 nM fluorescein-IkBa substrate (DRHDSGLDSMKDE), 20 μΜ ATP, 2 nM or 8 nM ΙΚΚε or TBK1 kinase respectively, and 3% DMSO in kinase assay buffer consisting of 50 mM HEPES (pH 7.5), 10 mM MgCl, 1 mM EGTA, 0.01% Brij-35.
Compound dilutions were prepared from 10 mM DMSO stocks by dilution into DMSO. Compound dilution series were further diluted in kinase assay buffer to give a 12% DMSO stock, the final concentration in the assay being 3% DMSO.
The kinase phosphorylation assay was initiated by the addition of the kinase and the reaction was allowed to proceed for 1 hr or 2.5 hr for ΙΚΚε and TBK-1 kinase respectively. Both conditions were within the linearity of the phosphorylation. The reaction was stopped by the addition of 10 mM EDTA, and phosphorylation was detected after a lhr incubation with 1.5 nM terbium-labelled antibody against phosphorylation at Serine 32 on the IkBa peptide, both in TR-FRET dilution buffer (Invitrogen).
The results of the testing are show below. B - Synthesis of inhibitors of IKKe or TBK-1
Abbreviations Used
DCM Dichloromethane
DMF N,N-Dimethylformamide
DMSO Dimethyl sulfoxide
EtOAc Ethyl acetate
LC-MS Liquid chromatography-mass spectrometry
MeCN Acetonitrile
MeOH Methanol
MgS04 Magnesium sulfate
NMP N-Methylpyrrolidinone
THF Tetrahydrofuran
Example DMX-A: Synthesis of 3-[4-(3-cyano-4-pyrrolidin-l-yl-phenyl)-pyrimidin-2- ylamino ]-benzamide
2 -pyrrolidin-l -yl-5-(4, 4, 5,5-tetramethyl-[l,3, 2 ] dioxaborolan-2 -yl)-benzonitrile
Figure imgf000044_0001
3-Cyano-4-fluorophenyl-boronic acid pinacol ester (250 mg, 1.01 mmol) was dissolved in
ΝΜΡ (4 mL). Pyrrolidine (415 μί, 5.05 mmol) was added and the mixture heated at 140 °C in the microwave (300W, stirring) for 5 minutes. The reaction was repeated 14 more times.
The 15 reaction mixtures were combined and the solvent evaporated in vacuo (Genevac™).
The residue was dissolved in EtOAc (200 mL) and the solution washed with saturated brine solution (2 x 75 mL). The organic phase was dried (MgS04), filtered and the solvent evaporated in vacuo. The crude product was purified by flash column chromatography (40-63 mesh silica gel, 90% isohexane-EtOAc) to provide the title compound as an off-white solid (2.84 g, 63%); LC-MS, R, = 3.33 min (MeOH-FA method), m/z 298 (MH+).
5-(2-chloro-pynmidin-4-yl)-2-pyrrolidin-l-yl-benzonitrile
Figure imgf000045_0001
Pyrrolidin-l-yl-5-(4,4,5,5-tetramethyl-[l,3,2]dioxaborolan-2-yl)-benzonitrile (200 mg, 0.67 mmol), 2,4-dichloropyrimidine (120 mg, 0.81 mmol),
tetrakis(triphenylphosphine)palladium(0) (78 mg, 0.07 mmol, 10 mol%) and sodium carbonate (213 mg, 2.01 mmol) were diluted with 1 : 1 1 ,4-dioxane-H20 (4.0 mL). The mixture was then heated at 100 °C in the microwave (300W, stirring) for 10 minutes. The reaction was repeated four more times. The reaction mixtures were combined and the solvents evaporated in vacuo (Genevac™). The residue was partitioned between DCM (150 mL) and H20 (50 mL). The organic phase was washed with water (50 mL) and saturated brine solution (75 mL). The organic phase was then dried (MgS04), filtered and the solvent evaporated in vacuo. The crude product was purified by column chromatography (40-63 mesh silica gel, 70% isohexane-EtOAc) to provide the title compound as a yellow solid (727 mg, 76%); LC-MS, Rt - 2.80 min (MeOH-FA method), m z 285, 287 (MH+).
3-[4-(3-cyano-4-pyrrolidin-l-yl-phenyl)-pyrimidin-2-ylamino]-benzamide (DMX-A)
Figure imgf000046_0001
DMX-A
5-(2-Chloro-pyrimidin-4-yl)-2-pyrrolidin-l-yl-benzonitrile (50 mg, 0.18 mmol) was dissolved in NMP (1 mL). 3-Aminobenzamide (100 mg, 0.734 mmol) was added and the mixture was heated at 140 °C in the microwave (300W, stirring) for 45 minutes. The solvent was evaporated in vacuo (Genevac™). The residue was dissolved in DMSO (1.5 mL) and the crude product purified by reversed phase preparative LC-MS. Fractions containing desired product were combined and the solvents evaporated in vacuo (Genevac™). The title compound was afforded as an off-white solid (15.3 mg, 23%); LC-MS, Rt = 8.00 min ( AN ALpH2_MeOH_QC) , m/z 400 (MH+). In addition to this procedure similar reactions may alternatively be accomplished by heating at 120-160°C for 15 to 360 minutes in a microwave reactor, with products purified by either reversed phase preparative LC-MS or flash chromatography on silica. Alternatively similar reactions may also be accomplished using potassium ieri-butoxide in THF at reflux for 16 hours in a sealed tube, or potassium carbonate in DMF in a sealed tube at 125 °C for 16 hours.
The compound DMX-A had an IC50 for inhibition of ΙΚΚε of <30nM; and an IC50 for inhibition of TBK-1 of <30nM.
The following compounds were made using analogous chemistry to that described for example DMX-A:
Figure imgf000047_0001
Analytical LC-MS Methods Used
AN ALpH2_MeOH_QC : Phenomenex Luna C18 (2) 5 μηι, 150 x 4.6 mm; 35°C; A = water + 0.1% formic acid; B = MeOH; 35°C; % B: 0 min 5%, 0.5 min 5%, 7.5 min 95%, 10 min 95%, 10.1 min 5%, 13 min 5%; 1.5 mL/min.
Method X: Xterra MS C18 2.5μηι, 4.6 x 50 mm; A = water + 0.1% formic acid; B = MeCN + 0.1% formic acid; 25°C; % B: 0 min 10%, 4 min 90%, 7.5 min 90%, 7.6 min 10%; 1.0 mL/min. In addition the compounds DMX-A, B and C were tested against a number of enzymes that are generally considered members of the PI3 kinase pathway. DMX-A, B and C showed high levels of selectivity for TBKl and ΙΚΚε over the other enzymes tested: i.e. PDK-1, mTOR, ΡΒΚα, ΡΒΚβ, PDKy and AKT-l.The assays were performed against ΙΚΚε and TBK-1 as described above. The counterscreening assays were performed using radiolabeled P33 ATP using the Kinase Hotspot Technology (Reaction Biology Corp). Screening was carried out at the Km for ATP for ΙΚΚ-ε and TBK-1 and at 10 μΜ ATP for the other enzymes, with the exception of screening of DMX-A against AKT-1 which was screened at ΙμΜ ATP.
C - PTEN synthetic lethal interactions experiments
A synthetic lethality screen was performed using an isogenic model of PTEN deficiency. HCT116 human tumour cells in which both alleles of PTEN had been deactivated by gene targeting (Lee et al., Cancer Res., 2004, 64 (1 ) 6906-14) were used alongside isogenic
PTEN wild type comparator HCT-116 cells. Both cell lines were screened with a siRNA library targeting -720 kinases and kinase related genes and the affects of each siRNA on cell viability were assessed.
Cell lines were transfected with SMARTpool siRNAs using Dharmafect 3 (DF3- Dharmacon). The siRNA library (siARRAY - targeting 714 known and putative human protein kinase genes) was obtained in nine 96 well plates from Dharmacon (USA). Each well in this library contained a SMARTpool of four distinct siRNA species targeting different sequences of the target transcript. Each plate was supplemented with siCONTROL (ten wells, Dharmacon (USA)).
Cells were transfected with the SMARTpool library, where each well of the 96 well-plate contained a pool of four different siRNAs (a SMARTpool) targeting one gene. After five population doublings, cell viability in each well was estimated by use of a highly sensitive luminescent assay measuring cellular ATP levels (Cell Titre Glo, Promega). To identify loss of viability/failure to proliferate effects in each cell line, luminescence readings from each well were log transformed and then centred by the plate median, to account for plate-to-plate variation common in high- throughput screens. Well position effects were identified and eliminated and the quality of data from each plate estimated by calculating Z' -factors (Zhang et al., J. Biomol. Screen, 1999, 4 (2), 67-73) based on positive (siRNA targeting PLK1) and negative (non-targeting siRNA) controls in each plate. The dynamic range of each screen was determined by calculating Z prime values (Zhang et al.; 1999); a threshold of Z prime >0.3 was used to define acceptable screens, based upon previous screens where Z prime >0.3 is predictive of reproducible data (Iorns, E., et al., Cancer Cell, 2008, 13 (2), 91 - 104; Turner, N. C, et al, EMBO J., 2008, 27 (9), 1368 - 77). Analysis of the siRNA viability data expressed as NPI scores (normalised percent inhibition) in the PTEN +/+ and PTEN -/- HCT 116 human tumour cell lines, wherein a NPI score < 1 indicates a loss of viability, identified that PTEN null cells were particularly sensitive to the knockdown of TBK-1. The NPI results of this experiment are shown in Figure 1. TBK- 1 siRNAs show a selective lethality towards (PTEN-/-)-HCTl 16 cells.
D - PTEN matched pair ΤΒΚ17ΙΚΚε dose response experiments
Survival assays
For measurement of sensitivity to inhibitor treatment, cells were plated in 96 well plates and exposed to the drug at the indicated concentrations. For DMX-A and DMX-B, cells were dosed at 24 and 96 hours. After 7 days, cell viability was measured using CellTiter Glo Luminescent Cell Viability Assay (Promega, USA). Surviving fractions were calculated and drug sensitivity curves plotted as previously described (Farmer et ah, Nature, 2005 434 (7035), 917-921). For DMX-C, cells were dosed every three days after initial plating. After 14 days, cell viability was measured using CellTiter Glo Luminescent Cell Viability Assay (Promega, USA). Results of these experiments are shown in Figure 2: Figure 2(a) shows the DMX-A results, Figure 2(b) shows the DMX-B results and Figure 2(c) shows the DMX-C results. A leftward shift for the (PTEN-/-) curve indicates a greater sensitivity of this cell type towards the inhibitors tested. In all cases an increased sensitivity of the PTEN null cells to TBK- 1 inhibition is indicated by a leftwards shift of the curve relative to the PTEN +/+ HCT 116 line.
E - Dose response selectivity of TBKl/IKKs inhibitors on breast tumour cell lines and an ovarian tumour cell lineFor measurement of sensitivity to inhibitor treatment, cells were plated in 96 well plates and exposed to the drug at the indicated concentrations. Cells were dosed at 24 hours after plating and then inhibitor and media replaced 3 days later before cell viability was measured using CellTiter Glo Luminescent Cell Viability Assay (Promega,
USA) at 7 days post plating. Surviving fractions were calculated and drug sensitivity curves plotted as previously described (Farmer et ah, 2005). See Figures 3(a) to 3(d) for results of dose response selectively in cells with PTEN mutations, and 3(e) for results of dose response selectively in cells with PTEN and PIK3CA mutations, Figures 4(a) to (c) for results of dose response selectively in cells with PIK3CA mutations, and Figures 5(a) to (e) for results of dose response selectively in cells with wild-type PTEN and PIK3CA. Figure 6(a) and (b) show SF80 (concentration at which 80% of cells survive) and SF50 (concentration at which 50% of cells survive) results for cell lines with PTEN and/or PIKCA mutations and those with wild type PTEN and PIKCA.
DMX-C shows enhanced sensitivity towards cell lines containing either a PTEN mutation or a PIK3CA mutation compared to cell lines that contain both PTEN and PIK3CA wild-type.
All cell lines were obtained from ATCC (USA) and maintained according to the supplier instructions. STR profiling (short tandem repeat) profiling was used to verify the origin of the panel of cell lines. 8 STR loci were simultaneously amplified in a multiplex PCR reaction (Promega PowerPlex 1.2 System) and compared with the ATCC database. Where appropriate PTEN gene mutation status was determined by Sanger sequencing and western blot analysis and PIK3CA mutation status defined by Sanger sequencing. All of the cell lines used in the panel were human breast cancer cell lines with the exception of A2780 which is a human ovarian cancer cell line.
F- TBKI/IKKE sensitising to BEZ235 T47D cell line was purchased from ATCC (USA) and maintained according to the suppliers instructions. The authenticity of the cell line was verified as described above for panel of tumour cell lines. The sensitivity of this cell line to the PI3 kinase inhibitor BEZ235 was measured in the presence or absence of TBK- 1 siRNA, shown in table 1 below. Cells were reverse transfected with siRNA and after 48 hours exposed to BEZ235. BEZ235 exposure was maintained for 7 days, with BEZ235 being replenished 3 days after the initial dosing. Two out of four TBK-1 siRNAs increased sensitivity to BEZ235, in a similar fashion to the positive control siRNA targeting PIK3CA.The siRNAs used were from Dharmacon siGenome® SMARTpool siRNA human protein kinase library. Results of this experiment are shown in Figure 7.
Table 1:
Figure imgf000051_0001
Qneg represents the negative control (i.e. no siRNA), PIK3CA represents a positive control targeting PI 3CA, and TBK_l-4 represents 4 different siRNAs targeting TBKl.

Claims

Claims:
1. A compound that is an inhibitor of one or both of TBKl and ΙΚΚε, or a down-regulator of the expression of one or both of TBKl and ΙΚΚε, for use in a method of treating a cancer that is dependent on the PDkinase pathway.
2. A compound as claimed in claim 1 wherein the cancer is deficient in PTEN activity.
3. A compound as claimed in claim 2 wherein the level of PTEN protein is measured, or wherein the level of PTEN function is assessed by measuring the level of phospho-PDKl present in the cancer cells.
4. A compound as claimed in any one of claim 1 to 3 wherein the cancer includes at least some cells carrying a loss of function mutation in the PTEN gene.
5. A compound as claimed in claim 4 wherein the presence or absence of loss of function mutations is assessed by determining the presence or absence of one or more mutations or polymorphisms in a nucleic acid sequence encoding at least a portion of the PTEN enzyme.
6. A compound as claimed in any one of claim 1 to 5 wherein the cancer has overactive PDkinase signalling.
7. A compound as claimed in claim 6 wherein the level of PDkinase signalling is assessed by measuring the level of phosphorylated PDkinase pathways enzymes, such as-PDKl, AKT and mTOR, present in the cancer cells
8. A compound as claimed in claim 6 or 7 wherein the cancer includes at least some cells carrying an activating PIK3CA mutation.
9. A compound as claimed in claim 8 wherein the presence or absence of an activating PIK3CA mutation is assessed by determining the presence or absence of one or more mutations or polymorphisms in a nucleic acid sequence encoding at least a portion of the PDkinase pi 10 catalytic subunit.
10. A compound as claimed in any one of claims 1 to 9 wherein the compound is an inhibitor of an inhibitor of one or both of TBKl and ΙΚΚε selected from selected from the group consisting of: (a) compounds of Formula (I):
Figure imgf000053_0001
(I)
in which:
R1 represents an aliphatic heterocyclyl group having 4, 5, 6 or 7 ring atoms, bonded to the phenyl group shown in formula I through a ring nitrogen atom, and optionally substituted by one or more substituents selected from halogen atoms; OH; =0; Ci_4alkyl, Ci_4alkoxy, Ci_4alkoxyCi_4alkyl, C2-4alkenyl and C2^alkynyl groups, each optionally substituted by one or more substituents independently selected from halogen atoms and NRaRb groups; NO2; CN; NRaRb; CORc; O.CO.Rc; C02Ra; NRa.CORc; NRaC02Rb; C(=NH)NH2; S02Rc; NRaS02Rc; and CH(CF3)NH2;
R2 represents a phenyl or heteroaryl group which is optionally substituted by one or more substituents independently selected from:
halogen atoms;
NRaRb;
Ci_4alkyl, Ci_4alkoxy, Ci_4alkoxyCi_4alkyl, C2^alkenyl and C2-4alkynyl groups, each optionally substituted by one or more substituents independently selected from halogen atoms and NRaRb groups; and
-(CH2)P-R' in which p is 0, 1, 2, 3 or 4 and R' represents one of the following substituents: OH; N02; CN; CORc; O.CO.Rc; C02Rb; NRa.CORc; NRaC02Rb;
C(=NH)NH2; S02Rc; NRaS02Rc; and CH(CF3)NH2;
and/or which is optionally substituted on adjacent ring atoms by a group
-NRa.CO.(CH2)n- or -(CH2)n.CO.NRa- forming a fused ring;
Ra represents a hydrogen atom or a Ci_4alkyl group;
Rb represents a hydrogen atom, a Ci-4alkyl group optionally substituted by a group
NRaRa, or a cycloalkyl group in which a C¾ moiety may be replaced by an oxygen atom or an NRa group; Rc represents a hydrogen atom, a group -NRaRb, or a Ci_4alkyl group optionally substituted by a group NRaRb;
or R and Rb together may, when attached to the same nitrogen atom, represent a -(CH2)m- group in which a C¾ moiety may be replaced by an oxygen atom or an -NRa- group;
m represents 4 or 5;
n represents 1 or 2; and
each of R3 and R4 independently represents a hydrogen atom or a Ci-4alkyl group; or a salt thereof;
(b) compounds of Formula (II):
Figure imgf000054_0001
(II) and pharmaceutically acceptable salts thereof, wherein:
Rl , R2, R3, and R5 are independently chosen from the following groups: alkyl, alkylene, alkenyl, alkenylene, alkynyl, carbocycle, cycloalkyl, cycloalkenyl, heterocycle, aryl, heteroaryl, halo, hydro, hydroxyl, alkoxy, alkynyloxy, cycloalkyloxy, heterocycloxy, aryloxy, heteroaryloxy, arylalkoxy, heteroarylalkoxy, mercapto, alkylthio, arylthio, cycloalkylthio, arylalkyl, heteroarylalkyl, heteroarylalkenyl, arylalkynyl, haloalkyl, aldehyde, thiocarbonyl, O-carboxy, C-carboxy, carboxylic acid, ester, C-carboxy salt, carboxyalkyl,
carboxyalkenylene, carboxyalkyl salt, carboxyalkoxy, carboxyalkoxyalkanoyl, amino, aminoalkyl, nitro, O-carbamyl, N- carbamyl, O-thiocarbamyl, N-thiocarbamyl, C-amido, N- amido, aminothiocarbonyl, hydroxyaminocarbonyl, alkoxyaminocarbonyl, cyano, nitrile, cyanato, isocyanato, thiocyanato, isothiocyanato, sulfinyl, sulfonyl, sulfonamide,
aminosulfonyl, aminosulfonyloxy, sulfonamidecarbonyl, alkanoylaminosulfonyl,
trihalomethylsulfonyl, or trihalomethylsulfonamide, wherein any of the foregoing groups are optionally substituted at least once with alkyl, alkylene, alkenyl, alkenylene, alkynyl, carbocycle, cycloalkyl, cycloalkenyl, heterocycle, aryl, heteroaryl, halo, hydro, hydroxyl, alkoxy, alkynyloxy, cycloalkyloxy, heterocycloxy, aryloxy, heteroaryloxy, arylalkoxy, heteroarylalkoxy, mercapto, alkylthio, arylthio, cycloalkylthio, arylalkyl, heteroarylalkyl, heteroarylalkenyl, arylalkynyl, haloalkyl, aldehyde, thiocarbonyl, O-carboxy, C-carboxy, carboxylic acid, ester, C-carboxy salt, carboxyalkyl, carboxyalkenylene, carboxyalkyl salt, carboxyalkoxy, carboxyalkoxyalkanoyl, amino, aminoalkyl, nitro, O-carbamyl, N-carbamyl, O-thiocarbamyl, N- thiocarbamyl, C-amido, N- amido, aminothiocarbonyl, hydroxyaminocarbonyl, alkoxyaminocarbonyl, cyano, nitrile, cyanato, isocyanato, thiocyanato, isothiocyanato, sulfinyl, sulfonyl, sulfonamide,
aminosulfonyl, aminosulfonyloxy, sulfonamidecarbonyl, alkanoylaminosulfonyl, trihalomethylsulfonyl, or trihalomethylsulfonamide, or, R2 and either Rl or R3, together with the carbon atoms to which they are bound, form an optionally-substituted cycloalkyl, heterocycle, aryl, or heteroaryl;
R4 is independently chosen from hydro, halo, and an optionally-substituted group chosen from lower alkyl, haloalkyl, alkoxy, arylalkoxy, heteroarylalkoxy, and heterocycloalkoxy; R6 and R7 are independently chosen from hydro, halo, and lower alkyl;
or R6, taken together with R7, form an aryl or heteroaryl ring;
(c) a compound of Formula (III):
Figure imgf000055_0001
(III)
(d) compounds of Formula (IV):
Figure imgf000056_0001
(IV) wherein n is from 2-6;
Ri is a 5 or 6 membered cyclic alkyl ring or aromatic cyclic ring, wherein optionally one or more carbons in the ring structure is substituted by an O, N or S;
R2 is a 5 or 6 membered cyclic alkyl ring or arromatic cyclic ring, wherein optionally one or more carbons in the ring structure is substituted by an O, N or S, or is a linear or branched substituted or unsubstituted Ci-Ce alkyl, Ci-C6 alkoxy or C2-C6 alkenyl, wherein when substituted the substituent group may be a C(=0)NH2;COOH; OH, NH2, N02, and R3 is a halo, H, CH3, CN, N02;
(e) compounds of Formula (V):
Figure imgf000056_0002
(V) wherein: R is C3_8-cycloalkyl; X is O, NR7 or C3_6-heterocycloalkyl;
R2 is aryl, heteroaryl, fused or unfused aryl-C3_6-heterocycloalkyl or fused or unfused heteroaryI-C3-6- heterocycloalkyl, each of which is optionally substituted by one or more substitutents selected from aryl, heteroaryl, Ci_6-alkyl, C3_7-cycloalkyl and a group A, wherein said Ci-6-alkyl group is optionally substituted by one or more substituents selected from aryl, heteroaryl, R10 and a group A, said heteroaryl group is optionally substituted by one or more R<10> groups; and wherein said C3-6- heterocycloalkyl group optionally contains one or more groups selected from oxygen, sulfur, nitrogen and CO;
R3 is Ci-6-alkyl optionally substituted by one or more substituents selected from aryl, heteroaryl, -NR4R5, -OR6, -NR7(CO)R6, -NR7(CO)NR4R5, -NR7S02R6, -NR7COOR7, - CONR4R5, C3_6-heterocycloalkyl and
Figure imgf000057_0001
wherein said aryl, heteroaryl and C3-6 -heterocycloalkyl groups are each optionally substituted by one or more substituents selected from -Ci-6-alkyl and a group A, wherein said -Ci-6-alkyl group is optionally substituted by one or more substituents selected from aryl, heteroaryl and a group A;
A is selected from halogen, hydroxyl, cyano, trifluoromethyl, alkoxy, -NO2, -NH2, -NR4R5, - OR6, -NR7(CO)R6, -NR7(CO)NR4R5, -NR7COOR7, -NR7 (S02)R6, - C02H, - NR7CS02)NR4R5, -COOR7, -C0NR4R5, COR6, S02NR4R5 and -S02CH3; each R4 and R5 is independently selected from hydrogen, C3_7-cycloalkyl, aryl, heteroaryl, Ci- 6-alkyl and a C3_6-heterocycloalkyl ring optionally further containing one or more groups selected from oxygen, sulfur, nitrogen and CO and optionally substituted by one or more R10 groups, wherein said Ci-6-alkyl is optionally substituted by one or more substituents selected from halogen, cyano, hydroxyl, aryl, heteroaryl, -NR8R9, -NR7 (CO)R6, NR7COOR6, -NR7 (S02)R6, -COOR6, -CONR8R9, OR10, -S02R6 and a C3_6-heterocycloalkyl ring optionally further containing one or more groups selected from oxygen, sulfur, nitrogen and CO and optionally substituted by one or more or R10 groups; or R4 and R5 together with the N to which they are attached form a C3_6-heterocycloalkyl ring optionally further containing one or more groups selected from oxygen, sulfur, nitrogen and CO, wherein said C3-6-heterocycloalkyl ring may be saturated or unsaturated and is optionally substituted with one or more groups selected from NR8R9 and R10 groups; each R6 is independently selected from Ci-6-alkyl, C3_7-cycloalkyl, C4_7-heterocycloalkyl, aryl and heteroaryl, each of which may be optionally substituted by one or more substituents selected from halogen, R10 and -NR8R9; each R7 is selected from hydrogen, Ci-6-alkyl and C3-7-cycloalkyl, wherein said Ci-6-alkyl is optionally substituted by one or more halogens; each of R8 and R9 is independently selected from hydrogen and Ci-6-alkyl, wherein said C1-6- alkyl group is optionally substituted by one or more halogens; or
R8 and R9 together with the N to which they are attached form a C4_6-heterocycloalkyl ring optionally further containing one or more heteroatoms selected from oxygen and sulfur, wherein said C4-6-heterocycloalkyl ring is optionally substituted by one or more R10 groups; and each R10 is selected from halogen, C3_7-cycloalkyl and Ci_6-alkyl optionally substituted by one or more halogens, wherein where R10 is Ci-6-alkyl and two or more R10 groups are attached to the same carbon atom, the R10 groups may be linked to form a spiroalkyl group;
(f) compounds of Formula (VI):
Figure imgf000058_0001
wherein: R1 is -NR7(CO)Ru;
R2 is aryl, heteroaryl, fused aryl-C3_6 -heterocycloalkyl or fused heteroaryl-C3_6- heterocycloalkyl, each of which is optionally substituted by one or more substitutents selected from aryl, heteroaryl, Ci_6-alkyl, C3_6 -heterocycloalkyl and a group A, wherein said Ci-6-alkyl group is in turn optionally substituted by one or more substituents selected from aryl, heteroaryl, C3-6 -heterocycloalkyl and a group A, said heteroaryl group is optionally substituted by one or more R10 groups; and wherein each C3-6 -heterocycloalkyl group is optionally substituted by one or more groups selected from Ci_6-alkyl, Ci_6-haloalkyl, and A, and which optionally contains one or more groups selected from oxygen, sulphur, nitrogen and CO;
R3 is H, halogen, cyano or Ci_6-alkyl; A is selected from halogen, hydroxyl, cyano, trifluoromethyl, -NO2, -NH2, -NR4R5, -OR6, - NR7 (CO)R6, -NR7(CO)NR4R5, -NR7COOR7, -NR7(S02)R6, -C02H, -NR7 (S02)NR4R5, - COOR7, -CONR4R5, COR6 and -S02CH3; each R4 and R5 is independently selected from hydrogen, C3_7-CycloaIkyI, aryl, heteroaryl, Ci-6-alkyl and a C3-6 -heterocycloalkyl ring optionally further containing one or more groups selected from oxygen, sulfur, nitrogen and CO, and optionally substituted by one or more R10 groups, wherein said Ci-6-alkyl is optionally substituted by one or more substituents selected from halogen, cyano, hydroxyl, aryl, heteroaryl, -NR8R9, -NR7(CO)R6, - NR7COOR6, -NR7 (S02)R6, -COOR6, -CONR8R9, OR10, -S02R6 and a C3-6-heterocycloalkyl ring optionally further containing one or more groups selected from oxygen, sulfur, nitrogen and CO and optionally substituted by one or more or R10 groups; or
R4 and R5 together with the N to which they are attached form a C3-6 -heterocycloalkyl ring optionally further containing one or more groups selected from oxygen, sulfur, nitrogen and CO, wherein said C3_6-heterocycloalkyl ring may be saturated or unsaturated and is optionally substituted with one or more groups selected from NR8R9 and R10; each R6 is independently selected from Ci_6-alkyl, C3_7-cycloalkyl, C4_7-heterocycloalkyl, aryl and heteroaryl, each of which may be optionally substituted by one or more substituents selected from halogen, R10 and -NR8R9; each R7 is selected from hydrogen, Ci_6-alkyl and C3_6-cycloalkyl, wherein said Ci_6-alkyl is optionally substituted by one or more halogens; each of R8 and R9 is independently selected from hydrogen and Ci-6-alkyl, wherein said C1-6- alkyl group is optionally substituted by one or more halogens; or
R8 and R9 together with the N to which they are attached form a C4-6-heterocycloalkyl ring optionally further containing one or more heteroatoms selected from oxygen and sulfur, wherein said C4_6 -heterocycloalkyl ring is optionally substituted by one or more R10 groups; and each R10 is selected from C3-7-cycloalkyl and Ci-6-alkyl optionally substituted by one or more halogens, wherein where R10 is Ci_6-alkyl and two or more R10 groups are attached to the same carbon atom, the R10 groups may be linked to form a spiroalkyl group; and each R11 is independently selected from Ci-6-alkyl, C3_7-cycloalkyl, Ci-6-alkyl-C3_7-cycloalkyl, C4-7- heterocycloalkyl, aryl and heteroaryl, each of which may be optionally substituted by one or more substituents selected from A;
Figure imgf000060_0001
(h) compound of Formula (VI
Figure imgf000061_0001
(VIII) (i) compounds of Formula (X):
Figure imgf000061_0002
(X)
wherein: n is 0, 1,2, 3, or 4 ;
each R1 which may be the same or different, independently represents H, halogen or a group (X)a(Y)bZ;
X represents -O- or -CONH-;
a is 0 or 1 ;
Y represents Ci_6alkylene-;
b is 0 or 1 ;
Z represents hydroxy, C1-6 alkyl, C1-6 haloalkyl, C5-7 heterocyclyl, C1-6 alkoxyalkyl, C1-6 haloalkoxyalkyl;
R2 represents a group -(X1)c(Y1)dZ1;
wherein X1 represents -Ci-12 alkylene-;
c is O or l;
Y1 represents -0-;
d is 0 or 1;
Z1 represents H, aryl or heteroaryl each of which contains 5-14 ring atoms, C5-7 heterocyclyl, C5-7 cycloalkyl, C5-7 cycloalkenyl, (each of which aryl, heteroaryl, heterocyclyl, cycloalkyl, cycloalkenyl may be optionally substituted by one or more substituents independently selected from C1-6 alkyl, Ci_6 haloalkyl, halogen, C1-6 alkoxy, Ci_6 haloalkoxy, S02R3, Ci_6 hydroxyalkyl); R3 represents H or C1-6 alkyl;
or pharmaceutically acceptable salts, solvates or physiologically functional derivatives thereof
Figure imgf000062_0001
(Xa)
where Z is aryl or heteroaryl each of which is optionally substituted by one of more substituents independently selected from halogen, CF3, CH2OH, SO2CH3, CH3, OCH3
(j) a compound of formula (XI)
Figure imgf000062_0002
(XI)
wherein R6 is Br, CN, CONH2 or CI;
wherein R7 represents
Figure imgf000063_0001
62 and wherein R re resents
Figure imgf000064_0001
or a pharmaceutically acceptable salt, solvate or physiologically functional derivative thereof; and
(k) compounds of the formula (XII)
Figure imgf000064_0002
(XII) in which R1 denotes A, -[C(R )( R4)]„Ar, -[C(R3)( R4)]„Het or --[C(R3)( R4)]„Cyc,
R2 denotes H, A, Hal, CN, -[C(R ) 2]„-Ar', -[C(R3) 2]„-Het', -[C(R3) 2]„-Cyc, OR3 or N(R ) 2,
R3 denotes H or A,
R4 denotes H, A, -[C(R3) 2]„OR3, -[C(R3) 2]„COOR3, -[C(R3) 2]„N(R3) 2 or -[C(R3) 2]„Het, A denotes unbranched or branched alkyl having 1-6 C atoms, in which one or two CH2 groups may be replaced by O, N and/or S atoms and/or by— CH=CH— groups and/or, in addition, 1-7 H atoms may be replaced by F,
Cyc denotes cycloalkyl having 3, 4, 5, 6 or 7 C atoms,
Ar denotes phenyl which is unsubstituted or mono-, di- or trisubstituted by Hal, A, -[C(R3) 2]„OR3, N(R3)2, N02, CN, COOR3, CON(R3)2, NR3COA, NR3S02A, COR3, S02N(R )2 and/or S(0)„A,
Ar' denotes phenyl which is unsubstituted or mono-, di- or trisubstituted by Hal, A, OR3, N(R3) 2, SR3, N02, CN, COOR3, CON(R3)2, NR3COA, NR3S02A, S02N(R3) 2, S(0)„A, CO- Heti, Heti, [C(R3) 2]„N(R3)2, [C(R3)2]„Heti, 0[C(R )2]„N(R3) 2, 0[C(R )2]„Heti, NHCOOA, NHCON(R3)2, NHCOO[C(R )2]„N(R3) 2, NHCOO[C(R3)2]¾Heti, NHCONH[C(R )2]„N(R3) 2, NHCONH[C(R3)2]„Heti, OCONH[C(R3)2]„N(R3) 2, OCONH[C(R3) 2]»Heti, CO-Heti, CHO and/or COA,
Het denotes a mono- or bicyclic saturated, unsaturated or aromatic heterocycle having 1 to 4 N, and/or O and/or S atoms which is unsubstituted or mono- or disubstituted by Hal, A, OR3, N(R3)2, N02, CN, COOR3, CON(R3)2, NR COA, NR3S02A, CORs, S02NR3 and/or S(0)„A, Het' denotes a mono-, bi- or tricyclic saturated, unsaturated or aromatic heterocycle having 1 to 4 N, O and/or S atoms which is unsubstituted or may be mono- or disubstituted by Hal, A, OR3, N(R3)2, SR3, N02, CN, COOR3, CON(R )2, NR COA, NR S02A, S02N(R3) 2, S(O)„ A, CO-Heti, Heti, [C(R3) 2]„N(R3) 2, [C(R3)
Figure imgf000065_0001
0[C(R3)2]„N(R3) 2, 0[C(R3)2]„Het!, NHCOOA, NHCON(R3)2, NHCOO[C(R3)2]„N(R3) 2, NHCOO[C(R3)2]„Het.sup.l,
NHCONH[C(R3)2]„N(R3) 2, NHCONH[C(R )2]„Heti, OCONH[C(R3)2]„N(R3) 2,
OCONH[C(R )2]„Heti, CO-Heti, CHO, COA, =S, =NH, =NA and/or =0 (carbonyl oxygen), Heti denotes a monocyclic saturated heterocycle having 1 to 2 N and/or O atoms, which may be mono- or disubstituted by A, OA, OH, Hal and/or =0 (carbonyl oxygen),
Hal denotes F, CI, Br or I,
N denotes 0, 1 or 2, and pharmaceutically usable salts, tautomers and stereoisomers thereof, including mixtures thereof in all ratios.
11. A compound as claimed in any one of claims 1 to 9 wherein the compound is a down- regulator of the expression of one or both of TBKl and ΙΚΚε, selected from the group consisting of siRNA reagents with the target sequence:
5 '-GAACGUAGAUUAGCUUAUA-3 ' (TBKl) [SEQ ID NO. 1],
5 -UG AC AG AG AUUU ACU AUC A- 3 ' (TBKl) [SEQ ID NO. 2],
5'-UAAAGUACAUCCACGUUAU-3' (TBKl) [SEQ ID NO.3],
5 -GGAUAUCGACAGCAGAUUA-3 ' (TBKl) [SEQ ID NO. 4];
5'-ACAAACAGUUCAAGAAGUC-3' (ΙΚΚε) [SEQ ID NO. 5],
5'-GGACCUGCUUCUCCACAUG-3' (ΙΚΚε) [SEQ ID NO. 6],
5 -GAAGGCGGCUGCAGAACUG-3 ' (ΙΚΚε) [SEQ ID NO. 7],
5'-GGCAGGAGCUAAUGUUUCG-3' (ΙΚΚε) [SEQ ID NO. 8].
12. A compound as claimed in any one of claims 1 to 11 that is used in combination with one or more further therapeutic agents.
13. A compound as claimed in claim 12 wherein a further therapeutic agent is selected from the group consisting of inhibitors of mTOR, inhibitors of PDkinase, inhibitors of both mTOR and PDkinase, inhibitors of AKT, and inhibitors of PDK1.
14. A compound that is an inhibitor of one or both of TBKl and ΙΚΚε, or a down-regulator of the expression of one or both of TBKl and ΙΚΚε, for use in a method of treating a cancer that is dependent on the PDkinase pathway comprising:
- identifying a cancer in the individual as being dependent on the PDkinase pathway, and
- administering the compound to the individual.
15. A compound as claimed in claim 14 wherein the cancer is as described in any one of claims 2 to 9, or the compound is as described in any one of claims 10 to 13.
16. A method of treating cancer in an individual in whom the cancer is dependent on the PDkinase pathway, comprising administering to the individual a compound that is an inhibitor of one or both of TBK1 and ΙΚΚε, or a down-regulator of the expression of one or both of TBK1 and lKKe.
17. A method as claimed in claim 16 wherein the cancer is as described in any one of claims 2 to 9, or the compound is as described in any one of claims 10 to 13.
18. A method of treating cancer in an individual comprising:
- identifying the cancer as dependent on the PDkinase pathway, and
- administering to the individual a compound that is an inhibitor of one or both of TBK1 and Ι Κε, or a down-regulator of the expression of one or both of TBK1 and ΙΚΚε.
19. A method as claimed in claim 18 wherein the cancer is as described in any one of claims 2 to 9, or the compound is as described in any one of claims 10 to 13.
20. A method of determining the susceptibility of a cancer in an individual to treatment with a compound that is an inhibitor of one or both of TBK1 and ΙΚΚε, or a down-regulator of the expression of one or both of TBK1 and ΙΚΚε, comprising determining whether the cancer is dependent on the PDkinase pathway, whereby a dependence on the PDkinase pathway indicates that the cancer in the individual has an increased susceptibility to treatment with a compound that is an inhibitor of one or both of TBK1 and ΙΚΚε, or a down-regulator of the expression of one or both of TBK1 and ΙΚΚε.
21. A method as claimed in claim 20 wherein the cancer is as described in any one of claims 2 to 9, or the compound is as described in any one of claims 10 to 13.
22. A method of determining a suitable treatment for an individual with a cancer comprising:
(i) determining whether the cancer is dependent on the PDkinase pathway; and
(ii) determining a suitable treatment, whereby a compound that is an inhibitor of one or both of TBK1 and ΙΚΚε, or a down-regulator of the expression of one or both of TBK1 and ΙΚΚε is suitable in the treatment of an individual with a cancer that is dependent on the PDkinase pathway.
23. A method as claimed in claim 22 wherein the cancer is as described in any one of claims 2 to 9, or the compound is as described in any one of claims 10 to 13.
24. A method of predicting the response of a cancer condition in an individual to a treatment with a compound that is an inhibitor of one or both of TBKl and ΙΚΚε, or a down-regulator of the expression of one or both of TBKl and ΙΚΚε, comprising:
- determining whether the cancer is dependent on the PDkinase pathway.
25. A method as claimed in claim 24 wherein the cancer is as described in any one of claims 2 to 9, or the compound is as described in any one of claims 10 to 13.
PCT/GB2012/051978 2011-08-15 2012-08-14 Compounds and their uses WO2013024282A2 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
GBGB1114051.4A GB201114051D0 (en) 2011-08-15 2011-08-15 Compounds and their uses
GB1114051.4 2011-08-15

Publications (2)

Publication Number Publication Date
WO2013024282A2 true WO2013024282A2 (en) 2013-02-21
WO2013024282A3 WO2013024282A3 (en) 2013-05-16

Family

ID=44764528

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/GB2012/051978 WO2013024282A2 (en) 2011-08-15 2012-08-14 Compounds and their uses

Country Status (2)

Country Link
GB (1) GB201114051D0 (en)
WO (1) WO2013024282A2 (en)

Cited By (23)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8598167B1 (en) 2011-02-28 2013-12-03 Epizyme, Inc. Substituted 6,5-fused bicyclic heteroaryl compounds
WO2014128486A1 (en) * 2013-02-21 2014-08-28 Domainex Limited Pyrimidine compounds useful in the treatment of diseases mediated by ikke and/or tbk1 mechanisms
US9186361B2 (en) 2013-03-15 2015-11-17 Novartis Ag Compounds and compositions for the treatment of parasitic diseases
US9233961B2 (en) 2013-03-15 2016-01-12 Novartis Ag Compounds and compositions for the treatment of parasitic diseases
US9296701B2 (en) 2012-04-24 2016-03-29 Vertex Pharmaceuticals Incorporated DNA-PK inhibitors
US9296754B2 (en) 2013-03-15 2016-03-29 Novartis Ag Compounds and compositions for the treatment of parasitic diseases
US9303034B2 (en) 2013-12-19 2016-04-05 Novartis Ag Compounds and compositions for the treatment of parasitic diseases
US9340557B2 (en) 2013-03-12 2016-05-17 Vertex Pharmaceuticals Incorporated Substituted quinoxaline DNA-PK inhibitors
WO2017102091A1 (en) 2015-12-18 2017-06-22 Bayer Pharma Aktiengesellschaft Heteroarylbenzimidazole compounds
US9783547B2 (en) 2014-01-15 2017-10-10 Centre National De La Recherche Scientifique (Cnrs) Water soluble 4-azapodophyllotoxin analogs
WO2017207534A1 (en) 2016-06-03 2017-12-07 Bayer Pharma Aktiengesellschaft Substituted heteroarylbenzimidazole compounds
DE102016113714A1 (en) 2016-07-26 2018-02-01 Rosa Karl Transfection procedure with non-viral gene delivery systems
US10040781B2 (en) 2014-09-26 2018-08-07 Gilead Sciences, Inc. Tank-binding kinase inhibitor compounds
US10039761B2 (en) 2013-10-17 2018-08-07 Vertex Pharmaceuticals Incorporated Co-crystals and pharmaceutical compositions comprising the same
US10072001B2 (en) 2014-06-03 2018-09-11 Gilead Sciences, Inc. Tank-binding kinase inhibitor compounds
US10253062B2 (en) 2014-12-23 2019-04-09 Margaret Anne Brimble Amino acid and peptide conjugates and uses thereof
US10316049B2 (en) 2015-12-17 2019-06-11 Gilead Sciences, Inc. Tank-binding kinase inhibitor compounds
EP3545956A1 (en) 2013-04-18 2019-10-02 Arrien Pharmaceuticals LLC 3,5-(un)substituted-1h-pyrrolo[2,3-b]pyridine, 1h-pyrazolo[3,4-b]pyridine and 5h- pyrrolo[2,3-b]pyrazine dual itk and jak3 kinase inhibitors
US10576144B2 (en) 2013-06-28 2020-03-03 Auckland Uniservices Limited Amino acid and peptide conjugates and conjugation process
CN110862380A (en) * 2019-10-24 2020-03-06 嘉兴特科罗生物科技有限公司 Small molecule compound
US11058686B2 (en) 2017-02-23 2021-07-13 Domainex Limited 5-(pyrimidin-4-yl)-2-(pyrrolidin-1-yl)nicotinonitrile compounds as IKKE, TBK1 and/or SIK2 kinases inhibitors
US11110108B2 (en) 2016-09-27 2021-09-07 Vertex Pharmaceuticals Incorporated Method for treating cancer using a combination of DNA-damaging agents and DNA-PK inhibitors
US11464853B2 (en) 2016-02-26 2022-10-11 Auckland Uniservices Limited Amino acid and peptide conjugates and conjugation process

Citations (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005075465A1 (en) 2004-02-09 2005-08-18 Glaxo Group Limited Benzimidazol substituted thiopene derivatives with atctivity on ikk3
US20070014951A1 (en) 2003-12-22 2007-01-18 Novo Nordisk A/S Transparent, flexible, impermeable plastic container for storage of pharmaceutical liquids
WO2009030890A1 (en) 2007-09-03 2009-03-12 University Court Of The University Of Dundee Pyrimidine compounds for the treatment of cancer, septic shock and/or primary open angle glaucoma
WO2009122180A1 (en) 2008-04-02 2009-10-08 Medical Research Council Pyrimidine derivatives capable of inhibiting one or more kinases
WO2010100431A1 (en) 2009-03-04 2010-09-10 Medical Research Council Technology Pyrrolopyrimidines used as kinase inhibitors
WO2010127754A1 (en) 2009-05-05 2010-11-11 Merck Patent Gmbh 3-([1,2,3]triazole-4-yl)-pyrrolo[2,3-b]pyridine derivates
US20110038862A1 (en) 2008-02-21 2011-02-17 Universite Libre De Bruxelles Method and kit for the detection of genes associated with pik3ca mutation and involved in pi3k/akt pathway activation in the er-postitive and her2-positive subtypes with clinical implications
WO2011046970A1 (en) 2009-10-12 2011-04-21 Myrexis, Inc. Amino - pyrimidine compounds as inhibitors of tbkl and/or ikk epsilon
WO2012010826A1 (en) 2010-07-19 2012-01-26 Domainex Limited Pyrimidine compounds as inhibitors of protein kinases ikk epsilon and/or tbk-1, processes for their preparation, and pharmaceutical compositions containing them
WO2012059171A1 (en) 2010-11-01 2012-05-10 Merck Patent Gmbh 7-([1,2,3]triazol-4-yl)-pyrrolo[2,3-b]pyrazine derivatives

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005034978A2 (en) * 2003-10-02 2005-04-21 Xantos Biomedicine Ag MEDICAL USE OF IKK&epsiv; OR OF INHIBITORS THEREOF
US20070149469A1 (en) * 2003-10-02 2007-06-28 Christian Korherr Medical use of tbk-1 or of inhibitors thereof

Patent Citations (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070014951A1 (en) 2003-12-22 2007-01-18 Novo Nordisk A/S Transparent, flexible, impermeable plastic container for storage of pharmaceutical liquids
WO2005075465A1 (en) 2004-02-09 2005-08-18 Glaxo Group Limited Benzimidazol substituted thiopene derivatives with atctivity on ikk3
WO2009030890A1 (en) 2007-09-03 2009-03-12 University Court Of The University Of Dundee Pyrimidine compounds for the treatment of cancer, septic shock and/or primary open angle glaucoma
US20110038862A1 (en) 2008-02-21 2011-02-17 Universite Libre De Bruxelles Method and kit for the detection of genes associated with pik3ca mutation and involved in pi3k/akt pathway activation in the er-postitive and her2-positive subtypes with clinical implications
WO2009122180A1 (en) 2008-04-02 2009-10-08 Medical Research Council Pyrimidine derivatives capable of inhibiting one or more kinases
WO2010100431A1 (en) 2009-03-04 2010-09-10 Medical Research Council Technology Pyrrolopyrimidines used as kinase inhibitors
WO2010127754A1 (en) 2009-05-05 2010-11-11 Merck Patent Gmbh 3-([1,2,3]triazole-4-yl)-pyrrolo[2,3-b]pyridine derivates
US20120053178A1 (en) 2009-05-05 2012-03-01 Merck Patent Gesellschaft Mit Beschrankter Haftung 3-(1,2,3-Triazol-4-yl) pyrrolo [2,3-b] pyridine derivatives
WO2011046970A1 (en) 2009-10-12 2011-04-21 Myrexis, Inc. Amino - pyrimidine compounds as inhibitors of tbkl and/or ikk epsilon
WO2012010826A1 (en) 2010-07-19 2012-01-26 Domainex Limited Pyrimidine compounds as inhibitors of protein kinases ikk epsilon and/or tbk-1, processes for their preparation, and pharmaceutical compositions containing them
WO2012059171A1 (en) 2010-11-01 2012-05-10 Merck Patent Gmbh 7-([1,2,3]triazol-4-yl)-pyrrolo[2,3-b]pyrazine derivatives

Non-Patent Citations (43)

* Cited by examiner, † Cited by third party
Title
ANDREWS ET AL., SCIENCE SIGNALLING, 2007, pages 407
ANGELL; BAULCOMBE, THE EMBO JOURNAL, vol. 16, no. 12, 1997, pages 3675 - 3684
AUSUBEL ET AL.: "Current Protocols in Molecular Biology", 1992, JOHN WILEY & SONS
BOARD ET AL., CLINICAL CHEMISTRY, vol. 54, no. 4, 2008, pages 757 - 760
BOEHM ET AL., CELL, vol. 129, 2007, pages 1065 - 1079
CHANG ET AL., LEUKEMIA RESEARCH, vol. 30, 2006, pages 262 - 265
CROOKE, ANN. REV. PHARMACOL. TOXICOL., vol. 32, 1992, pages 329 - 376
EHRLICH, SCIENCE, vol. 252, 1991, pages 1643 - 1650
EHRLICH: "PCR technology", 1989, STOCKTON PRESS
ELBASHIR ET AL., NATURE, vol. 411, 2001, pages 494 - 498
ENGLEMAN ET AL., NAT. MED., vol. 14, 2008, pages 1351 - 1356
ENGLEMAN, NATURE REVIEWS CANCER, vol. 9, 2009, pages 550 - 562
FARMER ET AL., NATURE, vol. 434, no. 7035, 2005, pages 917 - 921
FIRE ET AL., NATURE, vol. 391, 1998, pages 806 - 811
FRATTINI ET AL., BR. J. CANCER, vol. 97, no. 8, 2007, pages 1139 - 45
GUO ET AL., THE AMERICAN JOURNAL OF PATHOLOGY, vol. 175, 2009, pages 324 - 333
GUO ET AL., THE JOURNAL OF BIOLOGICAL CHEMISTRY, vol. 285, 2010, pages 3676 - 3684
HOLMES, NATURE REVIEWS DRUG DISCOVERY, vol. 10, 2011, pages 563 - 564
INNIS ET AL,: "PCR protocols; A Guide to Methods and Applications", 1990, ACADEMIC PRESS
J. BIOL. CHEM., vol. 284, no. 21, 2009, pages 14136 - 14146
KANG ET AL., PNAS, vol. 103, no. 5, 2006, pages 1289 - 1294
LEE ET AL., CANCER RES., vol. 64, no. 19, 2004, pages 6906 - 14
LORNS, E. ET AL., CANCER CELL, vol. 13, no. 2, 2008, pages 91 - 104
MULLIS, COLD SPRING HARBOR SYMP
NAGATA ET AL., CANCER CELL, vol. 6, no. 2, 2004, pages 117 - 27
OU ET AL., MOLECULAR CELL, vol. 41, 2011, pages 458 - 70
PEYMAN; ULMAN, CHEMICAL REVIEWS, vol. 90, 1990, pages 543 - 584
QUI ET AL., ONCOGENE, vol. 19, 2000, pages 5651 - 5661
SAMBROOK; RUSSELL: "Molecular Cloning: a Laboratory Manual: 3rd edition,", 2001, COLD SPRING HARBOR LABORATORY PRESS
SAMUELS ET AL., SCIENCE, vol. 304, no. 5670, 2004, pages 55
STEPHENS ET AL., CURRENT OPINION IN PHARMACOLOGY, vol. 5, 2005, pages 357 - 365
TURNER, N. C. ET AL., EMBO J., vol. 27, no. 9, 2008, pages 1368 - 77
VASUDEVAN ET AL., CANCER CELL, vol. 16, no. 1, 2009, pages 21 - 32
VOINNET; BAULCOMBE, NATURE, vol. 389, 1997, pages 553
WANG ET AL., BIORG. MED. CHEM. LETT, vol. 22, 2012, pages 2063 - 2068
WILLIAMS ET AL., BIOCHEM. SOC. TRANS, vol. 37, 2009, pages 615 - 626
XIE ET AL., DRUG DISCOVERY TODAY, vol. 11, no. 1-2, 2006, pages 67 - 73
XIE ET AL., PNAS, vol. 108, no. 16, 2011, pages 6474 - 6479
YING ET AL., CANCER DISCOVERY, vol. 1, no. 2, 2011, pages 158 - 169
ZAMORE ET AL., CELL, vol. 101, 2000, pages 25 - 33
ZAMORE, NATURE STRUCTURAL BIOLOGY, vol. 8, 2001, pages 746 - 750
ZHANG ET AL., J. BIOMOL. SCREEN, vol. 4, no. 2, 1999, pages 67 - 73
ZHANG ET AL., ONCOGENE, vol. 19, 2000, pages 4346 - 4353

Cited By (55)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9637472B2 (en) 2011-02-28 2017-05-02 Epizyme, Inc. Substituted 6,5-fused bicyclic heteroaryl compounds
US8598167B1 (en) 2011-02-28 2013-12-03 Epizyme, Inc. Substituted 6,5-fused bicyclic heteroaryl compounds
US8962620B2 (en) 2011-02-28 2015-02-24 Epizyme, Inc. Substituted 6,5-fused bicyclic heteroaryl compounds
US10273223B2 (en) 2011-02-28 2019-04-30 Epizyme, Inc. Substituted 6,5-fused bicyclic heteroaryl compounds
US9206157B2 (en) 2011-02-28 2015-12-08 Epizyme, Inc. Substituted 6,5-fused bicyclic heteroaryl compounds
US10501439B2 (en) 2012-04-24 2019-12-10 Vertex Pharmaceuticals Incorporated DNA-PK inhibitors
US9376448B2 (en) 2012-04-24 2016-06-28 Vertex Pharmaceuticals Incorporated DNA-PK inhibitors
US9878993B2 (en) 2012-04-24 2018-01-30 Vertex Pharmaceuticals Incorporated DNA-PK inhibitors for treatment of cancer
US11021465B2 (en) 2012-04-24 2021-06-01 Vertex Pharmaceuticals Incorporated DNA-PK inhibitors
US11008305B2 (en) 2012-04-24 2021-05-18 Vertex Pharmaceuticals Incorporated DNA-PK inhibitors
US10442791B2 (en) 2012-04-24 2019-10-15 Vertex Pharmaceuticals Incorporated DNA-PK inhibitors
US10076521B2 (en) 2012-04-24 2018-09-18 Vertex Pharamceuticals Incorporated DNA-PK inhibitors
US9296701B2 (en) 2012-04-24 2016-03-29 Vertex Pharmaceuticals Incorporated DNA-PK inhibitors
US9925188B2 (en) 2012-04-24 2018-03-27 Vertex Pharmaceuticals Incorporated DNA-PK inhibitors and uses thereof
US9592232B2 (en) 2012-04-24 2017-03-14 Vertex Pharmaceuticals Incorporated DNA-PK inhibitors
US10391095B2 (en) 2012-04-24 2019-08-27 Vertex Pharmaceuticals Incorporated DNA-PK inhibitors
US9433622B2 (en) 2013-02-21 2016-09-06 Case Western Reserve University Pyrimidine compounds useful in the treatment of diseases mediated by IKKE and/or TBK1 mechanisms
JP2016513120A (en) * 2013-02-21 2016-05-12 ドメイネクス リミテッド New pyrimidine compounds
WO2014128486A1 (en) * 2013-02-21 2014-08-28 Domainex Limited Pyrimidine compounds useful in the treatment of diseases mediated by ikke and/or tbk1 mechanisms
US9987284B2 (en) 2013-03-12 2018-06-05 Vertex Pharmaceuticals Incorporated Substituted benzooxadiazole DNA-PK inhibitors
US9359380B2 (en) 2013-03-12 2016-06-07 Vertex Pharmaceuticals Incorporated DNA-PK inhibitors
US11813267B2 (en) 2013-03-12 2023-11-14 Vertex Pharmaceuticals Incorporated DNA-PK inhibitors
US10786512B2 (en) 2013-03-12 2020-09-29 Vertex Pharmaceuticals Incorporated DNA-PK inhibitors
US10973830B2 (en) 2013-03-12 2021-04-13 Vertex Pharmaceuticals Incorporated Substituted quinoxaline DNA-PK inhibitors
US10258627B2 (en) 2013-03-12 2019-04-16 Vertex Pharmaceutical Incorporated DNA-PK inhibitors
US9340557B2 (en) 2013-03-12 2016-05-17 Vertex Pharmaceuticals Incorporated Substituted quinoxaline DNA-PK inhibitors
US9296754B2 (en) 2013-03-15 2016-03-29 Novartis Ag Compounds and compositions for the treatment of parasitic diseases
US9233961B2 (en) 2013-03-15 2016-01-12 Novartis Ag Compounds and compositions for the treatment of parasitic diseases
US9186361B2 (en) 2013-03-15 2015-11-17 Novartis Ag Compounds and compositions for the treatment of parasitic diseases
EP3545956A1 (en) 2013-04-18 2019-10-02 Arrien Pharmaceuticals LLC 3,5-(un)substituted-1h-pyrrolo[2,3-b]pyridine, 1h-pyrazolo[3,4-b]pyridine and 5h- pyrrolo[2,3-b]pyrazine dual itk and jak3 kinase inhibitors
US10576144B2 (en) 2013-06-28 2020-03-03 Auckland Uniservices Limited Amino acid and peptide conjugates and conjugation process
US10039761B2 (en) 2013-10-17 2018-08-07 Vertex Pharmaceuticals Incorporated Co-crystals and pharmaceutical compositions comprising the same
US10716789B2 (en) 2013-10-17 2020-07-21 Vertex Pharmaceuticals Incorporated Co-crystals and pharmaceutical compositions comprising the same
US10596175B2 (en) 2013-12-19 2020-03-24 Novartis Ag Compounds and compositions for the treatment of parasitic diseases
US9303034B2 (en) 2013-12-19 2016-04-05 Novartis Ag Compounds and compositions for the treatment of parasitic diseases
US10085989B2 (en) 2013-12-19 2018-10-02 Novartis Ag Compounds and compositions for the treatment of parasitic diseases
US11123348B2 (en) 2013-12-19 2021-09-21 Novartis Ag Compounds and compositions for the treatment of parasitic diseases
US9700559B2 (en) 2013-12-19 2017-07-11 Novartis Ag Compounds and compositions for the treatment of parasitic diseases
US9783547B2 (en) 2014-01-15 2017-10-10 Centre National De La Recherche Scientifique (Cnrs) Water soluble 4-azapodophyllotoxin analogs
US10259811B2 (en) 2014-06-03 2019-04-16 Gilead Sciences, Inc. Tank-binding kinase inhibitor compounds
US10072001B2 (en) 2014-06-03 2018-09-11 Gilead Sciences, Inc. Tank-binding kinase inhibitor compounds
US10253019B2 (en) 2014-09-26 2019-04-09 Gilead Sciences, Inc. Tank-binding kinase inhibitor compounds
US10040781B2 (en) 2014-09-26 2018-08-07 Gilead Sciences, Inc. Tank-binding kinase inhibitor compounds
US10253062B2 (en) 2014-12-23 2019-04-09 Margaret Anne Brimble Amino acid and peptide conjugates and uses thereof
US11014960B2 (en) 2014-12-23 2021-05-25 Auckland Uniservices Limited Amino acid and peptide conjugates and uses thereof
US10316049B2 (en) 2015-12-17 2019-06-11 Gilead Sciences, Inc. Tank-binding kinase inhibitor compounds
US10894784B2 (en) 2015-12-18 2021-01-19 Bayer Pharma Aktiengesellschaft Heteroarylbenzimidazole compounds
WO2017102091A1 (en) 2015-12-18 2017-06-22 Bayer Pharma Aktiengesellschaft Heteroarylbenzimidazole compounds
US11464853B2 (en) 2016-02-26 2022-10-11 Auckland Uniservices Limited Amino acid and peptide conjugates and conjugation process
WO2017207534A1 (en) 2016-06-03 2017-12-07 Bayer Pharma Aktiengesellschaft Substituted heteroarylbenzimidazole compounds
WO2018019341A1 (en) 2016-07-26 2018-02-01 Karl Rosa Transfection method comprising nonviral gene delivery systems
DE102016113714A1 (en) 2016-07-26 2018-02-01 Rosa Karl Transfection procedure with non-viral gene delivery systems
US11110108B2 (en) 2016-09-27 2021-09-07 Vertex Pharmaceuticals Incorporated Method for treating cancer using a combination of DNA-damaging agents and DNA-PK inhibitors
US11058686B2 (en) 2017-02-23 2021-07-13 Domainex Limited 5-(pyrimidin-4-yl)-2-(pyrrolidin-1-yl)nicotinonitrile compounds as IKKE, TBK1 and/or SIK2 kinases inhibitors
CN110862380A (en) * 2019-10-24 2020-03-06 嘉兴特科罗生物科技有限公司 Small molecule compound

Also Published As

Publication number Publication date
GB201114051D0 (en) 2011-09-28
WO2013024282A3 (en) 2013-05-16

Similar Documents

Publication Publication Date Title
WO2013024282A2 (en) Compounds and their uses
Bisserier et al. Mechanisms of resistance to EZH2 inhibitors in diffuse large B-cell lymphomas
Zhang et al. Discovery of a small molecule targeting ULK1-modulated cell death of triple negative breast cancer in vitro and in vivo
Huang et al. αB‐crystallin complexes with 14‐3‐3ζ to induce epithelial‐mesenchymal transition and resistance to sorafenib in hepatocellular carcinoma
JP2020169212A (en) Phosphatidylinositol-3-kinase pathway biomarkers
KR102106962B1 (en) Novel fgfr3 fusion product
WO2020093006A1 (en) Methods of treating cancer in biomarker-identified patients with non-covalent inhibitors of cyclin-dependent kinase 7 (cdk7)
ES2844200T3 (en) Method to predict the therapeutic efficacy of PI3K / AKT / mTOR inhibitor based on the expression of PHLDA1 or PIK3C2B
EP3365335A1 (en) 2-aryl- and 2-heteroaryl-substituted 2-pyridazin-3(2h)-one compounds as inhibitors of fgfr tyrosine kinases
Ren et al. Design, synthesis, and characterization of an orally active dual-specific ULK1/2 autophagy inhibitor that synergizes with the PARP inhibitor olaparib for the treatment of triple-negative breast cancer
US11696924B2 (en) Combination therapy using PDK1 and PI3K inhibitors
EP3350346B1 (en) Novel biomarkers and methods of treating cancer
Huang et al. SCCRO3 (DCUN1D3) antagonizes the neddylation and oncogenic activity of SCCRO (DCUN1D1)
Hou et al. The NuRD complex-mediated p21 suppression facilitates chemoresistance in BRCA-proficient breast cancer
Wang et al. MicroRNA-195 reverses the resistance to temozolomide through targeting cyclin E1 in glioma cells
Dienstmann et al. Recent developments in anti-cancer agents targeting PI3K, Akt and mTORC1/2
CN114901284A (en) Methods of treating cancer in patients identified by biomarkers with cyclin dependent kinase 7(CDK7) inhibitors
EP2760457A1 (en) Methods and pharmaceutical compositions for treating cancer
Chen et al. RBM4 dictates ESCC cell fate switch from cellular senescence to glutamine-addiction survival through inhibiting LKB1-AMPK-axis
CA2697887A1 (en) Modulating and/or detecting activation induced deaminase and methods of use thereof
US20230158034A1 (en) Co-treatment with cdk4/6 and cdk2 inhibitors to suppress tumor adaptation to cdk2 inhibitors
US20240034742A1 (en) Dosing regimens for cyclin-dependent kinase 7 (cdk7) inhibitors
Sak Aurka inhibition-based combination therapy approaches in glioblastoma.
Zhou et al. KIT mutations and expression: current knowledge and new insights for overcoming IM resistance in GIST
Tyler Exploring Mechanisms of Drug Resistance in ROS1-Gene Fusion Positive Non-Small Cell Lung Cancer Resistant to ROS1 Tyrosine Kinase Inhibitors

Legal Events

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

Ref document number: 12753800

Country of ref document: EP

Kind code of ref document: A2

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 12753800

Country of ref document: EP

Kind code of ref document: A2