WO2012146933A1 - Cyprodinil for use in medicine - Google Patents

Cyprodinil for use in medicine Download PDF

Info

Publication number
WO2012146933A1
WO2012146933A1 PCT/GB2012/050934 GB2012050934W WO2012146933A1 WO 2012146933 A1 WO2012146933 A1 WO 2012146933A1 GB 2012050934 W GB2012050934 W GB 2012050934W WO 2012146933 A1 WO2012146933 A1 WO 2012146933A1
Authority
WO
WIPO (PCT)
Prior art keywords
cancer
compound
cyprodinil
salt
compounds
Prior art date
Application number
PCT/GB2012/050934
Other languages
French (fr)
Inventor
Charles Roland Wolf
Anna Louise Westwood
Roger Neil Sleigh
Original Assignee
Cxr Biosciences 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 Cxr Biosciences Limited filed Critical Cxr Biosciences Limited
Publication of WO2012146933A1 publication Critical patent/WO2012146933A1/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
    • 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
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis

Definitions

  • the present invention relates to the medical use of the anilinopyrimidine compound Cyprodinil and particularly its use in the treatment of cancer.
  • Cancer (or neoplasm) covers a range of diseases in which a group of cells display uncontrolled growth (division beyond the normal limits), invasion (intrusion on and destruction of adjacent tissues), and sometimes metastasis (spread to other locations in the body via lymph or blood). Cancer affects people at all ages with the risk for most types increasing with age and caused about 13% of all human deaths in 2007. Deaths from cancer worldwide are projected to continue rising, with an estimated 12 million deaths in 2030. (WHO, February 2009)
  • cancer treatment is based around surgery, chemotherapy and radiotherapy.
  • the effectiveness of surgery is often limited by the propensity of cancers to invade adjacent tissue or to spread to distant sites by microscopic metastasis.
  • the effectiveness of chemotherapy is often limited by toxicity to other tissues in the body. Radiation can also cause damage to normal tissue.
  • targeted therapy has had a significant impact in the treatment of some types of cancer. This constitutes the use of agents specific for the deregulated proteins of cancer cells.
  • Small molecule targeted therapy drugs are generally inhibitors of enzymatic domains on mutated, overexpressed, or otherwise critical proteins within the cancer cell.
  • agents are selected as having a promising activity against a particular biological target thought to be important in disease; however, little will be known about the safety, toxicity, pharmacokinetics and metabolism of this agent in humans. Therefore, it is traditionally necessary to assess all of these parameters prior to human clinical trials in order to be able to recommend a dose and schedule to be used the first time.
  • much drug development work is required to establish the physicochemical properties of a new agent, such as its chemical makeup, stability, solubility. The process by which the chemical is made will be optimized and it will be further examined for its suitability to be made into capsules, tablets, aerosol, intramuscular injectable, subcuteneous injectable, or intravenous formulations.
  • Anilinopyrimidine fungicides are active against a broad spectrum of fungi (Kunz et a/. (1998), J. Phytopathology, 146, 231-238 and Knauf-Beiter et al. 1995, Plant Disease, 79(11), 1098-1103).
  • fungicides of this class are marketed, cyprodinil (Syngenta), mepanipyrim (Kumiai chemicals Industry Co) and pyrimethanil (AgrEvo GmbH).
  • Fungicidal mode of action is achieved via inhibition of methionine biosynthesis and secretion of hydrolytic enzymes.
  • Field monitoring of Cyprodinil efficacy has indicated that practical resistance to the compound is rare in the key target pathogens (botrytis and venturia). Resistance can be generated under controlled conditions but resistance to anilinopyrimidine fungicides does not correlate with resistance to other classes of fungicide (Myresiotis et al. 2007, Plant Disease, 91(4):407-413).
  • Cyprodinil was first introduced in France in 1993 by Ciba for use on cereals and is now used widely in Europe as a foliar fungicide on fruit, cereals, vegetables and ornamentals.
  • Cyprodinil may be useful in medicine, particularly in the treatment of cancer.
  • Cyprodinil or a salt or ester thereof for use in medicine.
  • Cyprodinil (CAS registration number 121552-61-2) is represented by the following structure:
  • Cyprodinil (4-cycloproplyl-6-methyl-N-phenyl-pyrimidinamine) is also marketed by Syngenta under the trade names UnixTM, ChorusTM, StereoTM, SwitchTM and VanguardTM. Cyprodinil has the CAS registration number 121552-61-2 and the development code was CGA219417. Synonyms for Cyprodinil
  • Cyprodinil is an inhibitor of PI kinase family members (PI 1, PIM2 and PIM3). PIM Kinase activity
  • PIM kinases are cytoplasmic serine/threonine kinases that are known to be involved in regulation of apoptosis and cellular metabolism. Certain PIM kinases have been shown to be upregulated in cancers and as such their inhibition represents a mechanism of action by which compounds such as cyprodinil can have an anti-tumour effect in conditions such as leukaemia, lymphoma, prostate cancer, colon cancer and pancreatic cancer. The studies shown below in Table 1 have shown this link:
  • Prostrate cancer Chen (2005), Mumenthaler (2009), He (2007), Xu (2005), Dai (2005) and Ron (2008) have shown PIM-1 overexpression in prostatic carcinoma.
  • Adipocyte tumours Nga (2010) has shown benign and malignant adipocytic tumours to have strong PIM-1 expression.
  • salts include acid addition salts and base addition salts.
  • Such salts may be formed by conventional means, for example by reaction of a free acid or a free base form of a compound of formula I with one or more equivalents of an appropriate acid or base, optionally in a solvent, or in a medium in which the salt is insoluble, followed by removal of said solvent, or said medium, using standard techniques (e.g. in vacuo, by freeze-drying or by filtration). Salts may also be prepared by exchanging a counter-ion of a compound of the invention in the form of a salt with another counter-ion, for example using a suitable ion exchange resin.
  • ester is included those formed with an alcohol of formula R 1 OH, wherein R represents aryl or alkyl; and those formed with a thiol of formula R SH, wherein R 1 is as hereinbefore defined (i.e. a thioester). It is preferred that the ester is not a thioester.
  • R 1 represents C 1-6 alkyl, for example alkyl (eg methyl).
  • Compounds of the invention may contain double bonds and may thus exist as E (entadel) and Z (zusammen) geometric isomers about each individual double bond. All such isomers and mixtures thereof are included within the scope of the invention.
  • Compounds of the invention may also exhibit tautomerism. All tautomeric forms and mixtures thereof are included within the scope of the invention. Compounds of the invention may also contain one or more asymmetric carbon atoms and may therefore exhibit optical and/or diastereoisomerism. Diastereoisomers may be separated using conventional techniques, e.g. chromatography or fractional crystallisation. The various stereoisomers may be isolated by separation of a racemic or other mixture of the compounds using conventional, e.g. fractional crystallisation or HPLC, techniques. Alternatively the desired optical isomers may be made by reaction of the appropriate optically active starting materials under conditions which will not cause racemisation or epimerisation (i.e.
  • a 'chiral pool' method by reaction of the appropriate starting material with a 'chiral auxiliary' which can subsequently be removed at a suitable stage, by derivatisation (i.e. a resolution, including a dynamic resolution), for example with a homochiral acid followed by separation of the diastereomeric derivatives by conventional means such as chromatography, or by reaction with an appropriate chiral reagent or chiral catalyst all under conditions known to the skilled person. All stereoisomers and mixtures thereof are included within the scope of the invention.
  • a pharmaceutical composition comprising cyprodinil or a salt or ester thereof and a pharmaceutically acceptable excipient, diluent or carrier.
  • a composition comprising between 10mg and 2000mg of an active ingredient per dosage unit, wherein the active ingredient is a compound of the invention or a derivative, salt or variant thereof.
  • dosage unit we mean the unit of medicament administered to a patient at one time.
  • the dosage unit, or single dose may be administered by a single capsule/tablet, single injection, or single intravenous infusion, a single subcutaneous injection, or by a single procedure using other routes of administration, as discussed below.
  • the single dose may be administered to the patient by two or more capsules/tablets or injections given simultaneously or sequentially to deliver the entire dose to the patient in the continuous, single and defined treatment period; by two or more intravenous infusions given simultaneously or sequentially to deliver the entire dose to the patient in the continuous, single and defined treatment; or by multiple procedures using other routes of administration as discussed below.
  • the single dose to be administered to the patient can be delivered by a combination of routes to deliver the entire dose to the patient in the continuous, single and defined treatment.
  • the dosage unit may then be repeated at intervals of time such as a few hours, days, weeks, or months later.
  • Dosage units can be administered to patients in such a way that the patient receives a loading dose followed by one or more maintenance doses.
  • the loading dose may be a high dose in order to quickly reach a desired plasma concentration and then subsequent maintenance doses are a lower dose than the loading dose in order to maintain the required plasma concentration.
  • active ingredient we mean the molecule having the desired effect.
  • variants and derivatives we mean any molecules of substantially identical chemical structure but including minor modifications that do not alter activity but may offer improved or alternative properties for formulation, such as formation into a salt.
  • the compound of the invention containing composition, and medicaments of the invention can be administered alone but will generally be administered in admixture with a suitable pharmaceutical excipient, diluent or carrier selected with regard to the intended route of administration and standard pharmaceutical practice.
  • the compound of the invention containing composition, and medicaments of the invention can be administered orally, buccally or sublingually in the form of tablets, capsules, ovules, elixirs, solutions or suspensions, which may contain flavouring or colouring agents, for immediate-, delayed- or controlled-release applications.
  • the compound of the invention containing composition, and medicaments of the invention may also be administered via intracavernosal injection.
  • Such tablets may contain excipients such as microcrystalline cellulose, lactose, sodium citrate, calcium carbonate, dibasic calcium phosphate and glycine, disintegrants such as starch (preferably corn, potato or tapioca starch), sodium starch glycollate, croscarmellose sodium and certain complex silicates, and granulation binders such as polyvinylpyrrolidone, hydroxypropylmethylcellulose (HPMC), hydroxy-propylcellulose (HPC), sucrose, gelatin and acacia. Additionally, lubricating agents such as magnesium stearate, stearic acid, glyceryl behenate and talc may be included.
  • compositions of a similar type may also be employed as fillers in gelatin capsules.
  • Preferred excipients in this regard include lactose, starch, cellulose, milk sugar or high molecular weight polyethylene glycols.
  • the compound of the invention containing composition, medicaments and pharmaceutical compositions of the invention may be combined with various sweetening or flavouring agents, colouring matter or dyes, with emulsifying and/or suspending agents and with diluents such as water, ethanol, propylene glycol and glycerin, and combinations thereof.
  • composition, and medicaments of the invention can also be administered parenterally, for example, intravenously, intra- arterially, intraperitoneally, intra-thecally, intraventricularly, intrasternally, intracranially, intra-muscularly or subcutaneously, or they may be administered by infusion techniques. They are best used in the form of a sterile aqueous solution which may contain other substances, for example, enough salts or glucose to make the solution isotonic with blood.
  • the aqueous solutions should be suitably buffered (preferably to a pH of from 3 to 9), if necessary.
  • suitable parenteral formulations under sterile conditions is readily accomplished by standard pharmaceutical techniques well-known to those skilled in the art.
  • Medicaments and pharmaceutical compositions suitable for parenteral administration include aqueous and non-aqueous sterile injection solutions which may contain antioxidants, 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 medicaments and pharmaceutical compositions 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 earner, for example water for injections, immediately prior to use.
  • Extemporaneous injection solutions and suspensions may be prepared from sterile powders, granules and tablets of the kind previously described.
  • the compound of the invention containing composition, and medicaments of the invention can also be administered intranasally or by inhalation and are conveniently delivered in the form of a dry powder inhaler or an aerosol spray presentation from a pressurised container, pump, spray or nebuliser with the use of a suitable propellant, e.g.
  • the dosage unit may be determined by providing a valve to deliver a metered amount.
  • the pressurised container, pump, spray or nebuliser may contain a solution or suspension of the active agent, e.g.
  • Capsules and cartridges (made, for example, from gelatin) for use in an inhaler or insufflator may be formulated to contain a powder mix of a compound of the invention containing composition, of the invention and a suitable powder base such as lactose or starch.
  • Aerosol or dry powder formulations are preferably arranged so that each metered dose or "puff contains an effective amount of an agent or polynucleotide of the invention for delivery to the patient. It will be appreciated that the overall daily dose with an aerosol will vary from patient to patient, and may be administered in a single dose or, more usually, in divided doses throughout the day.
  • the compound of the invention containing composition, and medicaments of the invention can be administered in the form of a suppository or pessary, or they may be applied topically in the form of a lotion, solution, cream, gel, ointment or dusting powder.
  • the compound of the invention containing composition, and medicaments of the invention may also be transdermal ⁇ administered, for example, by the use of a skin patch. They may also be administered by the ocular route, particularly for treating diseases of the eye.
  • the compound of the invention containing composition, and medicaments of the invention can be formulated as micronised suspensions in isotonic, pH adjusted, sterile saline, or, preferably, as solutions in isotonic, pH adjusted, sterile saline, optionally in combination with a preservative such as a benzylalkonium chloride.
  • a preservative such as a benzylalkonium chloride.
  • they may be formulated in an ointment such as petrolatum.
  • the compound of the invention containing composition, and medicaments of the invention can be formulated as a suitable ointment containing the active agent suspended or dissolved in, for example, a mixture with one or more of the following: mineral oil, liquid petrolatum, white petrolatum, propylene glycol, polyoxyethylene polyoxypropylene agent, emulsifying wax and water.
  • ком ⁇ онентs can be formulated as a suitable lotion or cream, suspended or dissolved in, for example, a mixture of one or more of the following: mineral oil, sorbitan monostearate, a polyethylene glycol, liquid paraffin, polysorbate 60, cetyl esters wax, cetearyl alcohol, 2- octyldodecanol, benzyl alcohol and water.
  • Formulations suitable for topical administration in the mouth include lozenges comprising the active ingredient in a flavoured basis, usually sucrose and acacia or tragacanth; pastilles comprising the active ingredient in an inert basis such as gelatin and glycerin, or sucrose and acacia; and mouth-washes comprising the active ingredient in a suitable liquid carrier.
  • oral or parenteral administration of the compound of the invention containing composition, medicaments and pharmaceutical compositions of the invention is the preferred route, being the most convenient.
  • the compound of the invention containing composition, and medicaments of the invention are administered as a suitably acceptable formulation in accordance with normal veterinary practice and the veterinary surgeon will determine the dosing regimen and route of administration which will be most appropriate for a particular animal.
  • the compound of the invention containing composition, as defined herein may be formulated as described in the accompanying Examples.
  • the composition may comprise any effective amount of active ingredient, this may be between 10mg and 2000mg of active ingredient per dosage unit, and preferably is between 50mg and 1000mg.
  • the dosage unit contains an amount of active ingredient per dosage unit selected from 10mg, 20mg, 25mg, 50mg, 100mg, 200mg, 300mg, 400mg, 450mg, 600mg, 750mg, 950mg, 1000mg and 1200mg.
  • the composition may comprise between 10-50mg, 10-75mg, 10-100mg, 10- 200mg, 10-300mg, 10-400mg, 10-600mg, 10-750mg, 10-950mg, 10-1000mg, 10- 1200mg, 50-75mg, 50-1 OOmg, 50-200mg, 50-300mg, 50-450mg, 50-600mg, 50-750mg, 50-950mg, 50-1000mg, 50-1200mg, 75-1 OOmg, 75-200mg, 75-300mg, 75-450mg, 75- 600mg, 75-750mg, 75-950mg, 75-1000mg, 75-1200mg, 100-200mg, 100-300mg, 100- 450mg, 100-600mg, 100-750mg, 100-950mg, 100-1000mg, 100-1200mg, 200-300mg, 200-450mg, 200-600mg, 200-750mg, 200-
  • composition is pharmaceutically acceptable, and may optionally contain a pharmaceutically acceptable excipient, diluent, carrier or filler.
  • a further aspect of the invention provides a method of treating cancer (such as cancers of the breast, colon, prostate, ovaries, brain and lung and their metastases) comprising administering to the patient an effective amount of cyprodinil or a salt or an ester thereof.
  • cancer such as cancers of the breast, colon, prostate, ovaries, brain and lung and their metastases
  • patient includes all animals including humans. Examples of patients include humans, cows, dogs, cats, goats, sheep, and pigs.
  • patienf means an animal having a disorder in need of treatment.
  • the patient is selected from humans, cows, dogs, cats, goats, sheep, and pigs. More preferably, the patient is a human.
  • the compounds of the invention may be used in cancer treatment either alone or in combination with well known anti-cancer agents.
  • Cancer treatments promote tumour regression by inhibiting tumour cell proliferation, inhibiting angiogenesis (growth of new blood vessels that is necessary to support tumour growth) and/or prohibiting metastasis by reducing tumour cell motility or invasiveness.
  • compositions of the invention may be effective in adult and pediatric oncology including in solid phase tumours/malignancies, locally advanced tumours, human soft tissue sarcomas, metastatic cancer, including lymphatic metastases, blood cell malignancies including multiple myeloma, acute and chronic leukemias, and lymphomas, head and neck cancers including mouth cancer, larynx cancer and thyroid cancer, lung cancers including small cell carcinoma and non-small cell cancers, breast cancers including small cell carcinoma and ductal carcinoma, gastrointestinal cancers including esophageal cancer, stomach cancer, colon cancer, colorectal cancer and polyps associated with colorectal neoplasia, pancreatic cancers, liver cancer, urologic cancers including bladder cancer and prostate cancer, malignancies of the female genital tract including ovarian carcinoma, uterine (including endometrial) cancers, and solid tumour in the ovarian follicle, kidney cancers including renal cell carcinoma, brain cancers including intrinsic brain tumours, neuroblastoma, as
  • Compounds of the invention may be administered to treat cancer.
  • the cancer to be treated is preferably selected from pancreatic cancer, ovarian cancer, breast cancer, prostate cancer, liver cancer, chondrosarcoma, lung cancer, head and neck cancer, colon cancer, sarcoma, leukaemia, myeloma, lymphoma, kidney cancer, thyroid cancer and brain cancers such as glioblastoma.
  • compositions can be administered in therapeutically effective dosages alone or in combination with adjuvant cancer therapy such as surgery, chemotherapy, radiotherapy, thermotherapy, and laser therapy, and may provide a beneficial effect, e.g. reducing tumour size, slowing rate of tumour growth, inhibiting metastasis, or otherwise improving overall clinical condition, without necessarily eradicating the cancer.
  • adjuvant cancer therapy such as surgery, chemotherapy, radiotherapy, thermotherapy, and laser therapy
  • composition can also be administered in therapeutically effective amounts as a portion of an anti-cancer cocktail.
  • An anti-cancer cocktail is a mixture of the compound or modulator of the invention with one or more anti-cancer drugs in addition to a pharmaceutically acceptable carrier for delivery. The use of anti-cancer cocktails as a cancer treatment is routine.
  • Anti-cancer drugs that are well known in the art and can be used as a treatment in combination with the compounds of the invention include: 17AAG, 17DMAG, 5FU, 7-hydroxystaurosporine (UCN-01), ABT888, Actinomycin D, Alsterpaullone, Axitinib, Aminoglutethimide, Amsacrine, Asparaginase, Azacitidine, AZD7762, Bay 11-7082, Belinostat, Bendamustine, Bexarotene, BIBW 2992, Bisindolylmaleimide I, Bleomycin, Bortezomib, Bosutinib, Busulfan, Canertinib , Capecitabine, Carboplatin, Carmustine, CDK4/6 IV, Chelerythrine Chloride, Chlorambucil, CHR-2863 (CHROMA), CHR-3531 (CHROMA), Chromomycin A3, CI-994, Cisplatin
  • compositions of the invention may be used for prophylactic treatment of cancer.
  • hereditary conditions and/or environmental situations e.g. exposure to carcinogens
  • In vitro models can be used to determine the effective doses of the compounds of the invention as a potential cancer treatment.
  • tumour cells in Boyden Chamber assays as described in Pilkington ef ai, Anticancer Res., 17: 4107-9 (1997), and angiogenesis assays such as induction of vascularization of the chick chorioallantoic membrane or induction of vascular endothelial cell migration as described in Ribatta et ai, Intl. J. Dev. Biol., 40: 1189-97 (1999) and Li et ai, Clin. Exp. Metastasis, 17:423-9 (1999), respectively.
  • Suitable tumour cells lines are available, e.g. from American Type Tissue Culture Collection catalogues.
  • the method, use or composition of the invention additionally comprises a further chemotherapeutic agent.
  • the further chemotherapeutic agent is selected from 17AAG, 17DMAG, 5FU, 7-hydroxystaurosporine (UCN-01), ABT888, Actinomycin D, Alsterpaullone, Axitinib, Aminoglutethimide, Amsacrine, Asparaginase, Azacitidine, AZD7762, Bay 11-7082, Belinostat, Bendamustine, Bexarotene, BIBW 2992, Bisindolylmaleimide I, Bleomycin, Bortezomib, Bosutinib, Busulfan, Canertinib , Capecitabine, Carboplatin, Carmustine, CDK4/6 IV, Chelerythrine Chloride, Chlorambucil, CHR-2863 (CHROMA), CHR-3531 (CHROMA), Chromomycin A3, CI-994, Cisplatin, Cladribine, Clofarabine, Clofibrate
  • the further chemotherapeutic agent is a HSP inhibitor, such as geldanamycin, 17-AAG or 17-DMAG. Due to the favourable toxicity and safety profiles of the compounds of the invention, the compounds may be used to treat patients who may not be amenable to conventional chemotherapeutic agents.
  • chemotherapies are generally immunosuppressive and so are withdrawn in certain situations, such as for patients who are pre-operative, post-operative, receiving radiation, terminally ill, elderly, or receiving adjuvant or neo-adjuvant thereapy.
  • the compounds of the invention may still be used in such situations.
  • the compounds of the invention are used to treat patients who may not be amenable to conventional chemotherapeutic agents, particularly those from whom conventional chemotherapeutic agents have been withdrawn.
  • a further aspect of the invention provides a kit of parts comprising: (i) Cyprodinil or a salt or ester thereof or a pharmaceutical composition comprising cyprodinil or a salt or ester thereof and a pharmaceutically acceptable excipient, diluent or carrier; (ii) apparatus for administering the compound or pharmaceutical composition; and (iii) instructions for use.
  • the kit of parts additionally comprises a further chemotherapeutic agent.
  • the further chemotherapeutic agent is selected from 17AAG, 17DMAG, 5FU, 7-hydroxystaurosporine (UCN-01), ABT888, Actinomycin D, Alsterpaullone, Axitinib, Aminoglutethimide, Amsacrine, Asparaginase, Azacitidine, AZD7762, Bay 11-7082, Belinostat, Bendamustine, Bexarotene, BIBW 2992, Bisindolylmaleimide I, Bleomycin, Bortezomib, Bosutinib, Busulfan, Canertinib , Capecitabine, Carboplatin, Carmustine, CDK4/6 IV, Chelerythrine Chloride, Chlorambucil, CHR-2863 (CHROMA), CHR-3531 (CHROMA), Chromomycin A3, CI-994, Ci
  • the further chemotherapeutic agent is a HSP inhibitor, such as geldanamycin, 17-AAG or 17-DMAG.
  • Figure 1 shows percentage survival of (a) OVCAR3 cells and (b) HepG2 cells in response to increasing doses of Cyprodinil.
  • Figure 2 Isobologram for Cyprodinil and Geldanamycin combination
  • Figure 2 shows the shift in IC 50 of Cyprodinil on PC3 cells when Cyprodinil is dosed in combination with the HSP90 inhibitor geldanamycin.
  • Figure 3 Core aniline-pyrimidine moiety of Cyprodinil shared with kinase inhibitors such as GleevecTM
  • Figure 3 shows the how the core aniline-pyrimidine moiety of Cyprodinil is shared with kinase inhibitors such as GleevecTM
  • Figure 4 shows % inhibition of the PIM Kinases with increasing dose of Cyprodinil.
  • Figure 5 Cyprodinil activity in further kinases
  • Figure 5 shows % inhibition of further kinases with increasing dose of Cyprodinil.
  • Figure 6 PC3 tumour volumes following treatment with Cyprodinil
  • Figure 6 shows PC3 tumour volumes over time when dosed with Cyprodinil versus control.
  • Figure 7 shows HT29 tumour volumes over time when dosed with Cyprodinil at various levels versus control.
  • Figure 8 shows PC3 tumour volumes over time when dosed with Cyprodinil (CXR6024) at various levels versus control.
  • Figure 9 shows ATP levels as a percentage of control with increasing dose of Cyprodinil and Pyrimethanil at 0, 45, 90 minutes and 4 hour time points.
  • Figure 10 shows NADH Dehydrogenase activity represented by percentage of control with increasing dose of Cyprodinil and Pyrimethanil at 0, 30, 60, 90, 180 and 270 minute time points.
  • Figure 11 shows the RNA microarray experimental structure using triplicates of each sample.
  • Example 1 selection of the compounds of the invention based on specific properties The following method details how the compounds of the invention were selected to arrive at those to be used for the treatment of diseases such as cancer.
  • Step 1 A database was produced of some 182 compounds registered for use as agro- or industrial chemicals with available dossiers regarding bioactivity and toxicity collated from sources including the UK's Advisory Committee on Pesticides (ACP), the US Environmental Protection Agency (EPA), the European Food Safety Authority (EFSA), the US Department of Health & Human Services' Agency for Toxic Substances and Disease Registry (ATSDR), Canada's Bureau of Chemical Safety and the International Programme on Chemical Safety (IPCS).
  • ACP Advisory Committee on Pesticides
  • EPA US Environmental Protection Agency
  • EFSA European Food Safety Authority
  • ATSDR US Department of Health & Human Services' Agency for Toxic Substances and Disease Registry
  • IPCS International Programme on Chemical Safety
  • Table 13 details the 182 compounds contained in the database.
  • Step 2 A panel of 144 chemically diverse compounds were selected as being readily available from the database produced in (1).
  • Table 14 details the 144 chemically diverse compounds.
  • Step 3 The compounds from (2) were tested against 15 human tumour cell lines for cytotoxicity. Effects on cell survival at 72 hours and at 7 days at a single compound concentration (100 ⁇ ) were measured. Tables 11 and 12 show cell proliferation data for the 144 compounds at 72 hours and 7 day exposure, respectively. The data is expressed as a percentage of untreated control cells, so 100 % means that there are the same number of cells as the control, less than 100 % indicates there are fewer cells than the control and greather than 100 % indicates that more cells than the control are present.
  • Test compounds were dissolved in D SO. Cells were seeded at a density of 500 cells per well in 75 ⁇ of DME (10% FBS/2 mM Glutamine) media for 7 day incubation in 96- well plates or 5000 cells per well for 72 hrs incubations. The following day, 25 ⁇ of test compounds, diluted 1 :25 in DMEM (10% FBS), were added to the wells to give a final compound concentrations of 100 ⁇ . Duplicate plates were assembled and then incubated for either 72 hrs or 7 days at 37°C/5% C0 2 . No media/drug replacement during the incubation period was undertaken.
  • Step 4 The cytotoxicity data from (3) was used to rank compounds with respect to (a) global cytotoxicity; (b) selective cytotoxicity; and (c) published literature (i.e. a lack of published literature linking the compounds with cytotoxicity).
  • Step 5 Panel of 48 compounds chosen as the top-ranked compounds using the results of (4).
  • Step 6 Compounds selected under (5) were tested for cytotoxicity with compound dose response (see table 10) and the published toxicology data reviewed for (a) bioavailability (see table 3); (b) rat LD50 (see table 5); (c) Therapeutic Index (NOEL (No Observable Effects Limit): IC50 ratio) (table 2).
  • Step 7 Panel from (6) screened for effects on protein kinase activities. The results are shown in Table 8.
  • Step 8 Compounds selected for further development on basis of meeting at least criteria (1) and (2) (but preferably more) of the 7 criteria listed below:
  • Step 9 (optional) Kinase inhibition (such as PI -Kinase) by dose response results can be used to select specific compounds for further development (see Example 2 and figure
  • Step 10 Compounds selected under (8) and (9) will be tested further in xenograft studies (see Example 4).
  • Toxicoloqv/Toxicitv Data must be available Toxicological testing is a significant barrier in the pre-clinical development of pharmaceuticals, both in terms of cost and risk. Selection of molecules already tested for toxicity is therefore an attractive alternative.
  • Favourable Therapeutic Index - NOELICsn Ratio must be greater than 5
  • An ideal pharmaceutical would exhibit efficacy in the absence of any toxicological or pharmacological side-effects at the therapeutic dose.
  • a compound with these desirable properties is described as having a favourable therapeutic index or therapeutic window.
  • Table 2 shows which of a selection of 48 compounds (as selected according to step 5 of the screening method) have an IC50/NOEL ratio of greater than five.
  • Myclobutanil 15 51.9 25.00 2.1 cyfluthrin 10 23.1 17.00 1.4
  • Oral bioavailability preferred to be greater than 20% Oral bioavailability is an attractive property for pharmaceutical compounds. Oral dosing will mean patients do not have to travel to the hospital situation regularly during their treatment. This will not only save considerable resources at the treatment centre but also reduce patient comfort by reducing travel and in-patient time. Oral bioavailability is calculated by measuring the plasma concentration of the compound over time after administering the drug both orally (po) and intravenously (IV). The area under the curve (AUC) is measured for both modes of administration and is corrected for different dose levels for the different routes. The following formula is used for calculating oral bioavailability (F):
  • Table 3 shows which of a selection of compounds has an oral bioavailability of >20%:
  • the LDgn be greater than 500 mg/kq
  • the selection approach described is to select compounds which have undergone extensive toxicological testing and have been deemed of low toxicity and safe for release to the environment as either agrochemicals or for use in other industries.
  • LD 50 Lethal Dose 50
  • MTD Maximum Tolerated Dose
  • the compounds selected are of no greater than "low toxicity", with oral LD50 values in the rat no lower than 500 mg/kg.
  • Table 4 details the different measures of toxicity of compounds.
  • Table 5 details which of a selection of compounds have an oral LD50 in the rat of over 500mg/kg.
  • Compounds are selected from a library partly on the basis of in vivo half-life measured during development of the compounds as agro-industrial chemicals.
  • In vivo half-life is a measure of the rate at which a compound is excreted or metabolised.
  • the change in plasma concentration of a compound following dosing is described by the equation:
  • tumour types may be best targeted with the molecule.
  • Some of this information can be learned from testing the compound for effects on cellular proliferation on a panel of tumour cell lines.
  • An alternative approach is to test the molecules for activity, inhibitory or activatory, on to use in vitro assays of molecular targets such as protein kinases, GPCRs, enzymes or other cellular machinery.
  • the compounds have a known mode of action.
  • One aspect of this could be inhibitory activity of a compound against a member(s) of a panel of protein kinases known to be implicated in cancer biology.
  • a panel could be comprised of the protein kinases shown in table 7:
  • Table 7 A panel of kinases known to be implicated in cancer biology Aurora A ERK2 JAK2 PIM3 S6K1
  • MAPK2 ERK2 assay MAPK/ERK2 (5-20 mU diluted in 50 mM Tris pH 7.5, 0.1 mM EGTA, 0.1 mM Na 3 V0 4 , 0.1% ⁇ -mercaptoethanol, 1 mg/ml BSA) is assayed against MBP in a final volume of 25.5 ⁇ in 25 mM Tris pH 7.5, 0.1 mM EGTA, 0.33 mg/ml MBP, 10 mM magnesium acetate and 0.05 mM [33 ⁇ - ⁇ - ⁇ ](500 -1000 cpm/pmole) and incubated for 30 min at room temperature. Assays are stopped by addition of 5 ⁇ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid.
  • MKK1 assay This is a two-step assay where inactive MAPK (0.06 mg/ml) is activated by MKK1 (diluted in 25 mM Tris, 0.1 mM EGTA, 0.1% ⁇ -mercaptoethanol, 0.01% Brij35, 1 mg/ml BSA) in 25.5 ⁇ containing 25 mM Tris, 0.1 mM EGTA, 0.01% Brij35, 10 mM magnesium acetate and 0.005 mM ATP.
  • PKCa assay PKCa (5-20 mU diluted in 20 mM Hepes pH 7.4, 0.03% Triton X-100) is assayed against Histone H1 in the presence of PtdSerine and DAG (0.1 mg/ml. and 10 pg/ml) and 0.1 mM CaCI2.
  • the assay is carried out in a final volume of 25.5 ⁇ containing 20 mM Hepes pH 7.4, 0.03% Triton X-100, 0.1 mg/ml Histone H1, 10 mM magnesium acetate and 0.02 mM[33 p -Y-ATP] (50-1000 cpm/pmole) and incubated for 30 min at room temperature.
  • Assays are stopped by addition of 5 ⁇ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid.
  • PDK1 (5-20 mU diluted in 50 mM Tris pH 7.5, 0.05% ⁇ -mercaptoethanol, 1 mg/ml BSA) is assayed against PDKtide (KTFCGTPEYLAPEVRREPRILSEEEQ-EMFRDFDYIADWC) in a final volume of 25.5 ⁇ containing 50 mM Tris pH 7.5, 0.05% ⁇ -mercaptoethanol, 100 ⁇ substrate peptide, 10mM magnesium acetate and 0.02 mM [33 ⁇ - ⁇ - ⁇ ] (50-1000 cpm/pmole) and incubated for 30 min at room temperature Assays are stopped by addition of 5 ⁇ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid.
  • APH-PKBa-S473D (5-20mU diluted in 50 mM Tris pH 7.5, 0.1 mM EGTA, 0.1% ⁇ - mercaptoethanol, 1 mg/ml BSA) is assayed against a modified Crosstide peptide GRPRTSSFAEGKK in a final volume of 25.5 ⁇ containing 50mM Tris pH 7.5, 0.05% ⁇ - mercaptoethanol, 30 ⁇ substrate peptide, 10 mM magnesium acetate and 0.005 mM [33 ⁇ - ⁇ - ⁇ ] (50-1000 cpm/pmole) and incubated for 30 min at room temperature. Assays are stopped by addition of 5 ⁇ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid. SGK assay
  • SGK (5-20mU diluted in 20 mM MOPS pH 7.5, 1mM EDTA, 0.01% Brij35, 5% glycerol, 0.1% ⁇ -mercaptoethanol, 1 mg/ml BSA) is assayed against a modified Crosstide peptide GRPRTSSFAEGKK in a final volume of 25.5 ⁇ containing 8 mM MOPS pH 7.0, 0.2 mM EDTA, 30 ⁇ substrate peptide, 10 mM magnesium acetate and 0.02 mM [33 ⁇ - ⁇ - ⁇ ] (50-1000 cpm/pmole) and incubated for 30 min at room temperature. Assays are stopped by addition of 5 ⁇ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid.
  • S6K1/ P70 S6K assay S6K1/P70 S6K (5-20 mU diluted in 20 mM MOPS pH 7.5, 1 mM EDTA, 0.01% Brij35, 5% glycerol, 0.1% ⁇ -mercaptoethanol, 1 mg/ml BSA) is assayed against substrate peptide (KKRNRTLTV) in a final volume of 25.5 ⁇ containing 8 mM MOPS pH 7.0, 0.2 mM EDTA, 0.1 mM substrate peptide, 10 mM magnesium acetate and 0.02 mM [33 ⁇ - ⁇ - ATP] (50-1000 cpm/pmole) and incubated for 30 min at room temperature. Assays are stopped by addition of 5 ⁇ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid.
  • ROCK-II (ROKa) assay ROCK-II (ROKa) (5-20 mU diluted in 50 mM Tris pH 7.5, 0.1 mM EGTA, 0.1% ⁇ - mercaptoethanol, 1 mg/ml BSA) is assayed against Long S6 substrate peptide (KEAKEKRQEQIAKRRRLSSLRASTSKSGGSQK) in a final volume of 25.5 ⁇ containing 50 mM Tris pH 7.5, 0.1 mM EGTA, 30 ⁇ Long S6 substrate peptide, 10 mM magnesium acetate and 0.02 mM [33 ⁇ - ⁇ - ⁇ ] (50-1000 cpm/pmole) and incubated for 30 min at room temperature. Assays are stopped by addition of 5 ⁇ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid.
  • CHK1 (5-20 mU diluted in 20 mM MOPS pH 7.5, 1 mM EDTA, 0.1% ⁇ -mercaptoethanol, 0.01% Brij-35, 5% glycerol, 1 mg/ml BSA) is assayed against CHKtide substrate peptide (KKKVSRSGLYRSPSMPENLNRPR) in a final volume of 25.5 ⁇ containing 8 mM MOPS pH 7.0, 0.2 mM EDTA, 200 ⁇ CHKtide, 10 mM magnesium acetate and 0.02 mM [33 p - ⁇ - ⁇ ](50-1000 cpm/pmole) and incubated for 30 min at room temperature Assays are stopped by addition of 5 ⁇ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid. LCK assay
  • LCK (5-20 mU diluted in 20 mM MOPS pH 7.5, 1 mM EDTA, 0.01% Brij35, 5% glycerol, 0.1% ⁇ -mercaptoethanol, 1 mg/ml BSA) is assayed against Cdc2 peptide (KVEKIGEGTYGWYK) in a final volume of 25.5 ⁇ containing 50 mM Tris pH 7.5, 0.1 mM EGTA, 0.1 mM Na3Vo4, Cdc2 peptide (0.25 mM), 10 mM magnesium acetate and 0.05mM [33 ⁇ - ⁇ - ⁇ ](500-1000 cpm/pmole) and incubated for 15 min at room temperature Assays are stopped by addition of 5 ⁇ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid.
  • Cdc2 peptide KVEKIGEGTYGWYK
  • CDK2/cyclin A assay CDK2/cyclin A (5-20 mU diluted in 50 mM Hepes pH 7.5, 1 mM DTT, 0.02% Brij35, 100 mM NaCI) is assayed against Histone H1 in a final volume of 25.5 ⁇ containing 50 mM Hepes pH7.5, 1 mM DTT, 0.02% Brij35, 100 mM NaCI, Histone H1 (1 mg/ml), 10 mM magnesium acetate and 0.02 mM [33 ⁇ - ⁇ - ⁇ ](500-1000 cpm/pmole) and incubated for 30 min at room temperature. Assays are stopped by addition of 5 ⁇ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid.
  • MAPKAP-K1 b/RSK2 assay MAPKAP-K1 b (5-20 mU diluted in 20 mM MOPS pH 7.5, 1 mM EDTA, 0.01% Brij35, 5% glycerol, 0.1% ⁇ -mercaptoethanol, 1 mg/ml BSA) is assayed against substrate peptide (KKLNRTLSVA) in a final volume of 25.51 containing 50 mM Na- ⁇ -glycerophosphate (pH 7.5), 0.5 mM EDTA, 30 ⁇ substrate peptide, 10 mM magnesium acetate and 0.05 mM [33 ⁇ - ⁇ - ⁇ ] (50-1000 cpm/pmole) and incubated for 30 min at room temperature. Assays are stopped by addition of 5 ⁇ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid.
  • IKKb 5-20mU of IKKb (diluted in 50mM Tris (pH 7.5), 0.1 mM EGTA, 1mg/ml BSA, 0.1% ⁇ - mercaptoethanol) is assayed against substrate peptide (LDDRHDSGLDSMKDEEY) in a final volume of 25.5 ⁇ containing 50mM Tris (pH 7.5), 0.1 mM EGTA, 0.1%, fjmercaptoethanol, 300 ⁇ substrate peptide, 10 mM magnesium acetate and 0.005 mM [33 ⁇ - ⁇ - ⁇ ]( 500-1000 cpm/pmole) and incubated for 30 mins at room temperature. Assays are stopped by addition of 5 ⁇ of 0.5M (3%) orthophosphoric acid. Assays are harvested onto P81 Unifilter plates using a wash buffer of 50mM orthophosphoric acid.
  • Aurora B (5-20mU diluted in 50 mM Tris pH 7.5, 0.1 mM EGTA, 0.1% ⁇ - mercaptoethanol, 1 mg/ml BSA) is assayed against a substrate peptide (LRRLSLGLRRLSLGLRRLSLGLRRLSLG) in a final volume of 25.5 ⁇ containing 50mM Tris pH 7.5, 0.05% B-mercaptoethanol, 300 ⁇ substrate peptide, 10 mM magnesium acetate and 0.02 mM [33 ⁇ - ⁇ ] (50-1000 cpm/pmole) and incubated for 30 min at room temperature. Assays are stopped by addition of 5 ⁇ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid.
  • LRRLSLGLRRLSLGLRRLSLGLRRLSLG substrate peptide
  • CHK2 (5-20 mU diluted in 20 mM MOPS pH 7.5, 1 mM EDTA, 0.1% ⁇ -mercaptoethanol, 0.01% Brij-35, 5% glycerol, 1 mg/ml BSA) is assayed against CHKtide substrate peptide (KKKVSRSGLYRSPSMPENLNRPR) in a final volume of 25.5 ⁇ containing 8 mM MOPS pH 7.0, 0.2 mM EDTA, 200 ⁇ CHKtide, 10 mM magnesium acetate and 0.02 mM [33 p - ⁇ - ⁇ ](50-1000 cpm/pmole) and incubated for 30 min at room temperature Assays are stopped by addition of 5 ⁇ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid.
  • CHKtide substrate peptide KKKVSRSGLYRSPSMPENLNRPR
  • Src assay Src (5-20mU diluted in 50 mM Tris pH 7.5, 0.1 mM EGTA, 0.1% ⁇ -mercaptoethanol, 1 mg/ml BSA) is assayed against a substrate peptide (KVEKIGEGTYGWYK) in a final volume of 25.5 ⁇ containing 50mM Tris pH 7.5, 0.05% ⁇ -mercaptoethanol, 300 ⁇ substrate peptide, 10 mM magnesium acetate and 0.05 mM [33 ⁇ - ⁇ - ⁇ ] (50-1000 cpm/pmole) and incubated for 30 min at room temperature. Assays are stopped by addition of 5 ⁇ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid. PLK1 assay
  • PLK1 (5-20mU diluted in 50 mM Tris pH 7.5, 0.1 mM EGTA, 0.1% ⁇ -mercaptoethanol, 1 mg ml BSA, 100 ⁇ Vanadate) is assayed against a substrate peptide (ISDELMDATFADQEAKKK) in a final volume of 25.5 ⁇ containing 50mM Tris pH 7.5, 0.05% ⁇ -mercaptoethanol, 10 ⁇ Vanadate, 300 ⁇ substrate peptide, 10 mM magnesium acetate and 0.005 mM [33 ⁇ - ⁇ - ⁇ ] (50-1000 cpm/pmole) and incubated for 30 min at room temperature. Assays are stopped by addition of 5 ⁇ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid
  • PIM1 (5-20mU diluted in 50 mM Tris pH 7.5, 0.1 mM EGTA, 0.1% ⁇ -mercaptoethanol, 1 mg/ml BSA) is assayed against a substrate peptide (RSRHSSYPAGT) in a final volume of 25.5 ⁇ containing 50mM Tris pH 7.5, 0.05% ⁇ -mercaptoethanol, 300 ⁇ substrate peptide, 10 mM magnesium acetate and 0.02 mM [33 ⁇ - ⁇ - ⁇ ] (50-1000 cpm/pmole) and incubated for 30 min at room temperature. Assays are stopped by addition of 5 ⁇ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid.
  • RSRHSSYPAGT substrate peptide
  • PIM3 (5-20mU diluted in 50 mM Tris pH 7.5, 0.1 mM EGTA, 0.1% ⁇ -mercaptoethanol, 1 mg/ml BSA) is assayed against a substrate peptide (RSRHSSYPAGT) in a final volume of 25.5 ⁇ containing 50mM Tris pH 7.5, 0.05% ⁇ -mercaptoethanol, 300 ⁇ substrate peptide, 10 mM magnesium acetate and 0.02 mM [33 ⁇ - ⁇ - ⁇ ] (50-1000 cpm/pmole) and incubated for 30 min at room temperature. Assays are stopped by addition of 5 ⁇ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid.
  • RSRHSSYPAGT substrate peptide
  • FGF-R1 (5-20mU diluted in 50 mM Tris pH 7.5, 0.1 mM EGTA, 1 mg/ml BSA) is assayed against a substrate peptide (Poly Glut Tyr) in a final volume of 25.5 ⁇ containing 50mM Tris pH 7.5, 1 mg/ml substrate peptide, 10 mM magnesium acetate and 0.02 mM [33 p -v- ATP] (50-1000 cpm/pmole) and incubated for 30 min at room temperature. Assays are stopped by addition of 5 ⁇ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid.
  • substrate peptide Poly Glut Tyr
  • EPH-A2 assay EPH-A2 (5-20mU diluted in 50 mM Tris pH 7.5, 0.1 mM EGTA, 1 mg/ml BSA) is assayed against a substrate peptide (Poly Glut Tyr) in a final volume of 25.5 ⁇ containing 50mM Tris pH 7.5, 0.1 mg/ml substrate peptide, 10 mM magnesium acetate and 0.05 mM [33 p - ⁇ - ⁇ ] (50-1000 cpm/pmole) and incubated for 30 min at room temperature. Assays are stopped by addition of 5 ⁇ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid.
  • a substrate peptide Poly Glut Tyr
  • IGF-1R (5-20mU diluted in 50 mM Tris pH 7.5, 0.1 mM EGTA, 1 mg/ml BSA) is assayed against a substrate peptide (KKKSPGEYVNIEFG) in a final volume of 25.5 ⁇ containing 50mM Tris pH 7.5, 300 ⁇ substrate peptide, 10 mM magnesium acetate and 0.005 mM [33 ⁇ - ⁇ - ⁇ ] (50-1000 cpm/pmole) and incubated for 30 min at room temperature. Assays are stopped by addition of 5 ⁇ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid.
  • VEG-FR (5-20mU diluted in 50 mM Tris pH 7.5, 0.1 mM EGTA, 1 mg/ml BSA) is assayed against a substrate peptide (KKKSPGEYVNIEFG) in a final volume of 25.5 ⁇ containing 50mM Tris pH 7.5, 300 ⁇ substrate peptide, 10 mM magnesium acetate and 0.02 mM [33 p -y-ATP] (50-1000 cpm/pmole) and incubated for 30 min at room temperature. Assays are stopped by addition of 5 ⁇ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid.
  • TBK1 (DU12569) (5-20mU diluted in 50 mM Tris pH 7.5, 0.1 mM EGTA, 1 mg/ml BSA) is assayed against a substrate peptide (KKKKERLLDDRHDSGLDSMKDEE) in a final volume of 25.5 ⁇ containing 50mM Tris pH 7.5, 300 ⁇ substrate peptide, 10 mM magnesium acetate and 0.05 mM [33 ⁇ - ⁇ - ⁇ ] (50-1000 cpm/pmole) and incubated for 30 min at room temperature. Assays are stopped by addition of 5 ⁇ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid.
  • IKKepsilon (DU14231)(diluted in 50mM Tris (pH 7.5), 0.1mM EGTA, 1mg/ml BSA) is assayed against MBP in a final volume of 25.5 ⁇ containing 50mM Tris (pH 7.5), 0.1 mM EGTA, 0.33mg/ml MBP, 10 mM magnesium acetate and 0.05 mM [33 ⁇ - ⁇ - ⁇ ]( 500-1000 cpm/pmole) and incubated for 30 mins at room temperature. Assays are stopped by addition of 5 ⁇ of 0.5M (3%) orthophosphoric acid. Assays are harvested onto P81 Unifilter plates using a wash buffer of 50mM orthophosphoric acid
  • HER4 assay HER4 (5-20 mU diluted in 50 mM Tris pH 7.5, 0.1 mM EGTA, 0.1% ⁇ -mercaptoethanol, 1 mg/ml BSA) is assayed against Poly Glut Tyr in a final volume of 25.5 ⁇ containing 50mM Tris pH 7.5, 0.1 mM EGTA, 1 mg/ml Poly Glut Tyr, 10 mM magnesium acetate and O.OOSmM [33 ⁇ - ⁇ - ⁇ ] (50-1000 cpm/pmole) and incubated for 30 min at room temperature. Assays are stopped by addition of 5 ⁇ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid.
  • Aurora A assay Aurora A (5-20 mU diluted in 50 mM Tris pH 7.5, 0.1 mM EGTA, 0.1% ⁇ - mercaptoethanol, 1 mg/ml BSA) is assayed against
  • LRRLSLGLRRLSLGLRRLSLGLRRLSLG in a final volume of 25.5 ⁇ containing 50mM Tris pH 7.5, 0.1 mM EGTA, 0.3mM LRRLSLGLRRLSLGLRRLSLGLRRLSLG, 10 mM magnesium acetate and 0.005mM [33 ⁇ - ⁇ - ⁇ ] (50-1000 cpm/pmole) and incubated for 30 min at room temperature. Assays are stopped by addition of 5 ⁇ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid.
  • JAK2 (5-20 mU diluted in 50 mM Tris pH 7.5, 0.1 mM EGTA, 0.05% ⁇ -mercaptoethanol, 1 mg/ml BSA) is assayed against PDKtide (KTFCGTPEYLAPEVRREPRILSEEEQ- EMFRDFDYIADWC) in a final volume of 25.5 ⁇ containing 50 mM Tris pH 7.5, 0.1 mM EGTA, 0.05% ⁇ -mercaptoethanol, 100 ⁇ substrate peptide, 10mM magnesium acetate and 0.005 mM [33 ⁇ - ⁇ - ⁇ ] (50-1000 cpm/pmole) and incubated for 30 min at room temperature Assays are stopped by addition of 5 ⁇ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid.
  • PDKtide KTFCGTPEYLAPEVRREPRILSEEEQ- EMFRDFDYIAD
  • TrkA assay TrkA (5-20mU diluted in 50 mM Tris pH 7.5, 0.1 mM EGTA, 10mM MnCI, 1 mg/ml BSA) is assayed against a substrate peptide (Poly Glut Tyr) in a final volume of 25.5 ⁇ containing 50mM Tris pH 7.5, 0.1 mM EGTA, 1 mg/ml substrate peptide, 10 mM magnesium acetate and 0.02 mM [33 ⁇ - ⁇ - ⁇ ] (50-1000 cpm/pmole) and incubated for 30 min at room temperature. Assays are stopped by addition of 5 ⁇ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid.
  • a substrate peptide Poly Glut Tyr
  • PKBAph SGK, MKK1, Aurora B, flocoumafen S6K1, CHK1 VEGFR, PKCa, SRC, TRKA, RSK2,
  • Potency be less than 200 uM on at least one cell line
  • Compounds must exhibit broad activity against a panel of cell lines from tumour types of varying origins. Effects on cellular proliferation were measured against 14 cell lines representing carcinomas of the lung, prostate, breast, liver, colon, pancreas as well as sarcomas. Cells were treated with 8 concentrations of compound 300, 100, 30, 10, 3, 1 , 0.3, 0.1, 0.03 and 0.01 ⁇ and cell number estimated at each concentration. Cell number was calculated as a percentage survival relative to untreated cells. EC 50 was calculated by fitting the data to the 4-parameter logistic model:
  • Table 9 details the transformed cell lines.
  • Table 10 shows the cytotoxicity data ( ⁇ EC50s) for selected compounds against 14 tumour cell lines over a 7 day exposure
  • Core anilino-pyrimidine moiety of CXR6024/Cyprodinil also features in some kinase inhibitors, notably the Abl inhibitor GleevecTM (see figure 3).
  • CXR6024/Cyprodinil was screened for kinase inhibition at 10 ⁇ and 100 ⁇ against a panel of 30 protein kinases. Data is presented as % inhibition. The most potent activity was against PIM3. Table 15 shows the % inhibition against the 30 kinases using Cyprodinil at 10 ⁇ and 100 ⁇ . The kinase assay methods are shown in Example 1 (criteria 6 of the selection criteria).
  • kinase inhibitors which feature aniline-pyrimidine moiety include molecules active against the following kinases:
  • Cyprodinil was part of a 144 compound library screened by CXR Biosciences for potential cytotoxic/cytostatic activity.
  • Test compounds were dissolved in DMSO and dilution series performed. Cells were seeded at a density of 500 cells per well in 75 ⁇ of DMEM (10% FBS/2 mM G!utamine) media for 7 day incubation in 96-well plates. The following day, 25 ⁇ of test compounds, diluted 1:25 in DMEM (10% FBS), were added to the wells to give final compound concentrations as detailed above. Duplicate plates were assembled and then incubated for 7 days at 37°C/5% C0 2 . No media/drug replacement during the 7 day period was undertaken.
  • CXR6024/Cyprodinil was tested in a number of cell lines for anti-proliferative and cytotoxic effects. Cells were seeded at 400 cells per well in 96 well dishes and maintained in the presence of CXR6024/Cyprodinil for 7 days without change of media. Cell number was estimated using the WST-1 dye and was presented as percentage inhibition of growth.
  • Figure 1 shows specimen dose-response curves for OVCAR3 and HepG2 cell lines.
  • CXR6024/Cyprodinil was active against all cell lines with EC 50 s varying by a factor of less than 10 across the tumour cell line panel.
  • the compounds showed little if any specificity for non-transformed dividing cell lines over dividing tumour cell lines, the EC 50 s against WI38 and hTERT lines being 19 and 13 ⁇ respectively, very similar to the 12 ⁇ median EC 50 of the dividing tumour cell lines.
  • non- transformed cell line hTERT when plated at near 100% confluence, shows marked resistance to CXR6024/Cyprodinil, having an EC 50 of 125 ⁇ , almost 10 fold the EC 50 on the same cell line when dividing, indicating the compound may have some specificity for dividing cells over non-dividing cells.
  • Combination Cell Culture PC3 cells were cultured and an IC 50 for CXR6024/Cyprodinil was calculated in the presence of a number of concentrations of the HSP90 inhibitor geldanamycin. As seen in the isobologram of figure 2, the lower concentrations of geldanamycin resulted in a left shift of the IC 50 for CXR6024/Cyprodinil indicating and increased efficacy of the compound.
  • Cyprodinil was selected for advancement to in vivo animal models based on in vitro efficacy, selectivity, kinase inhibitory activity and absorption, distribution, metabolism, and excretion (ADME) criteria as shown in Table 19.
  • Table 19
  • mice (6 - 8 weeks) female, athymic nude (nu/nu) mice were obtained from Charles River, UK. On arrival the mice were housed, up to 8 per cage, on sterile sawdust in sterile, solid-bottom, polypropylene cages. The cages were individually vented units attached to a Techniplast Slimline Air Handling Unit. This unit maintained 70-80 air changes per cage, per hour, through HEPA air filters. Bedding was changed once weekly in a laminar flow unit. The temperature was maintained within a target range of 19-23° C and relative humidity of the IVC within a range of 40-70%. Twelve-hour periods of light were cycled with twelve-hour periods of darkness.
  • mice Sterile RM1 diet (Special Diet Services Ltd., Stepfield, Witham, Essex, UK) was used. Deionised water was autoclaved prior to use and changed at least once a week. Mice were allowed water and diet ad libitum and were acclimatised for at least 5 days prior to the study start. The mice had SPF-status and the housing and changing system assured that the SPF- status was preserved during the study. Trained personnel under supervision handled the mice.
  • the animals were randomly allocated to groups on arrival, numbered appropriately and weighed prior to the start of the experiment.
  • the first mouse assigned to a cage was individually identified by tail tattooing with the lowest number for that cage; the second mouse was assigned the second (lowest) number and so on.
  • An experimental card was placed on each cage and showed the project licence code, test group, study number, sex and individual numbers of the mice within, and identified the Home Office Licensee. In addition, these cards were colour coded to correlate with the coding for the group.
  • PC3 and HT-29 cells were cultured in Dulbecco's Modified Eagle's Medium (DMEM) containing 10% heat inactivated foetal calf serum (FCS) supplemented with 2mM L- glutamine, penicillin (50 lU/ml) and streptomycin (50 ⁇ g/ml). Cultures were incubated in a humidified incubator at 37°C, 5% C0 2 , until sufficient cells were available to implant the mice. Cells were harvested, pooled, centrifuged, and re-suspended in cold medium. These were mixed with an equal volume of cold Matrigel, so that the tumour cell injection solution was a 50:50 mixture of tumour cells/medium and Matrigel.
  • DMEM Dulbecco's Modified Eagle's Medium
  • FCS heat inactivated foetal calf serum
  • Tumour cells were injected in a volume of 100 ⁇ in a single flank only.
  • HT-29 cells were injected at a density of 1.75 x 10 6 cells per flank.
  • PC3 cells were injected at a density of 2.5 x 10 6 cells per flank.
  • the cell/Matrigel suspension was aliquoted into pre-chilled Eppendorf tubes and kept on ice prior to injection. The period between preparation of the cell/Matrigel suspension and injection of tumour cells will not exceed two hours.
  • mice were administered Test Compounds 72 hours following implantation of tumour cells. Mice harbouring PC3 tumour cells were dosed with CXR6024 (Cyprodinil) in an ethanol/PEG200/water vehicle. HT-29 bearing mice were dosed with CXR6024(Cyprodinil) in a corn oil vehicle. Compounds were administered daily for 28 days by oral gavage in a dose volume of 10ml/kg.
  • the bodyweight of each mouse was recorded at the time of tail tattooing, before implantation and daily prior to dosing.
  • mice Prior to the start of the study, all mice were observed to ensure that they were physically normal and that they exhibited normal activity. Only normal mice were allocated to the study. Following cell inoculation, each mouse was observed twice weekly and a general assessment of condition recorded in the study diary. Animals were terminated if tumours become ulcerated or if the Home Office Project License moderate severity limit is exceeded.
  • tumour were cut in half and one half placed in 10 % neutral buffered formalin (NBF) then prepwered for paraffin embedding (section 6.4) for possible future histochemical analysis.
  • NBF neutral buffered formalin
  • cryovial 1 were labeled microarray and cryovial 2 were labeled
  • Cryovial 1 were labeled 'Microarray' and cryovial 2 were labeled 'MS'. These samples were stored at approximately - 80°C for possible future analysis. Following removal into tubes suitable for plasma preparation, venous blood samples were mixed on a roller for 10 minutes. Red blood cells were removed by centrifugation at 2,000 - 3000 rpm for 10 minutes at 8 - 10 °C. The supernatant (plasma) were transferred to a second tube and stored at approximately -70 °C until required for analysis.
  • PC3 cells grown subcutaneously were dosed orally daily with CXR6024/Cyprodinil at 150 mg/kg. Dosing started c 1 week after implantation of cells. a. 83 % mice developed tumours compared to 100 % in the control group.
  • HT29 cells were injected subcutaneously and dosed orally every day with CXR6024/Cyprodinil at 180, 250 and 400 mg/kg.
  • PC3 cells were injected subcutaneoualy and CXR6024/Cyprodinil was dosed orally every day at 250 and 400 mg/kg when tumour volume was c 100mm 3 .
  • PC3 cells were cultured and then Cyprodinil/CXR6024 was added and ATP levels were meaured a time intervals afterward. Cyprodinil/CXR6024 had no observable effect on levels of cellular ATP over 4 hours of exposure to cells (see Figure 9).
  • PC3 cells were plated and cyprodinil added to the culture media.
  • Cellular mitochondrial NADH utilising enzyme activities were subsequently estimated at various timepoints using the WST- reagent (as described in Example 3).
  • Cyprodinil/CXR6024 has a marked effect on the activity of these enzymes, in comparison to it's structural analogue pyrimethanil, suggesting that this activity may play some role in the cytotoxic/cytostatic activity of the moloecule.
  • This assay requires that the cells be incubated for around 60 - 90 minutes for the WST-1 dye to be metabolised by the enzymes and this probably explains the apparent activity at timepoint zero.
  • PC3 cells were treated for 24 hours with EC 20 and EC 50 concentrations (derived in 7 day exposure experiments) of CXR6024/Cyprodinil.
  • RNA was extracted and changes in gene expression levels using Agilent microarray. Methods for microarray studies
  • RNA microarray analysis involved labelled (one colour) RNA from triplicate samples of PC3 cell line treated vehicle, CXR6024 at EC 2 o and at EC 50 as shown in Figure 11. RNA integrity was checked using the Agilent bioanalyser and the RNA nano lab chip kit according to the CXR method sheet 'Set-up and Running of Nanochip Assay for RNA Quality Control'.
  • RNA was labelled prior to microarray hybridisation using the Quick Amp Labelling Kit - Two Color (Agilent* 5190-0442), according to the CXR method sheet entitled "Transcriptional Profiling using Standard Agilent 1 Colour Protocol” v2.
  • Results Tables 21 and 22 detail the genes identified as being changed beyond the threshold level of either 2-fold up or down regulation.
  • Example 6 Preferred pharmaceufcal formulations and modes and doses of administration.
  • the compounds of the present invention may be delivered using an injectable sustained- release drug delivery system. These are designed specifically to reduce the frequency of injections.
  • An example of such a system is Nutropin Depot which encapsulates recombinant human growth hormone (rhGH) in biodegradable microspheres that, once injected, release rhGH slowly over a sustained period.
  • the compounds of the present invention can be administered by a surgically implanted device that releases the drug directly to the required site.
  • Vitrasert releases ganciclovir directly into the eye to treat CMV retinitis.
  • the direct application of this toxic agent to the site of disease achieves effective therapy without the drug's significant systemic side-effects.
  • Electroporation therapy (EPT) systems can also be employed for administration.
  • a device which delivers a pulsed electric field to cells increases the permeability of the cell membranes to the drug, resulting in a significant enhancement of intracellular drug delivery.
  • Compounds of the invention can also be delivered by electroincorporation (El).
  • El occurs when small particles of up to 30 microns in diameter on the surface of the skin experience electrical pulses identical or similar to those used in electroporation. In El, these particles are driven through the stratum corneum and into deeper layers of the skin.
  • the particles can be loaded or coated with drugs or genes or can simply act as "bullets" that generate pores in the skin through which the drugs can enter.
  • ReGel injectable system that is thermosensitive. Below body temperature, ReGel is an injectable liquid while at body temperature it immediately forms a gel reservoir that slowly erodes and dissolves into known, safe, biodegradable polymers. The active drug is delivered over time as the biopolymers dissolve.
  • Trojan peptides are a class of polypeptides called penetratins which have translocating properties and are capable of carrying hydrophilic compounds across the plasma membrane. This system allows direct targeting of compounds to the cytoplasm and nucleus, and may be non-cell type specific and highly efficient (Derossi et al., 1998, Trends Cell Biol., 8, 84-87).
  • the pharmaceutical formulation of the present invention is a unit dosage containing a daily dose or unit, daily sub-dose or an appropriate fraction thereof, of the active ingredient.
  • the compounds of the invention can be administered by any parenteral route, in the form of a pharmaceutical formulation comprising the active ingredient, optionally in the form of a non-toxic organic, or inorganic, acid, or base, addition salt, in a pharmaceutically acceptable dosage form.
  • the compositions may be administered at varying doses.
  • the compounds of the invention can be administered alone but will generally be administered in admixture with a suitable pharmaceutical exipient diluent or carrier selected with regard to the intended route of administration and standard pharmaceutical practice.
  • the compounds of the invention can also be administered parenterally, for example, intravenously, intra-arterially, intraperitoneally, intra-thecally, intraventricularly, intrastemally, intracranially, intra-muscularly or subcutaneously, or they may be administered by infusion techniques. They are best used in the form of a sterile aqueous solution which may contain other substances, for example, enough salts or glucose to make the solution isotonic with blood.
  • the aqueous solutions should be suitably buffered (preferably to a pH of from 3 to 9), if necessary.
  • suitable parenteral formulations under sterile conditions is readily accomplished by standard pharmaceutical techniques well-known to those skilled in the art.
  • Formulations suitable 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 water for injections, immediately prior to use.
  • Extemporaneous injection solutions and suspensions may be prepared from sterile powders, granules and tablets of the kind previously described.
  • oral or parenteral administration of the compounds of the invention is the preferred route, being the most convenient.
  • the compounds of the invention are administered as a suitably acceptable formulation in accordance with normal veterinary practice and the veterinary surgeon will determine the dosing regimen and route of administration which will be most appropriate for a particular animal.
  • compositions of the invention may conveniently be presented in unit dosage form and may be prepared by any of the methods well known in the art of pharmacy. Such methods include the step of bringing into association the active ingredient 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.
  • Preferred unit dosage formulations are those containing a daily dose or unit, daily sub- dose or an appropriate fraction thereof, of an active ingredient.
  • a preferred delivery system of the invention may comprise a hydrogel impregnated with a polypeptides, polynucleotides and antibodies of the invention, which is preferably carried on a tampon which can be inserted into the cervix and withdrawn once an appropriate cervical ripening or other desirable affect on the female reproductive system has been produced.
  • formulations of this invention may include other agents conventional in the art having regard to the type of formulation in question.
  • compounds of the invention Whilst it is possible for compounds of the invention to be administered alone, it is preferable to present it as a pharmaceutical formulation, together with one or more acceptable carriers.
  • the carrier(s) must be "acceptable” in the sense of being compatible with the compound of the invention and not deleterious to the recipients thereof.
  • the carriers will be water or saline which will be sterile and pyrogen-free.
  • Example 7A Injectable Formulation Active ingredient 0.200 g
  • the active ingredient is dissolved in most of the phosphate buffer (35-40 ° C), then made up to volume and filtered through a sterile micropore filter into a sterile 10 ml amber glass vial (type 1) and sealed with sterile closures and overseals.
  • Example 7B Intramuscular injection Active ingredient 0.20 g
  • Tablets are prepared from the foregoing ingredients by wet granulation followed by compression.

Abstract

There is provided anilinopyrimidine compounds (such as Cyprodinil) for use in medicine, preferably for the treatment of cancer.

Description

CYPRODINIL FOR USE IN MEDICINE
The present invention relates to the medical use of the anilinopyrimidine compound Cyprodinil and particularly its use in the treatment of cancer.
Background
Cancer (or neoplasm) covers a range of diseases in which a group of cells display uncontrolled growth (division beyond the normal limits), invasion (intrusion on and destruction of adjacent tissues), and sometimes metastasis (spread to other locations in the body via lymph or blood). Cancer affects people at all ages with the risk for most types increasing with age and caused about 13% of all human deaths in 2007. Deaths from cancer worldwide are projected to continue rising, with an estimated 12 million deaths in 2030. (WHO, February 2009)
Traditionally cancer treatment is based around surgery, chemotherapy and radiotherapy. However, the effectiveness of surgery is often limited by the propensity of cancers to invade adjacent tissue or to spread to distant sites by microscopic metastasis. The effectiveness of chemotherapy is often limited by toxicity to other tissues in the body. Radiation can also cause damage to normal tissue.
More recently, targeted therapy has had a significant impact in the treatment of some types of cancer. This constitutes the use of agents specific for the deregulated proteins of cancer cells. Small molecule targeted therapy drugs are generally inhibitors of enzymatic domains on mutated, overexpressed, or otherwise critical proteins within the cancer cell.
The discovery and development of new agents for cancer treatment is both a very costly and very time consuming process. Traditionally agents are selected as having a promising activity against a particular biological target thought to be important in disease; however, little will be known about the safety, toxicity, pharmacokinetics and metabolism of this agent in humans. Therefore, it is traditionally necessary to assess all of these parameters prior to human clinical trials in order to be able to recommend a dose and schedule to be used the first time. In addition, much drug development work is required to establish the physicochemical properties of a new agent, such as its chemical makeup, stability, solubility. The process by which the chemical is made will be optimized and it will be further examined for its suitability to be made into capsules, tablets, aerosol, intramuscular injectable, subcuteneous injectable, or intravenous formulations.
Many aspects of drug development are focused on satisfying the regulatory requirements of drug licensing authorities. These generally constitute a number of tests designed to determine the major toxicities of a novel compound prior to first use in man. It is a legal requirement that an assessment of major organ toxicity be performed (effects on the heart and lungs, brain, kidney, liver and digestive system), as well as effects on other parts of the body that might be affected by the drug (e.g. the skin if the new drug is to be delivered through the skin). While, increasingly, these tests can be made using in vitro methods (e.g. with isolated cells), many tests can only be made by using experimental animals, since it is only in an intact organism that the complex interplay of metabolism and drug exposure on toxicity can be examined.
The process of drug development does not stop once a novel agent begins human clinical trials. In addition to the tests required to move a novel drug into the clinic for the first time it is also important to ensure that long-term or chronic toxicities are determined, as well as effects on systems not previously monitored (fertility, reproduction, immune system, etc).
If a compound emerges from these tests with an acceptable toxicity and safety profile, and it can further be demonstrated to have the desired effect in clinical trials, then it can be submitted for marketing approval in the various countries where it will be sold. However, most new agents fail during drug development, often because they have some unacceptable toxicity. Existing anti-cancer agents have considerable problems. A significant problem, which affects the majority of traditional anti-cancer agents, is the severe associated toxicities resulting in side effects such as depletion of the immune system, vomiting, malnutrition, hair loss, fatigue and anemia. Another significant problem is lack of efficacy. Current anti-cancer agents all have limited efficacy and cannot be guaranteed to cure a patient of disease. In some cancers a cure or halt of the disease can actually be very unlikely with the current anti-cancer drugs. Therefore, there remains a clear need to discover and develop new agents for use in the treatment of cancer which reduce the problems associated with current anti-cancer agents. To circumnavigate many of the developmental issues described above, the applicants have developed a method of screening compounds for potential pharmaceutical agents (preferably anti-cancer compounds) based on compounds for which bioactivity and toxicity data is already available. For example, such compounds might have previously been tested for use in unrelated fields, such as agrochemicals.
Here the applicants provide compounds that have been identified by this new screening method for use in medicine, and in particular, cancer treatment.
Anilinopyrimidine fungicides are active against a broad spectrum of fungi (Kunz et a/. (1998), J. Phytopathology, 146, 231-238 and Knauf-Beiter et al. 1995, Plant Disease, 79(11), 1098-1103). Currently three fungicides of this class are marketed, cyprodinil (Syngenta), mepanipyrim (Kumiai chemicals Industry Co) and pyrimethanil (AgrEvo GmbH). All have the same core structure and differ only in the substitution at the 4 position of the pyrimidine ring, where cyprodinil has a cyclopropyl, mepanipyrim a 1- propynyl and pyrimethanil a methyl substituent.
Fungicidal mode of action is achieved via inhibition of methionine biosynthesis and secretion of hydrolytic enzymes. Field monitoring of Cyprodinil efficacy has indicated that practical resistance to the compound is rare in the key target pathogens (botrytis and venturia). Resistance can be generated under controlled conditions but resistance to anilinopyrimidine fungicides does not correlate with resistance to other classes of fungicide (Myresiotis et al. 2007, Plant Disease, 91(4):407-413).
Cyprodinil was first introduced in France in 1993 by Ciba for use on cereals and is now used widely in Europe as a foliar fungicide on fruit, cereals, vegetables and ornamentals.
The applicant has now found that Cyprodinil may be useful in medicine, particularly in the treatment of cancer. In a first aspect of the invention, there is provided Cyprodinil or a salt or ester thereof for use in medicine. Cyprodinil (CAS registration number 121552-61-2) is represented by the following structure:
Figure imgf000005_0001
CXR6024/Cyprodinil (4-cycloproplyl-6-methyl-N-phenyl-pyrimidinamine) is also marketed by Syngenta under the trade names Unix™, Chorus™, Stereo™, Switch™ and Vanguard™. Cyprodinil has the CAS registration number 121552-61-2 and the development code was CGA219417. Synonyms for Cyprodinil
1 ) 4-cyclopropyl-6-methyl-N-phenylpyrimidin-2-amine
2) CYPRODINIL
3) CYPRODYNIL
4) 2-Pyrimidinamine, 4-cyclopropyl-6-methyl-N-phenyl-
5) Chorus™
6) 4-cyclopropyl-6-methyl-N-phenyl-2-pyrimidinamine
7) 4-cycloproplyl-6-methyl-N-phenyl-pyrimidinamine
8) Unix™
9) Stereo™
10) Switch™
It has now been found that Cyprodinil is an inhibitor of PI kinase family members (PI 1, PIM2 and PIM3). PIM Kinase activity
PIM kinases are cytoplasmic serine/threonine kinases that are known to be involved in regulation of apoptosis and cellular metabolism. Certain PIM kinases have been shown to be upregulated in cancers and as such their inhibition represents a mechanism of action by which compounds such as cyprodinil can have an anti-tumour effect in conditions such as leukaemia, lymphoma, prostate cancer, colon cancer and pancreatic cancer. The studies shown below in Table 1 have shown this link:
Table 1
Figure imgf000006_0001
Prostrate cancer: Chen (2005), Mumenthaler (2009), He (2007), Xu (2005), Dai (2005) and Ron (2008) have shown PIM-1 overexpression in prostatic carcinoma.
Breast cancer: Rho (2008) has shown PIM-1 overexpression to convert mammary epithelia cells to become tumourgenic.
Adipocyte tumours: Nga (2010) has shown benign and malignant adipocytic tumours to have strong PIM-1 expression.
PIM kinases are constitutively active and their activity as shown above and in Amaravadi (2005) and Shah (2008) supports in vitro and in vivo human cell growth and survival. Cyprodinil and salts and esters thereof are referred to herein as "the compounds of the invention":
Pharmaceutically-acceptable salts include acid addition salts and base addition salts. Such salts may be formed by conventional means, for example by reaction of a free acid or a free base form of a compound of formula I with one or more equivalents of an appropriate acid or base, optionally in a solvent, or in a medium in which the salt is insoluble, followed by removal of said solvent, or said medium, using standard techniques (e.g. in vacuo, by freeze-drying or by filtration). Salts may also be prepared by exchanging a counter-ion of a compound of the invention in the form of a salt with another counter-ion, for example using a suitable ion exchange resin.
By the term "ester" is included those formed with an alcohol of formula R1OH, wherein R represents aryl or alkyl; and those formed with a thiol of formula R SH, wherein R1 is as hereinbefore defined (i.e. a thioester). It is preferred that the ester is not a thioester. In embodiments R1 represents C1-6 alkyl, for example alkyl (eg methyl).
Compounds of the invention may contain double bonds and may thus exist as E (entgegen) and Z (zusammen) geometric isomers about each individual double bond. All such isomers and mixtures thereof are included within the scope of the invention.
Compounds of the invention may also exhibit tautomerism. All tautomeric forms and mixtures thereof are included within the scope of the invention. Compounds of the invention may also contain one or more asymmetric carbon atoms and may therefore exhibit optical and/or diastereoisomerism. Diastereoisomers may be separated using conventional techniques, e.g. chromatography or fractional crystallisation. The various stereoisomers may be isolated by separation of a racemic or other mixture of the compounds using conventional, e.g. fractional crystallisation or HPLC, techniques. Alternatively the desired optical isomers may be made by reaction of the appropriate optically active starting materials under conditions which will not cause racemisation or epimerisation (i.e. a 'chiral pool' method), by reaction of the appropriate starting material with a 'chiral auxiliary' which can subsequently be removed at a suitable stage, by derivatisation (i.e. a resolution, including a dynamic resolution), for example with a homochiral acid followed by separation of the diastereomeric derivatives by conventional means such as chromatography, or by reaction with an appropriate chiral reagent or chiral catalyst all under conditions known to the skilled person. All stereoisomers and mixtures thereof are included within the scope of the invention.
In a second aspect of the invention, there is provided a pharmaceutical composition comprising cyprodinil or a salt or ester thereof and a pharmaceutically acceptable excipient, diluent or carrier.
In one embodiment of the invention there is provided a composition comprising between 10mg and 2000mg of an active ingredient per dosage unit, wherein the active ingredient is a compound of the invention or a derivative, salt or variant thereof. By dosage unit we mean the unit of medicament administered to a patient at one time. For example, the dosage unit, or single dose may be administered by a single capsule/tablet, single injection, or single intravenous infusion, a single subcutaneous injection, or by a single procedure using other routes of administration, as discussed below. Altematively, the single dose may be administered to the patient by two or more capsules/tablets or injections given simultaneously or sequentially to deliver the entire dose to the patient in the continuous, single and defined treatment period; by two or more intravenous infusions given simultaneously or sequentially to deliver the entire dose to the patient in the continuous, single and defined treatment; or by multiple procedures using other routes of administration as discussed below. Alternatively, the single dose to be administered to the patient can be delivered by a combination of routes to deliver the entire dose to the patient in the continuous, single and defined treatment. The dosage unit may then be repeated at intervals of time such as a few hours, days, weeks, or months later.
Dosage units can be administered to patients in such a way that the patient receives a loading dose followed by one or more maintenance doses. For example the loading dose may be a high dose in order to quickly reach a desired plasma concentration and then subsequent maintenance doses are a lower dose than the loading dose in order to maintain the required plasma concentration.
By active ingredient we mean the molecule having the desired effect. In this case of this invention we primarily mean the compounds of the invention and derivatives, salts or variants thereof.
By variants and derivatives we mean any molecules of substantially identical chemical structure but including minor modifications that do not alter activity but may offer improved or alternative properties for formulation, such as formation into a salt.
In human therapy, the compound of the invention containing composition, and medicaments of the invention can be administered alone but will generally be administered in admixture with a suitable pharmaceutical excipient, diluent or carrier selected with regard to the intended route of administration and standard pharmaceutical practice.
For example, the compound of the invention containing composition, and medicaments of the invention can be administered orally, buccally or sublingually in the form of tablets, capsules, ovules, elixirs, solutions or suspensions, which may contain flavouring or colouring agents, for immediate-, delayed- or controlled-release applications. The compound of the invention containing composition, and medicaments of the invention may also be administered via intracavernosal injection. Such tablets may contain excipients such as microcrystalline cellulose, lactose, sodium citrate, calcium carbonate, dibasic calcium phosphate and glycine, disintegrants such as starch (preferably corn, potato or tapioca starch), sodium starch glycollate, croscarmellose sodium and certain complex silicates, and granulation binders such as polyvinylpyrrolidone, hydroxypropylmethylcellulose (HPMC), hydroxy-propylcellulose (HPC), sucrose, gelatin and acacia. Additionally, lubricating agents such as magnesium stearate, stearic acid, glyceryl behenate and talc may be included.
Solid compositions of a similar type may also be employed as fillers in gelatin capsules. Preferred excipients in this regard include lactose, starch, cellulose, milk sugar or high molecular weight polyethylene glycols. For aqueous suspensions and/or elixirs, the compound of the invention containing composition, medicaments and pharmaceutical compositions of the invention may be combined with various sweetening or flavouring agents, colouring matter or dyes, with emulsifying and/or suspending agents and with diluents such as water, ethanol, propylene glycol and glycerin, and combinations thereof.
The compound of the invention containing composition, and medicaments of the invention can also be administered parenterally, for example, intravenously, intra- arterially, intraperitoneally, intra-thecally, intraventricularly, intrasternally, intracranially, intra-muscularly or subcutaneously, or they may be administered by infusion techniques. They are best used in the form of a sterile aqueous solution which may contain other substances, for example, enough salts or glucose to make the solution isotonic with blood. The aqueous solutions should be suitably buffered (preferably to a pH of from 3 to 9), if necessary. The preparation of suitable parenteral formulations under sterile conditions is readily accomplished by standard pharmaceutical techniques well-known to those skilled in the art. Medicaments and pharmaceutical compositions suitable for parenteral administration include aqueous and non-aqueous sterile injection solutions which may contain antioxidants, 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 medicaments and pharmaceutical compositions 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 earner, for example water for injections, immediately prior to use. Extemporaneous injection solutions and suspensions may be prepared from sterile powders, granules and tablets of the kind previously described. The compound of the invention containing composition, and medicaments of the invention can also be administered intranasally or by inhalation and are conveniently delivered in the form of a dry powder inhaler or an aerosol spray presentation from a pressurised container, pump, spray or nebuliser with the use of a suitable propellant, e.g. dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoro-ethane, a hydrofluoroalkane such as 1,1,1,2-tetrafluoroethane (HFA 134A3 or 1,1,1,2,3,3,3- heptafluoropropane (HFA 227EA3), carbon dioxide or other suitable gas. In the case of a pressurised aerosol, the dosage unit may be determined by providing a valve to deliver a metered amount. The pressurised container, pump, spray or nebuliser may contain a solution or suspension of the active agent, e.g. using a mixture of ethanol and the propellant as the solvent, which may additionally contain a lubricant, e.g. sorbitan trioleate. Capsules and cartridges (made, for example, from gelatin) for use in an inhaler or insufflator may be formulated to contain a powder mix of a compound of the invention containing composition, of the invention and a suitable powder base such as lactose or starch.
Aerosol or dry powder formulations are preferably arranged so that each metered dose or "puff contains an effective amount of an agent or polynucleotide of the invention for delivery to the patient. It will be appreciated that the overall daily dose with an aerosol will vary from patient to patient, and may be administered in a single dose or, more usually, in divided doses throughout the day.
Alternatively, the compound of the invention containing composition, and medicaments of the invention can be administered in the form of a suppository or pessary, or they may be applied topically in the form of a lotion, solution, cream, gel, ointment or dusting powder. The compound of the invention containing composition, and medicaments of the invention may also be transdermal^ administered, for example, by the use of a skin patch. They may also be administered by the ocular route, particularly for treating diseases of the eye.
For ophthalmic use, the compound of the invention containing composition, and medicaments of the invention can be formulated as micronised suspensions in isotonic, pH adjusted, sterile saline, or, preferably, as solutions in isotonic, pH adjusted, sterile saline, optionally in combination with a preservative such as a benzylalkonium chloride. Alternatively, they may be formulated in an ointment such as petrolatum. For application topically to the skin, the compound of the invention containing composition, and medicaments of the invention can be formulated as a suitable ointment containing the active agent suspended or dissolved in, for example, a mixture with one or more of the following: mineral oil, liquid petrolatum, white petrolatum, propylene glycol, polyoxyethylene polyoxypropylene agent, emulsifying wax and water. Alternatively, they can be formulated as a suitable lotion or cream, suspended or dissolved in, for example, a mixture of one or more of the following: mineral oil, sorbitan monostearate, a polyethylene glycol, liquid paraffin, polysorbate 60, cetyl esters wax, cetearyl alcohol, 2- octyldodecanol, benzyl alcohol and water.
Formulations suitable for topical administration in the mouth include lozenges comprising the active ingredient in a flavoured basis, usually sucrose and acacia or tragacanth; pastilles comprising the active ingredient in an inert basis such as gelatin and glycerin, or sucrose and acacia; and mouth-washes comprising the active ingredient in a suitable liquid carrier.
Generally, in humans, oral or parenteral administration of the compound of the invention containing composition, medicaments and pharmaceutical compositions of the invention is the preferred route, being the most convenient.
For veterinary use, the compound of the invention containing composition, and medicaments of the invention are administered as a suitably acceptable formulation in accordance with normal veterinary practice and the veterinary surgeon will determine the dosing regimen and route of administration which will be most appropriate for a particular animal.
The compound of the invention containing composition, as defined herein may be formulated as described in the accompanying Examples. The composition may comprise any effective amount of active ingredient, this may be between 10mg and 2000mg of active ingredient per dosage unit, and preferably is between 50mg and 1000mg. Advantageously it is 1000mg. Conveniently, the dosage unit contains an amount of active ingredient per dosage unit selected from 10mg, 20mg, 25mg, 50mg, 100mg, 200mg, 300mg, 400mg, 450mg, 600mg, 750mg, 950mg, 1000mg and 1200mg. Alternatively, the composition may comprise between 10-50mg, 10-75mg, 10-100mg, 10- 200mg, 10-300mg, 10-400mg, 10-600mg, 10-750mg, 10-950mg, 10-1000mg, 10- 1200mg, 50-75mg, 50-1 OOmg, 50-200mg, 50-300mg, 50-450mg, 50-600mg, 50-750mg, 50-950mg, 50-1000mg, 50-1200mg, 75-1 OOmg, 75-200mg, 75-300mg, 75-450mg, 75- 600mg, 75-750mg, 75-950mg, 75-1000mg, 75-1200mg, 100-200mg, 100-300mg, 100- 450mg, 100-600mg, 100-750mg, 100-950mg, 100-1000mg, 100-1200mg, 200-300mg, 200-450mg, 200-600mg, 200-750mg, 200-950mg, 200-1 OOOmg, 200-1200mg, 300- 450mg, 300-600mg, 300-750mg, 300-950mg, 300-1 OOOmg, 300-1200mg, 400-600 mg, 400-750mg, 400-950mg, 400-1 OOOmg, 400-1200mg, 450-600mg, 450-750mg, 450- 950mg, 450-1000mg, 450-1200mg, 600-750mg, 600-950mg, 600-IOOOmg, 600-1200mg, 700-950mg, 700-1 OOOmg, 700-1200mg, 950-1 OOOmg, 950-1200mg and 1000-1200mg
Preferably there is 400-600mg of active ingredient. More preferably there is 400-1200mg of active ingredient. Most preferably there is 10OOmg of active ingredient.
Conveniently, the composition is pharmaceutically acceptable, and may optionally contain a pharmaceutically acceptable excipient, diluent, carrier or filler.
The examples describe some methods of producing pharmaceutical formulations, however the skilled person will appreciate that the most appropriate formulation will depend on a number of factors including route of administration, patient type (e.g. patient age, weight/size).
A further aspect of the invention provides a method of treating cancer (such as cancers of the breast, colon, prostate, ovaries, brain and lung and their metastases) comprising administering to the patient an effective amount of cyprodinil or a salt or an ester thereof.
The term "patient" includes all animals including humans. Examples of patients include humans, cows, dogs, cats, goats, sheep, and pigs. The term "patienf means an animal having a disorder in need of treatment.
Preferably, the patient is selected from humans, cows, dogs, cats, goats, sheep, and pigs. More preferably, the patient is a human.
The compounds of the invention may be used in cancer treatment either alone or in combination with well known anti-cancer agents. Cancer treatments promote tumour regression by inhibiting tumour cell proliferation, inhibiting angiogenesis (growth of new blood vessels that is necessary to support tumour growth) and/or prohibiting metastasis by reducing tumour cell motility or invasiveness. Therapeutic compositions of the invention may be effective in adult and pediatric oncology including in solid phase tumours/malignancies, locally advanced tumours, human soft tissue sarcomas, metastatic cancer, including lymphatic metastases, blood cell malignancies including multiple myeloma, acute and chronic leukemias, and lymphomas, head and neck cancers including mouth cancer, larynx cancer and thyroid cancer, lung cancers including small cell carcinoma and non-small cell cancers, breast cancers including small cell carcinoma and ductal carcinoma, gastrointestinal cancers including esophageal cancer, stomach cancer, colon cancer, colorectal cancer and polyps associated with colorectal neoplasia, pancreatic cancers, liver cancer, urologic cancers including bladder cancer and prostate cancer, malignancies of the female genital tract including ovarian carcinoma, uterine (including endometrial) cancers, and solid tumour in the ovarian follicle, kidney cancers including renal cell carcinoma, brain cancers including intrinsic brain tumours, neuroblastoma, astrocytic brain tumours, gliomas, metastatic tumour cell invasion in the central nervous system, bone cancers including osteomas, skin cancers including malignant melanoma, tumour progression of human skin keratinocytes, squamous cell carcinoma, basal cell carcinoma, hemangiopericytoma and Karposi's sarcoma.
Compounds of the invention may be administered to treat cancer. The cancer to be treated is preferably selected from pancreatic cancer, ovarian cancer, breast cancer, prostate cancer, liver cancer, chondrosarcoma, lung cancer, head and neck cancer, colon cancer, sarcoma, leukaemia, myeloma, lymphoma, kidney cancer, thyroid cancer and brain cancers such as glioblastoma.
Therapeutic compositions can be administered in therapeutically effective dosages alone or in combination with adjuvant cancer therapy such as surgery, chemotherapy, radiotherapy, thermotherapy, and laser therapy, and may provide a beneficial effect, e.g. reducing tumour size, slowing rate of tumour growth, inhibiting metastasis, or otherwise improving overall clinical condition, without necessarily eradicating the cancer.
The composition can also be administered in therapeutically effective amounts as a portion of an anti-cancer cocktail. An anti-cancer cocktail is a mixture of the compound or modulator of the invention with one or more anti-cancer drugs in addition to a pharmaceutically acceptable carrier for delivery. The use of anti-cancer cocktails as a cancer treatment is routine. Anti-cancer drugs that are well known in the art and can be used as a treatment in combination with the compounds of the invention include: 17AAG, 17DMAG, 5FU, 7-hydroxystaurosporine (UCN-01), ABT888, Actinomycin D, Alsterpaullone, Axitinib, Aminoglutethimide, Amsacrine, Asparaginase, Azacitidine, AZD7762, Bay 11-7082, Belinostat, Bendamustine, Bexarotene, BIBW 2992, Bisindolylmaleimide I, Bleomycin, Bortezomib, Bosutinib, Busulfan, Canertinib , Capecitabine, Carboplatin, Carmustine, CDK4/6 IV, Chelerythrine Chloride, Chlorambucil, CHR-2863 (CHROMA), CHR-3531 (CHROMA), Chromomycin A3, CI-994, Cisplatin, Cladribine, Clofarabine, Clofibrate, CP690550, CXR1002 (APFO), Cyclopamine, Cyclophosphamide, Cytarabine, Dacarbazine, Dactinomycin, Dasatinib, Daunorubicin, DBZ, Decitabine, dk2 inhibitor II, DMFO, Docetaxel, Doramapimod, Dovitinib, Doxorubicin, Entinostat, Epirubicin, Erlotinib, Estramustine, Etoposide (V16- 213), Everolimus, Flavopiridol, fludarabine phosphate, Flutamide, GDC-0449, Gefitanib, Geldanamycin, Go 6976, GSK-1904529A, GW2580, GW501516, HA14-1, Hexamethy!melamine, Hydroxyurea (hydroxycarbamide), Ifosfamide, Interferon Alpha- 2a, Interferon Alpha-2b, lnterleukin-2, Leuprolide acetate (LHRH-releasing factor analog), Idarubicin, Imatinib, Ispinesib mesilate, Kenpaullone, K0143, KU-0058948, Lapatinib, Lenalidomide, Lestaurtinib, Lomustine, LY 2157299, LY294002, Masitinib, Mechlorethamine HCI (nitrogen mustard), Melphalan, Mercaptopurine (6-MP), Mesna, MTX, Mitoguazone, Mitomycin C, MK 1775, Mocetinostat, Mitoxantrone HCI, Nelarabine, Nilotinib, Nobiletin, Nogalamycin, NSC 625987, Nutlin-3, NVP-AEW541, NVP-TAE684 - TAE 684, Obatoclax mesylate, Octreotide, Olaparib (KU0059436), Oxaliplatin, Pentostatin, Plicamycin, Procarbazine HCI, Paclitaxel, Panobinostat, Pazopanib, PD 98059, PD173074, Pemetrexed, Perifosine, perillic acid, PLX4032, PLX4720, PPP (Picropodophyllin), Puromycin, Radicicol, Raltitrexed, Rapamycin, Ridaforolimus, Semustine, Streptozocin, Saracatinib, SB 431542, SB202190, SB203580, Sorafenib, SP600125, Sunitinib, Suramin, Tamoxifen CITRATE, Teniposide, Tandutinib, Tegafur , Temodal, Thalidomide, Thioguanine (6-TG), Thiotepa, Topotecan hydrochloride hydrate, Tozasertib, Trichostatin A, Tyrphostin AG 490, Tyrphostin AG 538, Tyrphostin AG879, U0126, Valproic acid, Vandetanib, Vatalanib, Vinblastine sulfate salt, Vincristine, Vindesine sulphate, Vinolrelbine ditartrate salt hydrate, Wortmannin, XAV 939, YM155, ZSTK474,4HC,6-[4-(2-Piperidin-1-ylethoxy)phenyl]-3-pyridin-4-ylpyrazolo[1,5- ajpyrimidine (Dorsomorphin), Etoposide, Gemcitabine, Mitoxanthrone and Vorinostat. In addition, therapeutic compositions of the invention may be used for prophylactic treatment of cancer. There are hereditary conditions and/or environmental situations (e.g. exposure to carcinogens) known in the art that predispose an individual to developing cancers. Under these circumstances, it may be beneficial to treat these individuals with therapeutically effective doses of the compounds of the invention to reduce the risk of developing cancers. In vitro models can be used to determine the effective doses of the compounds of the invention as a potential cancer treatment. These in vitro models include proliferation assays of cultured tumour cells, growth of cultured tumour cells in soft agar (see Freshney, (1987) Culture of Animal Cells: A Manual of Basic Technique, Wily-Liss, New York, NY Ch 18 and Ch 21), tumour systems in nude mice as described in Giovanella et a/., J. Natl. Can. Inst, 52: 921-30 (1974), mobility and invasive potential of tumour cells in Boyden Chamber assays as described in Pilkington ef ai, Anticancer Res., 17: 4107-9 (1997), and angiogenesis assays such as induction of vascularization of the chick chorioallantoic membrane or induction of vascular endothelial cell migration as described in Ribatta et ai, Intl. J. Dev. Biol., 40: 1189-97 (1999) and Li et ai, Clin. Exp. Metastasis, 17:423-9 (1999), respectively. Suitable tumour cells lines are available, e.g. from American Type Tissue Culture Collection catalogues.
In one embodiment, the method, use or composition of the invention additionally comprises a further chemotherapeutic agent.
Preferably, the further chemotherapeutic agent is selected from 17AAG, 17DMAG, 5FU, 7-hydroxystaurosporine (UCN-01), ABT888, Actinomycin D, Alsterpaullone, Axitinib, Aminoglutethimide, Amsacrine, Asparaginase, Azacitidine, AZD7762, Bay 11-7082, Belinostat, Bendamustine, Bexarotene, BIBW 2992, Bisindolylmaleimide I, Bleomycin, Bortezomib, Bosutinib, Busulfan, Canertinib , Capecitabine, Carboplatin, Carmustine, CDK4/6 IV, Chelerythrine Chloride, Chlorambucil, CHR-2863 (CHROMA), CHR-3531 (CHROMA), Chromomycin A3, CI-994, Cisplatin, Cladribine, Clofarabine, Clofibrate, CP690550, CXR1002 (APFO), Cyclopamine, Cyclophosphamide, Cytarabine, Dacarbazine, Dactinomycin, Dasatinib, Daunorubicin, DBZ, Decitabine, dk2 inhibitor II, DMFO, Docetaxel, Doramapimod, Dovitinib, Doxorubicin, Entinostat, Epirubicin, Erlotinib, Estramustine, Etoposide (V16-213), Everolimus, Flavopiridol, fludarabine phosphate, Flutamide, GDC-0449, Gefitanib, Geldanamycin, Go 6976, GSK-1904529A, GW2580, GW501516, HA14-1, Hexamethylmelamine, Hydroxyurea (hydroxycarbamide), Ifosfamide, Interferon Alpha-2a, Interferon Alpha-2b, lnterleukin-2, Leuprolide acetate (LHRH-releasing factor analog), Idarubicin, Imatinib, Ispinesib mesilate, Kenpaullone, K0143, KU-0058948, Lapatinib, Lenalidomide, Lestaurtinib, Lomustine, LY 2157299, LY294002, Masitinib, Mechlorethamine HCI (nitrogen mustard), Melphalan, Mercaptopurine (6-MP), Mesna, MTX, itoguazone, Mitomycin C, MK 1775, Mocetinostat, Mitoxantrone HCI, Nelarabine, Nilotinib, Nobiletin, Nogalamycin, NSC 625987, Nutlin-3, NVP-AEW541, NVP-TAE684 - TAE 684, Obatoclax mesylate, Octreotide, Olaparib (KU0059436), Oxaiiplatin, Pentostatin, Plicamycin, Procarbazine HCI, Paclitaxel, Panobinostat, Pazopanib, PD 98059, PD173074, Pemetrexed, Perifosine, perillic acid, PLX4032, PLX4720, PPP (Picropodophyllin), Puromycin, Radicicol, Raltitrexed, Rapamycin, Ridaforolimus, Semustine, Streptozocin, Saracatinib, SB 431542, SB202190, SB203580, Sorafenib, SP600125, Sunitinib, Suramin, Tamoxifen CITRATE, Teniposide, Tandutinib, Tegafur , Temodal, Thalidomide, Thioguanine (6-TG), Thiotepa, Topotecan hydrochloride hydrate, Tozasertib, Trichostatin A, Tyrphostin AG 490, Tyrphostin AG 538, Tyrphostin AG879, U0126, Valproic acid, Vandetanib, Vatalanib, Vinblastine sulfate salt, Vincristine, Vindesine sulphate, Vinolrelbine ditartrate salt hydrate, Wortmannin, XAV 939, YM155, ZSTK474,4HC,6-[4-(2-Piperidin-1- ylethoxy)phenyl]-3-pyridin-4-ylpyrazolo[1 ,5-a]pyrimidine (Dorsomorphin), Etoposide, Gemcitabine, Mitoxanthrone and Vorinostat.
In one embodiment the further chemotherapeutic agent is a HSP inhibitor, such as geldanamycin, 17-AAG or 17-DMAG. Due to the favourable toxicity and safety profiles of the compounds of the invention, the compounds may be used to treat patients who may not be amenable to conventional chemotherapeutic agents.
Conventional chemotherapies are generally immunosuppressive and so are withdrawn in certain situations, such as for patients who are pre-operative, post-operative, receiving radiation, terminally ill, elderly, or receiving adjuvant or neo-adjuvant thereapy. However, the compounds of the invention may still be used in such situations.
Therefore, in a further aspect of the invention the compounds of the invention are used to treat patients who may not be amenable to conventional chemotherapeutic agents, particularly those from whom conventional chemotherapeutic agents have been withdrawn.
A further aspect of the invention provides a kit of parts comprising: (i) Cyprodinil or a salt or ester thereof or a pharmaceutical composition comprising cyprodinil or a salt or ester thereof and a pharmaceutically acceptable excipient, diluent or carrier; (ii) apparatus for administering the compound or pharmaceutical composition; and (iii) instructions for use.
In one embodiment, the kit of parts additionally comprises a further chemotherapeutic agent. Preferably the further chemotherapeutic agent is selected from 17AAG, 17DMAG, 5FU, 7-hydroxystaurosporine (UCN-01), ABT888, Actinomycin D, Alsterpaullone, Axitinib, Aminoglutethimide, Amsacrine, Asparaginase, Azacitidine, AZD7762, Bay 11-7082, Belinostat, Bendamustine, Bexarotene, BIBW 2992, Bisindolylmaleimide I, Bleomycin, Bortezomib, Bosutinib, Busulfan, Canertinib , Capecitabine, Carboplatin, Carmustine, CDK4/6 IV, Chelerythrine Chloride, Chlorambucil, CHR-2863 (CHROMA), CHR-3531 (CHROMA), Chromomycin A3, CI-994, Cisplatin, Cladribine, Clofarabine, Clofibrate, CP690550, CXR1002 (APFO), Cyclopamine, Cyclophosphamide, Cytarabine, Dacarbazine, Dactinomycin, Dasatinib, Daunorubicin, DBZ, Decitabine, dk2 inhibitor II, DMFO, Docetaxel, Doramapimod, Dovitinib, Doxorubicin, Entinostat, Epirubicin, Erlotinib, Estramustine, Etoposide (V16- 213), Everolimus, Flavopiridol, fludarabine phosphate, Flutamide, GDC-0449, Gefitanib, Geldanamycin, Go 6976, GSK-1904529A, GW2580, GW501516, HA14-1, Hexamethylmelamine, Hydroxyurea (hydroxycarbamide), Ifosfamide, Interferon Alpha- 2a, Interferon Alpha-2b, lnterleukin-2, Leuprolide acetate (LHRH-releasing factor analog), Idarubicin, Imatinib, Ispinesib mesilate, Kenpaullone, K0143, KU-0058948, Lapatinib, Lenalidomide, Lestaurtinib, Lomustine, LY 2157299, LY294002, Masitinib, Mechlorethamine HCI (nitrogen mustard), Melphalan, Mercaptopurine (6-MP), Mesna, MTX, Mitoguazone, Mitomycin C, MK 1775, Mocetinostat, Mitoxantrone HCI, Nelarabine, Nilotinib, Nobiletin, Nogalamycin, NSC 625987, Nutlin-3, NVP-AEW541, NVP-TAE684 - TAE 684, Obatoclax mesylate, Octreotide, Olaparib (KU0059436), Oxaliplatin, Pentostatin, Plicamycin, Procarbazine HCI, Paclitaxel, Panobinostat, Pazopanib, PD 98059, PD173074, Pemetrexed, Perifosine, perillic acid, PLX4032, PLX4720, PPP (Picropodophyllin), Puromycin, Radicicol, Raltitrexed, Rapamycin, Ridaforolimus, Semustine, Streptozocin, Saracatinib, SB 431542, SB202190, SB203580, Sorafenib, SP600125, Sunitinib, Suramin, Tamoxifen CITRATE, Teniposide, Tandutinib, Tegafur , Temodal, Thalidomide, Thioguanine (6-TG), Thiotepa, Topotecan hydrochloride hydrate, Tozasertib, Trichostatin A, Tyrphostin AG 490, Tyrphostin AG 538, Tyrphostin AG879, U0126, Valproic acid, Vandetanib, Vatalanib, Vinblastine sulfate salt, Vincristine, Vindesine sulphate, Vinolrelbine ditartrate salt hydrate, Wortmannin, XAV 939, YM155, ZSTK474,4HC,6-[4-(2-Piperidin-1-ylethoxy)phenyl]-3-pyridin-4-ylpyrazolo[1,5- a]pyrimidine (Dorsomorphin), Etoposide, Gemcitabine, Mitoxanthrone and Vorinostat.
In one embodiment the further chemotherapeutic agent is a HSP inhibitor, such as geldanamycin, 17-AAG or 17-DMAG.
EXAMPLES
The following examples embody various aspects of the invention. It will be appreciated that the specific compounds used in the examples serve to illustrate the principles of the invention and are not intended to limit its scope. The following examples are described with reference to the accompanying figures in which:
Figure 1 - Dose response curves in OVCAR3 and HepG2 cell lines
Figure 1 shows percentage survival of (a) OVCAR3 cells and (b) HepG2 cells in response to increasing doses of Cyprodinil. Figure 2 - Isobologram for Cyprodinil and Geldanamycin combination
Figure 2 shows the shift in IC50 of Cyprodinil on PC3 cells when Cyprodinil is dosed in combination with the HSP90 inhibitor geldanamycin.
Figure 3 - Core aniline-pyrimidine moiety of Cyprodinil shared with kinase inhibitors such as Gleevec™
Figure 3 shows the how the core aniline-pyrimidine moiety of Cyprodinil is shared with kinase inhibitors such as Gleevec™
Figure 4 - PIM Kinase specificity of Cyprodinil
Figure 4 shows % inhibition of the PIM Kinases with increasing dose of Cyprodinil. Figure 5 - Cyprodinil activity in further kinases
Figure 5 shows % inhibition of further kinases with increasing dose of Cyprodinil.
Figure 6 - PC3 tumour volumes following treatment with Cyprodinil Figure 6 shows PC3 tumour volumes over time when dosed with Cyprodinil versus control.
Figure 7 - HT29 tumour volumes following treatment with Cyprodinil
Figure 7 shows HT29 tumour volumes over time when dosed with Cyprodinil at various levels versus control.
Figure 8 - PC3 tumour volumes following treatment with increased dose of
Cyprodinil
Figure 8 shows PC3 tumour volumes over time when dosed with Cyprodinil (CXR6024) at various levels versus control.
Figure 9 - Effect of Cyprodinil on ATP levels
Figure 9 shows ATP levels as a percentage of control with increasing dose of Cyprodinil and Pyrimethanil at 0, 45, 90 minutes and 4 hour time points.
Figure 10 - Effect of Cyprodinil on NADH Dehydrogenase activity
Figure 10 shows NADH Dehydrogenase activity represented by percentage of control with increasing dose of Cyprodinil and Pyrimethanil at 0, 30, 60, 90, 180 and 270 minute time points.
Figure 11 - RNA microarray experimental structure
Figure 11 shows the RNA microarray experimental structure using triplicates of each sample.
Example 1: selection of the compounds of the invention based on specific properties The following method details how the compounds of the invention were selected to arrive at those to be used for the treatment of diseases such as cancer.
The compounds of the invention were selected using the following method: Step 1: A database was produced of some 182 compounds registered for use as agro- or industrial chemicals with available dossiers regarding bioactivity and toxicity collated from sources including the UK's Advisory Committee on Pesticides (ACP), the US Environmental Protection Agency (EPA), the European Food Safety Authority (EFSA), the US Department of Health & Human Services' Agency for Toxic Substances and Disease Registry (ATSDR), Canada's Bureau of Chemical Safety and the International Programme on Chemical Safety (IPCS).
Table 13 details the 182 compounds contained in the database.
Step 2: A panel of 144 chemically diverse compounds were selected as being readily available from the database produced in (1).
Table 14 details the 144 chemically diverse compounds.
Step 3: The compounds from (2) were tested against 15 human tumour cell lines for cytotoxicity. Effects on cell survival at 72 hours and at 7 days at a single compound concentration (100μΜ) were measured. Tables 11 and 12 show cell proliferation data for the 144 compounds at 72 hours and 7 day exposure, respectively. The data is expressed as a percentage of untreated control cells, so 100 % means that there are the same number of cells as the control, less than 100 % indicates there are fewer cells than the control and greather than 100 % indicates that more cells than the control are present.
Cell assay methods Cell Culture
Test compounds were dissolved in D SO. Cells were seeded at a density of 500 cells per well in 75 μΙ of DME (10% FBS/2 mM Glutamine) media for 7 day incubation in 96- well plates or 5000 cells per well for 72 hrs incubations. The following day, 25 μΙ of test compounds, diluted 1 :25 in DMEM (10% FBS), were added to the wells to give a final compound concentrations of 100μΜ. Duplicate plates were assembled and then incubated for either 72 hrs or 7 days at 37°C/5% C02. No media/drug replacement during the incubation period was undertaken.
WST-1 Assay
Cell number was estimated using WST-1 reagent from Roche Applied Bioscience according to the manufacturer's instructions. Briefly 10μΙ of reagent was added to 100 μΙ of media and incubated until colour developed. The plates were read at wavelength 450nm on an appropriate absorbance plate reader. Data Analysis
Data from the plate reader in text format was entered into an excel spreadsheet. The data was converted into a percentage value of the untreated control wells using the following formula:
Figure imgf000022_0001
Step 4: The cytotoxicity data from (3) was used to rank compounds with respect to (a) global cytotoxicity; (b) selective cytotoxicity; and (c) published literature (i.e. a lack of published literature linking the compounds with cytotoxicity).
Step 5: Panel of 48 compounds chosen as the top-ranked compounds using the results of (4).
Step 6: Compounds selected under (5) were tested for cytotoxicity with compound dose response (see table 10) and the published toxicology data reviewed for (a) bioavailability (see table 3); (b) rat LD50 (see table 5); (c) Therapeutic Index (NOEL (No Observable Effects Limit): IC50 ratio) (table 2).
Step 7: Panel from (6) screened for effects on protein kinase activities. The results are shown in Table 8.
Step 8: Compounds selected for further development on basis of meeting at least criteria (1) and (2) (but preferably more) of the 7 criteria listed below:
Step 9: (optional) Kinase inhibition (such as PI -Kinase) by dose response results can be used to select specific compounds for further development (see Example 2 and figure
Step 10: Compounds selected under (8) and (9) will be tested further in xenograft studies (see Example 4).
The seven criteria for compound selection
1. Available Toxicoloqv/Toxicitv Data must be available Toxicological testing is a significant barrier in the pre-clinical development of pharmaceuticals, both in terms of cost and risk. Selection of molecules already tested for toxicity is therefore an attractive alternative.
Sufficient publically available safety pharmacology, toxicology and toxicity data should be available, as outlined in the ICH Safety Guidelines S9 available from http://www.ich.org/products/guidelines/safety/article/safety-guidelines, to support the compound's use in clinical trials.
2. Favourable Therapeutic Index - NOELICsn Ratio must be greater than 5
Current anticancer drugs give rise to significant side-effects at the dose levels given to patients. This is due to the fact that these traditional molecules function as cellular toxins, their anti-cancer properties arising from the toxic nature. The methodology described herein of selecting compounds shown to be of low toxicity will overcome this limitation and give rise to anti-cancer agents with lower toxicology liabilities.
An ideal pharmaceutical would exhibit efficacy in the absence of any toxicological or pharmacological side-effects at the therapeutic dose. A compound with these desirable properties is described as having a favourable therapeutic index or therapeutic window.
As an indicator of compounds with a potentially favourable Therapeutic Index we use the ratio of the rat "No Observed Effect Level" (NOEL) from toxicology studies and the lowest IC50 value of the compound on our cell line panel as a proxy. Compounds which have a larger ratio are more likely to inhibit the growth of tumour cells whilst having no observable side-effects on the animal.
Table 2 shows which of a selection of 48 compounds (as selected according to step 5 of the screening method) have an IC50/NOEL ratio of greater than five.
Table 2
Figure imgf000023_0001
Rat Rat
Ratio
Compound NOEUNOAEL NOEUNOAEL Min IC50
(mg/kg) (M) IC50/NOEL
(M)
Fenbutatin oxide 5.2 4.9 0.30 16.5
Fluazinam 10 21.5 0.20 107.5
3-lodo-2-propynyl N-
20 70.9 1.40 50.7 butylcarbamate
fludioxonil 5 20.2 2.50 8.1
Thiodicarb 5 14.1 4.30 3.3
Buprofezin 3.5 11.5 10.00 1.1
Dinocap 18.2 50.0 1.90 26.3
Thiophanate-methyl 80 233.9 8.00 29.2
Tolylfluanid 37 106.6 1.20 88.9
Triazoxid 0.25 1.0 0.60 1.7
Tebufenpyrad 7 21.0 0.03 698.6
Triflusulfuron-methyl 7 14.2 5.00 2.8
Fenpiclonil 30 126.6 12.00 10.5
Cyprodinil 10 44.4 5.00 8.9 brodifacoum 0.001 0.0 10.00 0.0 flocoumafen 0.0014 0.0 9.60 0.0
Difenoconazol 3.5 8.6 2.20 3.9 kresoxim-methyl 159 508.0 3.00 169.3
Azamethiphos 1 2.9 1.60 1.8
Benfuracarb 1.3 3.2 10.00 0.3
Flusilazole 1.5 4.8 10.00 0.5 fenpropathrin 1.5 4.3 15.00 0.3
Fenbuconazol 5.2 15.4 15.00 1.0
Transfluthrin 15 40.4 22.00 1.8
Fenazaquin 5 16.3 0.26 62.8
Fenoxycarb 80 265.8 10.20 26.1
Metconazole 2.5 7.8 17.00 0.5
Folicur 10.8 35.1 26.00 1.3
Methiocarb 1.3 5.8 2.60 2.2
Myclobutanil 15 51.9 25.00 2.1 cyfluthrin 10 23.1 17.00 1.4
Bromuconazol 14 37.1 30.00 1.2
Dimethomorph 3.75 9.7 15.00 0.6
Triazophos 0.29 0.9 22.00 0.0 phorate 0.05 0.2 26.00 0.0 irgarol 9.7 38.3 22.00 1.7
Pyriproxifen 24 74.8 15.70 4.8 tau-fluvalinate 1 2.0 14.00 0.1
Fluoroglycofen-ethyl 84 200.0 10.00 20.0
Esfenvalerate 15 35.7 9.58 3.7 Rat Rat
Min IC50 Ratio
Compound NOEL/NOAEL NOEL/NOAEL
(mg/kg) (M) (M) IC50/NOEL
Fenoxaprop-ethyl 6 16.6 43.00 0.4
cyhalofop-butyl 3 8.4 35.00 0.2
Flufenoxuron 12.5 25.6 8.00 3.2
Dimefuron 20 59.0 27.00 2.2
Isoproturon 50 242.7 20.00 12.1
Ethoprophos 0.1 0.4 43.31 0.0
3. Oral bioavailability preferred to be greater than 20% Oral bioavailability is an attractive property for pharmaceutical compounds. Oral dosing will mean patients do not have to travel to the hospital situation regularly during their treatment. This will not only save considerable resources at the treatment centre but also reduce patient comfort by reducing travel and in-patient time. Oral bioavailability is calculated by measuring the plasma concentration of the compound over time after administering the drug both orally (po) and intravenously (IV). The area under the curve (AUC) is measured for both modes of administration and is corrected for different dose levels for the different routes. The following formula is used for calculating oral bioavailability (F):
[AUC]po * Dose! IV [AUC]IV * Dose,o
Table 3 shows which of a selection of compounds has an oral bioavailability of >20%: Table 3
Figure imgf000025_0001
Compound Bioavailability
Thiodicarb >30%
Buprofezin 15-20%
Dinocap 40%
Thiophanate-methyl -70%
Tolylfluanid >60%
Triazoxide 20-43%
Tebufenpyrad -80%
Triflusulfuron-methyl 50%
Fenpiclonil -70%
Cyprodinil -75% brodifacoum 75% flocoumafen 70%
Difenoconazole -20% kresoxim-methyl Up to 60%
Azamethiphos >60%
Benfuracarb >80%
Flusilazole 2%
fenpropathrin 50%
Fenbuconazole 7%
Transfluthrin -85%
Fenazaquin 17%
Fenoxycarb >25%
Metconazole >80%
Folicur 90%
Methiocarb 0%
yclobutanil -50% cyfluthrin >60%
Bromuconazole -25%
Dimethomorph 90%
Triazophos -75%
Phorate 30% irgarol 0%
Pyriproxifen 7%
tau-fluvalinate 20-40%
Fluoroglycofen-ethyl 20%
Esfenvalerate -50%
Fenoxaprop-P-ethyl -50% cyhalofop-butyl -90%
Flufenoxuron 50%
Dimefuron 30%
Isoproturon 85%
Ethoprophos 95% 4. When data is available it is preferred that the LDgn be greater than 500 mg/kq
The selection approach described is to select compounds which have undergone extensive toxicological testing and have been deemed of low toxicity and safe for release to the environment as either agrochemicals or for use in other industries.
One measure of toxicity is the Lethal Dose 50 (LD50). This is the concentration of compounds which, when dosed to a test population, causes the death of 50 % of the test population within a set time period. LD50 was introduced in the 1920's by Trevan (The error of determination of toxicity. Proc Roy Soc 1927;101B:483-514) as a measure of toxicity but has been largely replace the Maximum Tolerated Dose or MTD. MTD is the highest concentration at which the death rate in a dosed population is no greater than the death rate in a control population According to the classification of Hodge and Sterner (J Ind Hyg Toxicol. 1949 Mar;31 (2):79-92), the toxicity of molecules can be classified according to the table below. In contrast with current anti-cancer agents which are known toxic agents, the compounds selected are of no greater than "low toxicity", with oral LD50 values in the rat no lower than 500 mg/kg. Table 4 details the different measures of toxicity of compounds. Table 5 details which of a selection of compounds have an oral LD50 in the rat of over 500mg/kg.
Table 4
Figure imgf000027_0001
Table 5
Figure imgf000028_0001
Myclobutanil -2000
cyf!uthrin 600-1200 (r), >100 (d)
Bromuconazole 300-400
Dimethomorph 3900
Triazophos 65
Phorate 3
irgarol 2000
Pyriproxifen >5000
tau-fluvalinate >3000
Fluoroglycofen-ethyl 1480
Esfenvalerate -200
Fenoxaprop-P-ethyl -2500
cyhalofop-butyl >5000
F!ufenoxuron 3000
Dimefuron 2000
Isoproturon 1826
Ethoprophos 33
5. Preferred that in vivo half-life greater than 4 hours For a compound to exert its biological effect, it is clear that the compound must be exposed to its molecular target. Should a compound be metabolised or excreted rapidly then compound concentration and so pharmacological effect would naturally fall too. In order to circumvent these issues, medicinal chemistry manipulates the chemical structure of the molecule in order to improve resistance to metabolism and excretion.
Compounds are selected from a library partly on the basis of in vivo half-life measured during development of the compounds as agro-industrial chemicals. In vivo half-life is a measure of the rate at which a compound is excreted or metabolised. The change in plasma concentration of a compound following dosing is described by the equation:
•t - Where Ct is the concentration at a time t, C0 represents the concentration at t = 0, Ke is the rate constant of elimination and t is the chosen time point. The half-life can then be calculated using the relationship between rate constant and half-life using the formula: t1/2 = In2 / Ke
It is clearly desirable not to have the compound eliminated rapidly from the plasma in order to increase the likely efficacy of the compound in vivo. Our threshold of 4 hours means that if peak dosing were 4 fold of the IC50 level, then compound levels would be at least the IC50 level for 8 hours out of 24 and therefore dosing would be a convenient 3 times per day. Table 6 details which of a selection of compounds have a half-life of >4 hours.
Table 6
Figure imgf000030_0001
Compound Half-Life
Fenbuconazol 15 hrs
Transfluthrin < 48 hrs
Fenazaquin 25 hrs
Fenoxycarb ~24hrs
Metconazole ND
Folicur 40hrs
Methiocarb ND
Myclobutanil 5.3hrs, 25.7 hrs
cyfluthrin <24hrs
Bromuconazol 9 - 90 hrs
Dimethomorph 3 hrs
Triazophos ND
phorate >7 days
irgarol ND
Pyriproxifen 2-8hrs, 23-35hrs
tau-fluvalinate ~24hrs
Fluoroglycofen-ethyl 12-37hrs
Esfenvalerate ~12hrs
Fenoxaprop-ethyl 10 hrs, 3 days
cyhalofop-butyl ~3h
Flufenoxuron ND
Dimefuron < 72hrs
Isoproturon 8h
Ethoprophos < 6 hrs
6. Preferred that compounds exhibit activity on Kinase or other molecular target
In order to direct pre-clinical development it is useful to have an idea of which tumour types may be best targeted with the molecule. Some of this information can be learned from testing the compound for effects on cellular proliferation on a panel of tumour cell lines. An alternative approach is to test the molecules for activity, inhibitory or activatory, on to use in vitro assays of molecular targets such as protein kinases, GPCRs, enzymes or other cellular machinery.
It is preferable that the compounds have a known mode of action. One aspect of this could be inhibitory activity of a compound against a member(s) of a panel of protein kinases known to be implicated in cancer biology. Such a panel could be comprised of the protein kinases shown in table 7:
Table 7: A panel of kinases known to be implicated in cancer biology Aurora A ERK2 JAK2 PIM3 S6K1
Aurora B FGFR1 LCK PKB alpha SGK
CDK2/cyclinA HER4 MKK1 PKC alpha SRC
CHK1 IGF1R NEK2A PLK1 TBK1
CHK2 IKK beta PDK1 ROCKII TRKA
EPH-A2 IKK epsilon PIM1 RSK2 VEGFR1
Kinase assay methods
Kinase assays were performed as described below for each individual kinase except the test compound was dissolved in DMSO and added to reaction mix to give final concentrations of 10μΜ and 100μΜ
MAPK2 ERK2 assay MAPK/ERK2 (5-20 mU diluted in 50 mM Tris pH 7.5, 0.1 mM EGTA, 0.1 mM Na3V04, 0.1% β-mercaptoethanol, 1 mg/ml BSA) is assayed against MBP in a final volume of 25.5 μΙ in 25 mM Tris pH 7.5, 0.1 mM EGTA, 0.33 mg/ml MBP, 10 mM magnesium acetate and 0.05 mM [33ρ-γ-ΑΤΡ](500 -1000 cpm/pmole) and incubated for 30 min at room temperature. Assays are stopped by addition of 5 μΙ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid.
MKK1 assay This is a two-step assay where inactive MAPK (0.06 mg/ml) is activated by MKK1 (diluted in 25 mM Tris, 0.1 mM EGTA, 0.1% β-mercaptoethanol, 0.01% Brij35, 1 mg/ml BSA) in 25.5 μΙ containing 25 mM Tris, 0.1 mM EGTA, 0.01% Brij35, 10 mM magnesium acetate and 0.005 mM ATP. After incubating at room temperature for 30 min, 5 μΙ from the first reaction is pipetted into 20 μΙ of the second reaction mix containing (final concentration) 25 mM Tris pH 7.5, 0.1 mM EGTA, 0.1 mM Na3V04, 0.66 mg/ml myelin basic protein (MBP), 10 mM magnesium acetate and 0.05 mM [33ρ-γ-ΑΤΡ] (500 -1000 cpm/pmole) and incubated for 30 min at room temperature. Assays are stopped by addition of 5 μΙ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid.
PKCa assay PKCa (5-20 mU diluted in 20 mM Hepes pH 7.4, 0.03% Triton X-100) is assayed against Histone H1 in the presence of PtdSerine and DAG (0.1 mg/ml. and 10 pg/ml) and 0.1 mM CaCI2. The assay is carried out in a final volume of 25.5 μΙ containing 20 mM Hepes pH 7.4, 0.03% Triton X-100, 0.1 mg/ml Histone H1, 10 mM magnesium acetate and 0.02 mM[33p-Y-ATP] (50-1000 cpm/pmole) and incubated for 30 min at room temperature. Assays are stopped by addition of 5 μΙ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid.
PtdSer/DAG preparation:- PtdSer stock = 10 mg/ml in MeOH/Chloroform (1 :2). Dry down required amount. Resuspend in appropriate volume of 10 mM Hepes pH 7.4. Vortex and briefly sonicate. (2 x 10-15 seconds at 10-15 seconds apart). DAG stock = 10 mg/ml in MeOH/chloroform (1:2). Dry down required amount. Add sonicated PtdSer solution. Vortex and sonicate. PDK1 assay
PDK1 (5-20 mU diluted in 50 mM Tris pH 7.5, 0.05% β-mercaptoethanol, 1 mg/ml BSA) is assayed against PDKtide (KTFCGTPEYLAPEVRREPRILSEEEQ-EMFRDFDYIADWC) in a final volume of 25.5 μΙ containing 50 mM Tris pH 7.5, 0.05% β-mercaptoethanol, 100 μΜ substrate peptide, 10mM magnesium acetate and 0.02 mM [33ρ-γ-ΑΤΡ] (50-1000 cpm/pmole) and incubated for 30 min at room temperature Assays are stopped by addition of 5 μΙ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid. APH-PKBa-S473D assay
APH-PKBa-S473D (5-20mU diluted in 50 mM Tris pH 7.5, 0.1 mM EGTA, 0.1% β- mercaptoethanol, 1 mg/ml BSA) is assayed against a modified Crosstide peptide GRPRTSSFAEGKK in a final volume of 25.5 μΙ containing 50mM Tris pH 7.5, 0.05% β- mercaptoethanol, 30 μΜ substrate peptide, 10 mM magnesium acetate and 0.005 mM [33ρ-γ-ΑΤΡ] (50-1000 cpm/pmole) and incubated for 30 min at room temperature. Assays are stopped by addition of 5 μΙ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid. SGK assay
SGK (5-20mU diluted in 20 mM MOPS pH 7.5, 1mM EDTA, 0.01% Brij35, 5% glycerol, 0.1% β-mercaptoethanol, 1 mg/ml BSA) is assayed against a modified Crosstide peptide GRPRTSSFAEGKK in a final volume of 25.5 μΙ containing 8 mM MOPS pH 7.0, 0.2 mM EDTA, 30 μΜ substrate peptide, 10 mM magnesium acetate and 0.02 mM [33ρ-γ-ΑΤΡ] (50-1000 cpm/pmole) and incubated for 30 min at room temperature. Assays are stopped by addition of 5 μΙ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid.
S6K1/ P70 S6K assay S6K1/P70 S6K (5-20 mU diluted in 20 mM MOPS pH 7.5, 1 mM EDTA, 0.01% Brij35, 5% glycerol, 0.1% β-mercaptoethanol, 1 mg/ml BSA) is assayed against substrate peptide (KKRNRTLTV) in a final volume of 25.5 μΙ containing 8 mM MOPS pH 7.0, 0.2 mM EDTA, 0.1 mM substrate peptide, 10 mM magnesium acetate and 0.02 mM [33ρ-γ- ATP] (50-1000 cpm/pmole) and incubated for 30 min at room temperature. Assays are stopped by addition of 5 μΙ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid.
ROCK-II (ROKa) assay ROCK-II (ROKa) (5-20 mU diluted in 50 mM Tris pH 7.5, 0.1 mM EGTA, 0.1% β- mercaptoethanol, 1 mg/ml BSA) is assayed against Long S6 substrate peptide (KEAKEKRQEQIAKRRRLSSLRASTSKSGGSQK) in a final volume of 25.5 μΙ containing 50 mM Tris pH 7.5, 0.1 mM EGTA, 30 μΜ Long S6 substrate peptide, 10 mM magnesium acetate and 0.02 mM [33ρ-γ-ΑΤΡ] (50-1000 cpm/pmole) and incubated for 30 min at room temperature. Assays are stopped by addition of 5 μΙ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid.
CHK1 assay
CHK1 (5-20 mU diluted in 20 mM MOPS pH 7.5, 1 mM EDTA, 0.1% β-mercaptoethanol, 0.01% Brij-35, 5% glycerol, 1 mg/ml BSA) is assayed against CHKtide substrate peptide (KKKVSRSGLYRSPSMPENLNRPR) in a final volume of 25.5 μΙ containing 8 mM MOPS pH 7.0, 0.2 mM EDTA, 200 μΜ CHKtide, 10 mM magnesium acetate and 0.02 mM [33p- γ-ΑΤΡ](50-1000 cpm/pmole) and incubated for 30 min at room temperature Assays are stopped by addition of 5 μΙ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid. LCK assay
LCK (5-20 mU diluted in 20 mM MOPS pH 7.5, 1 mM EDTA, 0.01% Brij35, 5% glycerol, 0.1% β-mercaptoethanol, 1 mg/ml BSA) is assayed against Cdc2 peptide (KVEKIGEGTYGWYK) in a final volume of 25.5 μΙ containing 50 mM Tris pH 7.5, 0.1 mM EGTA, 0.1 mM Na3Vo4, Cdc2 peptide (0.25 mM), 10 mM magnesium acetate and 0.05mM [33ρ-γ-ΑΤΡ](500-1000 cpm/pmole) and incubated for 15 min at room temperature Assays are stopped by addition of 5 μΙ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid.
CDK2/cyclin A assay CDK2/cyclin A (5-20 mU diluted in 50 mM Hepes pH 7.5, 1 mM DTT, 0.02% Brij35, 100 mM NaCI) is assayed against Histone H1 in a final volume of 25.5 μΙ containing 50 mM Hepes pH7.5, 1 mM DTT, 0.02% Brij35, 100 mM NaCI, Histone H1 (1 mg/ml), 10 mM magnesium acetate and 0.02 mM [33ρ-γ-ΑΤΡ](500-1000 cpm/pmole) and incubated for 30 min at room temperature. Assays are stopped by addition of 5 μΙ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid.
NEK2a assay 5-20mU of NEK2a (diluted in 50mM Tris (pH 7.5), 0.1 mM EGTA, 1 mg/ml BSA, 0.1%, β- mercaptoethanol) is assayed against NEK2a peptide (RFRRSRRMI) in a final volume of 25.5μΙ containing 50mM Tris (pH 7.5), 0.1 mM EGTA, 0.01% Brij, 0.1%, β- mercaptoethanol, 300μΜ NEK2a peptide, 10 mM magnesium acetate and 0.05 mM [33p- γ-ΑΤΡ]( 500-1000 cpm/pmole) and incubated for 30 mins at room temperature. Assays are stopped by addition of 5μΙ of 0.5M (3%) orthophosphoric acid. Assays are harvested onto P81 Unifilter plates using a wash buffer of 50mM orthophosphoric acid.
MAPKAP-K1 b/RSK2 assay MAPKAP-K1 b (5-20 mU diluted in 20 mM MOPS pH 7.5, 1 mM EDTA, 0.01% Brij35, 5% glycerol, 0.1% β-mercaptoethanol, 1 mg/ml BSA) is assayed against substrate peptide (KKLNRTLSVA) in a final volume of 25.51 containing 50 mM Na- β-glycerophosphate (pH 7.5), 0.5 mM EDTA, 30 μΜ substrate peptide, 10 mM magnesium acetate and 0.05 mM [33ρ-γ-ΑΤΡ] (50-1000 cpm/pmole) and incubated for 30 min at room temperature. Assays are stopped by addition of 5 μΙ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid.
IKKb assay
5-20mU of IKKb (diluted in 50mM Tris (pH 7.5), 0.1 mM EGTA, 1mg/ml BSA, 0.1% β- mercaptoethanol) is assayed against substrate peptide (LDDRHDSGLDSMKDEEY) in a final volume of 25.5μΙ containing 50mM Tris (pH 7.5), 0.1 mM EGTA, 0.1%, fjmercaptoethanol, 300μΜ substrate peptide, 10 mM magnesium acetate and 0.005 mM [33ρ-γ-ΑΤΡ]( 500-1000 cpm/pmole) and incubated for 30 mins at room temperature. Assays are stopped by addition of 5μΙ of 0.5M (3%) orthophosphoric acid. Assays are harvested onto P81 Unifilter plates using a wash buffer of 50mM orthophosphoric acid.
Aurora B assay
Aurora B (5-20mU diluted in 50 mM Tris pH 7.5, 0.1 mM EGTA, 0.1% β- mercaptoethanol, 1 mg/ml BSA) is assayed against a substrate peptide (LRRLSLGLRRLSLGLRRLSLGLRRLSLG) in a final volume of 25.5 μΙ containing 50mM Tris pH 7.5, 0.05% B-mercaptoethanol, 300 μΜ substrate peptide, 10 mM magnesium acetate and 0.02 mM [33ρ-γ~ΑΤΡ] (50-1000 cpm/pmole) and incubated for 30 min at room temperature. Assays are stopped by addition of 5 μΙ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid.
CHK2 assay
CHK2 (5-20 mU diluted in 20 mM MOPS pH 7.5, 1 mM EDTA, 0.1% β-mercaptoethanol, 0.01% Brij-35, 5% glycerol, 1 mg/ml BSA) is assayed against CHKtide substrate peptide (KKKVSRSGLYRSPSMPENLNRPR) in a final volume of 25.5 μΙ containing 8 mM MOPS pH 7.0, 0.2 mM EDTA, 200 μΜ CHKtide, 10 mM magnesium acetate and 0.02 mM [33p- γ-ΑΤΡ](50-1000 cpm/pmole) and incubated for 30 min at room temperature Assays are stopped by addition of 5 μΙ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid.
Src assay Src (5-20mU diluted in 50 mM Tris pH 7.5, 0.1 mM EGTA, 0.1% β-mercaptoethanol, 1 mg/ml BSA) is assayed against a substrate peptide (KVEKIGEGTYGWYK) in a final volume of 25.5 μΙ containing 50mM Tris pH 7.5, 0.05% β-mercaptoethanol, 300 μΜ substrate peptide, 10 mM magnesium acetate and 0.05 mM [33ρ-γ-ΑΤΡ] (50-1000 cpm/pmole) and incubated for 30 min at room temperature. Assays are stopped by addition of 5 μΙ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid. PLK1 assay
PLK1 (5-20mU diluted in 50 mM Tris pH 7.5, 0.1 mM EGTA, 0.1% β-mercaptoethanol, 1 mg ml BSA, 100μΜ Vanadate) is assayed against a substrate peptide (ISDELMDATFADQEAKKK) in a final volume of 25.5 μΙ containing 50mM Tris pH 7.5, 0.05% β-mercaptoethanol, 10μΜ Vanadate, 300 μΜ substrate peptide, 10 mM magnesium acetate and 0.005 mM [33ρ-γ-ΑΤΡ] (50-1000 cpm/pmole) and incubated for 30 min at room temperature. Assays are stopped by addition of 5 μΙ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid
PIM1 assay
PIM1 (5-20mU diluted in 50 mM Tris pH 7.5, 0.1 mM EGTA, 0.1% β-mercaptoethanol, 1 mg/ml BSA) is assayed against a substrate peptide (RSRHSSYPAGT) in a final volume of 25.5 μΙ containing 50mM Tris pH 7.5, 0.05% β-mercaptoethanol, 300 μΜ substrate peptide, 10 mM magnesium acetate and 0.02 mM [33ρ-γ-ΑΤΡ] (50-1000 cpm/pmole) and incubated for 30 min at room temperature. Assays are stopped by addition of 5 μΙ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid.
PIM3 assay
PIM3 (5-20mU diluted in 50 mM Tris pH 7.5, 0.1 mM EGTA, 0.1% β-mercaptoethanol, 1 mg/ml BSA) is assayed against a substrate peptide (RSRHSSYPAGT) in a final volume of 25.5 μΙ containing 50mM Tris pH 7.5, 0.05% β-mercaptoethanol, 300 μΜ substrate peptide, 10 mM magnesium acetate and 0.02 mM [33ρ-γ-ΑΤΡ] (50-1000 cpm/pmole) and incubated for 30 min at room temperature. Assays are stopped by addition of 5 μΙ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid.
FGF-R1 assay
FGF-R1 (5-20mU diluted in 50 mM Tris pH 7.5, 0.1 mM EGTA, 1 mg/ml BSA) is assayed against a substrate peptide (Poly Glut Tyr) in a final volume of 25.5 μΙ containing 50mM Tris pH 7.5, 1 mg/ml substrate peptide, 10 mM magnesium acetate and 0.02 mM [33p-v- ATP] (50-1000 cpm/pmole) and incubated for 30 min at room temperature. Assays are stopped by addition of 5 μΙ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid.
EPH-A2 assay EPH-A2 (5-20mU diluted in 50 mM Tris pH 7.5, 0.1 mM EGTA, 1 mg/ml BSA) is assayed against a substrate peptide (Poly Glut Tyr) in a final volume of 25.5 μΙ containing 50mM Tris pH 7.5, 0.1 mg/ml substrate peptide, 10 mM magnesium acetate and 0.05 mM [33p- γ-ΑΤΡ] (50-1000 cpm/pmole) and incubated for 30 min at room temperature. Assays are stopped by addition of 5 μΙ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid.
IGF-1R assay
IGF-1R (5-20mU diluted in 50 mM Tris pH 7.5, 0.1 mM EGTA, 1 mg/ml BSA) is assayed against a substrate peptide (KKKSPGEYVNIEFG) in a final volume of 25.5 μΙ containing 50mM Tris pH 7.5, 300μΜ substrate peptide, 10 mM magnesium acetate and 0.005 mM [33ρ-γ-ΑΤΡ] (50-1000 cpm/pmole) and incubated for 30 min at room temperature. Assays are stopped by addition of 5 μΙ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid.
VEG-FR assay
VEG-FR (5-20mU diluted in 50 mM Tris pH 7.5, 0.1 mM EGTA, 1 mg/ml BSA) is assayed against a substrate peptide (KKKSPGEYVNIEFG) in a final volume of 25.5 μΙ containing 50mM Tris pH 7.5, 300μΜ substrate peptide, 10 mM magnesium acetate and 0.02 mM [33p-y-ATP] (50-1000 cpm/pmole) and incubated for 30 min at room temperature. Assays are stopped by addition of 5 μΙ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid.
TBK1 (DU12569) assay
TBK1 (DU12569) (5-20mU diluted in 50 mM Tris pH 7.5, 0.1 mM EGTA, 1 mg/ml BSA) is assayed against a substrate peptide (KKKKERLLDDRHDSGLDSMKDEE) in a final volume of 25.5 μΙ containing 50mM Tris pH 7.5, 300μΜ substrate peptide, 10 mM magnesium acetate and 0.05 mM [33ρ-γ-ΑΤΡ] (50-1000 cpm/pmole) and incubated for 30 min at room temperature. Assays are stopped by addition of 5 μΙ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid.
IKKepsilon (DU14231) assay
5-20mU of IKKepsilon (DU14231)(diluted in 50mM Tris (pH 7.5), 0.1mM EGTA, 1mg/ml BSA) is assayed against MBP in a final volume of 25.5μΙ containing 50mM Tris (pH 7.5), 0.1 mM EGTA, 0.33mg/ml MBP, 10 mM magnesium acetate and 0.05 mM [33ρ-γ-ΑΤΡ]( 500-1000 cpm/pmole) and incubated for 30 mins at room temperature. Assays are stopped by addition of 5μΙ of 0.5M (3%) orthophosphoric acid. Assays are harvested onto P81 Unifilter plates using a wash buffer of 50mM orthophosphoric acid
HER4 assay HER4 (5-20 mU diluted in 50 mM Tris pH 7.5, 0.1 mM EGTA, 0.1% β-mercaptoethanol, 1 mg/ml BSA) is assayed against Poly Glut Tyr in a final volume of 25.5μΙ containing 50mM Tris pH 7.5, 0.1 mM EGTA, 1 mg/ml Poly Glut Tyr, 10 mM magnesium acetate and O.OOSmM [33ρ-γ-ΑΤΡ] (50-1000 cpm/pmole) and incubated for 30 min at room temperature. Assays are stopped by addition of 5 μΙ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid.
Aurora A assay Aurora A (5-20 mU diluted in 50 mM Tris pH 7.5, 0.1 mM EGTA, 0.1% β- mercaptoethanol, 1 mg/ml BSA) is assayed against
LRRLSLGLRRLSLGLRRLSLGLRRLSLG in a final volume of 25.5μΙ containing 50mM Tris pH 7.5, 0.1 mM EGTA, 0.3mM LRRLSLGLRRLSLGLRRLSLGLRRLSLG, 10 mM magnesium acetate and 0.005mM [33ρ-γ-ΑΤΡ] (50-1000 cpm/pmole) and incubated for 30 min at room temperature. Assays are stopped by addition of 5 μΙ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid.
JAK2 assay
JAK2 (5-20 mU diluted in 50 mM Tris pH 7.5, 0.1 mM EGTA, 0.05% β-mercaptoethanol, 1 mg/ml BSA) is assayed against PDKtide (KTFCGTPEYLAPEVRREPRILSEEEQ- EMFRDFDYIADWC) in a final volume of 25.5 μΙ containing 50 mM Tris pH 7.5, 0.1 mM EGTA, 0.05% β-mercaptoethanol, 100 μΜ substrate peptide, 10mM magnesium acetate and 0.005 mM [33ρ-γ-ΑΤΡ] (50-1000 cpm/pmole) and incubated for 30 min at room temperature Assays are stopped by addition of 5 μΙ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid.
TrkA assay TrkA (5-20mU diluted in 50 mM Tris pH 7.5, 0.1 mM EGTA, 10mM MnCI, 1 mg/ml BSA) is assayed against a substrate peptide (Poly Glut Tyr) in a final volume of 25.5 μΙ containing 50mM Tris pH 7.5, 0.1 mM EGTA, 1 mg/ml substrate peptide, 10 mM magnesium acetate and 0.02 mM [33ρ-γ-ΑΤΡ] (50-1000 cpm/pmole) and incubated for 30 min at room temperature. Assays are stopped by addition of 5 μΙ of 0.5 M (3%) orthophosphoric acid and then harvested onto P81 Unifilter plates with a wash buffer of 50 mM orthophosphoric acid.
Kinase Data Analysis Raw data was converted into percent inhibition by using the following equation: P nercen *t I rnh I.i ·b I iti ·on _ = - 1m0n0— ( / cPm est ~ cPmbaek—3round * 1. 00 1 I
X0?111 control ~ cPmb ekgrOund f
The percent inhibition data was then modeled to the 4-parameter logistic equation: Bottom— Top
Y = Bottom
f ECSOmu
{Compound Concentration1*'1*
The values of Bottom, Top, Hill and EC50 were changed in order to give the best values to fit the experimentally obtained data.
Compounds were tested for activity at concentrations of 100μΜ and 10μΜ. Compounds were declared active if the activity at 100 μΜ exceeded a threshold of 50% above or below control level. Table 8 indicates which kinases are activated or inhibited by certain compounds.
Table 8
Figure imgf000041_0001
Compound Activated Kinases Inhibited Kinases
PDK1, PKBAph, MKK1, Aurora B, EPH brodifacoum S6K1, CHK1 A2 VEGFR, SRC, JAK2, TRKA, RSK2,
IKKb PLK1 (okadaic acid), HER4
PKBAph, SGK, MKK1, Aurora B, flocoumafen S6K1, CHK1 VEGFR, PKCa, SRC, TRKA, RSK2,
IKKb, PLK1 (okadaic acid), HER4
Difenoconazol Aurora B, RSK2
kresoxim-methyl
Azamethiphos
Benfuracarb SGK, S6K1
Flusilazole
CHK1, VEGFR, PIM3,
fenpropathrin FGF R1
Fenbuconazol S6K1
Transfluthrin CHK1 KK1
Fenazaquin
Fenoxycarb MKK1
etconazole
Folicur
Methiocarb
Myclobutanil
cyfluthrin CHK1, CHK2
Bromuconazol MKK1
Dimethomo h
Triazophos SGK VEGFR
phorate SGK RSK2
irgarol
CHK1, VEGFR,
Pyriproxifen FGF R1
tau-fluvalinate CHK1, VEGFR
Fluoroglycofen-ethyl CHK1, VEGFR
CHK1,
Esfenvalerate FGF R1
Fenoxaprop-ethyl
PDK1, SGK, S6K1,
cyhalofop-butyl CHK1, Aurora B,
PKCa, RSK2, PIM3
Flufenoxuron VEGFR
Dimefuron
Isoproturon
Ethoprophos SGK 7. In vitro Cytotoxic/Cytostatic Activity - Preferred that Potency be less than 200 uM on at least one cell line
Compounds must exhibit broad activity against a panel of cell lines from tumour types of varying origins. Effects on cellular proliferation were measured against 14 cell lines representing carcinomas of the lung, prostate, breast, liver, colon, pancreas as well as sarcomas. Cells were treated with 8 concentrations of compound 300, 100, 30, 10, 3, 1 , 0.3, 0.1, 0.03 and 0.01 μΜ and cell number estimated at each concentration. Cell number was calculated as a percentage survival relative to untreated cells. EC50 was calculated by fitting the data to the 4-parameter logistic model:
Figure imgf000043_0001
Table 9 details the transformed cell lines.
Table 9
Figure imgf000043_0002
Table 10 shows the cytotoxicity data (μΜ EC50s) for selected compounds against 14 tumour cell lines over a 7 day exposure
Table 10: Cytotoxicity data (μΜ) for selected compounds against 14 tumour cell lines
Figure imgf000044_0001
43
Figure imgf000045_0001
Figure imgf000046_0001
5
Table 11- 72 hour Cell Proliferation Data at 100 μΜ Compound Concentration (data shown as cell number as % of untreated control)
Figure imgf000047_0001
46
Figure imgf000048_0001
Figure imgf000049_0001
Figure imgf000050_0001
Figure imgf000051_0001
Figure imgf000052_0001
Figure imgf000053_0001
Figure imgf000054_0001
Figure imgf000055_0001
Figure imgf000056_0001
Figure imgf000057_0001
Table 12 - 7 day Cell Proliferation Data at 100 μΜ Compound Concentration (data shown as cell number as % of untreated control)
Figure imgf000058_0001
Figure imgf000059_0001
Figure imgf000060_0001
Figure imgf000061_0001
Figure imgf000062_0001
Figure imgf000063_0001
Figure imgf000064_0001
Figure imgf000065_0001
Figure imgf000066_0001
Figure imgf000067_0001
Figure imgf000068_0001
Table 13-182 compounds contained in the database according to screening step 1
Figure imgf000069_0001
Figure imgf000070_0001
Figure imgf000071_0001
Table 14 - A panel of 144 compounds (chemically diverse) from screening step 2
Figure imgf000072_0001
Figure imgf000073_0001
Example 2 - Kinase inhibition by cyprodinil
Core anilino-pyrimidine moiety of CXR6024/Cyprodinil also features in some kinase inhibitors, notably the Abl inhibitor Gleevec™ (see figure 3).
CXR6024/Cyprodinil was screened for kinase inhibition at 10μΜ and 100μΜ against a panel of 30 protein kinases. Data is presented as % inhibition. The most potent activity was against PIM3. Table 15 shows the % inhibition against the 30 kinases using Cyprodinil at 10μΜ and 100μΜ. The kinase assay methods are shown in Example 1 (criteria 6 of the selection criteria).
Table 15
Figure imgf000074_0001
Subsequent dose response of CXR6024/Cyprodinil on the PIM kinase produced the IC50s shown in table 16 and figure 4.
Table 16
Figure imgf000074_0002
Other kinase inhibitors which feature aniline-pyrimidine moiety include molecules active against the following kinases:
CDK1, c-Kit, EGFR, Flt1, JNK, KDR, PDGFR and Syk.
Dose responses were performed against these kinases to check kinase inhibitory activity (figure 5). The only activity seen was a weak inhibition of Syk kinase, with an IC50 of around 300μΜ. Example 3 - Cyprodinil cytotoxic/cytostatic activity
Cyprodinil was part of a 144 compound library screened by CXR Biosciences for potential cytotoxic/cytostatic activity.
Briefly, cells were incubated with the compounds in a primary screen at a single concentration of 100μΜ in a panel of 15 cell lines (see Example 1, tables 11 and 12). Compounds which showed promising activity were advanced and used to generate EC50 values on a panel of 14 tumour cell lines, 2 non-transformed cell lines and also for activity against a confluent (non-dividing) non-transformed cell line.
Methods
Cell Culture
Test compounds were dissolved in DMSO and dilution series performed. Cells were seeded at a density of 500 cells per well in 75 μΙ of DMEM (10% FBS/2 mM G!utamine) media for 7 day incubation in 96-well plates. The following day, 25 μΙ of test compounds, diluted 1:25 in DMEM (10% FBS), were added to the wells to give final compound concentrations as detailed above. Duplicate plates were assembled and then incubated for 7 days at 37°C/5% C02. No media/drug replacement during the 7 day period was undertaken.
For drug combination experiments and 72 hour cytoxicity testing on day one cells were seeded at a density of 5000 cells per well in 75 μΙ of DMEM (10% FBS/2 mM Glutamine) media in 96-well plates. The following day, 12.5 μΙ of each test compound, diluted 1 :50 in DMEM (10% FBS), was added to the wells to give final compound concentrations. Duplicate plates were assembled and then incubated for 3 days at 37°C/5% C02.
WST-1 Assay
Cell number was estimated using WST-1 reagent from Roche Applied Bioscience according to the manufacturer's instructions. Briefly 10μΙ of reagent was added to 100 μΙ of media and incubated until colour developed. The plates were read at wavelength 450nm on an appropriate absorbance plate reader. Data Analysis
Data from the plate reader in text format was entered into an excel spreadsheet. The data was converted into a percentage value of the untreated control wells using the following formula:
Figure imgf000076_0001
Control
The %Survival data was then modelled to the 4-parameter logistic equation:
Figure imgf000076_0002
^Compound Concentration' u The values of Bottom, Top, Hill and EC50 were changed manually in order to give the best values to fit the experimentally obtained data.
The cell lines employed in this study are shown in Table 17. Table 17 - CXR Biosciences Cell Line Panel
Figure imgf000076_0003
EC50S on Cell Line Panel CXR6024/Cyprodinil was tested in a number of cell lines for anti-proliferative and cytotoxic effects. Cells were seeded at 400 cells per well in 96 well dishes and maintained in the presence of CXR6024/Cyprodinil for 7 days without change of media. Cell number was estimated using the WST-1 dye and was presented as percentage inhibition of growth.
Figure 1 shows specimen dose-response curves for OVCAR3 and HepG2 cell lines.
Typically CXR6024/Cyprodinil was active against all cell lines with EC50s varying by a factor of less than 10 across the tumour cell line panel. The compounds showed little if any specificity for non-transformed dividing cell lines over dividing tumour cell lines, the EC50s against WI38 and hTERT lines being 19 and 13 μΜ respectively, very similar to the 12 μΜ median EC50 of the dividing tumour cell lines. Interestingly the non- transformed cell line hTERT, when plated at near 100% confluence, shows marked resistance to CXR6024/Cyprodinil, having an EC50 of 125 μΜ, almost 10 fold the EC50 on the same cell line when dividing, indicating the compound may have some specificity for dividing cells over non-dividing cells.
Table 18 - EC50s (μΜ) for CXR6024/Cyprodinil on the cell line panel.
Figure imgf000077_0001
Combination Cell Culture PC3 cells were cultured and an IC50 for CXR6024/Cyprodinil was calculated in the presence of a number of concentrations of the HSP90 inhibitor geldanamycin. As seen in the isobologram of figure 2, the lower concentrations of geldanamycin resulted in a left shift of the IC50 for CXR6024/Cyprodinil indicating and increased efficacy of the compound.
Example 4 - Cyprodinil In Vivo Animal Models (Xenografts)
Cyprodinil was selected for advancement to in vivo animal models based on in vitro efficacy, selectivity, kinase inhibitory activity and absorption, distribution, metabolism, and excretion (ADME) criteria as shown in Table 19. Table 19
Figure imgf000078_0001
Methods Adult (6 - 8 weeks) female, athymic nude (nu/nu) mice were obtained from Charles River, UK. On arrival the mice were housed, up to 8 per cage, on sterile sawdust in sterile, solid-bottom, polypropylene cages. The cages were individually vented units attached to a Techniplast Slimline Air Handling Unit. This unit maintained 70-80 air changes per cage, per hour, through HEPA air filters. Bedding was changed once weekly in a laminar flow unit. The temperature was maintained within a target range of 19-23° C and relative humidity of the IVC within a range of 40-70%. Twelve-hour periods of light were cycled with twelve-hour periods of darkness.
Sterile RM1 diet (Special Diet Services Ltd., Stepfield, Witham, Essex, UK) was used. Deionised water was autoclaved prior to use and changed at least once a week. Mice were allowed water and diet ad libitum and were acclimatised for at least 5 days prior to the study start. The mice had SPF-status and the housing and changing system assured that the SPF- status was preserved during the study. Trained personnel under supervision handled the mice.
The animals were randomly allocated to groups on arrival, numbered appropriately and weighed prior to the start of the experiment. The first mouse assigned to a cage was individually identified by tail tattooing with the lowest number for that cage; the second mouse was assigned the second (lowest) number and so on. An experimental card was placed on each cage and showed the project licence code, test group, study number, sex and individual numbers of the mice within, and identified the Home Office Licensee. In addition, these cards were colour coded to correlate with the coding for the group.
PC3 and HT-29 cells were cultured in Dulbecco's Modified Eagle's Medium (DMEM) containing 10% heat inactivated foetal calf serum (FCS) supplemented with 2mM L- glutamine, penicillin (50 lU/ml) and streptomycin (50μg/ml). Cultures were incubated in a humidified incubator at 37°C, 5% C02, until sufficient cells were available to implant the mice. Cells were harvested, pooled, centrifuged, and re-suspended in cold medium. These were mixed with an equal volume of cold Matrigel, so that the tumour cell injection solution was a 50:50 mixture of tumour cells/medium and Matrigel. Tumour cells were injected in a volume of 100μΙ in a single flank only. HT-29 cells were injected at a density of 1.75 x 106 cells per flank. PC3 cells were injected at a density of 2.5 x 106 cells per flank. The cell/Matrigel suspension was aliquoted into pre-chilled Eppendorf tubes and kept on ice prior to injection. The period between preparation of the cell/Matrigel suspension and injection of tumour cells will not exceed two hours.
Mice were administered Test Compounds 72 hours following implantation of tumour cells. Mice harbouring PC3 tumour cells were dosed with CXR6024 (Cyprodinil) in an ethanol/PEG200/water vehicle. HT-29 bearing mice were dosed with CXR6024(Cyprodinil) in a corn oil vehicle. Compounds were administered daily for 28 days by oral gavage in a dose volume of 10ml/kg.
The bodyweight of each mouse was recorded at the time of tail tattooing, before implantation and daily prior to dosing.
Prior to the start of the study, all mice were observed to ensure that they were physically normal and that they exhibited normal activity. Only normal mice were allocated to the study. Following cell inoculation, each mouse was observed twice weekly and a general assessment of condition recorded in the study diary. Animals were terminated if tumours become ulcerated or if the Home Office Project License moderate severity limit is exceeded.
Any mouse requiring euthanasia during the study were killed by an approved method and tissues harvested according to Terminal Procedures (section 5.5). Information on intercurrent deaths was entered into the study diary and were reported to the Project Licence Holder as soon as possible. Tumour growth was measured twice weekly for the duration of the study following cell implantation once tumours become palpable. Tumour diameters were measured at four different sites - two lengths and two widths -using a digital slide gauge. This was increased to three lengths and three widths if the tumour is an irregular shape.
Tumour volumes were calculated using the formula: 4/3
Figure imgf000080_0001
4
Where d = mean diameter (n
Animals were terminated if tumour volumes exceed 1.44cm3 or if they become ulcerated.
On the day of termination tumour volumes were recorded and the mice were weighed and transferred to the post mortem room. The animals were killed by exposure to a rising concentration of C02.
Blood were harvested by cardiac puncture and transferred to heparinised tubes for plasma preparation. Tumours and livers were processed as follows:
Tumours
· Each tumour was removed and the weights and gross morphology recorded.
• The tumour were cut in half and one half placed in 10 % neutral buffered formalin (NBF) then prepwered for paraffin embedding (section 6.4) for possible future histochemical analysis.
• The remaining half were cut in two and each piece was placed in a separate cryovial and flash frozen. Cryovial 1 were labeled microarray and cryovial 2 were labeled
MS. These samples were stored at approximately - 80°C for possible future analysis. Liver • The liver was removed and the weight recorded.
• 2 slices of liver (approximately 2 mm strips) were taken, one from the left lobe and one from the median lobe. These were placed in 10 % neutral buffered formalin (NBF) then prepwered for paraffin embedding (section 6.4) for possible future histochemical analysis.
• 2 pieces of liver will also be placed in separate cryovials and flash frozen. Cryovial 1 were labeled 'Microarray' and cryovial 2 were labeled 'MS'. These samples were stored at approximately - 80°C for possible future analysis. Following removal into tubes suitable for plasma preparation, venous blood samples were mixed on a roller for 10 minutes. Red blood cells were removed by centrifugation at 2,000 - 3000 rpm for 10 minutes at 8 - 10 °C. The supernatant (plasma) were transferred to a second tube and stored at approximately -70 °C until required for analysis.
Plasma was analyzed using a Cobas Integra 400 analyzer. Kits purchased from Roche Diagnostics GmbH were used to measure ALT (cat # 20764957322), AST (cat # 20764949322) and ALP (cat # 03333752190). Plasma samples were stored appropriately until analysis.
Results
1. PC3 cells grown subcutaneously were dosed orally daily with CXR6024/Cyprodinil at 150 mg/kg. Dosing started c 1 week after implantation of cells. a. 83 % mice developed tumours compared to 100 % in the control group.
b. Average tumour size was c 50% of control group.
The PC3 tumour volumes for Cyprodinil dosed versus control over the duration of the study are shown in Figure 6.
2. HT29 cells were injected subcutaneously and dosed orally every day with CXR6024/Cyprodinil at 180, 250 and 400 mg/kg.
a. An inverse dose response was seen where the lowest doses were effective at 70% and 80% T/C (Treated tumor volume/Control tumour volume) respectively, b. 400 mg/kg dose gave greater than 100% T/C. The HT29 tumour volumes for Cyprodinil at various doses versus control over the duration of the study are shown in Figure 7.
3. PC3 cells were injected subcutaneoualy and CXR6024/Cyprodinil was dosed orally every day at 250 and 400 mg/kg when tumour volume was c 100mm3.
a. 250 mg/kg dosing was ineffective (150 mg/kg has been effective in non- established tumours)
b. 400 mg/kg active with T/C of c 66%.
The PC3 tumour volumes for Cyprodinil at various doses versus control over the duration of the study are shown in Figure 8. Example 5 - Cyprodinil Mode of Actions Studies
Effect on ATP levels
Following exposure to Test Items, the CellTitre-Glo Luminescent Cell Viability Assay to measure ATP content is performed according to the manufacturer's detailed instructions (Promega Corporation, Technical Bulletin No. 288, and Cell notes, Issue 10, 2004).
PC3 cells were cultured and then Cyprodinil/CXR6024 was added and ATP levels were meaured a time intervals afterward. Cyprodinil/CXR6024 had no observable effect on levels of cellular ATP over 4 hours of exposure to cells (see Figure 9).
Effect on NADH Dehydrogenase activity
PC3 cells were plated and cyprodinil added to the culture media. Cellular mitochondrial NADH utilising enzyme activities were subsequently estimated at various timepoints using the WST- reagent (as described in Example 3).
As shown in Figure 10, Cyprodinil/CXR6024 has a marked effect on the activity of these enzymes, in comparison to it's structural analogue pyrimethanil, suggesting that this activity may play some role in the cytotoxic/cytostatic activity of the moloecule. This assay requires that the cells be incubated for around 60 - 90 minutes for the WST-1 dye to be metabolised by the enzymes and this probably explains the apparent activity at timepoint zero.
Effect of CXR6024/Cvprodinil on Gene expression in PC3 cells
PC3 cells were treated for 24 hours with EC20 and EC50 concentrations (derived in 7 day exposure experiments) of CXR6024/Cyprodinil. RNA was extracted and changes in gene expression levels using Agilent microarray. Methods for microarray studies
The RNA microarray analysis involved labelled (one colour) RNA from triplicate samples of PC3 cell line treated vehicle, CXR6024 at EC2o and at EC50 as shown in Figure 11. RNA integrity was checked using the Agilent bioanalyser and the RNA nano lab chip kit according to the CXR method sheet 'Set-up and Running of Nanochip Assay for RNA Quality Control'.
500ng RNA was labelled prior to microarray hybridisation using the Quick Amp Labelling Kit - Two Color (Agilent* 5190-0442), according to the CXR method sheet entitled "Transcriptional Profiling using Standard Agilent 1 Colour Protocol" v2.
Agilent whole genome human microarray 4x44K (G4122F were hybridised, washed and then scanned on an Agilent microarray scanner according to the CXR method sheet entitled, "Microarray hybridisation and scanning".
Images from the scanner were processed using Agilent feature extraction software version 10.1.7. The data files were loaded into Rosetta resolver 6 software for tertiary analysis. The data were analysed to generate a "signature" gene list comprising the genes that are significantly regulated (p<0.001) according to the CXR method sheet entitled, "Microarray Data Entry and Signature List Production for One colour and Two- colour data in Resolver 6 Software". Signature lists of significantly altered genes were generated for the comparisons shown in Table 20. The lists were not filtered by fold change. The lists were named with the denominator name first followed by the test name and the number of genes in the list. Table 20
Figure imgf000084_0001
Results Tables 21 and 22 detail the genes identified as being changed beyond the threshold level of either 2-fold up or down regulation.
Table 21 - Genes Expression changes at EC2o concentration of CXR6024/Cyprodinil
Figure imgf000085_0001
Table 22 - Genes Expression changes at EC50 concentration of CXR6024/Cyprodinil
Figure imgf000086_0001
Example 6 - Preferred pharmaceufcal formulations and modes and doses of administration.
The compounds of the present invention may be delivered using an injectable sustained- release drug delivery system. These are designed specifically to reduce the frequency of injections. An example of such a system is Nutropin Depot which encapsulates recombinant human growth hormone (rhGH) in biodegradable microspheres that, once injected, release rhGH slowly over a sustained period.
The compounds of the present invention can be administered by a surgically implanted device that releases the drug directly to the required site. For example, Vitrasert releases ganciclovir directly into the eye to treat CMV retinitis. The direct application of this toxic agent to the site of disease achieves effective therapy without the drug's significant systemic side-effects. Electroporation therapy (EPT) systems can also be employed for administration. A device which delivers a pulsed electric field to cells increases the permeability of the cell membranes to the drug, resulting in a significant enhancement of intracellular drug delivery. Compounds of the invention can also be delivered by electroincorporation (El). El occurs when small particles of up to 30 microns in diameter on the surface of the skin experience electrical pulses identical or similar to those used in electroporation. In El, these particles are driven through the stratum corneum and into deeper layers of the skin. The particles can be loaded or coated with drugs or genes or can simply act as "bullets" that generate pores in the skin through which the drugs can enter.
An alternative method of administration is the ReGel injectable system that is thermosensitive. Below body temperature, ReGel is an injectable liquid while at body temperature it immediately forms a gel reservoir that slowly erodes and dissolves into known, safe, biodegradable polymers. The active drug is delivered over time as the biopolymers dissolve.
Compounds of the invention can be introduced to cells by "Trojan peptides". These are a class of polypeptides called penetratins which have translocating properties and are capable of carrying hydrophilic compounds across the plasma membrane. This system allows direct targeting of compounds to the cytoplasm and nucleus, and may be non-cell type specific and highly efficient (Derossi et al., 1998, Trends Cell Biol., 8, 84-87).
Preferably, the pharmaceutical formulation of the present invention is a unit dosage containing a daily dose or unit, daily sub-dose or an appropriate fraction thereof, of the active ingredient. The compounds of the invention can be administered by any parenteral route, in the form of a pharmaceutical formulation comprising the active ingredient, optionally in the form of a non-toxic organic, or inorganic, acid, or base, addition salt, in a pharmaceutically acceptable dosage form. Depending upon the disorder and patient to be treated, as well as the route of administration, the compositions may be administered at varying doses.
In human therapy, the compounds of the invention can be administered alone but will generally be administered in admixture with a suitable pharmaceutical exipient diluent or carrier selected with regard to the intended route of administration and standard pharmaceutical practice.
The compounds of the invention can also be administered parenterally, for example, intravenously, intra-arterially, intraperitoneally, intra-thecally, intraventricularly, intrastemally, intracranially, intra-muscularly or subcutaneously, or they may be administered by infusion techniques. They are best used in the form of a sterile aqueous solution which may contain other substances, for example, enough salts or glucose to make the solution isotonic with blood. The aqueous solutions should be suitably buffered (preferably to a pH of from 3 to 9), if necessary. The preparation of suitable parenteral formulations under sterile conditions is readily accomplished by standard pharmaceutical techniques well-known to those skilled in the art.
Formulations suitable 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 water for injections, immediately prior to use. Extemporaneous injection solutions and suspensions may be prepared from sterile powders, granules and tablets of the kind previously described.
Generally, in humans, oral or parenteral administration of the compounds of the invention is the preferred route, being the most convenient.
For veterinary use, the compounds of the invention are administered as a suitably acceptable formulation in accordance with normal veterinary practice and the veterinary surgeon will determine the dosing regimen and route of administration which will be most appropriate for a particular animal.
The formulations of the pharmaceutical compositions of the invention may conveniently be presented in unit dosage form and may be prepared by any of the methods well known in the art of pharmacy. Such methods include the step of bringing into association the active ingredient 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.
Preferred unit dosage formulations are those containing a daily dose or unit, daily sub- dose or an appropriate fraction thereof, of an active ingredient. A preferred delivery system of the invention may comprise a hydrogel impregnated with a polypeptides, polynucleotides and antibodies of the invention, which is preferably carried on a tampon which can be inserted into the cervix and withdrawn once an appropriate cervical ripening or other desirable affect on the female reproductive system has been produced.
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. Example 7 - Exemplary pharmaceutical formulations
Whilst it is possible for compounds of the invention to be administered alone, it is preferable to present it as a pharmaceutical formulation, together with one or more acceptable carriers. The carrier(s) must be "acceptable" in the sense of being compatible with the compound of the invention and not deleterious to the recipients thereof. Typically, the carriers will be water or saline which will be sterile and pyrogen-free.
The following examples illustrate pharmaceutical formulations according to the invention in which the active ingredient is a compound of the invention.
Example 7A: Injectable Formulation Active ingredient 0.200 g
Sterile, pyrogen free phosphate buffer (pH7.0) to 10 ml
The active ingredient is dissolved in most of the phosphate buffer (35-40° C), then made up to volume and filtered through a sterile micropore filter into a sterile 10 ml amber glass vial (type 1) and sealed with sterile closures and overseals.
Example 7B: Intramuscular injection Active ingredient 0.20 g
Benzyl Alcohol 0.10 g
Glucofurol 75® 1.45 g
Water for Injection q.s. to 3.00 ml The active ingredient is dissolved in the glycofurol. The benzyl alcohol is then added and dissolved, and water added to 3 ml. The mixture is then filtered through a sterile micropore filter and sealed in sterile 3 ml glass vials (type 1).
Example 7C: Tablet
Active ingredient 100 mg
Lactose 200 mg
Starch 50 mg
Polyvinylpyrrolidone 5 mg
Magnesium stearate 4 mg
359 mg
Tablets are prepared from the foregoing ingredients by wet granulation followed by compression.
Example 7D: Ophthalmic Solution
Active ingredient 0.5 g
Sodium chloride, analytical grade 0.9 g
Thiomersal 0.001 g
Purified water to 100 ml
pH adjusted to7.5

Claims

Claims
1. A compound for use in medicine wherein the compound is Cyprodinil or a salt or ester thereof.
2. A pharmaceutical composition comprising Cyprodinil or a salt or ester thereof and a pharmaceutically acceptable excipient, diluent or carrier.
3. A method of treating cancer comprising administering to the patient an effective amount of Cyprodinil or a salt or an ester thereof.
4. A compound for use in the treatment of cancer wherein the compound is Cyprodinil or a salt or an ester thereof
5. The compound described in claim 4 comprising administering to a patient an effective amount of a Cyprodinil or a salt or an ester thereof.
6. Use of Cyprodinil or a salt or an ester thereof in the manufacture of a medicament for the treatment of cancer.
7. The method, compound or use of any of claims 3-6 wherein the cancer is selected from pancreatic cancer, ovarian cancer, breast cancer, prostate cancer, liver cancer, chondrosarcoma, lung cancer, head and neck cancer, colon cancer, sarcoma, leukaemia, lymphoma, myeloma, kidney cancer, thyroid cancer and brain cancers such as glioblastoma.
8. The method, compound or use of any of claims 1-7 wherein the patient is selected from humans, cows, dogs, cats, goats, sheep, and pigs.
9. The method, compound or use of any of claims 1-8 wherein the patient is a human.
10. The method, compound, use or composition of any previous claim additionally comprising a further chemotherapeutic agent.
11. The method, compound, use or composition of claim 10 wherein the further chemotherapeutic agent is selected from 17AAG, 17DMAG, 5FU, 7- hydroxystaurosporine (UCN-01), ABT888, Actinomycin D, Alsterpaulione, Axitinib, Aminoglutethimide, Amsacrine, Asparaginase, Azacitidine, AZD7762, Bay 11-7082, Belinostat, Bendamustine, Bexarotene, BIBW 2992, Bisindolylmaleimide I, Bleomycin, Bortezomib, Bosutinib, Busulfan, Canertinib , Capecitabine, Carboplatin, Carmustine, CDK4/6 IV, Chelerythrine Chloride, Chlorambucil, CHR-2863 (CHROMA), CHR-3531 (CHROMA), Chromomycin A3, CI-994, Cisplatin, Cladribine, Clofarabine, Clofibrate, CP690550, CXR1002 (APFO), Cyclopamine, Cyclophosphamide, Cytarabine, Dacarbazine, Dactinomycin, Dasatinib, Daunorubicin, DBZ, Decitabine, dk2 inhibitor II, DMFO, Docetaxel, Doramapimod, Dovitinib, Doxorubicin, Entinostat, Epirubicin, Erlotinib, Estramustine, Etoposide (V16-213), Everolimus, Flavopiridol, fludarabine phosphate, Flutamide, GDC-0449, Gefitanib, Geldanamycin, Go 6976, GSK-1904529A, GW2580, GW501516, HA14-1, Hexamethylmelamine, Hydroxyurea (hydroxycarbamide), Ifosfamide, Interferon Alpha-2a, Interferon Alpha-2b, lnterleukin-2, Leuprolide acetate (LHRH-releasing factor analog), Idarubicin, Imatinib, Ispinesib mesilate, Kenpaullone, K0143, KU-0058948, Lapatinib, Lenalidomide, Lestaurtinib, Lomustine, LY 2157299, LY294002, Masitinib, Mechlorethamine HCI (nitrogen mustard), Melphalan, Mercaptopurine (6-MP), Mesna, MTX, Mitoguazone, Mitomycin C, MK 1775, Mocetinostat, Mitoxantrone HCI, Nelarabine, Nilotinib, Nobiletin, Nogalamycin, NSC 625987, Nutlin-3, NVP-AEW541 , NVP-TAE684 - TAE 684, Obatoclax mesylate, Octreotide, Olaparib (KU0059436), Oxaliplatin, Pentostatin, Plicamycin, Procarbazine HCI, Paclitaxel, Panobinostat, Pazopanib, PD 98059, PD173074, Pemetrexed, Perifosine, perillic acid, PLX4032, PLX4720, PPP (Picropodophyllin), Puromycin, Radicicol, Raltitrexed, Rapamycin, Ridaforolimus, Semustine, Streptozocin, Saracatinib, SB 431542, SB202190, SB203580, Sorafenib, SP600125, Sunitinib, Suramin, Tamoxifen CITRATE, Teniposide, Tandutinib, Tegafur , Temodal, Thalidomide, Thioguanine (6-TG), Thiotepa, Topotecan hydrochloride hydrate, Tozasertib, Trichostatin A, Tyrphostin AG 490, Tyrphostin AG 538, Tyrphostin AG879, U0126, Valproic acid, Vandetanib, Vatalanib, Vinblastine sulfate salt, Vincristine, Vindesine sulphate, Vinolrelbine ditartrate salt hydrate, Wortmannin, XAV 939, YM155, ZSTK474,4HC,6-[4-(2-Piperidin-1- ylethoxy)phenyl]-3-pyridin-4-ylpyrazolo[1,5-a]pyrimidine (Dorsomorphin), Etoposide, Gemcitabine, Mitoxanthrone and Vorinostat.
12. The method, compound, use or composition of claim 10 wherein the further chemotherapeutic agent is a HSP inhibitor, such as geldanamycin, 17-AAG or 17-DMAG.
13. The compound, composition, method or use of claims 3-12 wherein the patient to be treated is not amenable to conventional chemotherapeutic agents.
14. A kit of parts comprising: (i) Cyprodinil or a salt or ester thereof or a pharmaceutical composition comprising Cyprodinil or a salt or ester thereof and a pharmaceutically acceptable excipient, diluent or carrier;
(ii) apparatus for administering the compound or pharmaceutical composition; and
(iii) instructions for use.
15. The kit of claim 14 additionally comprising a further chemotherapeutic agent.
16. The kit of claim 15 wherein the further chemotherapeutic agent is selected from 17AAG, 17DMAG, 5FU, 7-hydroxystaurosporine (UCN-01), ABT888, Actinomycin D, Alsterpaullone, Axitinib, Aminoglutethimide, Amsacrine, Asparaginase, Azacitidine, AZD7762, Bay 11-7082, Belinostat, Bendamustine, Bexarotene, BIBW 2992, Bisindolylmaleimide I, Bleomycin, Bortezomib, Bosutinib, Busulfan, Canertinib , Capecitabine, Carboplatin, Carmustine, CDK4/6 IV, Chelerythrine Chloride, Chlorambucil, CHR-2863 (CHROMA), CHR-3531 (CHROMA), Chromomycin A3, CI-994, Cisplatin, Cladribine, Clofarabine, Clofibrate, CP690550, CXR1002 (APFO), Cyclopamine, Cyclophosphamide, Cytarabine, Dacarbazine, Dactinomycin, Dasatinib, Daunorubicin, DBZ, Decitabine, dk2 inhibitor II, DMFO, Docetaxel, Doramapimod, Dovitinib, Doxorubicin, Entinostat, Epirubicin, Erlotinib, Estramustine, Etoposide (V16- 213), Everolimus, Flavopiridol, fludarabine phosphate, Flutamide, GDC-0449, Gefitanib, Geldanamycin, Go 6976, GSK-1904529A, GW2580, GW501516, HA14-1, Hexamethylmelamine, Hydroxyurea (hydroxycarbamide), Ifosfamide, Interferon Alpha- 2a, Interferon Alpha-2b, lnterleukin-2, Leuprolide acetate (LHRH-releasing factor analog), Idarubicin, Imatinib, Ispinesib mesilate, Kenpaullone, K01 3, KU-0058948, Lapatinib, Lenalidomide, Lestaurtinib, Lomustine, LY 2157299, LY294002, Masitinib, Mechlorethamine HCI (nitrogen mustard), Melphalan, Mercaptopurine (6-MP), Mesna, MTX, Mitoguazone, Mitomycin C, MK 1775, Mocetinostat, Mitoxantrone HCI, Nelarabine, Nilotinib, Nobiletin, Nogalamycin, NSC 625987, Nutlin-3, NVP-AEW541, NVP-TAE684 - TAE 684, Obatoclax mesylate, Octreotide, Olaparib (KU0059436), Oxaliplatin, Pentostatin, Plicamycin, Procarbazine HCI, Paclitaxel, Panobinostat, Pazopanib, PD 98059, PD173074, Pemetrexed, Perifosine, perillic acid, PLX4032, PLX4720, PPP (Picropodophyllin), Puromycin, Radicicol, Raltitrexed, Rapamycin, Ridaforolimus, Semustine, Streptozocin, Saracatinib, SB 431542, SB202190, SB203580, Sorafenib, SP600125, Sunitinib, Suramin, Tamoxifen CITRATE, Teniposide, Tandutinib, Tegafur , Temodal, Thalidomide, Thioguanine (6-TG), Thiotepa, Topotecan hydrochloride hydrate, Tozasertib, Trichostatin A, Tyrphostin AG 490, Tyrphostin AG 538, Tyrphostin AG879, U0126, Valproic acid, Vandetanib, Vatalanib, Vinblastine sulfate salt, Vincristine, Vindesine sulphate, Vinolrelbine ditartrate salt hydrate, Wortmannin, XAV 939, YM155, ZSTK474I4HC,6-[4-(2-Piperidin-1-ylethoxy)phenyl]-3-pyridin-4-ylpyrazolo[1,5- a]pyrimidine (Dorsomorphin), Etoposide, Gemcitabine, Mitoxanthrone and Vorinostat.
17. A compound substantially defined herein with reference to the description for use in medicine.
18. A compound substantially defined herein with reference to the description for use in the treatment of cancer.
19. A method of treating cancer substantially defined herein with reference to the description.
20. A pharmaceutical composition substantially defined herein with reference to the description
21. A kit of parts substantially defined herein with reference to the description.
PCT/GB2012/050934 2011-04-28 2012-04-27 Cyprodinil for use in medicine WO2012146933A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
GB1107197.4 2011-04-28
GBGB1107197.4A GB201107197D0 (en) 2011-04-28 2011-04-28 Compounds

Publications (1)

Publication Number Publication Date
WO2012146933A1 true WO2012146933A1 (en) 2012-11-01

Family

ID=44202958

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/GB2012/050934 WO2012146933A1 (en) 2011-04-28 2012-04-27 Cyprodinil for use in medicine

Country Status (2)

Country Link
GB (1) GB201107197D0 (en)
WO (1) WO2012146933A1 (en)

Cited By (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104394861A (en) * 2012-11-09 2015-03-04 中国中化股份有限公司 Use of benzonitrile compounds in preparing medicaments for treating tumor
US9200004B2 (en) 2013-01-15 2015-12-01 Incyte Holdings Corporation Thiazolecarboxamides and pyridinecarboxamide compounds useful as Pim kinase inhibitors
CN105267214A (en) * 2014-07-21 2016-01-27 沈阳化工研究院有限公司 Application of N-heteroaryl phenylamine compounds for preparation of antitumor drugs
US9278950B2 (en) 2013-01-14 2016-03-08 Incyte Corporation Bicyclic aromatic carboxamide compounds useful as Pim kinase inhibitors
US9540347B2 (en) 2015-05-29 2017-01-10 Incyte Corporation Pyridineamine compounds useful as Pim kinase inhibitors
US9556197B2 (en) 2013-08-23 2017-01-31 Incyte Corporation Furo- and thieno-pyridine carboxamide compounds useful as pim kinase inhibitors
US9580418B2 (en) 2014-07-14 2017-02-28 Incyte Corporation Bicyclic aromatic carboxamide compounds useful as Pim kinase inhibitors
EP3040073A4 (en) * 2013-08-27 2017-07-12 Sinochem Corporation Applications of substituent benzyloxy group containing ether compounds for preparing antitumor drugs
US9822124B2 (en) 2014-07-14 2017-11-21 Incyte Corporation Bicyclic heteroaromatic carboxamide compounds useful as Pim kinase inhibitors
US9862705B2 (en) 2015-09-09 2018-01-09 Incyte Corporation Salts of a pim kinase inhibitor
US9920032B2 (en) 2015-10-02 2018-03-20 Incyte Corporation Heterocyclic compounds useful as pim kinase inhibitors
CN110573154A (en) * 2017-02-06 2019-12-13 卡斯西部储备大学 Compositions and methods for modulating short-chain dehydrogenase activity
US10596161B2 (en) 2017-12-08 2020-03-24 Incyte Corporation Low dose combination therapy for treatment of myeloproliferative neoplasms
US11040027B2 (en) 2017-01-17 2021-06-22 Heparegenix Gmbh Protein kinase inhibitors for promoting liver regeneration or reducing or preventing hepatocyte death
JP2021520930A (en) * 2018-04-09 2021-08-26 ノボキュア ゲーエムベーハー Treatment of tumors with TTfields and Aurora kinase inhibitors
US11426420B2 (en) 2017-04-07 2022-08-30 Case Western Reserve University Inhibitors of short-chain dehydrogenase activity for treating coronary disorders
US11690847B2 (en) 2016-11-30 2023-07-04 Case Western Reserve University Combinations of 15-PGDH inhibitors with corticosteroids and/or TNF inhibitors and uses thereof

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005077181A1 (en) * 2004-02-12 2005-08-25 Bayer Cropscience Sa Fungicidal composition comprising a pyridylethylbenzamide derivative and a compound capable of inhibiting the spores germination or mycelium growth by acting on different metabolic routes
WO2010135581A1 (en) * 2009-05-20 2010-11-25 Cylene Pharmaceuticals, Inc. Pyrazolopyrimidines and related heterocycles as kinase inhibitors

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005077181A1 (en) * 2004-02-12 2005-08-25 Bayer Cropscience Sa Fungicidal composition comprising a pyridylethylbenzamide derivative and a compound capable of inhibiting the spores germination or mycelium growth by acting on different metabolic routes
WO2010135581A1 (en) * 2009-05-20 2010-11-25 Cylene Pharmaceuticals, Inc. Pyrazolopyrimidines and related heterocycles as kinase inhibitors

Non-Patent Citations (11)

* Cited by examiner, † Cited by third party
Title
"The error of determination of toxicity", PROC ROY SOC, vol. 101B, 1927, pages 483 - 514
DEROSSI ET AL., TRENDS CELL BIOL., vol. 8, 1998, pages 84 - 87
FRESHNEY: "Culture of Animal Cells: A Manual of Basic Technique", 1987, WILY-LISS
GIOVANELLA, J. NATL. CAN. INST., vol. 52, 1974, pages 921 - 30
HODGE; STERNER, J IND HYG TOXICOL., vol. 31, no. 2, March 1949 (1949-03-01), pages 79 - 92
KNAUF-BEITER ET AL., PLANT DISEASE, vol. 79, no. 11, 1995, pages 1098 - 1103
KUNZ ET AL., J. PHYTOPATHOLOGY, vol. 146, 1998, pages 231 - 238
LI ET AL., CLIN. EXP. METASTASIS, vol. 17, 1999, pages 423 - 9
MYRESIOTIS ET AL., PLANT DISEASE, vol. 91, no. 4, 2007, pages 407 - 413
PILKINGTON, ANTICANCER RES., vol. 17, 1997, pages 4107 - 9
RIBATTA, INTL. J. DEV. BIOI., vol. 40, 1999, pages 1189 - 97

Cited By (37)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104394861A (en) * 2012-11-09 2015-03-04 中国中化股份有限公司 Use of benzonitrile compounds in preparing medicaments for treating tumor
US9278950B2 (en) 2013-01-14 2016-03-08 Incyte Corporation Bicyclic aromatic carboxamide compounds useful as Pim kinase inhibitors
US9676750B2 (en) 2013-01-14 2017-06-13 Incyte Corporation Bicyclic aromatic carboxamide compounds useful as pim kinase inhibitors
US9849120B2 (en) 2013-01-15 2017-12-26 Incyte Holdings Corporation Thiazolecarboxamides and pyridinecarboxamide compounds useful as Pim kinase inhibitors
US11229631B2 (en) 2013-01-15 2022-01-25 Incyte Corporation Thiazolecarboxamides and pyridinecarboxamide compounds useful as Pim kinase inhibitors
US9550765B2 (en) 2013-01-15 2017-01-24 Incyte Holdings Corporation Thiazolecarboxamides and pyridinecarboxamide compounds useful as Pim kinase inhibitors
US9200004B2 (en) 2013-01-15 2015-12-01 Incyte Holdings Corporation Thiazolecarboxamides and pyridinecarboxamide compounds useful as Pim kinase inhibitors
US10265307B2 (en) 2013-01-15 2019-04-23 Incyte Corporation Thiazolecarboxamides and pyridinecarboxamide compounds useful as Pim kinase inhibitors
US10828290B2 (en) 2013-01-15 2020-11-10 Incyte Corporation Thiazolecarboxamides and pyridinecarboxamide compounds useful as pim kinase inhibitors
US10517858B2 (en) 2013-01-15 2019-12-31 Incyte Holdings Corporation Thiazolecarboxamides and pyridinecarboxamide compounds useful as PIM kinase inhibitors
US9556197B2 (en) 2013-08-23 2017-01-31 Incyte Corporation Furo- and thieno-pyridine carboxamide compounds useful as pim kinase inhibitors
US10000507B2 (en) 2013-08-23 2018-06-19 Incyte Corporation Furo- and thieno-pyridine carboxamide compounds useful as pim kinase inhibitors
EP3040073A4 (en) * 2013-08-27 2017-07-12 Sinochem Corporation Applications of substituent benzyloxy group containing ether compounds for preparing antitumor drugs
US9580418B2 (en) 2014-07-14 2017-02-28 Incyte Corporation Bicyclic aromatic carboxamide compounds useful as Pim kinase inhibitors
US9890162B2 (en) 2014-07-14 2018-02-13 Incyte Corporation Bicyclic aromatic carboxamide compounds useful as pim kinase inhibitors
US9822124B2 (en) 2014-07-14 2017-11-21 Incyte Corporation Bicyclic heteroaromatic carboxamide compounds useful as Pim kinase inhibitors
CN105267214A (en) * 2014-07-21 2016-01-27 沈阳化工研究院有限公司 Application of N-heteroaryl phenylamine compounds for preparation of antitumor drugs
US9802918B2 (en) 2015-05-29 2017-10-31 Incyte Corporation Pyridineamine compounds useful as Pim kinase inhibitors
US9540347B2 (en) 2015-05-29 2017-01-10 Incyte Corporation Pyridineamine compounds useful as Pim kinase inhibitors
US10336728B2 (en) 2015-09-09 2019-07-02 Incyte Corporation Salts of a Pim kinase inhibitor
US11066387B2 (en) 2015-09-09 2021-07-20 Incyte Corporation Salts of a Pim kinase inhibitor
US11505540B2 (en) 2015-09-09 2022-11-22 Incyte Corporation Salts of a Pim kinase inhibitor
US9862705B2 (en) 2015-09-09 2018-01-09 Incyte Corporation Salts of a pim kinase inhibitor
US9920032B2 (en) 2015-10-02 2018-03-20 Incyte Corporation Heterocyclic compounds useful as pim kinase inhibitors
US10450296B2 (en) 2015-10-02 2019-10-22 Incyte Corporation Heterocyclic compounds useful as Pim kinase inhibitors
US11053215B2 (en) 2015-10-02 2021-07-06 Incyte Corporation Heterocyclic compounds useful as Pim kinase inhibitors
US11690847B2 (en) 2016-11-30 2023-07-04 Case Western Reserve University Combinations of 15-PGDH inhibitors with corticosteroids and/or TNF inhibitors and uses thereof
US11040027B2 (en) 2017-01-17 2021-06-22 Heparegenix Gmbh Protein kinase inhibitors for promoting liver regeneration or reducing or preventing hepatocyte death
EP3576737A4 (en) * 2017-02-06 2021-04-21 Case Western Reserve University Compositions and methods of modulating short-chain dehydrogenase activity
JP2020514323A (en) * 2017-02-06 2020-05-21 ケース ウエスタン リザーブ ユニバーシティ Compositions and methods for modulating short chain dehydrogenase activity
US11718589B2 (en) 2017-02-06 2023-08-08 Case Western Reserve University Compositions and methods of modulating short-chain dehydrogenase
CN110573154A (en) * 2017-02-06 2019-12-13 卡斯西部储备大学 Compositions and methods for modulating short-chain dehydrogenase activity
US11426420B2 (en) 2017-04-07 2022-08-30 Case Western Reserve University Inhibitors of short-chain dehydrogenase activity for treating coronary disorders
US10596161B2 (en) 2017-12-08 2020-03-24 Incyte Corporation Low dose combination therapy for treatment of myeloproliferative neoplasms
US11278541B2 (en) 2017-12-08 2022-03-22 Incyte Corporation Low dose combination therapy for treatment of myeloproliferative neoplasms
JP7139448B2 (en) 2018-04-09 2022-09-20 ノボキュア ゲーエムベーハー Treatment of Tumors with TTFields and Aurora Kinase Inhibitors
JP2021520930A (en) * 2018-04-09 2021-08-26 ノボキュア ゲーエムベーハー Treatment of tumors with TTfields and Aurora kinase inhibitors

Also Published As

Publication number Publication date
GB201107197D0 (en) 2011-06-15

Similar Documents

Publication Publication Date Title
WO2012146933A1 (en) Cyprodinil for use in medicine
WO2012146936A1 (en) Pyrrolnitrin derivatives
Garcia-Echeverria et al. Drug discovery approaches targeting the PI3K/Akt pathway in cancer
Roh et al. p53-Reactivating small molecules induce apoptosis and enhance chemotherapeutic cytotoxicity in head and neck squamous cell carcinoma
KR101403100B1 (en) A synergistic pharmaceutical combination for the treatment of cancer
Das et al. Pro-survival autophagy: An emerging candidate of tumor progression through maintaining hallmarks of cancer
US20220110940A1 (en) Combinations for the treatment of neoplasms using quiescent cell targeting with egfr inhibitors
Fumarola et al. Expanding the arsenal of FGFR inhibitors: a novel chloroacetamide derivative as a new irreversible agent with anti-proliferative activity against FGFR1-amplified lung cancer cell lines
WO2016123054A2 (en) Kinase drug combinations and methods of use thereof
Favoni et al. Combined chemotherapy with cytotoxic and targeted compounds for the management of human malignant pleural mesothelioma
US20230011378A1 (en) Combination Therapy For Treatment Of Cancer
KR20210019422A (en) Cancer treatment method
Chen et al. Repressing MYC by targeting BET synergizes with selective inhibition of PI3Kα against B cell lymphoma
US11583509B2 (en) Compound for treating cancer and diabetes
US20220151976A1 (en) Targeting lasp1, eif4a1, eif4b and cxc4 with modulators and combinations thereof for cancer therapy
JP2020536890A (en) Combination of MDM2 Inhibitors and ERK Inhibitors to Treat Cancer
WO2018222135A1 (en) Cancer therapy
US20190117663A1 (en) Compositions and methods for treating cancer
FR2945747A1 (en) ANTITUMOR PHARMACEUTICAL COMPOSITION COMPRISING A CDKS INHIBITOR AND A CELL GROWTH INHIBITOR
CN115087463A (en) Combination drug
EP3200794A1 (en) A compound for anti-cancer therapy that acts by targeting gof mutant p53 and stimulates p73
A Pezuk et al. The antiproliferative and pro-apoptotic effects of methoxyamine on pediatric medulloblastoma cell lines exposed to ionizing radiation and chemotherapy
Raja GSK3B regulates epithelial-mesenchymal transition and cancer stem cell properties and is a novel drug target for triple-negative breast cancer
US20230226061A1 (en) Combination cancer therapy with dyrk1 inhibitors and inhibitors of the ras-raf-mek-erk (mapk) pathway
US10265294B2 (en) Compositions and methods for treating epithelial cancer

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: 12726471

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 12726471

Country of ref document: EP

Kind code of ref document: A1