WO2015166041A1 - Flavones as cyp1a1 inhibitors for the treatment of cancer - Google Patents

Flavones as cyp1a1 inhibitors for the treatment of cancer Download PDF

Info

Publication number
WO2015166041A1
WO2015166041A1 PCT/EP2015/059501 EP2015059501W WO2015166041A1 WO 2015166041 A1 WO2015166041 A1 WO 2015166041A1 EP 2015059501 W EP2015059501 W EP 2015059501W WO 2015166041 A1 WO2015166041 A1 WO 2015166041A1
Authority
WO
WIPO (PCT)
Prior art keywords
hydrogen
compound
alkoxy
aliphatic
compounds
Prior art date
Application number
PCT/EP2015/059501
Other languages
French (fr)
Inventor
Bhabatosh Chaudhuri
Original Assignee
De Montfort University
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 De Montfort University filed Critical De Montfort University
Publication of WO2015166041A1 publication Critical patent/WO2015166041A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D311/00Heterocyclic compounds containing six-membered rings having one oxygen atom as the only hetero atom, condensed with other rings
    • C07D311/02Heterocyclic compounds containing six-membered rings having one oxygen atom as the only hetero atom, condensed with other rings ortho- or peri-condensed with carbocyclic rings or ring systems
    • C07D311/04Benzo[b]pyrans, not hydrogenated in the carbocyclic ring
    • C07D311/22Benzo[b]pyrans, not hydrogenated in the carbocyclic ring with oxygen or sulfur atoms directly attached in position 4
    • C07D311/26Benzo[b]pyrans, not hydrogenated in the carbocyclic ring with oxygen or sulfur atoms directly attached in position 4 with aromatic rings attached in position 2 or 3
    • C07D311/28Benzo[b]pyrans, not hydrogenated in the carbocyclic ring with oxygen or sulfur atoms directly attached in position 4 with aromatic rings attached in position 2 or 3 with aromatic rings attached in position 2 only
    • C07D311/30Benzo[b]pyrans, not hydrogenated in the carbocyclic ring with oxygen or sulfur atoms directly attached in position 4 with aromatic rings attached in position 2 or 3 with aromatic rings attached in position 2 only not hydrogenated in the hetero ring, e.g. flavones
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Definitions

  • Cytochrome P450 (CYP) enzymes belong to a large family of detoxification enzymes that are present in different organs of the human body.
  • the human cytochrome P450 1 (CYP1 ) family consists of three members; namely CYP1A1 , CYP1A2 and CYP1 B1. While the mRNAs of CYP1 A1 and CYP1 B1 genes are known to be expressed in extra-hepatic tissues such as lung, ovary, prostate, kidney and mammary gland and the mRNA of CYP1 A2 gene is found in the liver amongst other tissues, protein expression of these genes is rarely seen in human tissues, under normal circumstances.
  • CYP1A1 expression is induced by poly-aromatic hydrocarbons (PAHs) which are found mainly in cigarette smoke, high-boiling fraction of crude oil, charred meat and vegetables.
  • PAHs poly-aromatic hydrocarbons
  • PAHs like 2, 3, 7, 8-tetrachlorodibenzo-p-dioxin (TCDD), benzo(a)pyrene, 7,12-dimethylbenz(a) anthracene (DMBA) can bind to the aromatic hydrocarbon receptor as ligands.
  • TCDD 7, 8-tetrachlorodibenzo-p-dioxin
  • DMBA 7,12-dimethylbenz(a) anthracene
  • the activated AhR performs the role of a transcription factor that is responsible for the induction of CYP1A1 genes.
  • PAHs also act as ideal substrates for CYP1A1 enzymes which efficiently hydroxylate the PAHs leading to the formation of carcinogenic entities from pro-carcinogenic molecules.
  • the PAHs appear to play a dominant role in the CYP1A1 mediated positive feedback mechanism that underlies the formation of carcinogenic substances capable of intercalating DNA. Hydroxylated PAHs are carcinogenic since they have great propensity to intercalate with double- stranded DNA and cause breaks in the double-stranded DNA. Hence all PAHs in general have tumour promoting properties.
  • CYP1A1 metabolises other xenobiotic compounds such as nitrogenous heterocyclics, caffeine, aromatic amines and an assortment of other compounds. Metabolism (biotransformation) of these compounds (i.e. pro-carcinogens) by CYP1A1 enzymes leads to the formation of carcinogenic substances.
  • CYP1A1 Induction of CYP1A1 therefore results in the biotransformation (metabolism) of PAHs which are potential pro-carcinogens to carcinogenic substances that can eventually lead to cancer.
  • CYP1A1 has been suggested to have a role in many cancers and appears to have a major role in the genesis of lung cancer.
  • Cigarette smoke which contains polyaromatic hydrocarbons (PAHs) and aromatic amine pro-carcinogenic compounds is particularly associated with the induction of CYP1A1 gene and the resultant metabolism of the PAHs in cigarette smoke is thought to be one of the primary causes of lung cancer.
  • PAHs polyaromatic hydrocarbons
  • aromatic amine pro-carcinogenic compounds is particularly associated with the induction of CYP1A1 gene and the resultant metabolism of the PAHs in cigarette smoke is thought to be one of the primary causes of lung cancer.
  • CYP1A1 gene induction occurs at the transcriptional level which, in turn, leads to the induction of CYP1A1 enzyme activity.
  • CYP1A1 enzyme is also known as aromatic hydrocarbon hydroxylase (AHH) since it metabolises PAHs through hydroxylation to form carcinogens.
  • AHH aromatic hydrocarbon hydroxylase
  • For induction of CYP1A1 gene transcription a transcription factor composed of two proteins is required.
  • Ligand (PAH)-bound aromatic (aryl) hydrocarbon receptor (AhR) forms a complex with the aryl hydrocarbon receptor nuclear translocator (Arnt) to form the active transcription factor.
  • AhR AhR is involved in various signalling pathways critical to cells' normal homeostasis, which includes physiological processes such as cell proliferation and differentiation, gene regulation, cell motility and migration, inflammation and others. Dysregulation of these processes is known to contribute to events such as tumour initiation, promotion, and progression.
  • R 1 to R 9 are independently selected from hydrogen, aliphatic, alkoxy, thioalkyi, alkylamino, halogen, hydroxy, cyano, nitro, hydroxyalkyl, alkylcarbonyloxy, alkoxycarbonyl, alkylcarbonyl, haloalkyl,
  • R 1 to R 9 are independently selected from hydrogen, aliphatic, halogen, hydroxy, alkoxy, thioalkyi, alkylamino or cyano, preferably hydrogen, hydroxyl, halogen or alkoxy, more preferably hydrogen, hydroxyl or alkoxy.
  • R 1 , R 5 and R 9 are hydrogen and one or more of R 2 , R 3 , R 4 , R 6 , R 7 and R 8 are selected from aliphatic, alkoxy, thioalkyi, alkylamino, halogen, hydroxy, cyano, nitro, hydroxyalkyl, alkylcarbonyloxy, alkoxycarbonyl, alkylcarbonyl, haloalkyl,
  • R x is hydrogen, aliphatic or aryl, with the remaining groups being hydrogen.
  • R 2 , R 3 , R 4 , R 6 , R 7 and R 8 are independently selected from aliphatic, halogen, hydroxy, alkoxy, thioalkyi, alkylamino or cyano, preferably hydroxyl, halogen or alkoxy, more preferably hydroxyl or alkoxy with the remaining groups being hydrogen.
  • the phenyl ring is preferably mono- or di-substituted, that is one or two of R 2 , R 3 and R 4 are preferably selected from aliphatic, alkoxy, thioalkyi, alkylamino, halogen, hydroxy, cyano, nitro, hydroxyalkyl, alkylcarbonyloxy, alkoxycarbonyl, alkylcarbonyl, haloalkyl, alkylsulfonylamino NH 2 , N0 2 , S0 2 R x , SOR x and COOR x , where R x is hydrogen, aliphatic or aryl, preferably one or two of R 2 , R 3 and R 4 are independently aliphatic, halogen, hydroxy, alkoxy, thioalkyi, alkylamino or cyano, preferably hydroxyl, halogen or alkoxy, more preferably hydroxyl or alkoxy and the remainder are hydrogen.
  • R 3 is selected from aliphatic, alkoxy, thioalkyi, alkylamino, halogen, hydroxy, cyano, nitro, hydroxyalkyl, alkylcarbonyloxy,
  • R x is hydrogen, aliphatic or aryl, preferably R 3 is selected from aliphatic, halogen, hydroxy, alkoxy, thioalkyi, alkylamino or cyano, preferably hydroxyl, halogen or alkoxy, more preferably hydroxyl or alkoxy; and R 2 and R 4 are independently both hydrogen.
  • R 2 and R 3 are independently selected from aliphatic, alkoxy, thioalkyi, alkylamino, halogen, hydroxy, cyano, nitro, hydroxyalkyl, alkylcarbonyloxy,
  • R x is hydrogen, aliphatic or aryl, preferably R 2 and R 3 are independently aliphatic, halogen, hydroxy, alkoxy, thioalkyi, alkylamino or cyano, preferably hydroxyl, halogen or alkoxy, more preferably hydroxyl or alkoxy; and R 4 is hydrogen.
  • R 2 is hydrogen and R 3 and R 4 are independently selected from aliphatic, alkoxy, thioalkyi, alkylamino, halogen, hydroxy, cyano, nitro, hydroxyalkyl,
  • the fused phenyl ring is preferably di- or tri-substituted, that is two or three of R 6 , R 7 and R 8 are independently selected from aliphatic, alkoxy, thioalkyi, alkylamino, halogen, hydroxy, cyano, nitro, hydroxyalkyl, alkylcarbonyloxy, alkoxycarbonyl, alkylcarbonyl, haloalkyl, alkylsulfonylamino NH 2 , N0 2 , S0 2 R x , SOR x and COOR x , where R x is hydrogen, aliphatic or aryl, preferably two or three of R 6 , R 7 and R 8 are independently aliphatic, halogen, hydroxy, alkoxy, thioalkyi, alkylamino or cyano, preferably hydroxyl, halogen or alkoxy, more preferably hydroxyl or alkoxy with the remainder of R 6 , R 7
  • the one or more alkoxy group is particularly one or more C 1-6 alkoxy group, more particularly one or more C 1-4 alkoxy group, more particularly a methoxy, ethoxy, propoxy or butoxy group, for example a methoxy or ethoxy group, most particularly methoxy.
  • R 1 to R 9 are independently selected from hydrogen, hydroxy or alkoxy.
  • R 1 , R 5 and R 9 are hydrogen.
  • the phenyl ring is preferably mono- or di-substituted, that is one or two of R 2 , R 3 and R 4 are preferably selected from alkoxy or hydroxy, and the remainder are hydrogen.
  • R 3 is alkoxy or hydroxyl, preferably alkoxy and R 2 and R 4 are selected from hydrogen, alkoxy or hydroxy, particularly R 2 and R 4 are independently both hydrogen or R 2 is alkoxy or hydroxyl and R 4 is hydrogen or R 2 is hydrogen and R 4 is alkoxy or hydroxyl.
  • R 1 to R 9 are independently selected from hydrogen, hydroxy or methoxy.
  • R 1 , R 5 and R 9 are hydrogen.
  • the phenyl ring is preferably mono- or di-substituted, that is one or two of R 2 , R 3 and R 4 are preferably selected from methoxy or hydroxy, and the remainder are hydrogen.
  • R 3 is methoxy or hydroxyl, preferably methoxy and R 2 and R 4 are selected from hydrogen, methoxy or hydroxy, particularly R 2 and R 4 are independently both hydrogen or R 2 is methoxy or hydroxyl and R 4 is hydrogen or R 2 is hydrogen and R 4 is methoxy or hydroxyl.
  • the fused phenyl ring is preferably di- or tri-substituted, that is two or three of R 6 , R 7 and R 8 are alkoxy or hydroxyl with the remainder of R 6 , R 7 , R 8 being hydrogen.
  • Preferably two or three of R 6 , R 7 and R 8 are methoxy or hydroxyl with the remainder of R 6 , R 7 or R 8 being hydrogen.
  • R 6 , R 7 and R 8 are each hydroxyl or alkoxy.
  • R 6 , R 7 and R 8 are each hydroxyl or alkoxy.
  • all three groups are methoxy as illustrated below.
  • R 1 to R 5 are as disclosed above.
  • aryl includes for example optionally substituted unsaturated monocyclic, bicyclic or tricyclic rings of up to 14 carbon atoms, such as phenyl, naphthyl and phenanthroline.
  • aryl may include partially saturated bicyclic rings such as tetrahydro-naphthyl.
  • the aryl group is phenyl, naphthy or phenanthroline.
  • aliphatic refers to a straight or branched chain hydrocarbon which is completely saturated or contains one or more units of unsaturation.
  • aliphatic may be alkyl, alkenyl or alkynyl, preferably having 1 to 12 carbon atoms, up to 6 carbon atoms or up to 4 carbon atoms.
  • alkyl relates to both straight chain and branched alkyl radicals of 1 to 12 carbon atoms, preferably 1 to 8 carbon atoms and most preferably 1 to 4 carbon atoms including but not limited to methyl, ethyl, n-propyl, isopropyl, n-butyl, sec- butyl, isobutyl, tert-butyl n-pentyl, n-hexyl, n-heptyl, n-octyl.
  • alkyl therefore relates to radicals comprising 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1 or 12 carbon atoms.
  • alkyl also encompasses cycloalkyl radicals of 3 to 12 carbon atoms, preferably 4 to 8 carbon atoms, and most preferably 5 to 6 carbon atoms including but not limited to cyclopropyl, cyclobutyl, CH 2 -cyclopropyl, CH 2 -cyclobutyl, cyclopentyl or cyclohexyl. Cycloalkyl groups may be optionally substituted or fused to one or more carbocyclyl or heterocyclyl group.
  • Haloalkyl relates to an alkyl radical preferably having 1 to 8 carbon atoms, preferably 1 to 4 carbon atoms substituted with one or more halide atoms for example CH 2 CH 2 Br, CF 3 or CCI 3 .
  • alkenyl means a straight chain or branched alkylenyl radical of 2 to 12 carbon atoms, preferably 2 to 6 carbon atoms and most preferably 2 to 4 carbon atoms, and containing one or more carbon-carbon double bonds and includes but is not limited to ethylene, n-propyl-1-ene, n-propyl-2-ene, isopropylene, etc..
  • alkynyl means a straight chain or branched alkynyl radical of 2 to 12 carbon atoms, preferably 2 to 6 carbon atoms and most preferably 2 to 4 carbon atoms, and containing one or more carbon-carbon triple bonds and includes but is not limited to ethynyl, 2-methylethynyl etc.
  • alkenyl and alkynyl therefore encompass radicals comprising 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1 or 12 carbon atoms.
  • alkoxy refers to an oxy group that is bonded to an alkyl group as defined herein.
  • An alkoxy is preferably a "C 1-8 alkoxy group", even more preferably a "Ci-6 alkoxy group” and more preferably a "Ci -4 alkoxy group”.
  • the alkoxy group particularly includes 1 , 2, 3 or 4 carbon atoms.
  • Particularly preferably alkoxy groups include methoxy, ethoxy, propyloxy and butyloxy.
  • Halogen means F, CI, Br or I, preferably F.
  • the compounds of formula (I) are one or more of
  • Preferred compounds of the invention listed above extend to the tautomers thereof, as well as (but not limited to) pharmaceutically acceptable salts, esters, amides, carbamates, carbonates, ureides or prodrugs thereof or a derivative optionally with one or more lipid groups (natural or synthetic) attached.
  • a prodrug is any compound that may be converted under physiological conditions or by solvolysis to any of the compounds of the invention or to a pharmaceutically acceptable salt of the compounds of the invention.
  • a prodrug may be inactive when administered to a subject but is converted in vivo to an active compound of the invention.
  • the compounds of the invention may contain one or more stereogenic (asymmetric) carbon atoms and may exist in racemic and optically active forms (enantiomers or diastereoisomers).
  • the first aspect of the invention includes all such enantiomers or diastereoisomers and mixtures thereof, including racemic mixtures.
  • Examples of pharmaceutically acceptable salts of the compounds of formulae (I), (IA) or (IB) include those derived from organic acids such as methanesulphonic acid, benzenesulphonic acid and p-toluenesulphonic acid, mineral acids such as hydrochloric and sulphuric acid and the like, giving methanesulphonate, benzenesulphonate, p- toluenesulphonate, hydrochloride and sulphate, and the like, respectively or those derived from bases such as organic and inorganic bases.
  • organic acids such as methanesulphonic acid, benzenesulphonic acid and p-toluenesulphonic acid
  • mineral acids such as hydrochloric and sulphuric acid and the like
  • methanesulphonate, benzenesulphonate, p- toluenesulphonate, hydrochloride and sulphate, and the like respectively or those derived from bases such as organic
  • suitable inorganic bases for the formation of salts of compounds for this invention include the hydroxides, carbonates, and bicarbonates of ammonia, lithium, sodium, calcium, potassium, aluminium, iron, magnesium, zinc and the like. Salts can also be formed with suitable organic bases.
  • bases suitable for the formation of pharmaceutically acceptable base addition salts with compounds of the present invention include organic bases which are nontoxic and strong enough to form salts.
  • Such organic bases are already well known in the art and may include amino acids such as arginine and lysine, mono-, di-, or trihydroxyalkylamines such as mono-, di-, and triethanolamine, choline, mono-, di-, and trialkylamines, such as methylamine, dimethylamine, and trimethylamine, guanidine; N- methylglucosamine; N-methylpiperazine; morpholine; ethylenediamine; N- benzylphenethylamine; tris(hydroxymethyl) aminomethane; and the like.
  • amino acids such as arginine and lysine, mono-, di-, or trihydroxyalkylamines such as mono-, di-, and triethanolamine, choline, mono-, di-, and trialkylamines, such as methylamine, dimethylamine, and trimethylamine, guanidine; N- methylglucosamine; N-methylpiperazine; morpholine; ethylenediamine; N
  • Salts may be prepared in a conventional manner using methods well known in the art. Acid addition salts of said basic compounds may be prepared by dissolving the free base compounds according to the first or second aspects of the invention in aqueous or aqueous alcohol solution or other suitable solvents containing the required acid. Where a compound of formula (I), (IA) or (IB) contain an acidic function, a base salt of said compound may be prepared by reacting said compound with a suitable base. The acid or base salt may separate directly or can be obtained by concentrating the solution eg. by evaporation. The compounds of this invention may also exist in solvated or hydrated forms. The compounds of the invention are provided for the prevention and/or treatment of cancer.
  • the compounds are provided for the prevention and/or treatment of cancer of the lung, colon, pancreas, liver and/or kidney.
  • the prevention and/or treatment of cancer means any effect which mitigates any damage, to any extent.
  • treatment means any amelioration of a disorder, disease, syndrome, condition, pain or a combination of two or more thereof.
  • prevention means to prevent the condition from occurring, lessening the severity of the condition or to prevent from deteriorating or getting worse for example by halting the progress of the disease without necessary ameliorating the condition.
  • the compounds of the first aspect of the invention are provided as inhibitors of CYP1 A1 which has been implicated in cancer of the lung, colon, pancreas, liver and/or kidney. Inhibition of CYP1A1 by the claimed compounds will allow the prevention and/or treatment of cancer.
  • R 1 , R 5 and R 9 are hydrogen, R 3 , R 6 and R 8 are alkoxy and R 2 , R 4 and R 7 are hydrogen, hydroxyl or alkoxy, preferably hydrogen or alkoxy.
  • R 1 , R 5 and R 9 are hydrogen
  • R 3 , R 6 and R 8 are methoxy
  • R 2 and R 7 are hydrogen, hydroxyl or methoxy, preferably hydrogen or methoxy.
  • Particularly preferred compounds of formula (IB) include
  • a second aspect of the invention provides a composition comprising a compound, in particular a novel compound according to the first aspect of the invention, in combination with a pharmaceutically acceptable excipient.
  • the pharmaceutically acceptable excipient may comprise a pharmaceutically acceptable carrier and/or pharmaceutically acceptable diluent. Suitable carriers and/or diluents are well known in the art and include
  • composition may be a mixed preparation of a composition or may be a combined preparation for simultaneous, separate or sequential use (including administration).
  • a pharmaceutical composition may be provided in unit dosage form, will generally be provided in a sealed container and may be provided as part of a kit. Such a kit would normally (although not necessarily) include instructions for use. It may include a plurality of said unit dosage forms.
  • compositions according to the invention for use in the aforementioned indications may be administered by any convenient method, for example by oral (including by inhalation), parenteral, mucosal (e.g. buccal, sublingual, nasal), rectal or transdermal administration and the compositions adapted accordingly.
  • Such compositions may be prepared by any method known in the art of pharmacy, for example by admixing the active ingredient with a carrier(s) or excipient(s) under sterile conditions.
  • the compounds can be formulated as liquids or solids, for example solutions, syrups, suspensions or emulsions, tablets, capsules and lozenges.
  • compositions adapted for oral administration may be presented as discrete units such as capsules or tablets; as powders or granules; as solutions, syrups or suspensions (in aqueous or non-aqueous liquids; or as edible foams or whips; or as emulsions).
  • suitable excipients for tablets or hard gelatine capsules include lactose, starch including maize starch or derivatives thereof, stearic acid or salts thereof, such as
  • a composition in the form of a capsule can be prepared using routine encapsulation procedures.
  • powders, granules or pellets containing the active ingredient can be prepared using standard carriers and then filled into a hard gelatin capsule; alternatively, a dispersion or suspension can be prepared using any suitable pharmaceutical carrier(s), for example aqueous gums, celluloses, silicates or oils and the dispersion or suspension then filled into a soft gelatin capsule.
  • suitable excipients for use with soft gelatine capsules include for example vegetable oils, waxes, fats, semi-solid, or liquid polyols etc.
  • compositions for oral administration may be designed to protect the active ingredient against degradation as it passes through the alimentary tract, for example by an outer coating of the formulation on a tablet or capsule.
  • a liquid formulation such as a solution or a syrup will generally consist of a suspension or solution of the compound or physiologically acceptable salt in a suitable aqueous or non- aqueous liquid carrier(s) for example water, ethanol, glycerine, sugars, polyethylene glycol or an oil.
  • a suitable aqueous or non- aqueous liquid carrier(s) for example water, ethanol, glycerine, sugars, polyethylene glycol or an oil.
  • suspensions oils e.g. vegetable oils
  • the formulation may also contain a suspending agent, preservative, flavouring or colouring agent.
  • compositions for nasal or oral administration may conveniently be formulated as aerosols, drops, gels and powders.
  • Aerosol formulations typically comprise a solution or fine suspension of the active substance in a physiologically acceptable aqueous or nonaqueous solvent and are usually presented in single or multidose quantities in sterile form in a sealed container, which can take the form of a cartridge or refill for use with an atomising device.
  • the sealed container may be a unitary dispensing device such as a single dose nasal inhaler or an aerosol dispenser fitted with a metering valve which is intended for disposal once the contents of the container have been exhausted.
  • the dosage form comprises an aerosol dispenser, it will contain a pharmaceutically acceptable propellant.
  • the aerosol dosage forms can also take the form of a pump- atomiser.
  • compositions adapted for nasal administration wherein the carrier is a solid include a coarse powder having a particle size for example in the range 20 to 500 microns which is administered in the manner in which snuff is taken, i.e. by rapid inhalation through the nasal passage from a container of the powder held close up to the nose.
  • suitable compositions wherein the carrier is a liquid, for administration as a nasal spray or as nasal drops, include aqueous or oil solutions of the active ingredient.
  • compositions adapted for topical administration may be formulated as ointments, creams, suspensions, lotions, powders, solutions, pastes, gels, sprays, aerosols or oils.
  • the compositions are preferably applied as a topical ointment or cream.
  • the active ingredient may be employed with either a paraffinic or a water-miscible ointment base.
  • the active ingredient may be formulated in a cream with an oil- in-water cream base or a water-in-oil base.
  • compositions adapted for topical administration to the eye include eye drops wherein the active ingredient is dissolved or suspended in a suitable carrier, especially an aqueous solvent.
  • Pharmaceutical compositions adapted for topical administration in the mouth include lozenges, pastilles and mouth washes.
  • Pharmaceutical compositions adapted for rectal administration may be presented as suppositories or enemas.
  • compositions for rectal or vaginal administration are conveniently in the form of suppositories (containing a conventional suppository base such as cocoa butter), pessaries, vaginal tabs, foams or enemas.
  • compositions suitable for buccal or sublingual administration include tablets, lozenges and pastilles, wherein the active ingredient is formulated with a carrier such as sugar and acacia, tragacanth, or gelatin and glycerin.
  • a carrier such as sugar and acacia, tragacanth, or gelatin and glycerin.
  • compositions suitable for transdermal administration include ointments, gels, patches and injections including powder injections.
  • compositions adapted for parenteral administration include aqueous and non-aqueous sterile injection solution which may contain anti-oxidants, buffers, bacteriostats and solutes which render the formulation substantially isotonic with the blood of the intended recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents.
  • Excipients which may be used for injectable solutions include water, alcohols, polyols, glycerine and vegetable oils, for example.
  • 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 (lyophilized) condition requiring only the addition of the sterile liquid carried, for example water for injections, immediately prior to use.
  • sterile liquid carried, for example water for injections, immediately prior to use.
  • Extemporaneous injection solutions and suspensions may be prepared from sterile powders, granules and tablets.
  • compositions may contain preserving agents, solubilising agents, stabilising agents, wetting agents, emulsifiers, sweeteners, colourants, odourants, salts, buffers, coating agents or antioxidants. They may also contain an adjuvant and/or therapeutically active agents in addition to the substance of the present invention.
  • Dosages of the substance of the present invention can vary between wide limits, depending upon a variety of factors including the disease or disorder to be treated, the age, weight and condition of the individual to be treated, the route of administration etc. and a physician will ultimately determine appropriate dosages to be used.
  • the dosage adopted for each route of administration when a compound of the invention is administered to adult humans is 0.001 to 500 mg/kg.
  • Such a dosage may be given, for example, from 1 to 5 times daily by bolus infusion, infusion over several hours and/or repeated administration.
  • the compositions may be administered in conjunction with one or more other therapeutically active agents, especially those effective for treating cancers (i.e. a chemotherapeutic agent).
  • chemotherapeutic agent may be, for example, mitoxantrone, Vinca alkaloids, such as vincristine and vinblastine, anthracycline antibiotics such as daunorubicin and doxorubicin, alkylating agents such as chlorambucil and melphalan, taxanes such as paclitaxel, anti-folates such as methotrexate and tomudex, epipodophyllotoxins such as etoposide, camptothecins such as irinotecan and its active metabolite SN-38 and DNA methylation inhibitors.
  • Vinca alkaloids such as vincristine and vinblastine
  • anthracycline antibiotics such as daunorubicin and doxorubicin
  • alkylating agents such as chlorambucil and melphalan
  • taxanes such as paclitaxel
  • anti-folates such as methotrexate and tomudex
  • the other active compound(s) may be incorporated in the same composition as the compounds of the present invention or they may be administered alongside the compounds of the present invention, e.g. simultaneously or sequentially.
  • the invention provides a kit of parts comprising a compound of the invention and another chemotherapeutic agent, optionally with instructions for use.
  • the compound of the first aspect of the invention may be administered by their addition to a food or drink.
  • the compounds of the first aspect of the invention are formulated into a powder or liquid for addition to food or drink and administration by these means.
  • the compounds of the first aspect will be formulated with an excipient or diluent but such excipient or diluent does not need to be pharmaceutically acceptable but instead should be acceptable for consumption.
  • a third aspect of the invention provides a process for the manufacture of a composition according to the second aspect of the invention.
  • the manufacture can be carried out by standard techniques well known in the art and involves combining a compound according to the first aspect of the invention and a pharmaceutically acceptable carrier or diluent.
  • the composition may be in any form including a tablet, a liquid, a capsule, and a powder or in the form of a food product, e.g. a functional food. In the latter case the food product itself may act as the pharmaceutically acceptable carrier.
  • the fourth aspect of the invention provides a method of preventing or treating cancer comprising administering a compound of the first aspect of the invention to a patient in need thereof.
  • the compounds of the present invention inhibit the conversion of pre- carcinogens into carcinogenic compounds thereby reducing or removing the risk of cancer.
  • the patient in need thereof does not therefore need to be suffering from cancer but can instead wish to reduce his or her risk of cancer.
  • the fourth aspect of the invention provides a method of reducing the risk of developing cancer, comprising administering a compound of the first aspect of the invention.
  • a person wishing to reduce this or her risk of cancer may be a person who is genetically predisposed to cancer or who is at risk of cancer due to environmental factors (i.e. smoking, pollution, exposure to toxins etc.).
  • the compound of the first aspect of the invention can be provided in combination with one or more other therapeutic agents, especially those effective for treating cancers (i.e. a chemotherapeutic agent) as described in the second aspect of the invention.
  • the fifth aspect of the invention relates to the use of the compounds of the first aspect of the invention in the manufacture of a medicament for the prevention and/or treatment of cancer.
  • the compounds of the present invention inhibit the conversion of pre- carcinogens into carcinogenic compounds thereby reducing or removing the risk of cancer.
  • the medicament can therefore be provided to patient who is not suffering from cancer but instead wishes to reduce his or her risk of cancer.
  • the compound of the first aspect of the invention can be provided in combination with one or more other therapeutic agents, especially those effective from treating cancers (i.e. a chemotherapeutic agent) as described in the second aspect of the invention.
  • Figures 1 and 2 show the restoration of yeast cell growth by DMU 1 10;
  • Figure 3 shows the restoration of HEK-293 cell growth by DMU 1 10
  • Figure 4 shows a plasmid map of pSYE263/BamHI-Xbal/h_CYP1A1 (with restriction sites that cut the plasmid only once);
  • Figure 5 shows a plasmid map of pcDNA3.1/BamHI-Xbal/h_CYP1A1 (with restriction sites that cut the plasmid only once).
  • IC50 values the concentration at which 50 % of the enzyme activity is inhibited
  • An IC50 assay includes microsomes which either contain cytochrome P450 enzymes, a chosen chemical compound in six serial dilutions, DMSO, 96-well flat-bottomed microtitre plate, substrates such as ER or CEC or EOMCC or DBF (which form fluorescent compounds upon CYP metabolism) and a fluorescent plate reader which ultimately determines IC50 values via endpoint fluorescence assays.
  • the computer was switched on and the KC4 software (on the BioTek plate reader) was opened to select the assay parameters and plate layout.
  • the plate reader machine was warmed at 37°C.
  • Compounds were serially diluted to six different concentrations with 10% DMSO in a Sero-Wel white microplate. Serial dilutions were made with a dilution factor of 1 :20. 45 ⁇ of regenerating system was prepared and pre-warmed at 37°C as detailed below:
  • Table 1 The constitution of the regenerating system used per reaction in each single well for different CYPs was as follows.
  • Tris-acetonitrile was added to all wells using an 8-channel multi-pipette to stop the reaction; after that 50 ⁇ of enzyme/substrate reaction was added into the background well.
  • the plate was left to shake for 10 seconds and an endpoint assay was run using appropriate settings.
  • Example 2 Comparing the IC50 values of flavonoids in CYP1A1 , CYP1 B1 and CYP1A2 enzyme assays (using CYP1A1 , CYP1 B1 and
  • CYP1A2 yeast microsomes obtained from CYP Design Ltd
  • Table: 3 IC 50 values obtained by screening flavonoids on CYP1A1 , CYP1 B1 and
  • Example 3 IC50 values of DMU 110 in CYP1A1 , CYP1 B1 , CYP1A2, CYP2D6 and CYP3A4 enzyme assays (using CYP1A1 , CYP1 B1 , CYP1A2, CYP2D6 and CYP3A4 yeast microsomes) from CYP Design Ltd
  • Table:4 IC 50 values obtained by screening DMU 1 10 on CYP1A1 , CYP1 B1 , CYP1A2, CYP2D6 and CYP3A4 bearing microsomes
  • DMU 1 10 is a relatively potent CYP1-specific inhibitor
  • Tetramethyl luteolin ether Tetramethyl luteolin ether
  • IC 50 values obtained by screening them on CYP1A1 and CYP1 B1 enzymes from CYP Design Ltd.
  • the assays provide a rapid and inexpensive method of determining the inhibitory potential of compounds.
  • the assays could also be used to determine the expression levels of a particular CYP from different clones.
  • the cells can be grown and expressed at various time points and the metabolism of a fluorescence substrate can be analysed to determine the relative amounts of a CYP that is produced from different clones.
  • the cell-based enzyme inhibition assays were carried out to find if the earlier results obtained from the in vitro enzyme assays (using isolated microsomes) have any bearing in the cellular context. This can be achieved by comparing results from the in vitro assays with those obtained from cellular assays. As observed with microsomes, P450 activity is inhibited by certain compounds. However, it is important to consider if live cells expressing CYP1A1 , CYP1 B1 and CYP1A2 enzymes have the potential to take up the compounds of interest through the yeast cell wall.
  • the live cell procedures include the use of 96-well flat-bottomed microplates, the substrates and a multi-mode filter plate reader to obtain fluorescence outputs that help in determining IC50 values.
  • yeast strains were streaked out for growth on SD-minimal medium agar plates that contained the required nutrients and 2% glucose. The plates were then incubated at 30°C for 3 days. A loop-full of cells, from one of the many colonies that grew on the SD-minimal medium agar plate, were taken and were inoculated in 10 ml of autoclaved minimal medium broth that contained 0.02% casamino acids (SW6 broth) in a sterile conical flask. The broth was incubated in a shaking incubator at 30°C at 220 rpm for 16 hours. The culture was then diluted 1 :10 and optical density was measured at 600 nm.
  • Table 6 Outline of kinetic assay parameters used for analysing cytochrome P450 enzymes using live cells and the Bio-Tek Synergy HT fluorescent plate reader.
  • Table 7 Compounds screened with yeast strain that expresses the CYP1A1 enzyme via live cell based inhibition assays. Comparison of CYP1A1 IC 50 values of selected compounds obtained from microsomal and live cell assays.
  • Example 6 Restoration of HEK-293 cell growth by DMU 110 after recombinant human cells were treated with benzo(a)pyrene and TCDD

Abstract

The present application relates to a compound of formula (I) for use in the prevention and/or treatment of cancer, wherein R1 to R9 are independently selected from hydrogen, aliphatic, alkoxy, thioalkyi, alkylamino, halogen, hydroxy, cyano, nitro, hydroxyalkyl, alkylcarbonyloxy, alkoxycarbonyl, alkylcarbonyl, haloalkyl, alkylsulfonylamino NH2, NO2, SO2Rx, SORx and COORx, where Rx is hydrogen, aliphatic or aryl. The compounds are particularly provided for the prevention and/or treatment of cancer of the lung, colon, pancreas, liver and/or kidney.

Description

FLAVONES AS CYP1A1 INHIBITORS FOR THE TREATMENT OF CANCER
Cytochrome P450 (CYP) enzymes belong to a large family of detoxification enzymes that are present in different organs of the human body. The human cytochrome P450 1 (CYP1 ) family consists of three members; namely CYP1A1 , CYP1A2 and CYP1 B1. While the mRNAs of CYP1 A1 and CYP1 B1 genes are known to be expressed in extra-hepatic tissues such as lung, ovary, prostate, kidney and mammary gland and the mRNA of CYP1 A2 gene is found in the liver amongst other tissues, protein expression of these genes is rarely seen in human tissues, under normal circumstances. Expression of CYP1A1 , CYP1 B1 and CYP1A2 are however implicated in cancer. In particular, CYP1A1 expression is induced by poly-aromatic hydrocarbons (PAHs) which are found mainly in cigarette smoke, high-boiling fraction of crude oil, charred meat and vegetables. PAHs like 2, 3, 7, 8-tetrachlorodibenzo-p-dioxin (TCDD), benzo(a)pyrene, 7,12-dimethylbenz(a) anthracene (DMBA) can bind to the aromatic hydrocarbon receptor as ligands. The activated AhR performs the role of a transcription factor that is responsible for the induction of CYP1A1 genes.
Induction leads to increased levels of CYP1A1 enzymes. The PAHs also act as ideal substrates for CYP1A1 enzymes which efficiently hydroxylate the PAHs leading to the formation of carcinogenic entities from pro-carcinogenic molecules. The PAHs appear to play a dominant role in the CYP1A1 mediated positive feedback mechanism that underlies the formation of carcinogenic substances capable of intercalating DNA. Hydroxylated PAHs are carcinogenic since they have great propensity to intercalate with double- stranded DNA and cause breaks in the double-stranded DNA. Hence all PAHs in general have tumour promoting properties.
Besides PAHs and its derivatives, CYP1A1 metabolises other xenobiotic compounds such as nitrogenous heterocyclics, caffeine, aromatic amines and an assortment of other compounds. Metabolism (biotransformation) of these compounds (i.e. pro-carcinogens) by CYP1A1 enzymes leads to the formation of carcinogenic substances.
Induction of CYP1A1 therefore results in the biotransformation (metabolism) of PAHs which are potential pro-carcinogens to carcinogenic substances that can eventually lead to cancer. CYP1A1 has been suggested to have a role in many cancers and appears to have a major role in the genesis of lung cancer.
Cigarette smoke which contains polyaromatic hydrocarbons (PAHs) and aromatic amine pro-carcinogenic compounds is particularly associated with the induction of CYP1A1 gene and the resultant metabolism of the PAHs in cigarette smoke is thought to be one of the primary causes of lung cancer.
CYP1A1 gene induction occurs at the transcriptional level which, in turn, leads to the induction of CYP1A1 enzyme activity. CYP1A1 enzyme is also known as aromatic hydrocarbon hydroxylase (AHH) since it metabolises PAHs through hydroxylation to form carcinogens. For induction of CYP1A1 gene transcription a transcription factor composed of two proteins is required. Ligand (PAH)-bound aromatic (aryl) hydrocarbon receptor (AhR) forms a complex with the aryl hydrocarbon receptor nuclear translocator (Arnt) to form the active transcription factor.
Recent animal and human data suggested that AhR is involved in various signalling pathways critical to cells' normal homeostasis, which includes physiological processes such as cell proliferation and differentiation, gene regulation, cell motility and migration, inflammation and others. Dysregulation of these processes is known to contribute to events such as tumour initiation, promotion, and progression.
While natural products such as flavonoids, chalcones and stilbenes have a general ability to modulate the activity of cytochrome P450 (CYP) enzymes, until now no potent CYP 1A1 -specific inhibitor has been identified. There is therefore a need in the art for compounds that inhibit CYP1A1 and which have the potential to prevent conversion of pro-carcinogenic substances to carcinogens and thereby act as chemo-preventive agents in the treatment of cancer. The first aspect of the invent d of formula (I)
Figure imgf000003_0001
for use in the prevention and/or treatment of cancer, wherein R1 to R9 are independently selected from hydrogen, aliphatic, alkoxy, thioalkyi, alkylamino, halogen, hydroxy, cyano, nitro, hydroxyalkyl, alkylcarbonyloxy, alkoxycarbonyl, alkylcarbonyl, haloalkyl,
alkylsulfonylamino NH2, N02, S02Rx, SORx and COORx, where Rx is hydrogen, aliphatic or aryl.
In particular, R1 to R9 are independently selected from hydrogen, aliphatic, halogen, hydroxy, alkoxy, thioalkyi, alkylamino or cyano, preferably hydrogen, hydroxyl, halogen or alkoxy, more preferably hydrogen, hydroxyl or alkoxy.
Preferably, R1, R5 and R9 are hydrogen and one or more of R2, R3, R4, R6, R7 and R8 are selected from aliphatic, alkoxy, thioalkyi, alkylamino, halogen, hydroxy, cyano, nitro, hydroxyalkyl, alkylcarbonyloxy, alkoxycarbonyl, alkylcarbonyl, haloalkyl,
alkylsulfonylamino NH2, N02, S02Rx, SORx and COORx, where Rx is hydrogen, aliphatic or aryl, with the remaining groups being hydrogen. In particular, one or more of R2, R3, R4, R6, R7 and R8 are independently selected from aliphatic, halogen, hydroxy, alkoxy, thioalkyi, alkylamino or cyano, preferably hydroxyl, halogen or alkoxy, more preferably hydroxyl or alkoxy with the remaining groups being hydrogen. The phenyl ring is preferably mono- or di-substituted, that is one or two of R2, R3 and R4 are preferably selected from aliphatic, alkoxy, thioalkyi, alkylamino, halogen, hydroxy, cyano, nitro, hydroxyalkyl, alkylcarbonyloxy, alkoxycarbonyl, alkylcarbonyl, haloalkyl, alkylsulfonylamino NH2, N02, S02Rx, SORx and COORx, where Rx is hydrogen, aliphatic or aryl, preferably one or two of R2, R3 and R4 are independently aliphatic, halogen, hydroxy, alkoxy, thioalkyi, alkylamino or cyano, preferably hydroxyl, halogen or alkoxy, more preferably hydroxyl or alkoxy and the remainder are hydrogen. In a particularly preferred feature of the first aspect, R3 is selected from aliphatic, alkoxy, thioalkyi, alkylamino, halogen, hydroxy, cyano, nitro, hydroxyalkyl, alkylcarbonyloxy,
alkoxycarbonyl, alkylcarbonyl, haloalkyl, alkylsulfonylamino NH2, N02, S02Rx, SORx and COORx, where Rx is hydrogen, aliphatic or aryl, preferably R3 is selected from aliphatic, halogen, hydroxy, alkoxy, thioalkyi, alkylamino or cyano, preferably hydroxyl, halogen or alkoxy, more preferably hydroxyl or alkoxy; and R2 and R4 are independently both hydrogen. Alternatively, R2 and R3 are independently selected from aliphatic, alkoxy, thioalkyi, alkylamino, halogen, hydroxy, cyano, nitro, hydroxyalkyl, alkylcarbonyloxy,
alkoxycarbonyl, alkylcarbonyl, haloalkyl, alkylsulfonylamino NH2, N02, S02Rx, SORx and COORx, where Rx is hydrogen, aliphatic or aryl, preferably R2 and R3 are independently aliphatic, halogen, hydroxy, alkoxy, thioalkyi, alkylamino or cyano, preferably hydroxyl, halogen or alkoxy, more preferably hydroxyl or alkoxy; and R4 is hydrogen. Alternatively, R2 is hydrogen and R3 and R4 are independently selected from aliphatic, alkoxy, thioalkyi, alkylamino, halogen, hydroxy, cyano, nitro, hydroxyalkyl,
alkylcarbonyloxy, alkoxycarbonyl, alkylcarbonyl, haloalkyl, alkylsulfonylamino NH2, N02, S02Rx, SORx and COORx, where Rx is hydrogen, aliphatic or aryl, preferably R3 and R4 are independently aliphatic, halogen, hydroxy, alkoxy, thioalkyi, alkylamino or cyano, preferably hydroxyl, halogen or alkoxy, more preferably hydroxyl or alkoxy.
The fused phenyl ring is preferably di- or tri-substituted, that is two or three of R6, R7 and R8 are independently selected from aliphatic, alkoxy, thioalkyi, alkylamino, halogen, hydroxy, cyano, nitro, hydroxyalkyl, alkylcarbonyloxy, alkoxycarbonyl, alkylcarbonyl, haloalkyl, alkylsulfonylamino NH2, N02, S02Rx, SORx and COORx, where Rx is hydrogen, aliphatic or aryl, preferably two or three of R6, R7 and R8 are independently aliphatic, halogen, hydroxy, alkoxy, thioalkyi, alkylamino or cyano, preferably hydroxyl, halogen or alkoxy, more preferably hydroxyl or alkoxy with the remainder of R6, R7, R8 being hydrogen. Preferably all three of R6, R7 and R8 are independently selected from aliphatic, alkoxy, thioalkyi, alkylamino, halogen, hydroxy, cyano, nitro, hydroxyalkyl,
alkylcarbonyloxy, alkoxycarbonyl, alkylcarbonyl, haloalkyl, alkylsulfonylamino NH2, N02, S02Rx, SORx and COORx, where Rx is hydrogen, aliphatic or aryl, preferably R6, R7 and R8 are independently aliphatic, halogen, hydroxy, alkoxy, thioalkyi, alkylamino or cyano, preferably hydroxyl, halogen or alkoxy, more preferably hydroxyl or alkoxy.
Where one or more of R1 to R9 are alkoxy, the one or more alkoxy group is particularly one or more C1-6 alkoxy group, more particularly one or more C1-4 alkoxy group, more particularly a methoxy, ethoxy, propoxy or butoxy group, for example a methoxy or ethoxy group, most particularly methoxy.
In a particular feature of the first aspect of the invention, R1 to R9 are independently selected from hydrogen, hydroxy or alkoxy.
Preferably, R1, R5 and R9 are hydrogen.
The phenyl ring is preferably mono- or di-substituted, that is one or two of R2, R3 and R4 are preferably selected from alkoxy or hydroxy, and the remainder are hydrogen. In a particularly preferred feature of the first aspect, R3 is alkoxy or hydroxyl, preferably alkoxy and R2 and R4 are selected from hydrogen, alkoxy or hydroxy, particularly R2 and R4 are independently both hydrogen or R2 is alkoxy or hydroxyl and R4 is hydrogen or R2 is hydrogen and R4 is alkoxy or hydroxyl.
In a particular feature of the first aspect of the invention, R1 to R9 are independently selected from hydrogen, hydroxy or methoxy.
Preferably, R1, R5 and R9 are hydrogen.
The phenyl ring is preferably mono- or di-substituted, that is one or two of R2, R3 and R4 are preferably selected from methoxy or hydroxy, and the remainder are hydrogen. In a particularly preferred feature of the first aspect, R3 is methoxy or hydroxyl, preferably methoxy and R2 and R4 are selected from hydrogen, methoxy or hydroxy, particularly R2 and R4 are independently both hydrogen or R2 is methoxy or hydroxyl and R4 is hydrogen or R2 is hydrogen and R4 is methoxy or hydroxyl.
The fused phenyl ring is preferably di- or tri-substituted, that is two or three of R6, R7 and R8 are alkoxy or hydroxyl with the remainder of R6, R7, R8 being hydrogen. Preferably two or three of R6, R7 and R8 are methoxy or hydroxyl with the remainder of R6, R7 or R8 being hydrogen. In a particularly preferred feature of the first aspect R6, R7 and R8 are each hydroxyl or alkoxy. Alternatively R6, R7 and R8 are each hydroxyl or alkoxy. Preferably all three groups are methoxy as illustrated below.
Figure imgf000006_0001
where R1 to R5 are as disclosed above.
For the purposes of the present invention, the term aryl includes for example optionally substituted unsaturated monocyclic, bicyclic or tricyclic rings of up to 14 carbon atoms, such as phenyl, naphthyl and phenanthroline. Alternatively, the term aryl may include partially saturated bicyclic rings such as tetrahydro-naphthyl. Preferably, the aryl group is phenyl, naphthy or phenanthroline.
The term "aliphatic" as used herein refers to a straight or branched chain hydrocarbon which is completely saturated or contains one or more units of unsaturation. Thus, aliphatic may be alkyl, alkenyl or alkynyl, preferably having 1 to 12 carbon atoms, up to 6 carbon atoms or up to 4 carbon atoms.
For the purposes of this invention, alkyl relates to both straight chain and branched alkyl radicals of 1 to 12 carbon atoms, preferably 1 to 8 carbon atoms and most preferably 1 to 4 carbon atoms including but not limited to methyl, ethyl, n-propyl, isopropyl, n-butyl, sec- butyl, isobutyl, tert-butyl n-pentyl, n-hexyl, n-heptyl, n-octyl. The term alkyl therefore relates to radicals comprising 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1 or 12 carbon atoms. The term alkyl also encompasses cycloalkyl radicals of 3 to 12 carbon atoms, preferably 4 to 8 carbon atoms, and most preferably 5 to 6 carbon atoms including but not limited to cyclopropyl, cyclobutyl, CH2-cyclopropyl, CH2-cyclobutyl, cyclopentyl or cyclohexyl. Cycloalkyl groups may be optionally substituted or fused to one or more carbocyclyl or heterocyclyl group. Haloalkyl relates to an alkyl radical preferably having 1 to 8 carbon atoms, preferably 1 to 4 carbon atoms substituted with one or more halide atoms for example CH2CH2Br, CF3 or CCI3.
The term "alkenyl" means a straight chain or branched alkylenyl radical of 2 to 12 carbon atoms, preferably 2 to 6 carbon atoms and most preferably 2 to 4 carbon atoms, and containing one or more carbon-carbon double bonds and includes but is not limited to ethylene, n-propyl-1-ene, n-propyl-2-ene, isopropylene, etc.. The term "alkynyl" means a straight chain or branched alkynyl radical of 2 to 12 carbon atoms, preferably 2 to 6 carbon atoms and most preferably 2 to 4 carbon atoms, and containing one or more carbon-carbon triple bonds and includes but is not limited to ethynyl, 2-methylethynyl etc. The term alkenyl and alkynyl therefore encompass radicals comprising 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1 or 12 carbon atoms.
The term "alkoxy" as used herein refers to an oxy group that is bonded to an alkyl group as defined herein. An alkoxy is preferably a "C1-8 alkoxy group", even more preferably a "Ci-6 alkoxy group" and more preferably a "Ci-4 alkoxy group". The alkoxy group particularly includes 1 , 2, 3 or 4 carbon atoms. Particularly preferably alkoxy groups include methoxy, ethoxy, propyloxy and butyloxy. Halogen means F, CI, Br or I, preferably F.
In a particularly preferred aspect, the compounds of formula (I) are one or more of
Figure imgf000008_0001
(Tetramethyl luteolin ether)
Figure imgf000008_0002
Figure imgf000009_0001
Preferred compounds of the invention listed above extend to the tautomers thereof, as well as (but not limited to) pharmaceutically acceptable salts, esters, amides, carbamates, carbonates, ureides or prodrugs thereof or a derivative optionally with one or more lipid groups (natural or synthetic) attached.
Methods of obtaining or synthesising flavonoids are known in the art and such methods can be adapted or modified by the skilled person to provide the compounds of the first aspect of the invention.
The invention extends to prodrugs of the aforementioned compounds. A prodrug is any compound that may be converted under physiological conditions or by solvolysis to any of the compounds of the invention or to a pharmaceutically acceptable salt of the compounds of the invention. A prodrug may be inactive when administered to a subject but is converted in vivo to an active compound of the invention.
The compounds of the invention may contain one or more stereogenic (asymmetric) carbon atoms and may exist in racemic and optically active forms (enantiomers or diastereoisomers). The first aspect of the invention includes all such enantiomers or diastereoisomers and mixtures thereof, including racemic mixtures.
Examples of pharmaceutically acceptable salts of the compounds of formulae (I), (IA) or (IB) include those derived from organic acids such as methanesulphonic acid, benzenesulphonic acid and p-toluenesulphonic acid, mineral acids such as hydrochloric and sulphuric acid and the like, giving methanesulphonate, benzenesulphonate, p- toluenesulphonate, hydrochloride and sulphate, and the like, respectively or those derived from bases such as organic and inorganic bases. Examples of suitable inorganic bases for the formation of salts of compounds for this invention include the hydroxides, carbonates, and bicarbonates of ammonia, lithium, sodium, calcium, potassium, aluminium, iron, magnesium, zinc and the like. Salts can also be formed with suitable organic bases. Such bases suitable for the formation of pharmaceutically acceptable base addition salts with compounds of the present invention include organic bases which are nontoxic and strong enough to form salts. Such organic bases are already well known in the art and may include amino acids such as arginine and lysine, mono-, di-, or trihydroxyalkylamines such as mono-, di-, and triethanolamine, choline, mono-, di-, and trialkylamines, such as methylamine, dimethylamine, and trimethylamine, guanidine; N- methylglucosamine; N-methylpiperazine; morpholine; ethylenediamine; N- benzylphenethylamine; tris(hydroxymethyl) aminomethane; and the like.
Salts may be prepared in a conventional manner using methods well known in the art. Acid addition salts of said basic compounds may be prepared by dissolving the free base compounds according to the first or second aspects of the invention in aqueous or aqueous alcohol solution or other suitable solvents containing the required acid. Where a compound of formula (I), (IA) or (IB) contain an acidic function, a base salt of said compound may be prepared by reacting said compound with a suitable base. The acid or base salt may separate directly or can be obtained by concentrating the solution eg. by evaporation. The compounds of this invention may also exist in solvated or hydrated forms. The compounds of the invention are provided for the prevention and/or treatment of cancer. In particular, the compounds are provided for the prevention and/or treatment of cancer of the lung, colon, pancreas, liver and/or kidney. Throughout this text, the prevention and/or treatment of cancer means any effect which mitigates any damage, to any extent. The term "treatment" means any amelioration of a disorder, disease, syndrome, condition, pain or a combination of two or more thereof. The term "prevention" means to prevent the condition from occurring, lessening the severity of the condition or to prevent from deteriorating or getting worse for example by halting the progress of the disease without necessary ameliorating the condition.
The compounds of the first aspect of the invention are provided as inhibitors of CYP1 A1 which has been implicated in cancer of the lung, colon, pancreas, liver and/or kidney. Inhibition of CYP1A1 by the claimed compounds will allow the prevention and/or treatment of cancer.
It is therefore a particular desire of the invention to provide compounds which selectively inhibit CYP1A1 when compared with other CYPs. The present invention therefore particular provides a compound of formula (IB)
Figure imgf000011_0001
(IB)
wherein R1, R5 and R9 are hydrogen, R3, R6 and R8 are alkoxy and R2, R4 and R7 are hydrogen, hydroxyl or alkoxy, preferably hydrogen or alkoxy.
In particular, wherein R1, R5 and R9 are hydrogen, R3, R6 and R8 are methoxy and R2, and R7 are hydrogen, hydroxyl or methoxy, preferably hydrogen or methoxy.
Particularly preferred compounds of formula (IB) include
Compound Chemical structure
Figure imgf000012_0001
Figure imgf000012_0002
A second aspect of the invention provides a composition comprising a compound, in particular a novel compound according to the first aspect of the invention, in combination with a pharmaceutically acceptable excipient. The pharmaceutically acceptable excipient may comprise a pharmaceutically acceptable carrier and/or pharmaceutically acceptable diluent. Suitable carriers and/or diluents are well known in the art and include
pharmaceutical grade starch, mannitol, lactose, magnesium stearate, sodium saccharin, talcum, cellulose, glucose, sucrose, (or other sugar), magnesium carbonate, gelatin, oil, alcohol, detergents, emulsifiers or water (preferably sterile). The composition may be a mixed preparation of a composition or may be a combined preparation for simultaneous, separate or sequential use (including administration). A pharmaceutical composition may be provided in unit dosage form, will generally be provided in a sealed container and may be provided as part of a kit. Such a kit would normally (although not necessarily) include instructions for use. It may include a plurality of said unit dosage forms.
The compounds according to the invention for use in the aforementioned indications may be administered by any convenient method, for example by oral (including by inhalation), parenteral, mucosal (e.g. buccal, sublingual, nasal), rectal or transdermal administration and the compositions adapted accordingly. Such compositions may be prepared by any method known in the art of pharmacy, for example by admixing the active ingredient with a carrier(s) or excipient(s) under sterile conditions.
For oral administration, the compounds can be formulated as liquids or solids, for example solutions, syrups, suspensions or emulsions, tablets, capsules and lozenges.
Pharmaceutical compositions adapted for oral administration may be presented as discrete units such as capsules or tablets; as powders or granules; as solutions, syrups or suspensions (in aqueous or non-aqueous liquids; or as edible foams or whips; or as emulsions). Suitable excipients for tablets or hard gelatine capsules include lactose, starch including maize starch or derivatives thereof, stearic acid or salts thereof, such as
magnesium stearate, sucrose or microcrystalline cellulose. A composition in the form of a capsule can be prepared using routine encapsulation procedures. For example, powders, granules or pellets containing the active ingredient can be prepared using standard carriers and then filled into a hard gelatin capsule; alternatively, a dispersion or suspension can be prepared using any suitable pharmaceutical carrier(s), for example aqueous gums, celluloses, silicates or oils and the dispersion or suspension then filled into a soft gelatin capsule. Suitable excipients for use with soft gelatine capsules include for example vegetable oils, waxes, fats, semi-solid, or liquid polyols etc.
Compositions for oral administration may be designed to protect the active ingredient against degradation as it passes through the alimentary tract, for example by an outer coating of the formulation on a tablet or capsule.
A liquid formulation, such as a solution or a syrup will generally consist of a suspension or solution of the compound or physiologically acceptable salt in a suitable aqueous or non- aqueous liquid carrier(s) for example water, ethanol, glycerine, sugars, polyethylene glycol or an oil. For the preparation of suspensions oils (e.g. vegetable oils) may be used to provide oil-in-water or water in oil suspensions. The formulation may also contain a suspending agent, preservative, flavouring or colouring agent.
Compositions for nasal or oral administration may conveniently be formulated as aerosols, drops, gels and powders. Aerosol formulations typically comprise a solution or fine suspension of the active substance in a physiologically acceptable aqueous or nonaqueous solvent and are usually presented in single or multidose quantities in sterile form in a sealed container, which can take the form of a cartridge or refill for use with an atomising device. Alternatively the sealed container may be a unitary dispensing device such as a single dose nasal inhaler or an aerosol dispenser fitted with a metering valve which is intended for disposal once the contents of the container have been exhausted. Where the dosage form comprises an aerosol dispenser, it will contain a pharmaceutically acceptable propellant. The aerosol dosage forms can also take the form of a pump- atomiser.
Pharmaceutical compositions adapted for nasal administration wherein the carrier is a solid include a coarse powder having a particle size for example in the range 20 to 500 microns which is administered in the manner in which snuff is taken, i.e. by rapid inhalation through the nasal passage from a container of the powder held close up to the nose. Suitable compositions wherein the carrier is a liquid, for administration as a nasal spray or as nasal drops, include aqueous or oil solutions of the active ingredient.
Pharmaceutical compositions adapted for topical administration may be formulated as ointments, creams, suspensions, lotions, powders, solutions, pastes, gels, sprays, aerosols or oils. For application to the eye or other external tissues, for example mouth and skin, the compositions are preferably applied as a topical ointment or cream. When formulated in an ointment, the active ingredient may be employed with either a paraffinic or a water-miscible ointment base. Alternatively, the active ingredient may be formulated in a cream with an oil- in-water cream base or a water-in-oil base. Pharmaceutical compositions adapted for topical administration to the eye include eye drops wherein the active ingredient is dissolved or suspended in a suitable carrier, especially an aqueous solvent. Pharmaceutical compositions adapted for topical administration in the mouth include lozenges, pastilles and mouth washes. Pharmaceutical compositions adapted for rectal administration may be presented as suppositories or enemas. Pharmaceutical compositions adapted for vaginal
administration may be presented as pessaries, tampons, creams, gels, pastes, foams or spray formulations. Compositions for rectal or vaginal administration are conveniently in the form of suppositories (containing a conventional suppository base such as cocoa butter), pessaries, vaginal tabs, foams or enemas.
Compositions suitable for buccal or sublingual administration include tablets, lozenges and pastilles, wherein the active ingredient is formulated with a carrier such as sugar and acacia, tragacanth, or gelatin and glycerin.
Compositions suitable for transdermal administration include ointments, gels, patches and injections including powder injections.
Conveniently the composition is in unit dose form such as a tablet, capsule or ampoule. Pharmaceutical compositions adapted for parenteral administration include aqueous and non-aqueous sterile injection solution which may contain anti-oxidants, buffers, bacteriostats and solutes which render the formulation substantially isotonic with the blood of the intended recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents. Excipients which may be used for injectable solutions include water, alcohols, polyols, glycerine and vegetable oils, for example. The 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 (lyophilized) condition requiring only the addition of the sterile liquid carried, for example water for injections, immediately prior to use. Extemporaneous injection solutions and suspensions may be prepared from sterile powders, granules and tablets.
The pharmaceutical compositions may contain preserving agents, solubilising agents, stabilising agents, wetting agents, emulsifiers, sweeteners, colourants, odourants, salts, buffers, coating agents or antioxidants. They may also contain an adjuvant and/or therapeutically active agents in addition to the substance of the present invention.
Dosages of the substance of the present invention can vary between wide limits, depending upon a variety of factors including the disease or disorder to be treated, the age, weight and condition of the individual to be treated, the route of administration etc. and a physician will ultimately determine appropriate dosages to be used. Typically, however, the dosage adopted for each route of administration when a compound of the invention is administered to adult humans is 0.001 to 500 mg/kg. Such a dosage may be given, for example, from 1 to 5 times daily by bolus infusion, infusion over several hours and/or repeated administration. The compositions may be administered in conjunction with one or more other therapeutically active agents, especially those effective for treating cancers (i.e. a chemotherapeutic agent). Another chemotherapeutic agent may be, for example, mitoxantrone, Vinca alkaloids, such as vincristine and vinblastine, anthracycline antibiotics such as daunorubicin and doxorubicin, alkylating agents such as chlorambucil and melphalan, taxanes such as paclitaxel, anti-folates such as methotrexate and tomudex, epipodophyllotoxins such as etoposide, camptothecins such as irinotecan and its active metabolite SN-38 and DNA methylation inhibitors. The other active compound(s) may be incorporated in the same composition as the compounds of the present invention or they may be administered alongside the compounds of the present invention, e.g. simultaneously or sequentially. Thus, the invention provides a kit of parts comprising a compound of the invention and another chemotherapeutic agent, optionally with instructions for use.
Alternatively, the compound of the first aspect of the invention may be administered by their addition to a food or drink. In an alternative feature of the second aspect of the invention, the compounds of the first aspect of the invention are formulated into a powder or liquid for addition to food or drink and administration by these means. In this feature of the second aspect, the compounds of the first aspect will be formulated with an excipient or diluent but such excipient or diluent does not need to be pharmaceutically acceptable but instead should be acceptable for consumption.
A third aspect of the invention provides a process for the manufacture of a composition according to the second aspect of the invention. The manufacture can be carried out by standard techniques well known in the art and involves combining a compound according to the first aspect of the invention and a pharmaceutically acceptable carrier or diluent. The composition may be in any form including a tablet, a liquid, a capsule, and a powder or in the form of a food product, e.g. a functional food. In the latter case the food product itself may act as the pharmaceutically acceptable carrier.
The fourth aspect of the invention provides a method of preventing or treating cancer comprising administering a compound of the first aspect of the invention to a patient in need thereof.
As discussed above, the compounds of the present invention inhibit the conversion of pre- carcinogens into carcinogenic compounds thereby reducing or removing the risk of cancer. The patient in need thereof does not therefore need to be suffering from cancer but can instead wish to reduce his or her risk of cancer. Thus the fourth aspect of the invention provides a method of reducing the risk of developing cancer, comprising administering a compound of the first aspect of the invention. A person wishing to reduce this or her risk of cancer may be a person who is genetically predisposed to cancer or who is at risk of cancer due to environmental factors (i.e. smoking, pollution, exposure to toxins etc.).
The compound of the first aspect of the invention can be provided in combination with one or more other therapeutic agents, especially those effective for treating cancers (i.e. a chemotherapeutic agent) as described in the second aspect of the invention.
The fifth aspect of the invention relates to the use of the compounds of the first aspect of the invention in the manufacture of a medicament for the prevention and/or treatment of cancer. As discussed above, the compounds of the present invention inhibit the conversion of pre- carcinogens into carcinogenic compounds thereby reducing or removing the risk of cancer. The medicament can therefore be provided to patient who is not suffering from cancer but instead wishes to reduce his or her risk of cancer. The compound of the first aspect of the invention can be provided in combination with one or more other therapeutic agents, especially those effective from treating cancers (i.e. a chemotherapeutic agent) as described in the second aspect of the invention.
All preferred features of the first to fifth aspects of the invention relate to all other aspects of the invention mutandis mutandi. In particular, cancer according to the fourth and fifth aspects of the invention is as defined in the first aspect of the invention.
Brief description of the figures Figures 1 and 2 show the restoration of yeast cell growth by DMU 1 10;
Figure 3 shows the restoration of HEK-293 cell growth by DMU 1 10;
Figure 4 shows a plasmid map of pSYE263/BamHI-Xbal/h_CYP1A1 (with restriction sites that cut the plasmid only once); Figure 5 shows a plasmid map of pcDNA3.1/BamHI-Xbal/h_CYP1A1 (with restriction sites that cut the plasmid only once).
The invention will now be described with reference to the following non-limiting examples. Examples
Example 1. Determination of IC50 values
This method is used to measure the IC50 values (the concentration at which 50 % of the enzyme activity is inhibited) of the compounds. IC50 values, which effectively reflect the inhibitory potential of a compound, also provide evidence of at the possible effectiveness of a compound in a biological process.
An IC50 assay includes microsomes which either contain cytochrome P450 enzymes, a chosen chemical compound in six serial dilutions, DMSO, 96-well flat-bottomed microtitre plate, substrates such as ER or CEC or EOMCC or DBF (which form fluorescent compounds upon CYP metabolism) and a fluorescent plate reader which ultimately determines IC50 values via endpoint fluorescence assays.
Procedure for IC50 determination
The computer was switched on and the KC4 software (on the BioTek plate reader) was opened to select the assay parameters and plate layout. The plate reader machine was warmed at 37°C. Compounds were serially diluted to six different concentrations with 10% DMSO in a Sero-Wel white microplate. Serial dilutions were made with a dilution factor of 1 :20. 45 μΙ of regenerating system was prepared and pre-warmed at 37°C as detailed below:
Table 1 : The constitution of the regenerating system used per reaction in each single well for different CYPs was as follows.
Figure imgf000018_0001
CYP1A1 5 μΐ 1 μΐ 5 μΐ 39 μ1 0.2 Μ -
CYP1B1 5 μΐ 1 μΐ 5 μΐ 39 μ1 0.2 Μ -
CYP1A2 5 μΐ 1 μΐ 5 μΐ 20 μΐ 0.5 Μ 19 μΐ
CYP2D6 5 μΐ 1 μΐ 5 μΐ 25 μΐ 0.2 Μ 14 μΐ
CYP3A4 5 μΐ 1 μΐ 5 μΐ 25 μΐ 0.2 Μ
14 μΙ
Meanwhile, 50 μΙ of enzyme substrate mix reaction was prepared and kept for incubation at 37°C for 10 minutes (Table 2).
Table 2: Enzyme-Substrate mixtures per reaction in each well were as follows.
Figure imgf000019_0001
In wells of a 96-well flat-bottomed microplate, 45 μΙ of regenerating system, 5 μΙ serial dilutions of inhibitor were added from the dilution plate and 50 μΙ of enzyme/substrate was added except in control well (positive control); for this well, instead of inhibitor 5 μΙ of 10% DMSO was added. In the background well (negative control), only 45 μΙ regenerating system and 5 μΙ 10% DMSO were added but no enzyme and then microplate was vortexed for few seconds. The microplate was incubated for 10 minutes. After 10 minutes, 75 μΙ of Tris-acetonitrile was added to all wells using an 8-channel multi-pipette to stop the reaction; after that 50 μΙ of enzyme/substrate reaction was added into the background well. The plate was left to shake for 10 seconds and an endpoint assay was run using appropriate settings.
Calculation of IC50 values
• Step 1 : Calculated percentage inhibition of the samples
• Step 2: Subtracted the value below 50% by 50 = A
• Step 3: Chose the value above 50% and below 50% and subtract the values = B · Step 4: Divided A by B = C
• Step 5: Multiplied C by ( high concentration obtained - minus lower concentration obtained) = D
• Step 6: Added lower concentration than D which probably will lead to IC50 value
• Step 7: IC50 = (50- low percentage below 50%) x (higher concentration - lower concentration) + lower concentration.
Example 2. Comparing the IC50 values of flavonoids in CYP1A1 , CYP1 B1 and CYP1A2 enzyme assays (using CYP1A1 , CYP1 B1 and
CYP1A2 yeast microsomes) obtained from CYP Design Ltd
In the compound library that was screened initially for CYP1 A1 inhibition, a number of compounds were flavonoids. Out of these two compounds were found to inhibit CYP1 A1 potently when compared with CYP1 B1 and CYP1A2 enzyme inhibition. These compounds were DMU 1 14 and DMU 1 10 which were shown to have IC50 values of 2 μΜ and 397 nM as shown in Table 3. These flavonoids (natural products) had been synthesised previously within the Chemistry group at the Leicester School of Pharmacy (De Montfort University) and are also commercially available.
Table: 3: IC50 values obtained by screening flavonoids on CYP1A1 , CYP1 B1 and
CYP1A2 bearing microsomes
Figure imgf000020_0001
Figure imgf000021_0001
Example 3. IC50 values of DMU 110 in CYP1A1 , CYP1 B1 , CYP1A2, CYP2D6 and CYP3A4 enzyme assays (using CYP1A1 , CYP1 B1 , CYP1A2, CYP2D6 and CYP3A4 yeast microsomes) from CYP Design Ltd
Table:4: IC50 values obtained by screening DMU 1 10 on CYP1A1 , CYP1 B1 , CYP1A2, CYP2D6 and CYP3A4 bearing microsomes
Figure imgf000021_0002
The results indicate that DMU 1 10 is a relatively potent CYP1-specific inhibitor
Table 5: Chemical structure of commercial flavonoids (analogues of DMU 1 10;
Tetramethyl luteolin ether) and the IC50 values obtained by screening them on CYP1A1 and CYP1 B1 enzymes from CYP Design Ltd.
Figure imgf000021_0003
Figure imgf000022_0001
Diosmetin 447nM 152nM
OMe
Example 4. Cell-based enzyme inhibition assays
The assays provide a rapid and inexpensive method of determining the inhibitory potential of compounds. The assays could also be used to determine the expression levels of a particular CYP from different clones. The cells can be grown and expressed at various time points and the metabolism of a fluorescence substrate can be analysed to determine the relative amounts of a CYP that is produced from different clones.
The cell-based enzyme inhibition assays were carried out to find if the earlier results obtained from the in vitro enzyme assays (using isolated microsomes) have any bearing in the cellular context. This can be achieved by comparing results from the in vitro assays with those obtained from cellular assays. As observed with microsomes, P450 activity is inhibited by certain compounds. However, it is important to consider if live cells expressing CYP1A1 , CYP1 B1 and CYP1A2 enzymes have the potential to take up the compounds of interest through the yeast cell wall.
Recombinant yeast cells that harbour the CYP1A1, CYP1B1 and CYP1A2 genes and are activated by a modified human P450 reductase, AhRDM, were grown. Assays were carried out using selected compounds which had already shown specificity towards microsomal CYP1A1 , CYP1 B1 and CYP1A2 enzymes during in vitro IC50 determinations of these compounds.
The live cell procedures include the use of 96-well flat-bottomed microplates, the substrates and a multi-mode filter plate reader to obtain fluorescence outputs that help in determining IC50 values.
Procedure for live cell assays
From frozen glycerol stocks yeast strains were streaked out for growth on SD-minimal medium agar plates that contained the required nutrients and 2% glucose. The plates were then incubated at 30°C for 3 days. A loop-full of cells, from one of the many colonies that grew on the SD-minimal medium agar plate, were taken and were inoculated in 10 ml of autoclaved minimal medium broth that contained 0.02% casamino acids (SW6 broth) in a sterile conical flask. The broth was incubated in a shaking incubator at 30°C at 220 rpm for 16 hours. The culture was then diluted 1 :10 and optical density was measured at 600 nm. Once the optical density of SW6 broth culture reached OD6oo between 5 and 6, 10 ml of SW6 pre-culture was transferred to a sterile 10 ml tube and centrifuged at 3000 g for 10 minutes, the pellet obtained was transferred to a sterile conical flask containing 10 ml of SE medium which contained yeast nitrogen base, 2% ethanol, 12.5 ml/litre L-adenine, 8.3 ml/litre of L-histidine, L-leucine, and L-tryptophan. 10 ml of cell culture was incubated at 30°C at 220 rpm for 4 hours. After 4 hours, approximately 0.4 X 107 cells per ml were aliquoted into sterile Eppendorf tubes and centrifuged at 13,000 rpm. The supernatant was poured off and cell pellet was washed with TE buffer 3 times. Finally, the cell pellet was re-suspended in 450 μΙ of TE buffer (0.5M Tris-HCI, pH7.4 and 0.1 M EDTA).
Meanwhile, serial dilutions of selected compounds were made to yield different concentrations of compound. In each well of black microplate, 50 μΙ of cell suspension (containing a specific CYP), 5 μΙ of potential inhibitor (selected chemical compounds) and 50 μΙ of substrate mixture (containing CYP-specific substrates) were added. In the control wells, 45μΙ of cell suspension (contain a specific CYP) was mixed with 5 μ1 10% DMSO and 50μΙ of CYP-specific substrate. After preparation of all wells, the microplate was vortexed for 10 seconds so that contents were mixed well in each well and kinetic assay was carried out for 30 minutes at 30°C using appropriate settings (see Table 6).
Table 6: Outline of kinetic assay parameters used for analysing cytochrome P450 enzymes using live cells and the Bio-Tek Synergy HT fluorescent plate reader.
Figure imgf000024_0001
Table 7 Compounds screened with yeast strain that expresses the CYP1A1 enzyme via live cell based inhibition assays. Comparison of CYP1A1 IC50 values of selected compounds obtained from microsomal and live cell assays.
Figure imgf000025_0001
The above results show that besides screening potential inhibitors of a CYP isozyme in microsomal assays, compounds could also be screened conventionally in live cells that express a CYP isoyme and that there is a strict correlation between the two forms of assays. Example 5. Restoration of yeast cell growth by DMU 110 after recombinant yeast cells were treated with benzo(a)pyrene and TCDD
Yeast transformed with ADH2p-CYP1A1 plasmid (figure 4) was treated with
benzo(a)pyrene [BaP; 1 and 5μΜ] and 2,3,7,8-Tetrachlorodibenzo-p-dioxin [TCDD; 1 and 5μΜ] for 16 hours. Cells were blocked in growth as determined by measurement of optical density at 600nm (OD600). Treatment with DMU 1 10 (4μΜ) restored growth (confirmed through measurement of OD600, Figure 1 & Figure 2). This indicates that the CYP1A1 specific inhibitor, DMU 1 10, can prevent CYP1A1 -mediated biotransformation of BaP and TCDD into toxic substances in yeast cells.
Example 6. Restoration of HEK-293 cell growth by DMU 110 after recombinant human cells were treated with benzo(a)pyrene and TCDD
Human embryonic kidney HEK-293 cells transformed with pcDNA3.1/h_CYP1A1 plasmid (figure 5) were treated with benzo(a)pyrene [BaP; 0.5μΜ] and 2,3,7,8-Tetrachlorodibenzo- p-dioxin [TCDD; 50nM]. Cells were blocked in growth. Treatment with DMU 1 10 (4μΜ) restored growth. This indicates that the CYP1A1 specific inhibitor, DMU 1 10, can prevent CYP1A1-mediated biotransformation of BaP and TCDD into toxic substances in human cells.

Claims

A compound of formula (I)
Figure imgf000027_0001
for use in the prevention and/or treatment of cancer, wherein R1 to R9 are independently selected from hydrogen, aliphatic, alkoxy, thioalkyi, alkylamino, halogen, hydroxy, cyano, nitro, hydroxyalkyl, alkylcarbonyloxy, alkoxycarbonyl, alkylcarbonyl, haloalkyl,
alkylsulfonylamino NH2, N02, S02Rx, SORx and COORx, where Rx is hydrogen, aliphatic or aryl.
2. The compound as claimed in claim 1 wherein R1 to R9 are independently selected from hydrogen, aliphatic, halogen, hydroxy, alkoxy, thioalkyi, alkylamino or cyano, preferably hydroxyl, halogen or alkoxy.
3. The compound as claimed in claim 1 or claim 2 wherein R1, R5 and R9 are hydrogen.
4. The compound as claimed in any one of claims 1 to 3 wherein one or two of R2, R3 and R4 are selected from aliphatic, alkoxy, thioalkyi, alkylamino, halogen, hydroxy, cyano, nitro, hydroxyalkyl, alkylcarbonyloxy, alkoxycarbonyl, alkylcarbonyl, haloalkyl,
alkylsulfonylamino NH2, N02, S02Rx, SORx and COORx, where Rx is hydrogen, aliphatic or aryl with the remainder or R2, R3 and R4 being hydrogen.
5. The compound as claimed in any preceding claim wherein two or three of R6, R7 and R8 are independently selected from aliphatic, alkoxy, thioalkyi, alkylamino, halogen, hydroxy, cyano, nitro, hydroxyalkyl, alkylcarbonyloxy, alkoxycarbonyl, alkylcarbonyl, haloalkyl, alkylsulfonylamino NH2, N02, S02Rx, SORx and COORx, where Rx is hydrogen, aliphatic or aryl with the remainder of R6, R7, R8 being hydrogen.
6. The compound as claimed in claim 1 wherein R1 to R9 are independently selected from hydrogen, hydroxy or methoxy.
7. The compound of claim 6 wherein R1, R5 and RM are hydrogen.
8. The compound of claim 6 or 7 wherein one or two of R2, R3 and R4 are selected from methoxy or hydroxyl.
9. The compound as claimed in any one of claims 6 to 8 wherein two or three of R6, R7 and R8 are methoxy or hydroxyl.
10. The compound as claimed in any one of claims 6 to 9 having the structure
Figure imgf000028_0001
where R1 to R5 are as claimed in any one of claims 6 to 9.
1 1 . The compound of any preceding claim, selected from
Figure imgf000028_0002
Figure imgf000029_0001
12. The compound of any preceding claim for the prevention and/or treatment of cancer of the lung, colon, pancreas, liver and/or kidney.
13. The compound of any preceding claim having the formula (IB)
Figure imgf000030_0001
(IB)
wherein R1, R5 and R9 are hydrogen, R3, R6 and R8 are methoxy and R2, R4 and R7 are hydrogen, hydroxyl or methoxy
14. The compounds of formula (IB), as claimed in claim 13 selected from
Figure imgf000030_0002
Figure imgf000030_0003
Figure imgf000031_0001
15 A composition comprising a compound as claimed in any one of claims 1 to 14, in combination with a pharmaceutically acceptable carrier or diluent.
16. A process for the manufacture of a composition as claimed in claim 15 comprising combining a compound as claimed in any one of claims 1 to 14 and a pharmaceutically acceptable carrier or diluent.
17. A method of preventing and/or treating cancer comprising administering a compound as claimed in any one of claims 1 to 14 to a patient in need thereof.
18. The use of a compound as claimed in any one of claims 1 to 14 in the manufacture of a medicament for the prevention and/or treatment of cancer.
PCT/EP2015/059501 2014-05-01 2015-04-30 Flavones as cyp1a1 inhibitors for the treatment of cancer WO2015166041A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
GBGB1407694.7A GB201407694D0 (en) 2014-05-01 2014-05-01 Compound
GB1407694.7 2014-05-01

Publications (1)

Publication Number Publication Date
WO2015166041A1 true WO2015166041A1 (en) 2015-11-05

Family

ID=50980425

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2015/059501 WO2015166041A1 (en) 2014-05-01 2015-04-30 Flavones as cyp1a1 inhibitors for the treatment of cancer

Country Status (2)

Country Link
GB (1) GB201407694D0 (en)
WO (1) WO2015166041A1 (en)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN110981844A (en) * 2019-12-19 2020-04-10 江南大学 Preparation method of sulfoxide flavonoid and sulfone flavonoid
CN111039998A (en) * 2019-12-19 2020-04-21 江南大学 Preparation method of 8-benzenesulfonyl substituted flavone glucose glycoside
CN112830917A (en) * 2021-01-18 2021-05-25 五邑大学 Hydroxylated polymethoxyflavone and preparation method thereof
CN113350331A (en) * 2021-05-21 2021-09-07 广州医科大学附属第一医院(广州呼吸中心) Application of hesperetin and/or derivatives thereof in preparation of drugs or drugs for preventing and/or treating renal cell carcinoma

Citations (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPH09301915A (en) * 1996-05-08 1997-11-25 Sankyo Co Ltd Flavone and naphthalene derivative
WO2001058410A2 (en) * 2000-02-11 2001-08-16 Musc Foundation For Research Development A method of treating colon cancer by administering apigenin, luteolin, diosmetin and crysin
JP2007291040A (en) * 2006-04-27 2007-11-08 Hamamatsu Kagaku Gijutsu Kenkyu Shinkokai 4-site catechol estrogen production inhibitor
KR20090061358A (en) * 2007-12-11 2009-06-16 건국대학교 산학협력단 Composition for preventing and treating breast cancer comprising acacetin
CN101486703A (en) * 2009-03-03 2009-07-22 中国药科大学 Flavone compound with antineoplastic activity, preparation thereof and uses thereof
WO2010112510A1 (en) * 2009-03-31 2010-10-07 Nestec S.A. Use of flavonoids to increase the bioavailability of hesperetin
CN101863867A (en) * 2009-04-14 2010-10-20 清华大学深圳研究生院 Derivatives of flavonoids compounds and preparation method and application thereof
CN101948458A (en) * 2010-09-07 2011-01-19 中国药科大学 Baicalein derivatives with antitumor activity and application thereof in medicines
JP2011057574A (en) * 2009-09-07 2011-03-24 Meijo Univ Androgen receptor antagonist and androgen receptor-binding inhibitor
CN102344454A (en) * 2011-08-01 2012-02-08 广东药学院 Wikstroemia indica (L.) C.A.Mey extract as well as preparation method and application thereof
CN102382092A (en) * 2011-07-15 2012-03-21 北京大学 Novel isopentene flavone compounds and application thereof

Patent Citations (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPH09301915A (en) * 1996-05-08 1997-11-25 Sankyo Co Ltd Flavone and naphthalene derivative
WO2001058410A2 (en) * 2000-02-11 2001-08-16 Musc Foundation For Research Development A method of treating colon cancer by administering apigenin, luteolin, diosmetin and crysin
JP2007291040A (en) * 2006-04-27 2007-11-08 Hamamatsu Kagaku Gijutsu Kenkyu Shinkokai 4-site catechol estrogen production inhibitor
KR20090061358A (en) * 2007-12-11 2009-06-16 건국대학교 산학협력단 Composition for preventing and treating breast cancer comprising acacetin
CN101486703A (en) * 2009-03-03 2009-07-22 中国药科大学 Flavone compound with antineoplastic activity, preparation thereof and uses thereof
WO2010112510A1 (en) * 2009-03-31 2010-10-07 Nestec S.A. Use of flavonoids to increase the bioavailability of hesperetin
CN101863867A (en) * 2009-04-14 2010-10-20 清华大学深圳研究生院 Derivatives of flavonoids compounds and preparation method and application thereof
JP2011057574A (en) * 2009-09-07 2011-03-24 Meijo Univ Androgen receptor antagonist and androgen receptor-binding inhibitor
CN101948458A (en) * 2010-09-07 2011-01-19 中国药科大学 Baicalein derivatives with antitumor activity and application thereof in medicines
CN102382092A (en) * 2011-07-15 2012-03-21 北京大学 Novel isopentene flavone compounds and application thereof
CN102344454A (en) * 2011-08-01 2012-02-08 广东药学院 Wikstroemia indica (L.) C.A.Mey extract as well as preparation method and application thereof

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN110981844A (en) * 2019-12-19 2020-04-10 江南大学 Preparation method of sulfoxide flavonoid and sulfone flavonoid
CN111039998A (en) * 2019-12-19 2020-04-21 江南大学 Preparation method of 8-benzenesulfonyl substituted flavone glucose glycoside
CN112830917A (en) * 2021-01-18 2021-05-25 五邑大学 Hydroxylated polymethoxyflavone and preparation method thereof
CN113350331A (en) * 2021-05-21 2021-09-07 广州医科大学附属第一医院(广州呼吸中心) Application of hesperetin and/or derivatives thereof in preparation of drugs or drugs for preventing and/or treating renal cell carcinoma
CN113350331B (en) * 2021-05-21 2023-08-29 广州医科大学附属第一医院(广州呼吸中心) Application of hesperetin and/or derivative thereof in preparation of or application of hesperetin and/or derivative thereof in preparation of medicines for preventing and/or treating renal cell carcinoma

Also Published As

Publication number Publication date
GB201407694D0 (en) 2014-06-18

Similar Documents

Publication Publication Date Title
CN116370479B (en) Use of compound ATV014 in the preparation of a product for the treatment of novel coronavirus infection
EP3781564B1 (en) Pyridazine derivatives for the treatment of cancer
JP6496296B2 (en) Inhibitors of human EZH2 and methods of use thereof
EP3212189B1 (en) Substituted chromanes and method of use
WO2015166041A1 (en) Flavones as cyp1a1 inhibitors for the treatment of cancer
EP3148543B1 (en) Small molecule transcription modulators of bromodomains
JP5124443B2 (en) Novel isoindole derivatives, compositions containing them, their preparation and their pharmaceutical use, in particular as inhibitors of chaperone protein HSP90 activity
Potze et al. Betulinic acid kills colon cancer stem cells
EP2763983A1 (en) L-pyrazolyl-3- (4- ( (2 -anilinopyrimidin- 4 - yl) oxy) napththalen- 1 - yl) ureas as p3i map kinase inhibitors
CA2612979A1 (en) Parp modulators and treatment of cancer
KR102338568B1 (en) Novel quinazoline derivative
JP7237383B2 (en) Compositions for inhibition of androgen-dependent or -independent prostate cancer cells and pharmaceutical formulations for prostate cancer containing same
WO2015166043A1 (en) Compounds
US9879020B2 (en) GABAA agonists and methods of using to control airway hyperresponsiveness and inflammation in asthma
WO2015166042A1 (en) Stilbene derivatives and their use for the treatment of cancer
WO2015166040A2 (en) Compound
WO2015157382A1 (en) Treatment of neurodegenerative diseases with asparagine endopeptidase (aep) inhibitors and compositions related thereto
KR101149878B1 (en) New compounds having a spiro chiral carbon, process for preparing the same and pharmaceutical composition comprising the same
AU2007289232A1 (en) Therapeutic methods using WRN binding molecules
CN100535000C (en) Method of preparing cyclopamine from jervine
KR101510995B1 (en) Pharmaceutical composition for treating or preventing microRNA-31 related diseases
WO2018204987A1 (en) Carbonic anhydrase inhibitors
EP4340887A1 (en) Bifunctional protac-type compounds targeting pxr, method for preparing same, and therapeutic use thereof
CN111808056A (en) Dopamine D2Receptor selective agonists and their use in the treatment of disease
KR100874209B1 (en) 3,10-dihydro-1h-[1,4]diazepino[5,6-b]indol-2-one derivatives and pharmaceutically acceptable salt thereof, process for preparation thereof and use thereof

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: 15719230

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: 15719230

Country of ref document: EP

Kind code of ref document: A1