US20070203217A1 - Substituted stilbenes and their reactions - Google Patents

Substituted stilbenes and their reactions Download PDF

Info

Publication number
US20070203217A1
US20070203217A1 US11/744,405 US74440507A US2007203217A1 US 20070203217 A1 US20070203217 A1 US 20070203217A1 US 74440507 A US74440507 A US 74440507A US 2007203217 A1 US2007203217 A1 US 2007203217A1
Authority
US
United States
Prior art keywords
compound
alkyl
group
compounds
independently selected
Prior art date
Legal status (The legal status 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 status listed.)
Abandoned
Application number
US11/744,405
Inventor
John Hadfield
Alan McGown
Stephen Mayalarp
Edward Land
Ian Hamblett
Keira Gaukroger
Nicholas Lawrence
Lucy Hepworth
John Butler
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Universiteit Utrecht Holding BV
Original Assignee
Universiteit Utrecht Holding BV
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from GB0031262A external-priority patent/GB0031262D0/en
Priority claimed from GB0100295A external-priority patent/GB0100295D0/en
Application filed by Universiteit Utrecht Holding BV filed Critical Universiteit Utrecht Holding BV
Priority to US11/744,405 priority Critical patent/US20070203217A1/en
Publication of US20070203217A1 publication Critical patent/US20070203217A1/en
Assigned to UNIVERSITEIT UTRECHT UU HOLDING B.V. reassignment UNIVERSITEIT UTRECHT UU HOLDING B.V. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: KVERKA, MILOSLAV, TLASKALOVA-HOGENOVA, HELENA, VAN DER ZEE, RUURD, VAN EDEN, WILLEM
Priority to US13/588,331 priority patent/US8853270B2/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F7/00Compounds containing elements of Groups 4 or 14 of the Periodic System
    • C07F7/02Silicon compounds
    • C07F7/08Compounds having one or more C—Si linkages
    • C07F7/18Compounds having one or more C—Si linkages as well as one or more C—O—Si linkages
    • C07F7/1804Compounds having Si-O-C linkages
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C205/00Compounds containing nitro groups bound to a carbon skeleton
    • C07C205/27Compounds containing nitro groups bound to a carbon skeleton the carbon skeleton being further substituted by etherified hydroxy groups
    • C07C205/35Compounds containing nitro groups bound to a carbon skeleton the carbon skeleton being further substituted by etherified hydroxy groups having nitro groups and etherified hydroxy groups bound to carbon atoms of six-membered aromatic rings of the carbon skeleton
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C205/00Compounds containing nitro groups bound to a carbon skeleton
    • C07C205/27Compounds containing nitro groups bound to a carbon skeleton the carbon skeleton being further substituted by etherified hydroxy groups
    • C07C205/35Compounds containing nitro groups bound to a carbon skeleton the carbon skeleton being further substituted by etherified hydroxy groups having nitro groups and etherified hydroxy groups bound to carbon atoms of six-membered aromatic rings of the carbon skeleton
    • C07C205/36Compounds containing nitro groups bound to a carbon skeleton the carbon skeleton being further substituted by etherified hydroxy groups having nitro groups and etherified hydroxy groups bound to carbon atoms of six-membered aromatic rings of the carbon skeleton to carbon atoms of the same non-condensed six-membered aromatic ring or to carbon atoms of six-membered aromatic rings being part of the same condensed ring system
    • C07C205/37Compounds containing nitro groups bound to a carbon skeleton the carbon skeleton being further substituted by etherified hydroxy groups having nitro groups and etherified hydroxy groups bound to carbon atoms of six-membered aromatic rings of the carbon skeleton to carbon atoms of the same non-condensed six-membered aromatic ring or to carbon atoms of six-membered aromatic rings being part of the same condensed ring system the oxygen atom of at least one of the etherified hydroxy groups being further bound to an acyclic carbon atom
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C205/00Compounds containing nitro groups bound to a carbon skeleton
    • C07C205/44Compounds containing nitro groups bound to a carbon skeleton the carbon skeleton being further substituted by —CHO groups
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C271/00Derivatives of carbamic acids, i.e. compounds containing any of the groups, the nitrogen atom not being part of nitro or nitroso groups
    • C07C271/06Esters of carbamic acids
    • C07C271/08Esters of carbamic acids having oxygen atoms of carbamate groups bound to acyclic carbon atoms
    • C07C271/10Esters of carbamic acids having oxygen atoms of carbamate groups bound to acyclic carbon atoms with the nitrogen atoms of the carbamate groups bound to hydrogen atoms or to acyclic carbon atoms
    • C07C271/22Esters of carbamic acids having oxygen atoms of carbamate groups bound to acyclic carbon atoms with the nitrogen atoms of the carbamate groups bound to hydrogen atoms or to acyclic carbon atoms to carbon atoms of hydrocarbon radicals substituted by carboxyl groups
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C43/00Ethers; Compounds having groups, groups or groups
    • C07C43/02Ethers
    • C07C43/20Ethers having an ether-oxygen atom bound to a carbon atom of a six-membered aromatic ring
    • C07C43/215Ethers having an ether-oxygen atom bound to a carbon atom of a six-membered aromatic ring having unsaturation outside the six-membered aromatic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C43/00Ethers; Compounds having groups, groups or groups
    • C07C43/02Ethers
    • C07C43/20Ethers having an ether-oxygen atom bound to a carbon atom of a six-membered aromatic ring
    • C07C43/23Ethers having an ether-oxygen atom bound to a carbon atom of a six-membered aromatic ring containing hydroxy or O-metal groups
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C46/00Preparation of quinones
    • C07C46/02Preparation of quinones by oxidation giving rise to quinoid structures
    • C07C46/06Preparation of quinones by oxidation giving rise to quinoid structures of at least one hydroxy group on a six-membered aromatic ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C50/00Quinones
    • C07C50/26Quinones containing groups having oxygen atoms singly bound to carbon atoms
    • C07C50/30Quinones containing groups having oxygen atoms singly bound to carbon atoms with polycyclic non-condensed quinoid structure
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/60Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D213/62Oxygen or sulfur atoms
    • C07D213/63One oxygen atom
    • C07D213/68One oxygen atom attached in position 4

Definitions

  • the present invention relates to novel compounds, and more particularly to stilbene and quinone compounds related to combretastatin A-4 and their possible use as anticancer compounds and prodrugs.
  • the present invention relates to the photochemical reactions of some of these compounds, in the photochemical isomerisation of the compounds and/or the photochemical release of an active compound from a protected compound (prodrug).
  • the stilbene cis-combretastatin A-4 Z-1 isolated from the African bush willow, Combretum caffrum shows exciting potential as an anticancer agent, binding strongly to tubulin and displaying potent and selective toxicity toward tumour vasculature (U.S. Pat. No: 4,996,237, Arizona Board of Regents, Pettit et al, Experimentia, 1989, 45, 209; Lin et al, Mol. Pharmacol., 1988, 34, 200; Grosios et al, Brit. J. Cancer, 1999, 81, 1318; Lin et al, Biochemistry, 1989, 28, 6984; Woods et al, Brit. J.
  • Cis-combretastatin A-4 Z-1 is able to inhibit cell growth at low concentrations (IC 50 , P388 murine leukaemia cell line 2.6 nM).
  • the potency of trans-combretastatin A-4 E-1 is much lower and inhibits cell growth in the ⁇ M range.
  • it is the ability of Z-1 and Z-2 to destroy tumour blood vessels, effectively starving tumours of nutrients, which makes them such exciting molecules.
  • Tumour vasculature and the formation of neovasculature were first identified as a target for cancer therapy by Judah Folkman some 30 years ago. The work of Folkman and others has clearly identified angiogenesis and blood supply as necessary requirements for primary tumour growth, invasiveness and metastasis. It is now becoming clear that the selective destruction of tumour vasculature will have a significant impact on the clinical treatment of cancer.
  • Angiogenesis is subject to a complex process of regulation and thereby offers a multitude of molecular targets for drug design.
  • Z-1 as a clinically useful anticancer agent has been severely hampered by its poor water solubility (Brown et al, J. Chem. Soc., Perkin Trans. 1, 1995, 577).
  • the phosphate salt Z-2 is more soluble in water than Z-1 and is soon to enter phase II clinical trials (Pettit et al, Anti - Cancer Drug Des., 1995, 10, 299). Nevertheless, both Z-1 and Z-2 are not targeted towards cancer cells and their therapeutic efficacy would be improved if their selectivity were better.
  • No: 5,561,122 discloses the sodium and potassium salts of cis-combretastatin A-4 and a hemisuccinic acid ester derivative
  • WO99/35150 discloses the lithium, caesium, magnesium, calcium, manganese and zinc salts of cis-combretastatin A-4, and ammonium cation salts with imidazole, morpholine, piperazine, piperidine, pyrazole, pyridine, adenosine, cinchonine, glucosamine, quinine, quinidine, tetracycline and verapamil.
  • both compounds target tubulin, binding strongly at or close to the colchicine (3) binding site, preventing polymerisation of ⁇ , ⁇ -tubulin heterodimer to microtubules.
  • Their inhibition of microtubule formation prevents mitosis and is important in disrupting the growth of new vascular epithelial cells.
  • disruption of the intracellular microtubule networks by combretastatin A4 leads to the destruction of microvessels within the tumour.
  • This antivascular activity offers exciting therapeutic possibilities as the destruction of microvessels results in the death of all tumour cells which depend on the vessel for nutrients and oxygen.
  • the multi-functional role of tubulin in both healthy and cancer cells highlights the need for selectively targeted drugs.
  • the present invention relates to novel compounds and more particularly to stilbene and quinone compounds related to combretastatin A-4.
  • the synthesis of new compounds is disclosed herein, together with experiments demonstrating their activity in vitro and in vivo, supporting their use as anticancer compounds and prodrugs.
  • the compounds include those with an alkyl group on the double bond of cis or trans-stilbenes, compounds with one or more (and preferably 2 or 3) alkyl group substituents on the stilbene A ring, compounds with an alkoxy group other than methoxy at position 3, 4, and/or 5 of the stilbene A ring, compounds (or prodrugs) in which BOC amino acid esters are formed with the phenolic hydroxyl at the 3-position of the B ring and compounds (or prodrugs) based on a benzoquinone B ring.
  • the present invention relates to the photochemical reactions of stilbene compounds, either the above compounds disclosed for the first time herein or compounds based on prior art stilbenes.
  • These reactions include the photochemical release of an active form of the compound from a prodrug conjugate and the photochemical isomerisation of the compounds, especially from a trans to cis form of compounds.
  • the reactions can be used alone or in combination to convert inactive or comparatively less active forms of the compounds to more active forms, thereby allowing the compounds to be selectively targeted, e.g. activating them at the site of a tumour.
  • the present invention provides a compound represented by the structural formula: wherein:
  • alkyl or alkoxy substituents are substituted or unsubstituted C 1-10 alkyl or alkoxy groups.
  • the alkyl chain can be straight chain or branched.
  • Preferred alkyl substituents are methyl or ethyl.
  • Preferred alkoxy substituents are methoxy or ethoxy.
  • Halogen substituents can be fluorine, chlorine, bromine or iodine, and are preferably fluorine.
  • the haloalkyl groups are fluoroalkyl, and most preferably is a CF 3 group.
  • the O-ester group is represented by the formula O-phosphate, OCO-alkyl, OCO-aryl, OCO-heteroaryl, OCO-amino acid, OCO-peptide, OCO-polymer, OCO-sugar or OCO—CHR—NH—BOC, where BOC represents a t-butoxycarbonyl group.
  • R and R′ are substituted or unsubstituted C 1-10 alkyl groups.
  • R′′ is preferably selected from substituted or unsubstituted alkyl (e.g. C 1-10 ), aryl or heteroaryl groups.
  • the present invention provides compounds in which there are one or more alkyl groups present on the double bond linking the stilbene A and B rings.
  • the present invention provides compounds represented by the structural formula: wherein:
  • the compounds in this aspect of the invention may be either the cis or Z-isomer, i.e. be related to combretastatin A4, or the trans or E-isomer. Examples of the synthesis of both isomers are proved below.
  • the alkyl group R 4 and/or R 5 is a methyl or ethyl group.
  • the present invention provides compounds in which one or more of the methoxy groups on the A ring of combretastatin is replaced by an alkyl group.
  • the present invention provides compounds represented by the structural formula: wherein:
  • two or more preferably all three of the groups are alkyl groups.
  • Exemplary compounds include those with methyl, ethyl or propyl groups.
  • R 6 , R 7 and R 8 are methyl groups.
  • the present invention provides compounds in which one or more of the methoxy groups on the A ring of combretastatin is replaced by a higher alkoxy group, i.e. an ethoxy or longer chain group.
  • the present invention provides compounds represented by the structural formula: wherein:
  • two or all three of the groups is replaced by an alkoxy group other than methoxy.
  • the present invention relates to compounds in which the phenolic hydroxyl group on the B ring of the combretastatin is derivatised to form a t-BOC-amino acid ester.
  • These compounds may be prodrugs capable of releasing combretastatin, or a variant thereof, e.g. by the action of an enzyme capable of hydrolysing the BOC-amino acid ester, e.g. an esterase enzyme.
  • the present invention provides compounds represented by the structural formula: wherein:
  • the BOC amino acid ester may include a naturally occurring or synthetic amino acid, in either the D or L-isoform.
  • Examples of compounds of the aspect of the invention include those where the amino acid is Phe, Ile, Gly, Trp, Met, Leu, Ala, His, Pro, D-Met, D-Trp, or Tyr, e.g. when in compound 33 the amino acid is Phe, the A group is —CH 2 Ph etc.
  • the present invention provides compounds in which the B ring of combretastatin is replaced by a substituted or unsubstituted benzoquinone ring.
  • These quinone compounds may act as prodrugs of combretastatin and be activated in vivo by enzymes such as DT-diaphorase.
  • the present invention provides compounds represented by the structural formula: wherein:
  • the present invention also includes compositions comprising one or more of the above defined compounds.
  • the present invention provides the compounds for use in a method of medical treatment and the use of the compounds for the preparation of a medicament for the treatment of a condition that responds to the medicament, and in particular for the treatment of cancer.
  • the compounds may act directly or be prodrugs capable of releasing an active form of the compound upon hydrolysis or reduction, e.g. as mediated in situ by an enzyme.
  • the present invention provides a prodrug comprising a compound conjugated to a photocleavable group, wherein the prodrug is represented by the general formula: wherein:
  • the compounds conjugated to the photocleavable group to form the prodrug may be the new combretastatin derivatives disclosed herein or may be a known combretastatin which has not be conjugated in this way in the prior art.
  • the prodrugs can be activated by exposure to electromagnetic radiation, especially ultraviolet-visible light (e.g. having a wavelength of between about 190-1000 nm), to remove the protecting photocleavable group and cause the release of the compound.
  • electromagnetic radiation especially ultraviolet-visible light (e.g. having a wavelength of between about 190-1000 nm)
  • the prodrugs can be used to provide selective activation of the active form of the compound, e.g. at the site of a tumour, by administering the compound and exposing to light the site at which activation is required.
  • Particularly preferred compounds are those which can be exposed to light to release combretastatin, and especially cis-combretastatin A4.
  • Examples of preferred compounds include those which Z, the photocleavable group is selected from:
  • R is the photoprotected group
  • R 9 and R 10 are independently selected from alkyl, aryl or heteroaryl
  • X is any functional group. Examples of photoactivatable groups are also provided on pages 54-59.
  • the present invention provides the compounds as defined herein for use in a method of medical treatment.
  • the present invention provides the use of the compounds defined herein for the preparation of a medicament for the treatment of a condition that is ameliorated by administration of the activated or released form of the compound.
  • the activated form of the compound has significantly greater activity than the protected form of the compound, e.g. making it possible to obtain selectivity in the delivery and activation of the compound, e.g. to a target tissue.
  • the compounds are employed in medicaments for the treatment of cancer.
  • the present invention provides a process for providing the compound at a site, the process comprising exposing a prodrug represented by the above formula to light to release the compound at the site.
  • the light is in the visible range, e.g. from about 350-800 nm.
  • the present invention provides a process for isomerising a compound represented by the general formula: wherein:
  • the present invention provides a process for producing the photoactivatable compounds defined herein, the process comprising linking a photoactivatable group to the Y group of a precursor compound to produce photoactivatable compounds as defined above.
  • the nitro vanillin derivative was chosen since it has been used as a photo-cleavable linker for solid phase synthesis applications and its synthesis is relatively simple.
  • These prodrugs have been successfully cleaved in both the E and Z series (E-6 and Z-6 respectively) and have produced highly cytotoxic agents in vitro upon in situ exposure to ultra violet radiation.
  • the cleavage of the water solubilising group appears to be faster than the E ⁇ Z isomerisation, at least under ex situ irradiation.
  • Z-6 has some merit. Indeed it forms the prototype for systems that do not rely on any special photochemical features of the molecule to be delivered. This provides a more general approach to the site-specific photochemical activation of prodrugs.
  • the photocleavable water solubilising group can be engineered, so that cleavage occurs at longer wavelength and rapidly.
  • FIG. 1 shows the anti-tumour activity of compound 97-64H in an in vivo tumour implant experiment in mice.
  • FIG. 2 shows the anti-tumour activity of compound 97-96 in an in vivo tumour implant experiment in mice.
  • the compounds of the invention may be derivatised in various ways.
  • “derivatives” of the compounds includes salts, esters such as in vivo hydrolysable esters, free acids or bases, hydrates, prodrugs or coupling partners.
  • esters such as in vivo hydrolysable esters, free acids or bases, hydrates, prodrugs or coupling partners.
  • the derivatives are soluble in water and/or saline or can be hydrolysed to provide physiologically active agents.
  • Salts of the compounds of the invention are preferably physiologically well tolerated and non toxic. Many examples of salts are known to those skilled in the art.
  • Compounds having acidic groups can form salts with alkaline or alkaline earth metals such as Na, K, Mg and Ca, and with organic amines such as triethylamine and Tris (2-hydroxyethyl)amine. Salts can be formed between compounds with basic groups, e.g. amines, with inorganic acids such as hydrochloric acid, phosphoric acid or sulfuric acid, or organic acids such as acetic acid, citric acid, benzoic acid, fumaric acid, or tartaric acid.
  • Compounds having both acidic and basic groups can form internal salts.
  • Esters can be formed between hydroxyl or carboxylic acid groups present in the compound and an appropriate carboxylic acid or alcohol reaction partner, using techniques well known in the art. Examples of esters include those formed between the phenolic hydroxyl of the substituted stilbenes and carboxylic acids, hemisuccinic acid esters, phosphate esters, BOC esters, sulphate esters and selenate esters.
  • prodrugs which as prodrugs of the compounds are convertible in vivo or in vitro into one of the parent compounds. Typically, at least one of the biological activities of compound will be reduced in the prodrug form of the compound, and can be activated by conversion of the prodrug to release the compound or a metabolite of it.
  • prodrugs include combretastatin A1 phosphate, combretastatin A4 phosphate and RH1.
  • Coupled derivatives include coupling partners of the compounds in which the compounds is linked to a coupling partner, e.g. by being chemically coupled to the compound or physically associated with it.
  • Examples of coupling partners include a label or reporter molecule, a supporting substrate, a carrier or transport molecule, an effector, a drug, an antibody or an inhibitor.
  • Coupling partners can be covalently linked to compounds of the invention via an appropriate functional group on the compound such as a hydroxyl group, a carboxyl group or an amino group.
  • the compounds described herein or their derivatives can be formulated in pharmaceutical compositions, and administered to patients in a variety of forms, in particular to treat conditions which are ameliorated by the activation of the compound.
  • compositions for oral administration may be in tablet, capsule, powder, cream, liquid form or encapsulated by liposomes.
  • a tablet may include a solid carrier such as gelatin or an adjuvant or an inert diluent.
  • Liquid pharmaceutical compositions generally include a liquid carrier such as water, petroleum, animal or vegetable oils, mineral oil or synthetic oil. Physiological saline solution, or glycols such as ethylene glycol, propylene glycol or polyethylene glycol may be included.
  • Such compositions and preparations generally contain at least 0.1 wt % of the compound.
  • Parental administration includes administration by the following routes: intravenous, cutaneous or subcutaneous, nasal, intramuscular, intraocular, transepithelial, intraperitoneal and topical (including dermal, ocular, rectal, nasal, inhalation and aerosol), and rectal systemic routes.
  • intravenous, cutaneous or subcutaneous injection, or injection at the site of affliction the active ingredient will be in the form of a parenterally acceptable aqueous solution which is pyrogen-free and has suitable pH, isotonicity and stability.
  • suitable solutions using, for example, solutions of the compounds or a derivative thereof, e.g. in physiological saline, a dispersion prepared with glycerol, liquid polyethylene glycol or oils.
  • compositions can comprise one or more of a pharmaceutically acceptable excipient, carrier, buffer, stabiliser, isotonicizing agent, preservative or anti-oxidant or other materials well known to those skilled in the art. Such materials should be non-toxic and should not interfere with the efficacy of the active ingredient.
  • a pharmaceutically acceptable excipient such materials should be non-toxic and should not interfere with the efficacy of the active ingredient.
  • the precise nature of the carrier or other material may depend on the route of administration, e.g. orally or parentally.
  • Liquid pharmaceutical compositions are typically formulated to have a pH between about 3.0 and 9.0, more preferably between about 4.5 and 8.5 and still more preferably between about 5.0 and 8.0.
  • the pH of a composition can be maintained by the use of a buffer such as acetate, citrate, phosphate, succinate, Tris or histidine, typically employed in the range from about 1 mM to 50 mM.
  • the pH of compositions can otherwise be adjusted by using physiologically acceptable acids or bases.
  • Preservatives are generally included in pharmaceutical compositions to retard microbial growth, extending the shelf life of the compositions and allowing multiple use packaging.
  • preservatives include phenol, meta-cresol, benzyl alcohol, para-hydroxybenzoic acid and its esters, methyl paraben, propyl paraben, benzalconium chloride and benzethonium chloride.
  • Preservatives are typically employed in the range of about 0.1 to 1.0% (w/v).
  • the pharmaceutically compositions are given to an individual in a “prophylactically effective amount” or a “therapeutically effective amount” (as the case may be, although prophylaxis may be considered therapy), this being sufficient to show benefit to the individual. Typically, this will be to cause a therapeutically useful activity providing benefit to the individual.
  • the actual amount of the compounds administered, and rate and time-course of administration, will depend on the nature and severity of the condition being treated. Prescription of treatment, e.g. decisions on dosage etc, is within the responsibility of general practitioners and other medical doctors, and typically takes account of the disorder to be treated, the condition of the individual patient, the site of delivery, the method of administration and other factors known to practitioners.
  • compositions are preferably administered to patients in dosages of between about 0.01 and 100 mg of active compound per kg of body weight, and more preferably between about 0.5 and 10 mg/kg of body weight.
  • the compounds may be used in the treatment of cancer and other conditions involving abnormal proliferation of vasculature including diabetic retinopathy, psoriasis and endometriosis.
  • the ethyl derivative (45) has also been synthesised.
  • This method was also used to synthesise 98-40, 98-23, 98-33 and 98-24.
  • quinones may act as prodrugs producing the active hydroquinones on reduction by enzymes.
  • 97-64H is orally bio-available when administered at 200 mg/kg in 5% dimethylacetamide in arachis oil, 97-64H showed a Cmax of 1.49 ug/ml, an absorption half life 15 mins, and an elimination half life 32 mins.
  • the 3-position of the B-ring can also be substituted with larger groups such as boc-amino acid esters, pyridyl esters, and ethers.
  • the amino acid esters (33-44) are prodrugs which, upon the action of esterase, release the active agent.
  • the activity of these agents is related to the rate of hydrolysis of these agents by esterase. The most active compounds being those which are most readily hydrolysed. The activity of these compounds indicates that ester linked polymers and peptides would be good prodrugs for agents of this type (see Table 1).
  • the pyridyl esters (96-167 and 97-07) are potent inhibitors of tubulin assembly and can displace 3 H-colchicine from tubulin without the action of esterase, showing that the 3-position of the B-ring can be substituted with bulky side groups. Potent growth inhibitory activity and good activity in the shape-change assay can also be seen for the tetrafluoropyridyl ether (97-13) and other ethers (98-29) which are not a substrate for esterase.
  • prodrugs capable of being activated by DT-diaphorase, an enzyme over-expressed in a wide range of human cancers and in endothelial cells of the vasculature were synthesised.
  • the rationale for this is that these agents will be activated in situ by the tumours over-expressing the enzyme, thus giving rise to a high local concentration of active drug. This confers selectivity to this agent.
  • quinone prodrugs (97-96 and 98-23) were tested for their ability to act as substrates for DT-diaphorase.
  • RH1 2,5,-diaziridinyl-3-(hydroxymethyl)-6-methyl-1,4-benzoquinone
  • RH1 2,5,-diaziridinyl-3-(hydroxymethyl)-6-methyl-1,4-benzoquinone
  • An analysis of the growth inhibitory properties of these agents shows that 97-96 is 16-fold more active in the H460 human lung cell line which expresses active DT-diaphorase than in the diaphorase null H596 cell line 97-96 is active in an H460 human lung tumour xenograft, a tumour which expresses active DT-diaphorase.
  • This agent may therefore have a role in the treatment of tumours that over-express DT-diaphorase.
  • these agents maybe selectively activated in the endothelial cells of the vasculature, thereby conferring a selective anti-vascular effect.
  • IC 50 are in nM.
  • the pale pink solid 4 was used without further purification (2.7 g, 82%). m.p. 69° C. [lit. (D. L. McMinn and M. M. Greenberg, Tetrahedron, 1996, 52, 3827) m.p.
  • the desired compound 7 was furnished as a yellow oil.
  • the oil was then purified via column chromatography (SiO 2 , hexane:EtOAc, 2:1 v/v) thus providing 7 as a yellow powder (580 mg, 64%). m.p.
  • Table 3 shows the retention times, by GC for the individual components of the reaction mixture.
  • Table 4 shows the retention times, by GC for the individual components of the reaction mixture.
  • TABLE 3 Time (mins) % trans-stilbene % cis-stilbene % phenanthrene 0 100 0 0 0.5 100 0 0 1.5 100 0 0 3.0 87 13 0 5.0 69 31 0 6.5 55 45 0 8.0 53 47 0 10.0 44 53 0 12.5 32 68 0 15.0 29 71 0 17.5 22 74 4 20.0 23 73 4
  • Table 5 shows the outcome of photoisomerisation of trans-CA-4 (99.7% E) and the corresponding IC 50 values.
  • the cytotoxicity (IC 50 value) of the reaction mixture at each time point is shown in Table 3, showing an impressive increase in the cytotoxicity of the mixture over time.
  • K562 cells were dosed with a known concentration of trans-CA-4, the resulting solutions were then irradiated using an ultra violet lamp consisting of 2 ⁇ 7 W ultra violet tubes for given lengths of time. Following irradiation, the cells were incubated in the normal way and the IC 50 values were determined.
  • trans-CA-4 was dissolved in benzene and the resulting solution was irradiated for a total of 20 minutes with aliquots removed at given time points to determine the isomeric ratios and the IC 50 value.
  • the results obtained are detailed in Table 8.
  • K562 cells were dosed with a known concentration of coupled trans-CA-4 E-8 and subsequently exposed to ultra violet radiation for specific lengths of time and the IC 50 value determined.
  • a series of control experiments were performed simultaneously to verify that any positive results obtained were due to the effect of ultra violet light on the drug candidate and not its effect on the cells. These consisted of i) exposing KS562 cells only (i.e.

Abstract

The present invention relates to stilbene and quinine compounds related to combretastatin A-4 and their use as anticancer compounds and prodrugs. The compounds include those with an alkyl group on the double bond of cis or trans-stilbenes, compounds with one or more (and preferably 2 or 3) alkyl group substituents on the stilbene a ring, compounds with an alkoxy group other than methoxy at position 3, 4 and/or 5 of the stilbene A ring, compounds (or prodrugs) in which BOC amino acid esters are formed with the phenolic hydroxyl at the 3-position of the B ring and compounds (or prodrugs) based on a benzoquinone B ring. The present invention further relates to the photochemical reactions of stilbene compounds, either the above compounds disclosed for the first time herein or compounds based on prior art stilbenes. These reactions include the photochemical release of an active form of the compound from a prodrug conjugate and the photochemical isomerisation of the compounds, especially from a trans to cis form of compounds. The reactions can be used alone or in combination to convert inactive or comparatively less active forms of the compounds to more active forms, thereby allowing the compounds to be selectively targeted, e.g., activating them at the site of a tumour.

Description

    FIELD OF THE INVENTION
  • The present invention relates to novel compounds, and more particularly to stilbene and quinone compounds related to combretastatin A-4 and their possible use as anticancer compounds and prodrugs. In further aspects, the present invention relates to the photochemical reactions of some of these compounds, in the photochemical isomerisation of the compounds and/or the photochemical release of an active compound from a protected compound (prodrug).
  • BACKGROUND OF THE INVENTION
  • The stilbene cis-combretastatin A-4 Z-1, isolated from the African bush willow, Combretum caffrum shows exciting potential as an anticancer agent, binding strongly to tubulin and displaying potent and selective toxicity toward tumour vasculature (U.S. Pat. No: 4,996,237, Arizona Board of Regents, Pettit et al, Experimentia, 1989, 45, 209; Lin et al, Mol. Pharmacol., 1988, 34, 200; Grosios et al, Brit. J. Cancer, 1999, 81, 1318; Lin et al, Biochemistry, 1989, 28, 6984; Woods et al, Brit. J. Cancer, 1995, 71, 705; McGown et al, Cancer Chemother. Pharmacol., 1990, 26, 79; El-Zayat et al, Anti-Cancer Drugs, 1993, 4, 19; Dark et al, Cancer Research, 1997, 57, 1829.).
    Figure US20070203217A1-20070830-C00001
  • Cis-combretastatin A-4 Z-1 is able to inhibit cell growth at low concentrations (IC50, P388 murine leukaemia cell line 2.6 nM). The potency of trans-combretastatin A-4 E-1 is much lower and inhibits cell growth in the μM range. Arguably, it is the ability of Z-1 and Z-2 to destroy tumour blood vessels, effectively starving tumours of nutrients, which makes them such exciting molecules. Tumour vasculature and the formation of neovasculature were first identified as a target for cancer therapy by Judah Folkman some 30 years ago. The work of Folkman and others has clearly identified angiogenesis and blood supply as necessary requirements for primary tumour growth, invasiveness and metastasis. It is now becoming clear that the selective destruction of tumour vasculature will have a significant impact on the clinical treatment of cancer. Angiogenesis is subject to a complex process of regulation and thereby offers a multitude of molecular targets for drug design.
  • The use of Z-1 as a clinically useful anticancer agent has been severely hampered by its poor water solubility (Brown et al, J. Chem. Soc., Perkin Trans. 1, 1995, 577). The phosphate salt Z-2 is more soluble in water than Z-1 and is soon to enter phase II clinical trials (Pettit et al, Anti-Cancer Drug Des., 1995, 10, 299). Nevertheless, both Z-1 and Z-2 are not targeted towards cancer cells and their therapeutic efficacy would be improved if their selectivity were better. The low solubility of cis-combretastatin A-4 in water and saline has led to attempts in the art to make related compounds or prodrugs which retain the activity of cis-combretastatin A-4 as an anticancer agent and which have enhanced solubility. These attempts focus on forming salts or derivatives at the phenolic hydroxyl group of combretastatin. By way of example, U.S. Pat. No: 5,561,122 (Arizona Board of Regents) discloses the sodium and potassium salts of cis-combretastatin A-4 and a hemisuccinic acid ester derivative, and WO99/35150 (Arizona Board of Regents) discloses the lithium, caesium, magnesium, calcium, manganese and zinc salts of cis-combretastatin A-4, and ammonium cation salts with imidazole, morpholine, piperazine, piperidine, pyrazole, pyridine, adenosine, cinchonine, glucosamine, quinine, quinidine, tetracycline and verapamil.
  • At the molecular level, both compounds target tubulin, binding strongly at or close to the colchicine (3) binding site, preventing polymerisation of α,β-tubulin heterodimer to microtubules. Their inhibition of microtubule formation prevents mitosis and is important in disrupting the growth of new vascular epithelial cells. In addition, disruption of the intracellular microtubule networks by combretastatin A4 leads to the destruction of microvessels within the tumour. This antivascular activity offers exciting therapeutic possibilities as the destruction of microvessels results in the death of all tumour cells which depend on the vessel for nutrients and oxygen. The multi-functional role of tubulin in both healthy and cancer cells highlights the need for selectively targeted drugs.
  • We have previously investigated the tubulin-binding properties of agents related to Z-1 and 3 and as part of this effort, we have designed many related compounds that behave in a similar fashion to Z-1 (Ducki et al, Bioorg. Med. Chem. Lett., 1998, 8, 1051; Zhao et al, Eur. J. Nuc. Medicine, 1999, 26, 231; Aleksandrzak et al, Anti-Cancer Drugs, 1998, 9, 545). However, it remains a problem in the art in designing effective compounds and especially those which can be selectively targeted.
  • SUMMARY OF THE INVENTION
  • In a first group of aspects, the present invention relates to novel compounds and more particularly to stilbene and quinone compounds related to combretastatin A-4. The synthesis of new compounds is disclosed herein, together with experiments demonstrating their activity in vitro and in vivo, supporting their use as anticancer compounds and prodrugs. The compounds include those with an alkyl group on the double bond of cis or trans-stilbenes, compounds with one or more (and preferably 2 or 3) alkyl group substituents on the stilbene A ring, compounds with an alkoxy group other than methoxy at position 3, 4, and/or 5 of the stilbene A ring, compounds (or prodrugs) in which BOC amino acid esters are formed with the phenolic hydroxyl at the 3-position of the B ring and compounds (or prodrugs) based on a benzoquinone B ring.
  • In a further group of aspects, the present invention relates to the photochemical reactions of stilbene compounds, either the above compounds disclosed for the first time herein or compounds based on prior art stilbenes. These reactions include the photochemical release of an active form of the compound from a prodrug conjugate and the photochemical isomerisation of the compounds, especially from a trans to cis form of compounds. The reactions can be used alone or in combination to convert inactive or comparatively less active forms of the compounds to more active forms, thereby allowing the compounds to be selectively targeted, e.g. activating them at the site of a tumour.
  • Accordingly, in a first aspect, the present invention provides a compound represented by the structural formula:
    Figure US20070203217A1-20070830-C00002

    wherein:
      • X is selected from hydroxyl, nitro, amino, aryl, heteroaryl, alkyl, alkoxy, CHO, COR, halogen, haloalkyl, NH2, NHR, NRR′, SR, CONH2, CONHR, CONHRR′, O-aryl, O-heteroaryl or O-ester;
      • R1 is selected from alkyl, CHO, alkoxy, NH2, NHR, NRR′, SR, CF3 or halogen;
      • R2 and R3 are independently selected from hydrogen, alkyl, alkoxy, hydroxyl NH2, NHR, NRR′, SR, haloalkyl or halogen;
      • R4 and R5 are independently selected from hydrogen, alkyl, CH2NHCOR″ or CH2CONHR″; and,
      • R6, R7 and R8 are independently selected from hydrogen, alkyl or alkoxy;
      • or a salt or derivative thereof.
  • In all aspect of the invention, preferably, the substituents are chosen according to the following lists of preferred groups.
  • Preferably, alkyl or alkoxy substituents are substituted or unsubstituted C1-10 alkyl or alkoxy groups. In either case, the alkyl chain can be straight chain or branched.
  • Preferred alkyl substituents are methyl or ethyl. Preferred alkoxy substituents are methoxy or ethoxy.
  • Halogen substituents can be fluorine, chlorine, bromine or iodine, and are preferably fluorine. Preferably, the haloalkyl groups are fluoroalkyl, and most preferably is a CF3 group.
  • Preferably, the O-ester group is represented by the formula O-phosphate, OCO-alkyl, OCO-aryl, OCO-heteroaryl, OCO-amino acid, OCO-peptide, OCO-polymer, OCO-sugar or OCO—CHR—NH—BOC, where BOC represents a t-butoxycarbonyl group.
  • As used herein, preferably R and R′ are substituted or unsubstituted C1-10 alkyl groups. R″ is preferably selected from substituted or unsubstituted alkyl (e.g. C1-10), aryl or heteroaryl groups.
  • In a further aspect, the present invention provides compounds in which there are one or more alkyl groups present on the double bond linking the stilbene A and B rings. Thus, in this aspect, the present invention provides compounds represented by the structural formula:
    Figure US20070203217A1-20070830-C00003

    wherein:
      • the zigzag line indicates that the compound can be cis or trans;
      • X is selected from hydroxyl, nitro, amino, aryl, heteroaryl, alkyl, alkoxy, CHO, COR, halogen, haloalkyl, NH2, NHR, NRR′, SR, CONH2, CONHR, CONHRR′, O-aryl, O-heteroaryl or O-ester;
      • R1 is selected from alkyl, CHO, alkoxy, NH2, NHR, NRR′, S, CF3 or halogen;
      • R2 and R3 are independently selected from hydrogen, alkyl, alkoxy, hydroxyl, NH2, NHR, NRR′, SR, haloalkyl or halogen;
      • R4 and R5 are independently selected from hydrogen, alkyl, CH2NHCOR″ or CH2CONHR″; and,
      • R6, R7 and R8 are independently selected from hydrogen, alkyl or alkoxy;
        wherein at least one of the substituents R4 and R5 is an allcyl group.
      • or a salt or derivative thereof.
  • As defined above, the compounds in this aspect of the invention may be either the cis or Z-isomer, i.e. be related to combretastatin A4, or the trans or E-isomer. Examples of the synthesis of both isomers are proved below. Preferably, the alkyl group R4 and/or R5 is a methyl or ethyl group.
  • In a further aspect, the present invention provides compounds in which one or more of the methoxy groups on the A ring of combretastatin is replaced by an alkyl group. Thus, in this aspect, the present invention provides compounds represented by the structural formula:
    Figure US20070203217A1-20070830-C00004

    wherein:
      • X is selected from hydroxyl, nitro, amino, aryl, heteroaryl, alkyl, alkoxy, CHO, COR, halogen, haloalkyl, NH2, NHR, NRR′, SR, CONH2, CONHR, CONHRR′, O-aryl, O-heteroaryl or O-ester;
      • R1 is selected from alkyl, CHO, alkoxy, NH2, NHR, NRR′, SR, CF3 or halogen;
      • R2 and R3 are independently selected from hydrogen, alkyl, alkoxy, hydroxyl, NH2, NHR, NRR′, SR, haloalkyl or halogen;
      • R4 and R5 are independently selected from hydrogen, alkyl, CH2NHCOR″ or CH2CONHR″; and,
      • wherein R6, R7 and R8 are independently selected from hydrogen, alkyl or alkoxy such that at least one of these substituents is an alkyl group;
      • or a salt or derivative thereof.
  • In preferred embodiment, two or more preferably all three of the groups are alkyl groups. Exemplary compounds include those with methyl, ethyl or propyl groups. In a preferred embodiment, R6, R7 and R8 are methyl groups.
  • In a further aspect, the present invention provides compounds in which one or more of the methoxy groups on the A ring of combretastatin is replaced by a higher alkoxy group, i.e. an ethoxy or longer chain group. Thus, in this aspect, the present invention provides compounds represented by the structural formula:
    Figure US20070203217A1-20070830-C00005

    wherein:
      • X is selected from hydroxyl, nitro, amino, aryl, heteroaryl, alkyl, alkoxy, CHO, COR, halogen, haloalkyl, NH2, NHR, NRR′, SR, CONH2, CONHR, CONHRR′, O-aryl, O-heteroaiyl, or O-ester;
      • R1 is selected from alkyl, CHO, alkoxy, NH2, NHR, NRR′, SR, CF3 or halogen;
      • R2 and R3 are independently selected from hydrogen, alkyl, alkoxy, hydroxyl, NH2, NHR, NRR′, SR, haloalkyl or halogen;
      • R4 and R5 are independently selected from hydrogen, alkyl, CH2NHCOR″ or CH2CONHR″; and,
      • wherein R6, R7 and R8 are independently selected from hydrogen, alkyl or alkoxy such that at least one of these substituents is an alkoxy group other than methoxy group;
      • or a salt or derivative thereof.
  • Preferably, two or all three of the groups is replaced by an alkoxy group other than methoxy.
  • In a further aspect, the present invention relates to compounds in which the phenolic hydroxyl group on the B ring of the combretastatin is derivatised to form a t-BOC-amino acid ester. These compounds may be prodrugs capable of releasing combretastatin, or a variant thereof, e.g. by the action of an enzyme capable of hydrolysing the BOC-amino acid ester, e.g. an esterase enzyme. Thus, in this aspect, the present invention provides compounds represented by the structural formula:
    Figure US20070203217A1-20070830-C00006

    wherein:
      • R1 is selected from alkyl, alkoxy, NH2, NHR, NRR′, SR, CF3, CHO or halogen;
      • R2 and R3 are independently selected from hydrogen, alkyl, alkoxy, hydroxyl, NH2, NHR, NRR′, SR, haloalkyl or halogen;
      • R4 and R5 are independently selected from hydrogen, alkyl, CH2NHCOR″ or CH2CONHR″; and,
      • R6, R7 and R8 are independently selected from hydrogen, alkyl or alkoxy; and,
      • or a salt or derivative thereof;
      • wherein X is a group represented by:
        Figure US20070203217A1-20070830-C00007

        wherein BOC represents a t-butoxycarbonyl group and the A group is an amino acid side chain.
  • The BOC amino acid ester may include a naturally occurring or synthetic amino acid, in either the D or L-isoform. Examples of compounds of the aspect of the invention include those where the amino acid is Phe, Ile, Gly, Trp, Met, Leu, Ala, His, Pro, D-Met, D-Trp, or Tyr, e.g. when in compound 33 the amino acid is Phe, the A group is —CH2Ph etc.
  • In a further aspect, the present invention provides compounds in which the B ring of combretastatin is replaced by a substituted or unsubstituted benzoquinone ring. These quinone compounds may act as prodrugs of combretastatin and be activated in vivo by enzymes such as DT-diaphorase. Thus, in this aspect, the present invention provides compounds represented by the structural formula:
    Figure US20070203217A1-20070830-C00008

    wherein:
      • the dotted line indicates a single or double covalent bond and the zigzag line indicates that the compound can be cis or trans;
      • R1, R2 and R3 are independently selected from hydrogen, allyl, CHO, COR, alkoxy, hydroxyl, NH2, NHR, NRR′, SR, haloalkyl or halogen;
      • R4 and R5 are independently selected from hydrogen, alkyl, CH2NHCOR″ or CH2CONHR″; and,
      • R6, R7 and R8 are independently selected from hydrogen, alkyl or alkoxy; or
      • a salt or derivative thereof.
  • The present invention also includes compositions comprising one or more of the above defined compounds. In other aspects, the present invention provides the compounds for use in a method of medical treatment and the use of the compounds for the preparation of a medicament for the treatment of a condition that responds to the medicament, and in particular for the treatment of cancer. The compounds may act directly or be prodrugs capable of releasing an active form of the compound upon hydrolysis or reduction, e.g. as mediated in situ by an enzyme.
  • In the second group of aspects, the present invention provides a prodrug comprising a compound conjugated to a photocleavable group, wherein the prodrug is represented by the general formula:
    Figure US20070203217A1-20070830-C00009

    wherein:
      • R1 is selected from alkyl, CHO, alkoxy, NH2, NHR, NRR′, SR, CF3 or halogen;
      • R2 and R3 are independently selected from hydrogen, alkyl, alkoxy, hydroxyl, NH2, NHR, NRR′, SR, haloalkyl or halogen;
      • R4 and R5 are independently selected from hydrogen, allcyl, CH2NHCOR″ or CH2CONHR″; and,
      • R6, R7 and R8 are independently selected from hydrogen, alkyl or alkyoxy;
      • Y is selected from O, S, Se, NH; and,
      • Z is a photocleavable group;
      • or a salt or derivative thereof.
  • The compounds conjugated to the photocleavable group to form the prodrug may be the new combretastatin derivatives disclosed herein or may be a known combretastatin which has not be conjugated in this way in the prior art.
  • The prodrugs can be activated by exposure to electromagnetic radiation, especially ultraviolet-visible light (e.g. having a wavelength of between about 190-1000 nm), to remove the protecting photocleavable group and cause the release of the compound. Thus, the prodrugs can be used to provide selective activation of the active form of the compound, e.g. at the site of a tumour, by administering the compound and exposing to light the site at which activation is required.
  • Particularly preferred compounds (prodrugs) are those which can be exposed to light to release combretastatin, and especially cis-combretastatin A4.
  • Examples of preferred compounds include those which Z, the photocleavable group is selected from:
    Figure US20070203217A1-20070830-C00010
      • D. H. R Barton, Y. L. Chow, A. Cox, and G. W. Kirby, J. Chem. Soc., 1965, 3571.
        Figure US20070203217A1-20070830-C00011
      • T. Kishi, T. Tsuchiya and S. Umezawa, Bull. Chem. Soc. Jpn., 1979, 52, 3015.
        Figure US20070203217A1-20070830-C00012
      • L. D. Cama and B. G. Christensen, J. Am. Chem. Soc., 1978, 100, 8006.
        Figure US20070203217A1-20070830-C00013
  • Patchornik, A.; Amit, B.; Woodward R. B. J. Am. Chem. Soc., 1970, 92, 6333-6335 B. Amit, E. Hazum, M. Fridkin, and A. Patchornik, Int. J. Pept. Protein Res., 1977, 9, 91.
  • In the above formulae, R is the photoprotected group, R9 and R10 are independently selected from alkyl, aryl or heteroaryl and X is any functional group. Examples of photoactivatable groups are also provided on pages 54-59.
  • In a further aspect, the present invention provides the compounds as defined herein for use in a method of medical treatment. In preferred embodiments, the present invention provides the use of the compounds defined herein for the preparation of a medicament for the treatment of a condition that is ameliorated by administration of the activated or released form of the compound. In such uses, it is preferred that the activated form of the compound has significantly greater activity than the protected form of the compound, e.g. making it possible to obtain selectivity in the delivery and activation of the compound, e.g. to a target tissue. In preferred embodiments of the invention, the compounds are employed in medicaments for the treatment of cancer.
  • In a further aspect, the present invention provides a process for providing the compound at a site, the process comprising exposing a prodrug represented by the above formula to light to release the compound at the site. In this embodiment of the invention, preferably the light is in the visible range, e.g. from about 350-800 nm.
  • In a further aspect, the present invention provides a process for isomerising a compound represented by the general formula:
    Figure US20070203217A1-20070830-C00014

    wherein:
      • R1 is selected from alkyl, alkoxy, CHO, NH2, NHR, NRR′, SR, CF3 or halogen;
      • R2 and R3 are independently selected from hydrogen, alkyl, alkoxy, hydroxyl, NH2, NHR, NRR′, SR, haloalkyl or halogen;
      • R4 and R5 are independently selected from hydrogen, alkyl, CH2NHCOR″ or CH2CONHR″; and,
      • R6, R7 and R8 are independently selected from hydrogen, alkyl or alkoxy;
      • X is selected from hydroxyl, nitro, amino, aryl, heteroaryl, alkyl, alkoxy, CHO, COR, halogen, haloalkyl, NH2, NHR, NRR′, SR, CONH2, CONHR, CONHRR′, O-aryl, O-heteroaryl, O-ester, or the group Y-Z as defined above;
      • or a salt or derivative thereof;
      • the process comprising exposing the compound to light so that it isomerises from the E-isomer to the Z-isomer. This process might be carried out separately or in conjunction with the light activated release of the compound from a prodrug as defined above.
  • In a further aspect, the present invention provides a process for producing the photoactivatable compounds defined herein, the process comprising linking a photoactivatable group to the Y group of a precursor compound to produce photoactivatable compounds as defined above.
  • The work disclosed herein arises from the findings that the inactive trans isomer of combretastatin A-4 E-1 can be converted to the active cis-isomer Z-1 by the action of ultraviolet light ex situ in a photochemical reactor. The irradiation of E-1 in this manner leads to an impressive and rapid increase in activity. Further we have found that only after a long period of irradiation is the formation of the phenanthrene (which is only moderately active, as measured by its ability to inhibit cancer cell growth in vitro, IC50 0.7 μM) evident. We have prepared phenanthrene, by irradiation of E-1, in good yield when an oxidant (I2) is present (to oxidize the first-formed cyclization product). This provides an opportunity to exploit the hypoxic nature of solid tumours and increase the selectivity of irradiated E-1. In other words, healthy cells may provide an oxidative pathway for the formation of the less toxic phenanthrene and effectively decrease the lifetime of Z-1.
  • The same result can be achieved in situ in the presence of cultured cancer cells (K562 human myelogeneous leukaemia cell line. These experiments showed that within 2 seconds of exposure to ultraviolet light the activity of the E-combretastatin A-4 (IC50 originally 5 μM) increases to 2 nM, providing a rapid thousand-fold increase in activity. Moreover, the cells in the absence of the drug are not affected by exposure to the radiation and grow normally over the 5 days of the assay. To increase the water solubility of the drug we have produced prodrugs with a photo-cleavable group attached to the B-ring phenolic OH group. The nitro vanillin derivative was chosen since it has been used as a photo-cleavable linker for solid phase synthesis applications and its synthesis is relatively simple. These prodrugs have been successfully cleaved in both the E and Z series (E-6 and Z-6 respectively) and have produced highly cytotoxic agents in vitro upon in situ exposure to ultra violet radiation. The cleavage of the water solubilising group appears to be faster than the E→Z isomerisation, at least under ex situ irradiation. Thus, the use of Z-6 has some merit. Indeed it forms the prototype for systems that do not rely on any special photochemical features of the molecule to be delivered. This provides a more general approach to the site-specific photochemical activation of prodrugs. Moreover, the photocleavable water solubilising group can be engineered, so that cleavage occurs at longer wavelength and rapidly.
  • Embodiments of the present invention will now be described by way of example and not limitation with reference to the accompanying figures.
  • BRIEF DESCRIPTION OF THE FIGURES
  • FIG. 1 shows the anti-tumour activity of compound 97-64H in an in vivo tumour implant experiment in mice.
  • FIG. 2 shows the anti-tumour activity of compound 97-96 in an in vivo tumour implant experiment in mice.
  • DETAILED DESCRIPTION
  • Pharmaceutical Compositions
  • The compounds of the invention may be derivatised in various ways. As used herein “derivatives” of the compounds includes salts, esters such as in vivo hydrolysable esters, free acids or bases, hydrates, prodrugs or coupling partners. In the case of compounds which are combretastatin or analogues thereof, preferably the derivatives are soluble in water and/or saline or can be hydrolysed to provide physiologically active agents.
  • Examples in the prior art of salts or prodrugs of cis-combretastatin A-4 focus on forming salts or derivatives at the phenolic hydroxyl group of combretastatin. These include sodium phosphate salts, sodium and potassium salts (U.S. Pat. No. 5,561,122), lithium, caesium, magnesium, calcium, manganese and zinc salts of cis-combretastatin A-4, and ammonium cation salts with imidazole, morpholine, piperazine, piperidine, pyrazole, pyridine, adenosine, cinchonine, glucosamine, quinine, quinidine, tetracycline and verapamil (WO99/35150).
  • Salts of the compounds of the invention are preferably physiologically well tolerated and non toxic. Many examples of salts are known to those skilled in the art. Compounds having acidic groups, can form salts with alkaline or alkaline earth metals such as Na, K, Mg and Ca, and with organic amines such as triethylamine and Tris (2-hydroxyethyl)amine. Salts can be formed between compounds with basic groups, e.g. amines, with inorganic acids such as hydrochloric acid, phosphoric acid or sulfuric acid, or organic acids such as acetic acid, citric acid, benzoic acid, fumaric acid, or tartaric acid. Compounds having both acidic and basic groups can form internal salts.
  • Esters can be formed between hydroxyl or carboxylic acid groups present in the compound and an appropriate carboxylic acid or alcohol reaction partner, using techniques well known in the art. Examples of esters include those formed between the phenolic hydroxyl of the substituted stilbenes and carboxylic acids, hemisuccinic acid esters, phosphate esters, BOC esters, sulphate esters and selenate esters.
  • Derivatives which as prodrugs of the compounds are convertible in vivo or in vitro into one of the parent compounds. Typically, at least one of the biological activities of compound will be reduced in the prodrug form of the compound, and can be activated by conversion of the prodrug to release the compound or a metabolite of it. Example of prodrugs include combretastatin A1 phosphate, combretastatin A4 phosphate and RH1.
  • Other derivatives include coupling partners of the compounds in which the compounds is linked to a coupling partner, e.g. by being chemically coupled to the compound or physically associated with it. Examples of coupling partners include a label or reporter molecule, a supporting substrate, a carrier or transport molecule, an effector, a drug, an antibody or an inhibitor. Coupling partners can be covalently linked to compounds of the invention via an appropriate functional group on the compound such as a hydroxyl group, a carboxyl group or an amino group.
  • The compounds described herein or their derivatives can be formulated in pharmaceutical compositions, and administered to patients in a variety of forms, in particular to treat conditions which are ameliorated by the activation of the compound.
  • Pharmaceutical compositions for oral administration may be in tablet, capsule, powder, cream, liquid form or encapsulated by liposomes. A tablet may include a solid carrier such as gelatin or an adjuvant or an inert diluent. Liquid pharmaceutical compositions generally include a liquid carrier such as water, petroleum, animal or vegetable oils, mineral oil or synthetic oil. Physiological saline solution, or glycols such as ethylene glycol, propylene glycol or polyethylene glycol may be included. Such compositions and preparations generally contain at least 0.1 wt % of the compound.
  • Parental administration includes administration by the following routes: intravenous, cutaneous or subcutaneous, nasal, intramuscular, intraocular, transepithelial, intraperitoneal and topical (including dermal, ocular, rectal, nasal, inhalation and aerosol), and rectal systemic routes. For intravenous, cutaneous or subcutaneous injection, or injection at the site of affliction, the active ingredient will be in the form of a parenterally acceptable aqueous solution which is pyrogen-free and has suitable pH, isotonicity and stability. Those of relevant skill in the art are well able to prepare suitable solutions using, for example, solutions of the compounds or a derivative thereof, e.g. in physiological saline, a dispersion prepared with glycerol, liquid polyethylene glycol or oils.
  • In addition to one or more of the compounds, optionally in combination with other active ingredient, the compositions can comprise one or more of a pharmaceutically acceptable excipient, carrier, buffer, stabiliser, isotonicizing agent, preservative or anti-oxidant or other materials well known to those skilled in the art. Such materials should be non-toxic and should not interfere with the efficacy of the active ingredient. The precise nature of the carrier or other material may depend on the route of administration, e.g. orally or parentally.
  • Liquid pharmaceutical compositions are typically formulated to have a pH between about 3.0 and 9.0, more preferably between about 4.5 and 8.5 and still more preferably between about 5.0 and 8.0. The pH of a composition can be maintained by the use of a buffer such as acetate, citrate, phosphate, succinate, Tris or histidine, typically employed in the range from about 1 mM to 50 mM. The pH of compositions can otherwise be adjusted by using physiologically acceptable acids or bases.
  • Preservatives are generally included in pharmaceutical compositions to retard microbial growth, extending the shelf life of the compositions and allowing multiple use packaging. Examples of preservatives include phenol, meta-cresol, benzyl alcohol, para-hydroxybenzoic acid and its esters, methyl paraben, propyl paraben, benzalconium chloride and benzethonium chloride. Preservatives are typically employed in the range of about 0.1 to 1.0% (w/v).
  • Preferably, the pharmaceutically compositions are given to an individual in a “prophylactically effective amount” or a “therapeutically effective amount” (as the case may be, although prophylaxis may be considered therapy), this being sufficient to show benefit to the individual. Typically, this will be to cause a therapeutically useful activity providing benefit to the individual. The actual amount of the compounds administered, and rate and time-course of administration, will depend on the nature and severity of the condition being treated. Prescription of treatment, e.g. decisions on dosage etc, is within the responsibility of general practitioners and other medical doctors, and typically takes account of the disorder to be treated, the condition of the individual patient, the site of delivery, the method of administration and other factors known to practitioners. Examples of the techniques and protocols mentioned above can be found in Remington's Pharmaceutical Sciences, 16th edition, Osol, A. (ed), 1980. By way of example, and the compositions are preferably administered to patients in dosages of between about 0.01 and 100 mg of active compound per kg of body weight, and more preferably between about 0.5 and 10 mg/kg of body weight. The compounds may be used in the treatment of cancer and other conditions involving abnormal proliferation of vasculature including diabetic retinopathy, psoriasis and endometriosis.
  • General
  • Proton nuclear magnetic resonance (1H NMR) spectra were recorded on a Brüker AC 300 (300 MHz) or AC 400 (400 MHz) NMR spectrometer. Chemical shifts, δ, for all NMR spectra are given in ppm, relative to tetramethylsilane, and, unless otherwise stated, using CDCl3 as both solvent and internal standard. Coupling constants (J) were measured in Hz. Melting points were determined on a Gallenkamp melting point apparatus and are uncorrected. The UV/VIS spectra were determined using a Hewlett-Packard HP8452 diode-array spectrophotometer. Extinction coefficients(ε) are presented as their natural logarithms. Microanalyses were carried out by the laboratories of the Departments of Chemistry of the University of Manchester and UMIST. High resolution mass spectroscopy was determined using a Kratos Concept 15 mass spectrometer. Thin layer chromatography (tlc) was performed using precoated aluminium-backed silica gel plates (60 F254) with 0.2 mm thickness (Merck), with observation under UV when necessary. Gas chromatography was carried out using an SE 54 column at 195-225 kPa at 1.5 kPa/min. The oven temperature was 180-280° C. at 5° C./min.
  • EXAMPLE 1 Synthesis of Combretastatins with Alkyl Groups on the Double Bond Z- and E-1-(3′-t-Butyldimethylsilyloxy-4′-methoxyphenyl)-2-(3″,4″,5″-trimetboxyphenyl)propene, 17a, 17b
  • Figure US20070203217A1-20070830-C00015
  • To a slurry of 3-t-butyldimethylsilyloxy-4-methoxybenzylphosphonium bromide, 18, (1 g, 1.69 mmol) in THF (10 ml) was added n-butyllithium (1.16 ml of 1.6 M solution, 1.86 mmol) at −15° C. The red anion was stirred for 20 minutes and 3,4,5-trimethoxyacetophenone (355 mg, 1.69 mmol) added. The resultant solution was stirred at room temperature for 1 hour and water (10 ml) carefully added. The aqueous layer was separated and extracted with ether (3×10 ml). The combined organic layers were washed with water (2×10 ml) and brine (10 ml), dried (MgSO4) and concentrated in vacuo.
  • Following flash column chromatography (SiO2 petrol:EtOAc 19:1) the Z stilbene, 17a, was isolated as a colourless oil (109 mg, 15%). Rf=0.72 (SiO2 petrol:EtOAc 1:1); δH (300 M) 0.20 [6H, s, (CH3)2], 1.03 [9H, s, (CH3)3], 2.28 (3H, d, J=1.1, CH3), 3.85 (3H, s, OCH3), 3.89 (3H, s, OCH3), 3.93 [6H, s, (OCH3)2], 6.69 (1H, q, J=1.1, olefinic H), 6.72 (2H, s, H-2′,6′) 6.87 (1H, d, J=8.3, H-5″), 6.91 (1H, q, J=2.3, H-2″), 6.95 (1H, dd, J=8.3, 2.3, H-6″); λmax (MeOH)=270 (ε=7,607); M+, found 444.2329; C25H36O5Si requires M+444.2332.
  • Further elution afforded the E stilbene, 17b, as a colourless oil (258 mg, 34%). Rf=0.67 (SiO2 petrol:EtOAc 1:1); δH (300 MHz) 0.01 [6H, s, (CH3)2], 0.92 [9H, s, (CH3)3], 2.17 (3H, d, J=1.5, CH3), 3.75 [6H, s, (OCH3)2], 3.76 (3H, s, OCH3), 3.87 (3H, s, OCH3), 6.35 (1H, q, J=1.5, olefinic H), 6.42 (2H, s, H-2′,6′) 6.52 (1H, d, J=1.9, H-2″), 6.62 (1H, dd, J=8.3, 1.9, H-6″), 6.67 (1H, d, J=8.3, H-5″); λmax (MeOH)=296 (e=16,541). M+, found 444.2333; C25H36O5Si requires M+444.2332.
  • Z-(3′-Hydroxy-4′-methoxyphenyl)-2-(3″,4″,5″-trimethoxyphenyl)propene, 19
  • Figure US20070203217A1-20070830-C00016
  • To a stirred solution of Z-(3′-t-butyldimethylsilyloxy-4′-methoxyphenyl)-2-(3″,4″,5″-trimethoxyphenyl)propene, 17a, (111 mg, 0.250 mmol) in dry THF (5 ml) was added tetra-n-butylammonium fluoride (700 ml of 1 M solution, 0.7 mmol). The resulting yellow solution was stirred for 20 minutes and then treated with water (2 ml). The aqueous layer was separated and extracted with chloroform (3×10 ml). The combined organic layers were washed with water (2×10 ml) and brine (10 ml), dried (MgSO4) and concentrated in vacuo. Flash column chromatography (SiO2 petrol:EtOAc 2:1) afforded Z-(3′-hydroxy-4′-methoxyphenyl)-2-(3″,4″,5″-trimethoxyphenyl)propene, 19, as a fine white powder (49 mg, 0.148 mmol, 60%). m.p. 156-8° C.; Rf=0.36 (SiO2 petrol:EtOAc 2:1); δH (300 MHz) 2.18 (3H, d, J=1.5, CH3), 3.75 [6H, s, (OCH3)2], 3.84 (3H, s, OCH3), 3.88 (3H, s, OCH3), 5.42 (1H, s, OH), 6.36 (1H, q, J=1.5, olefinic H), 6.43 (2H, s, H-2′,6′) 6.48 (1H, dd, J=8.3, 2.3, H-6″), 6.62 (1H, d, J=8.3, H-5″), 6.63 (1H, d, J=2.3, H-2″); λmax (MeOH)=270 (e=11,524); M+, found 330.1469; C19H22O5 requires M+330.1467.
  • Z-1-(3′,4′,5′-trimethoxyphenyl)-2-(3″-hydroxy-4″-methoxyphenyl)propene, 20
  • Figure US20070203217A1-20070830-C00017
  • A mixture of cis-1-(3′,4′,5′-trimethoxyphenyl)propene 21 (0.42 g, 2 mmol), 5-iodo-2-methoxyphenol 22 (1 g, 4 mmol), triethylamine (0.51 g, 5 mmol), palladium acetate (9 mg, 0.04 mmol) and triphenylphosphine (21 mg, 0.08 mmol) were heated at 100° C. To the cooled reaction mixture was added aqueous hydrochloric acid (45 ml of a 2.7 M solution). After stirring for 10 min, the liquid was decanted off and the solid residue extracted with several portions of hot hexane. The combined hot hexane fractions were filtered. The cooled hexane solution was washed with water (2×10 ml), brine (10 ml), dried over magnesium sulfate, filtered and the solvent evaporated. Flash column chromatography (SiO2 petrol:EtOAc 15:1) afforded the stilbene (20) as a white crystalline solid (109 mg, 16%). m.p. 99-100° C.; Rf=0.34 (SiO2 petrol:EtOAc 1:1); δH (300 MHz) 2.28 (3H, d, J=1.1, CH3), 3.89 (3H, s, OCH3), 3.90 (6H, s, 2×OCH3), 3.94 (3H, s, OCH3), 5.61 (1H, s, OH), 6.59 (2H, s, H-2′,6′), 6.74 (1H, q, J=1.1, olefinic H), 6.87 (1H, d, J=8.7, H-5″), 7.04 (1H, dd, J=8.7, 2.3, H-6″), 7.14 (1H, d, J=2.3, H-2″); Found C, 69.13; H, 6.71; C19H22O5 requires C, 69.07; H, 6.71%; M+, found 331.1541; C19H22O5 (+H) requires 331.1545; λmax (MeOH)=296 (ε=14,126).
  • The ethyl derivative (45) has also been synthesised.
    Figure US20070203217A1-20070830-C00018
  • Z- and E-1-(4′-Methoxyphenyl)-2-(3″,4″,5″-trimethoxyphenyl)propene, 23a, 23b
  • Figure US20070203217A1-20070830-C00019
  • To a slurry of 4-methoxybenzylphosphonium chloride (598 mg, 1.43 mmol) in THF (8 ml) was added n-butyllithium (990 ml of 1.6 M solution, 1.58 mmol) at −15° C. The red anion was stirred for 20 minutes and 3,4,5-trimethoxyacetophenone (300 mg, 1.43 mmol) added. The resultant solution was stirred at room temperature for 1 hour and water (10 ml) carefully added. The aqueous layer was separated and extracted with ether (3×10 ml). The combined organic layers were washed with water (2×10 ml) and brine (10 ml), dried (MgSO4) and concentrated in vacuo.
  • The nmr of the crude reaction product showed that the Z:E ratio was 1:1.5. Following flash column chromatography (SiO2 petrol:EtOAc 9:1) the Z stilbene, 23a, was isolated as white needles (45 mg, 0.143 mmol, 10%). m.p. 73-5° C.; Rf=0.46 (SiO2 petrol:EtOAc 3:1); δH (300 MHz) 2.19 (1H, d, J=1.5, CH3), 3.74 [6H, s, (OCH3)2], 3.76 (3H, s, OCH3), 3.88 (3H, s, OCH3), 6.40 (1H, q, J=1.5, olefinic H), 6.42 (2H, s, H-2′,6′), 6.69 (1H, dt, J=8.7, 2.3, H-3″,5″), 6.93 (1H, dt, J=8.7, 2.3, H-2″,6″); λmax (MeOH)=273 (ε=14,926).
  • Further elution afforded the E stilbene, 23b, as an off white solid (44 mg, 0.140 mmol, 9.8%). m.p. 80-2° C.; Rf=0.41 (SiO2 petrol:EtOAc 3:1); δH (300 Mz) 2.28 (1H, d, J=1.5, CH3), 3.86 (3H, s, OCH3), 3.90 (3H, s, OCH3), 3.94 [6H, s, (OCH3)2], 6.74 (2H, s, H-2′,6′), 6.75 (1H, q, J=1.5, olefinic H), 6.94 (1H, dt, J=8.7, 2.3, H-3″,5″), 7.34 (1H, dt, J=8.7, 2.3, H-2″,6″); λmax (MeOH)=287 (ε=21,822).
  • EXAMPLE 2 Synthesis of Combretastatins with Alkyl Groups Replacing the Methoxy Groups on the A Ring E-2-(3′,4′,5′-trimethylphenyl)-3-(3″-(2′″,3′″,5′″,6′″-tetrafluoropyridoxy)-4″-methoxyphenyl)prop-2-enoic acid, 24
  • Figure US20070203217A1-20070830-C00020
  • A mixture of 3-(2′,3′,5′,6′-tetrafluoropyridoxy)-4-benzaldehyde 25 (2 g, 6.64 mmol), 3,4,5-trimethylphenylacetic acid 26 (2.37 g, 13.3 mmol) acetic anhydride (6 ml) and triethylamine (3 ml) were heated under reflux for 3 h. After acidification with concentrated hydrochloric acid (9 ml), the solid was filtered off and recrystallised from ethanol to give E-2-(3′,4′,5-trimethyl)-3-(3″-(2′″,3′″,5′″,6′″-tetrafluoropyridoxy)-4″-methoxyphenyl)prop-2-enoic acid 24 as a yellow crystalline solid (700 mg, 1.52 mmol, 23%). m.p. 184-6° C. δH (300 MHz, DMSO) 2.11, (3H, s, CH3), 2.14 (6H, s, (CH3)2), 3.83 (3H, s, OCH3), 6.61 (1H, d, J=1.5, H-2″), 6.71 (2H, s, H-2′,6′), 7.13 (1H, d, J=8.7, H-5″), 7.19 (1H, dd, J=8.7, 1.5, H-6″), 7.61, (1H, s, olefinic H), 12.52, (1H, s OH).
  • (Z)-1-(3′,4′,5′-trimethylphenyl)-2-(3″-(2′″,3′″,5′″,6′″-tetrafluoropyridoxy)-4″-methoxyphenyl)ethene, 27
  • Figure US20070203217A1-20070830-C00021
  • (E)-2-(3′,4′,5 ′-trimethylphenyl)-3-(3″-(2′″,3′″,5′″,6′″-tetrafluoropyridoxy)-4″-methoxyphenyl)prop-2-enoic acid 24 (700 mg, 1.52 mmol) was added to powdered copper (500 mg, 7.81 mmol) in quinoline (5.5 ml, 6.02 g, 28.2 mmol) and the resulting mixture was heated at 200° C. for 2 h. Upon cooling, ether was added and the copper filtered off through celite. The filtrate was washed with 1 M hydrochloric acid (2×20 ml) and the aqueous layer separated and extracted with ether (3×50 ml). The combined organic layers were washed with saturated sodium carbonate (50 ml), water (2×50 ml) and brine (50 ml), dried (MgSO4) and concentrated in vacuo. Flash column chromatography (SiO2 petrol:EtOAc 9:1) afforded (Z)-1-(3′,4′,5′-trimethylphenyl)-2-(3″-(2′″,3′″,5′″,6′″-tetrafluoropyridoxy)-4″-methoxyphenyl)ethene 27 as a yellow oil (224 mg, 0.538 mmol, 35%). Rf=0.48 (SiO2 petrol:EtOAc 9:1); δH (300 MHz) 2.15 (3H, s, CH3), 2.19 [6H, s, (CH3)2], 3.84 (3H, s, OCH3), 6.41 (1H, d, J=12.1, olefinic H), 6.51 (1H, d, J=12.1, olefinic H), 6.87 (2H, s, H-2′,6′), 6.89 (1H, d, J=8.7, H-5″), 6.98 (1H, d, J=2.3, H-2″), 7.11 (1H, dd, J=8.7, 2.3, H-6″).
  • (Z)-1-(3′,4′,5′-trimethylphenyl)-2-(3″-hydroxy-4″-methoxyphenyl)ethene, 28
  • Figure US20070203217A1-20070830-C00022
  • To a solution of the (Z)-1-(3′,4′,5′-trimethylphenyl)-2-(3″-(2′″,3′″,3′″,5′″,6′″-tetrafluoropyridoxy)-4″-methoxyphenyl)ethene (100 mg, 0.24 mmol) 27 in dry DMF (600 ml) and dichloromethane (115 ml) at 0° C. was added sodium methoxide (25 mg, 0.463 mmol). After stirring overnight, the mixture was partitioned between ether (5 ml) and 1 M sulfuric acid (5 ml). The organic phase was washed with water (5 ml), dried (MgSO4) and concentrated in vacuo. Flash column chromatography (SiO2 petrol:EtOAc 9:1) and recrystallisation from petrol afforded (Z)-1-(3′,4′,5′-trimethylphenyl)-2-(3″-hydroxy-4″-methoxyphenyl)ethene as a white crystalline solid (31 mg, 0.116 mmol, 48%). m.p. 60-1° C. Rf=0.39 (SiO2petrol:EtOAc 4:1); δH (300 Mz) 2.16 (3H, s, CH3), 2.21 [6H, s, (CH3)2], 3.90 (3H, s, OCH3), 5.50 (1H, s, OH), 6.40 (1H, d, J=12.4, olefinic H), 6.45 (1H, d, J=12.4, olefinic H), 6.72 (1H, d, J=8.3, H-5″), 6.82 (1H, dd, J=8.3, 2.3, H-6″), 6.91 (1H, d, J=2.3, H-2″), 6.96 (2H, s, H-2′, 6′).
  • EXAMPLE 3 Synthesis of Combretastatins with a 3,4,5 Trialkoxy Group Z- and E-1-(3′,4′,5′-triethoxyphenyl)-2-(3″-t-butyldimethylsilyloxy-4″-methoxyphenyl)ethene, 30a, 30b
  • To a slurry of 3,4,5-triethoxybenzylphosphonium bromide 29 (2 g, 3.24 mmol) in THF (30 ml) was added n-butyllithium (2.5 ml of 1.6M solution in hexanes, 4 mmol) at −15° C. under argon. The red anion was stirred for 20 min and 3-O-t butyldimethylsilyl-4-methoxybenzaldehyde 6 (0.86 g, 3.24 mmol) added. The resultant solution was stirred for 1 h at room temperature and water (10 ml) was carefully added. The aqueous layer was separated and extracted with ethyl acetate (3×100 ml). The combined organic layers were washed with water (2×100 ml), brine (100 ml), dried (MgSO4) and concentrated in vacuo. Flash column chromatography afforded the cis stilbene 30a as a colourless oil (0.23 g, 15%). Rf=0.65 (petrol:ethyl acetate 9:1); δH (300 MHz) 0.08 (6H, s, Si(CH3) 2), 0.95 (9H, s, 3×CH3), 1.35 (9H m, 3×OCH2CH 3), 3.79 (3H, s, OCH3), 3.91 (4H, q, J=6.8, CH2), 4.06 (2H, q, J=7.2, CH2), 6.40 (1H, d, J=12.1, olefinic H), 6.43 (1H, d, J=12.1, olefinic H), 6.74 (2H, s, ArH 2, 6), 6.83 (1H, dd, J=8.0, 2.1, ArHpara to OSi), 6.84 (1H, d, J=8.0, ArH ortho to OMe), 6.88 (1H, d, J=2.1, ArH ortho to OSi).
  • Further elution gave the trans stilbene 30b as white crystals (0.27 g, 17.6%). Rf=0.75; δH (300 MHz) 0.20 (6H, s, Si(CH3) 2), 1.03 (9H, s, 3×CH3), 1.37 (3H, t, J=7.5, CH2CH 3), 1.46 (6H, t, J=7.2, 2×CH2CH 3), 3.85 (3H, s, OCH3), 4.12 (6H, m, 3×CH2), 6.73 (2H, s, ArH 2, 6), 6.84 (1H, dd, J=7.8, 1.98, ArH para to OSi), 6.90 (1H, d, J=15.1, olefinic H), 7.04 (1H, d, J=7.8, ArH ortho to OMe), 7.05 (1H, d, J=15.1, olefinic H), 7.66 (1H, d, J=2.0, ArH ortho to OSi).
  • Z- and E-1-(3′,4′,5′-triethoxyphenyl)-2-(3″-hydroxy-4″-methoxyphenyl)ethene, 31a, 31b
  • To a stirred mixture of cis and trans-1-(3′,4′,5′-triethoxyphenyl)-2-(3″-tert-butyldimethylsiloxy-4″-methoxy)ethene 30a, 30b (0.23 g, 0.48 mmol-cis isomer; 0.27 g, 0.57 mmol-trans isomer) in dry THF (17.5 ml) was added tetra-n-butylammonium fluoride (1.46 ml of 1 M solution-in THF). The resulting yellow solution was stirred for twenty min and treated with water (50 ml). The aqueous layer was separated and extracted with chloroform (3×50 ml). The combined organic layers were washed with water (2×50 ml) and brine (50 ml), dried (MgSO4) and concentrated in vacuo.
  • Flash column chromatography (petrol:ethyl acetate 4:1) afforded Z-1-(3′,4′,5′-triethoxyphenyl)-2-(3″-hydroxy-4″-methoxyphenyl)ethene 31a as a colourless oil (0.08 g, 46%). Rf=0.24. δH (300 MHz) 1.34 (9H, m, 3×CH2CH 3), 3.87 (3H, s, OCH3), 3.92 (4H, q, J=7.2, 2×CH2), 4.06 (2H, q, J=7.2, CH2), 6.40 (1H, d, J=12.43, olefinic H), 6.45 (1H, d, J=12.4, olefinic H), 6.50 (2H, s, ArH 2,6), 6.75 (1H, d, J=8.1, ArH ortho to OMe), 6.79 (1H, dd, J=8.7, 1.88, ArH para to OH), 6.91 (1H, d, J=1.9, ArH ortho to OH). M+, 358.
    Figure US20070203217A1-20070830-C00023
  • Further elution afforded E-1-(3′,4′,5′-triethoxyphenyl)-2-(3″-hydroxy-4″-methoxyphenyl)ethene 31b (0.09 g, 46.6%) as white crystals. mp: 91-92° C.; Rf=0.11. δH (300 MHz) 1.37 (3H, t, J=6.0, CH3), 1.46 (6H, t, J=6.4, 2×CH3), 3.92 (3H, s, OCH3), 4.12 (6H, m, 3×CH2), 6.71 (2H, s, H2, 6), 6.84 (1H, d, J=7.8, ArH ortho to OMe), 6.85 (1H, d, J=15.5, olefinic H), 6.91 (1H, dd, J=7.9, 2.26, ArH para to OH), 7.11 (1H, d, J=15.5, olefinic H), 7.13 (1H, d, J=2.3, ArH ortho to OH). M+, 358.
  • Z- and E-1-(3′,4′,5′-triethoxyphenyl)-2-(3″-fluoro-4″-methoxyphenyl)ethene, 32a, 32b
  • To a slurry of 3,4,5-triethoxybenzylphosphonium bromide 29 (2 g, 3.24 mmol) in THF (30 ml) was added n-butyllithium (2.5 ml of 1.6 M solution in hexanes, 4 mmol) at −15° C. under argon. The red anion was stirred for 20 min and 3-fluoro-4-methoxybenzaldehyde (0.50 g, 3.24 mmol) was added. The resultant solution was stirred for 1 h and water (10 ml) was carefully added. The aqueous layer was separated and extracted with ethyl acetate (3×100 ml). The combined organic layers were washed with water (2×100 ml) and brine (100 ml), dried (MgSO4) and concentrated in vacuo.
  • Flash column chromatography (SiO2, petrol:etliyl acetate 20:1) afforded the cis-stilbene 32a as a pale yellow oil (0.35 g, 29%), Rf=0.16; δH (300 MHz) 1.35 (9H, m, 3×CH3), 3.87 (3H, s, OCH3), 3.92 (4H, q, J=6.8, 2×CH2), 4.07 (2H, q, J=7.2, CH2), 6.41 (1H, d, J=12.4, olefinic H), 6.47 (1H, d, J=12.4, olefinic H), 6.47 (2H, s, ArH 2,6), 6.84 (1H, t, J=8.7, ArH ortho to OMe), 7.00 (1H, dd, J=8.7, 1.5, ArH para to F), 7.05 (1H, d, J=12.1, 1.5, ArH ortho to F). M+, 360.
  • Further elution afforded the trans isomer 32b (0.27 g, 23%) as white needles mp:97-99° C.; Rf=0.22; δH (300 MHz) 1.38 (6H, t, J=7.2, 2×CH3), 1.46 (3H, t, J=7.2, CH3) 3.92 (3H, s, OCH3), 4.12 (6H, m, 3×CH2), 6.71 (2H, s, H 2,6) 6.91 (1H, d, J=15.5, olefinic H), 6.94 (1H, t, J=8.7, ArH ortho to OMe), 6.95 (1H, d, J=15.5, olefinic H), 7.17 (1H, dd, J=8.7, 2.3 ArH para to F), 7.25 (1H, dd, J=12.0, 2.3, ArH ortho to F). M+, 360.
  • EXAMPLE 4 Boc-Combretastatin Compounds Boc-Phenylalanine combretastatin A-4, 33
  • To a stirred solution of t-butoxycarbonyl-phenylalanine (168 mg, 0.634 mmol), dicyclohexylcarbodiimide (157 mg, 0.76 mmol), N,N-4-dimethylaminopyridine (8 mg, 61 μmol) in dichloromethane (15 ml) under nitrogen at room temperature in the dark was added combretastatin A-4 (1) (200 mg, 0.633 mmol). After stirring for 48 h, the mixture was filtered, evaporated and the residue chromatographed on silica using petroleum (bp 40-60° C.)/ethyl acetate 4:1 to afford the title ester (33) as a clear gum (143 mg, 40%). vmax 3364 (NH); 1766, 1716 (C═O). δH 7.22 (5H, m, phenylalanine ArHs); 7.11 (1H, dd, J=8.1, 2.0, Hpara to O ester); 6.95 (1H, d, J=2.0, H ortho to O ester); 6.83 (1H, d, J=2.0, H meta to O ester); 6.40, 6.50 (4H, 2 s, olefinic Hs, A-ring Hs); 5.07 (1H, broad, NH); 4.80 (1H, m, H-α); 3.80, 3.70, 3.66 (12H, 3×s, 4×OMe); 3.35-3.08 (2H, m, CH2); 1.40 (9H, s, 3×CH3). λmax 241; 271; 305. m/z 463 (35%, M+H-BOC); 317 (100).
  • The other BOC compounds 34-44 were made using the above method.
    Figure US20070203217A1-20070830-C00024
    33 R = Phe 39 R = Ala
    34 R = Ile 40 R = His
    35 R = Gly 41 R = Pro
    36 R = Trp 42 R = D-Met
    37 R = Met 43 R = D-Trp
    38 R = Leu 44 R = Tyr
  • EXAMPLE 5 Benzoguinone Compounds 2-Methoxy-5-[(Z)-2-(3′,4′,5′-trimethoxyphenyl)-vinyl]-[1,4]benzoquinone 97-96
  • To a mixture of Aliquat 336 (0.181 ml, 1.25 equiv) and NaH2PO4.H2O (323 mg, 2.34 mmol) in water (100 ml) was added a solution of combretastatin A4 (1) (100 mg, 0.316 mmol) in dichloromnethane (7 ml). Fremy's salt (potassium nitrosodisulfonate) (212 mg, 0.8 mmol) was added and the mixture shaken for 30 min. (Colour changes from mauve to red). The dichloromethane was collected and the aqueous fraction extracted with dichloromethane (3×10 ml). The combined organic phases were washed with water (3×10 ml), brine (10 ml) and dried over magnesium sulfate. Evaporation of the solvent followed by flash chromatography of the residue using petrol:ethyl acetate (65:35) as eluent afforded the quinone as a red crystalline solid (51 mg, 49%) mp 130-2° C., δH (acetone d6) 3.70, 3.75, 3.85 (12H, 3 s, 4×OMe); 6.08 (1H, s, quinone-H ortho to OMe); 6.43 (1H, dd, J12.5, 0.5, olefnic-H next to quinone ring); 6.67 (1H, d, J0.5, quinone-H meta to OMe); 6.73 (2H, s, ArHs); 6.96 (1H, d, J, 12.5, olefinic-H next to Ar ring). λmax 296 (ε12,708); 470 (2038). νmax 1666, 1647 cm−1. M+, 330 (40%); 315 (M-Me, 60); 69 (100).
  • This method was also used to synthesise 98-40, 98-23, 98-33 and 98-24.
    Figure US20070203217A1-20070830-C00025
  • These quinones may act as prodrugs producing the active hydroquinones on reduction by enzymes.
  • EXAMPLE 6 Biological Activity of Compounds
  • The following assays (1-5) were carried out as described in our paper (Woods, et al, British Journal of Cancer, 71, 705-711 (1995)). In addition to the cell lines described in this paper, other established human cell lines (K562, HUVEC, H460, BE, H529, and HT29) were used in the cytotoxicity/growth inhibition assay.
    • (1) Inhibition of tubulin assembly. The figure quoted is the concentration required to reduce assembly of tubulin by 50%. Tubulin assembly is monitored by light scatter/absorption at 350 nm.
    • (2) Competition for the colchicine binding site on tubulin. The figure quoted represents the % of 3H-colchicine bound following co-incubation of test compound and 3H-colchicine with isolated tubulin. Where ester pro-drugs were used the experiments were carried out in the presence and absence of porcine liver esterase.
    • (3) Cytotoxicity/growth inhibition assay. This was carried out by the MTT assay.
    • (4) Permeability (shape-change) assay in endothelial cells. This was carried out using a method based on that of Watts et al. (Anticancer Res., 17, 71-75, (1997)). This involved the diffusion of a fluorescently labelled dextran through a barrier of endothelial cells (HUVEC) grown to confluence on a porous membrane. The effect of agents to alter the shape of these cells results in an increase in the permeability of the cells to the dye. The figure represents the increase in permeability over control cultures when drug is added (30 minutes, 1 mM.).
    • (5) Experiments to study the anti-vascular effects of these agents were carried out as described previously by our group. (Zhao et al, European Journal of Nuclear Medicine, 26, 231-238 (1999)). The anti-vascular effects of the agents were monitored by either positron emission tomography (PET) or by histologogical examination following treatment of mice bearing T50:80 murine breast tumours, or H460 human lung tumours.
    • (6) Anti-tumour efficacy of agents was determined in xenografts of H460 human lung cancer. Animals (n=5) were treated either with 5 daily injections and tumour size measured with time following treatment.
    • (7) The pharmacolinetic profile of agents was determined in mice following injection with drug. Blood was taken at various times following treatment and analysed by HPLC with UV detection. The HPLC conditions were isocratic using a 5 micron C18 BDS column (150×4.6) and eluting with 75% MeOH in water with a detection wavelength of 290 nm. The retention time of 6.6 mins for 97-64H.
    • (8) Activation of quinone prodrugs by DT-diaphorase. (NAD(P)H:quinone oxidoreductase, EC 1.6.99.2). DT-diaphorase is an enzyme over-expressed in a number of human tumours. We have recently shown this enzyme to be highly expressed in the endothelial cells of blood vessels. This two-electron reducing enzyme can be used to selectively activate prodrugs within cancers which over-express the enzyme. We have therefore synthesised quinone pro-drugs (97-96, 98-40, 98-23, 98-33, 98-24) which, upon reduction by the enzyme, produces an agent (a hydroquinone stilbene) which is cytotoxic.
      Figure US20070203217A1-20070830-C00026

      Results
  • The in vitro studies reported in Table 11 illustrate the structural requirements necessary for biological activity. Modifications of the A ring have shown that the methoxy groups can be replaced by alkoxy or alkyl (28,31a,32a) whilst retaining their ability to inhibit assembly of isolated tubulin. Similarly, molecules with alkyl substituents on the ethene bridge (19,20,46) retain activity both as inhibitors of tubulin assembly and are growth inhibitory in vitro. The pharmacophore consists of a stilbene in a cis configuration with a small alkyl or O-alkyl substituent at the 4-position of the B ring. Substitution at the 3-position of the B-ring with F results in highly active compounds that are potent inhibitors of tubulin assembly, and are potently growth inhibitory (97-64H, and 98-35). These agents show good activity in the shape-change (permeability) assay. This test is used as an in vitro assay of vascular damage. 97-64H also shows anti-tumour activity in vivo in H460 human lung cancer xenografts. 97-64H was given at either ⅔ of the maximum tolerated dose (MTD=200 mg/kg) or at ¾ of the MTD to mice bearing liver metastatic T50:80 tumours. Tumours were removed and examined for evidence of vascular damage at 2 hr and 4 hr following drug administration. Similarly a dose equivalent to ¾ of the MTD was administered and the tumours examined 24 hr and 48 hr following administration of drug. Examination of these tumours showed tumour necrosis, blocked vessels, infiltration with red blood cells, consistent with damage to the vasculature. These effects are seen within 2 hours. 97-64H is orally bio-available when administered at 200 mg/kg in 5% dimethylacetamide in arachis oil, 97-64H showed a Cmax of 1.49 ug/ml, an absorption half life 15 mins, and an elimination half life 32 mins. The concentration achieved in vivo (1.49 mg/ml=4.6 mM) is far in excess of the concentration necessary to inhibit the growth of HLUVEC cells in vitro (0.001 mM) indicating that this agent is bio-available when administered orally.
  • The 3-position of the B-ring can also be substituted with larger groups such as boc-amino acid esters, pyridyl esters, and ethers. The amino acid esters (33-44) are prodrugs which, upon the action of esterase, release the active agent. The activity of these agents is related to the rate of hydrolysis of these agents by esterase. The most active compounds being those which are most readily hydrolysed. The activity of these compounds indicates that ester linked polymers and peptides would be good prodrugs for agents of this type (see Table 1).
  • The pyridyl esters (96-167 and 97-07) are potent inhibitors of tubulin assembly and can displace 3H-colchicine from tubulin without the action of esterase, showing that the 3-position of the B-ring can be substituted with bulky side groups. Potent growth inhibitory activity and good activity in the shape-change assay can also be seen for the tetrafluoropyridyl ether (97-13) and other ethers (98-29) which are not a substrate for esterase.
  • A series of prodrugs capable of being activated by DT-diaphorase, an enzyme over-expressed in a wide range of human cancers and in endothelial cells of the vasculature were synthesised. The rationale for this is that these agents will be activated in situ by the tumours over-expressing the enzyme, thus giving rise to a high local concentration of active drug. This confers selectivity to this agent. These quinone prodrugs (97-96 and 98-23) were tested for their ability to act as substrates for DT-diaphorase. It can be seen that these compounds are good substrates for the enzyme, and are comparable to RH1 (2,5,-diaziridinyl-3-(hydroxymethyl)-6-methyl-1,4-benzoquinone) a novel alkylating agent which is activated by DT-diaphorase and is currently about to enter clinical trial. An analysis of the growth inhibitory properties of these agents shows that 97-96 is 16-fold more active in the H460 human lung cell line which expresses active DT-diaphorase than in the diaphorase null H596 cell line 97-96 is active in an H460 human lung tumour xenograft, a tumour which expresses active DT-diaphorase. This agent may therefore have a role in the treatment of tumours that over-express DT-diaphorase. Similarly, these agents maybe selectively activated in the endothelial cells of the vasculature, thereby conferring a selective anti-vascular effect.
    TABLE 1
    Colchicine Colchicine
    IC50 Inhibition of displacement displacement
    IC50 (nM) tubulin protein:drug protein:drug
    Com- (nM) A2780/ assembly 1:10 (with 1:10 (without
    pound A2780 ADR (IC50 μM) esterase) esterase)
    1 0.72 0.84 2.4 NA 12
    33 2.8 4.0 3.6* 2 92
    34 >60 >60 6.2* 2 65
    35 4.3 5.0 7.5* 4 65
    36 1.7 2.0 4.0* ND ND
    37 6.7 9.1 6.0* 2 88
    38 14.0 27.0 7.8* ND 72
    39 2.7 4.6 8.0 2 77
    40 2.2 4.3 7.0* 4  2
    41 29.0 39.0 9.0* 2 78
    42 2.9 2.9 3.5* 2 ND
    43 3.3 3.8 3.0* 2 ND
    44 4.5 6.4 ND ND ND
  • TABLE 2
    Compound P388 A2780 H460 H596 H596/H469
    A-4 2.6 0.72 1.51
    97-96 570 190 38 620 16
    98-40 >5000 4630 >5000 2190 <0.44
    98-23 2180 2150 4370 2280 0.52
    98-33 <780 950 4110 1490 0.36
    98-24 17050 1630 3080 1950 0.63
  • IC50 are in nM.
    Figure US20070203217A1-20070830-C00027
    • 97-96 R=OMe (cis) 98-24
    • 98-40 R=OMe (trans)
    • 98-23 R=Me (cis)
    • 98-33 R=Me (trans)
    EXAMPLE 7 Synthesis of Combrestatatin A-4 Bearing a Photocleavable Group Methyl 4-(4′-formyl-2′-methoxyphenoxy) butanoate 4 (see D. L. McMinn and M. M. Greenberg, Tetrahedron, 1996, 52, 3827)
  • Figure US20070203217A1-20070830-C00028
  • To a stirred solution of vanillin (3)(2 g, 13.16 mmol) and methyl 4-bromobutyrate (2.38 g, 13.16 mmol) in DMF (20 cm3) was added freshly ground anhydrous potassium carbonate (2.03 g, 14.47 mmol). The resulting pale pink suspension was heated at 100° C. for 90 mins, after which time the suspension has become milky pink in appearance. The mixture was then poured into water (50 cm3) and extracted with Et2O (3×30 cm3). The combined organic extracts were washed with water (2×30 cm3), then 1M HCl (2×30 cm3) before being dried (MgSO4) and concentrated in vacuo. The pale pink solid 4 was used without further purification (2.7 g, 82%). m.p. 69° C. [lit. (D. L. McMinn and M. M. Greenberg, Tetrahedron, 1996, 52, 3827) m.p. 68-69° C.]; Found C, 62.0; H, 6.3; C13H16O5 requires C, 62.0; H, 6.4%; Rf 0.44 (SiO2, hexane:EtOAc, 2:1 v/v); νmax (KBr disc)/cm−1 3100-2700 (m), 1740 (s), 1680 (s); δH (300 MHz, CDCl3) 2.19 (2H, q, J6.6 Hz, CH2), 2.56 (2H, t, J6.6 Hz, CH2), 3.68 (3H, s, OCH3), 3.91 (3H, s, OCH3), 4.15 (2H, t, J6.6 Hz, CH2), 6.97 (1H, d, J 8.0Hz, H-6′), 7.40 (1H, dd, J8.0 Hz, J1.8 Hz, H-5′), 7.44 (1H, d, J1.8 Hz, H-3′), 9.84 (1H, s, CHO) ppm; δc (100 MHz, CDCl3) 24.1 (CH2), 30.2 (CH2), 51.5 (OCH3), 55.8 (OCH3), 67.3 (CH2), 109.2 (CH), 111.5 (CH), 126.6 (CH), 130.0, 149.7, 153.7, 173.3 (COOCH3), 190.8 (CHO) ppm; m/z (FAB) 253 [(MH+), 50%].
  • Methyl 4-(4′-formyl-2′-methoxy-5′-nitrophenoxy) butanoate 5 (see D. L. McMinn and M. M. Greenberg, Tetrahedron, 1996, 52, 3827.)
  • Figure US20070203217A1-20070830-C00029
  • To a pale pink solution of methyl 4-(4′-formyl-2-methoxyphenoxy) butanoate (4) (3 g, 12.3 mmol) in DCM (20 cm3) was added dropwise, at 0° C. fuming nitric acid (1.5 cm3, 37.0 mmol). The resulting green solution was stirred at 0° C. for a flirther 30 mins before being allowed to spontaneously warm to r.t., where it remained for 3 hours. The subsequent bright yellow suspension was poured onto iced water (50 cm3) and extracted with DCM (3×30 cm3). The combined organic phase was washed with sat. NaHCO3 solution (2×50 cm3), followed by water (2×50 cm3) before being dried and concentrated in vacuo providing a bright yellow solid. The desired compound 5 was used without furter purification (2.48 g, 68%). A small amount of material (250 mg, 0.84 mmol) was purified via column chromatography (SiO2, CH2Cl2) so that complete characterisation data could be obtained. m.p. 75° C. [lit. (D. L. McMinn and M. M. Greenberg, Tetrahedron, 1996, 52, 3827.) m.p. 76-78° C.]; Found C, 52.7; H, 5.2; N, 4.8; C3H15NO7 requires C, 52.5; H, 5.1; N, 4.7%; Rf 0.44 (SiO2, CH2Cl2); νmax (KBr disc)/cm−1 3100-2700 (m), 1735 (s), 1690 (s), 1290, 1220; δH (200 MHz, CDCl3) 2.23 (2H, q, J7.1 Hz, CH2), 2.57 (2H, t, J7.1 Hz, CH2), 3.71 (3H, s, OCH3), 3.99 (3H, s, OCH3), 4.21 (2H, t, J7.1 Hz, CH2), 7.40 (1H, s, H-3′), 7.61 (1H, s, H-6′), 10.44 (1H, s, CHO) ppm; δc (100 MHz, CDCl3) 24.1 (CH2), 30.2 (CH2), 51.8 (OCH3), 56.6 (OCH3), 59.6 (CH2), 108.1 (CH), 109.9 (CH), 125.5, 143.8, 151.7, 153.4, 173.2 (COOCH3), 187.8 (CHO) ppm; m/z (FAB) 298 [(MH+), 40%].
  • Methyl 4-[4′-(hydroxymethyl)-2′-methoxy-5′-nitrophenoxy] butanoate 6 (see D. L. McMinn and M. M. Greenberg, Tetrahedron, 1996, 52, 3827.)
  • Figure US20070203217A1-20070830-C00030
  • To a clear yellow solution of methyl 4-(4′-formyl-2′-methoxy-5′-nitrophenoxy) butanoate (5)(1 g, 3.36 mmol) in THF (10 cm3) was added, portion wise, sodium borohydride (128 mg, 3.36 mmol). The solution quickly changed appearance, becoming deep orange after about 5 mins. The mixture was stirred for a furher 55 mins before water was added (25 cm3), the subsequent yellow mixture was extracted with ether (3×30 cm3) and the combined organic extracts were dried (MgSO4) and concentrated in vacuo providing a pale yellow solid. The solid, 6 was used without further purification (875 mg, 87.5%). A small amount of material (200 mg, 0.67 mmol) was purified via column chromatography (SiO2, hexane:EtOAc 2:1 v/v) so that complete characterisation data could be obtained. m.p. 100° C. [lit. (D. L. McMinn and M. M. Greenberg, Tetrahedron, 1996, 52, 3827.) m.p. 98-100° C.]; Found C, 52.2; H, 5.7; N, 4.7; C13H17NO7 requires C, 52.2; H, 5.7; N, 4.7%; Rf 0.24 (SiO2, hexane:EtOAc 2:1 v/v); νmax (KBr disc)/cm−1 3400-3100 (br), 3000-2800 (m), 1730 (s), 1280, 1220; δH (200 MHz, CDCl3) 2.20 (2H, q, J7.3 Hz, CH2), 2.56 (2H, t, J7.3 Hz, CH2), 2.61 (1H, br, OH), 3.70 (3H, s, OCH3), 3.98 (3H, s, OCH3), 4.13 (2H, t, J 7.3 Hz, CH2), 4.95 (2H, s, CH2), 7.15 (1H, s, H-3′), 7.71 (1H, s, H-6′) ppm; δc (100 MHz, CDCl3) 24.3 (CH2), 30.4 (CH2), 51.7 (OCH3), 56.4 (OCH3), 62.8 (CH2), 68.3 (CH2), 109.5 (CH), 111.2 (CH), 132.4, 139.6, 147.2, 154.3, 173.4 (COOCH3) ppm; m/z (FAB) 299 [(M+), 15%], 282 [(M-OH), 40%].
  • Methyl 4-[4′-(bromomethyl)-2′-methoxy-5′-nitrophenoxy] butanoate 7
  • Figure US20070203217A1-20070830-C00031
  • To a solution of methyl 4-[4′-(hydroxymethyl)-2′-methoxy-5′-nitrophenoxy] butanoate (6) (750 mg, 2.52 mmol) in anhydrous THF (10 cm3) was added PBr3 (0.24 cm3, 2.52 mmol). The resulting deep yellow solution was refluxed for 3 hours, after which time no visible changes had occurred. The reaction mixture was cooled then poured onto ice. The resulting aqueous solution was extracted with ether (3×30 cm3). The combined organic extracts were washed with 5% NaHCO3 solution (2×30 cm3) and water (2×30 cm3) before being dried (MgSO4) and concentrated in vacuo. The desired compound 7 was furnished as a yellow oil. The oil was then purified via column chromatography (SiO2, hexane:EtOAc, 2:1 v/v) thus providing 7 as a yellow powder (580 mg, 64%). m.p. 68-70° C.; Found C, 43.2; H, 4.5; N, 4.0; Br, 21.9; C13H16NO6Br requires C, 43.1; H, 4.4; N, 3.9; Br, 22.1%; Rf 0.63 (SiO2, hexane:EtOAc 2:1 v/v); νmax (KBr disc)/cm−1 3000-2800 (m), 1720 (s), 1610, 1580, 1530, 1280, 1220; δH (200 MHz, CDCl3) 2.20 (2H, q, J6.7 Hz, CH2), 2.56 (2H, t, J 6.7 Hz, CH2), 3.70 (3H, s, OCH3), 3.97 (3H, s, OCH3), 4.13 (2H, t, J6.7 Hz, CH2), 4.86 (2H, s, CH2), 6.92 (1H, s, H-3′), 7.67 (1H, s, H-6′) ppm; δc (100 MHz, CDCl3) 24.1 (CH2), 30.1 (CH2), 51.6 (OCH3), 56.4 (OCH3), 68.0 (CH2), 68.3 (CH2), 109.7 (CH), 113.8 (CH), 127.3, 140.0, 148.1, 153.5, 173.2 (COOCH3) ppm; m/z (FAB) 362 [(MH+), 100%].
  • Methyl 4-[2′-methoxy-5′-nitro-4′-(phenoxymethyl)phenoxy] butanoate 9
  • Figure US20070203217A1-20070830-C00032
  • To a suspension of phenol (130 mg, 138 mmol) and methyl 4-[4′-(bromomethyl)-2′-methoxy-5′-nitrophenoxy] butanoate (7) (500 mg, 138 mmol) in anhydrous methanol (5 cm3) was added KtOBu (186 mg, 166 mmol). The resulting green mixture was stirred at r.t. for 30 mins after which time a white precipitate had formed. The precipitate was filtered and recrystallised from methanol affording the desired compound 9 as a pure white solid (319 mg, 61%). m.p. 96-98° C.; Found C, 61.0; H, 5.8; N, 4.0; C19H21NO7 requires C, 60.8; H, 5.6; N, 3.7%; Rf 0.64 (SiO2, hexane:EtOAc 2:1 v/v); νmax (KBr disc)/cm−1 3000-2800 (m), 1720 (s), 1610, 1580, 1520, 1280, 1220; δH (300 MHz, CDCl3) 2.21 (2H, q, J6.7 Hz, CH2), 2.57 (2H, t, J 6.7 Hz, CH2), 3.71 (3H, s, OCH3), 3.91 (3H, s, OCH3), 4.14 (2H, t, J6.7 Hz, CH2), 5.50 (2H, s, CH2), 6.98-7.02 (3H, m, ar), 7.29-7.34 (3H, m, ar), 7.77 (1H, s, ar) ppm; δc (100 MHz, CDCl3) 24.7 (CH2), 30.4 (CH2), 51.7 (OCH3), 56.3 (OCH3), 67.0 (CH2), 68.2 (CH2), 109.4 (CH), 109.6 (CH), 115.0 (CH), 121.5 (CH), 129.6 (CH), 129.7 (CH), 138.9, 147.0, 154.3, 158.1, 173.2 (COOCH3) ppm; m/z (FAB) 376 [(MH+), 20%], 282 [(M-C6H5O), 80%].
  • Methyl 4-[2′-methoxy-4′-({2″-methoxy-5″-[(Z)-2′″-3′″,4′″,5′″-trimethoxyphenyl)ethenyl]phenoxy}-methyl)-5′-nitrophenoxy]butanoate Z-8
  • Figure US20070203217A1-20070830-C00033
  • To a suspension of methyl 4-[4′-(bromomethyl)-2′-methoxy-5′-nitrophenoxy] butanoate (7) (1.5 g, 4.14 mmol) and cis-CA-4 (1.6 g, 5.0 mmol) in anhydrous methanol (20 cm3) was added KtOBu (0.58 g, 5.2 mmol). The resulting yellow mixture was stirred at r.t. for 30 mins after which time a yellow precipitate had formed. The precipitate was filtered and recrystallised from methanol furnishing the desired compound Z-8 as a pale yellow solid (1.62 g, 69%). m.p. 132-134° C.; Found C, 65.2; H, 6.5; N, 2.4; C31H35NO11 requires C, 65.6; H, 6.2; N, 2.5%; Accurate mass; found M+597.2201; C31H35NO11 requires M+597.2210; Rf 0.60 (SiO2, hexane:EtOAc 2:1 v/v); νmax (KBr disc)/cm−1 3000-2800 (m), 1730 (s), 1610, 1580, 1520, 1320, 1280, 1220, 1130, 880; λmax(MeCN)/nm 222 (ε35050), 242 (ε29263) and 293 (ε14071); δH (300 MHz, CDCl3) 2.21 (2H, q, J6.7 Hz, CH2), 2.58 (2H, t, J6.7 Hz, CH2), 3.69 (6H, s, 2×OCH3), 3.71 (3H, s, OCH3), 3.83 (3H, s, OCH3), 3.89 (3H, s, OCH3), 3.95 (3H, s, OCH3), 4.51 (2H, t, J6.7 Hz, CH2), 5.41 (2H, s, CH2), 6.45 (1H, d, J12.4 Hz, olefmic H), 6.49 (1H, d, J12.4 Hz, olefinic H), 6.50 (2H, s, 2′″-H and 6′″-H), 6.82 (1H, d, J8.3 Hz, H-3″), 6.93-6.97 (2H, m, H-4″ and H-6″), 7.46 (1H, s, H-3′), 7.75 (1H, s, H-6′) ppm; δc (100 MHz, CDCl3) 24.3 (CH2), 30.4 (CH2), 51.7 (OCH3), 55.9 (OCH3), 56.0 (OCH3), 56.2 (OCH3), 60.9 (OCH3), 68.2 (CH2), 68.4 (CH2), 105.9 (CH), 109.3 (CH), 109.7 (CH), 111.6 (CH), 115.6 (CH), 123.0 (CH), 129.2 (CH), 129.4 (CH), 129.6, 130.3, 132.6, 137.2, 138.8, 147.0, 147.3, 149.0, 152.9, 154.3, 173.4 (COOCH3) ppm; m/z (FAB) 376 [(M+), 40%].
  • 4-[2′-methoxy-4′-({2″-methoxy-5″-[(Z)-2′″-3′″,4′″,5′″-trimethoxyphenyl)ethenyl]phenoxy}methyl)-5′-nitrophenoxy]butanoic acid Z-11
  • Figure US20070203217A1-20070830-C00034
  • In a foil wrapped flask a suspension of the methyl ester Z-8 (250 mg, 0.42 mmol) in 1M aqueous NaOH (0.84 ml, 0.84 mmol) was prepared. The yellow suspension was then heated at reflux for 40 mins, after which time water (5 cm3) was added. The resulting orange solution was then acidified to pH 1 using conc. HCl and the subsequent yellow precipitate was filtered. The precipitate was purified by recrystallisation from ethanol, which provided the desired compound as a pale yellow solid (214 mg, 81%). m.p. 138° C.; Found C, 61.6; H, 5.4; N, 2.3; C30H33NO11 requires C, 61.7; H, 5.7; N, 2.4%; Rf 0.10 (SiO2, hexane:EtOAc 1:1 v/v); νmax (KBr disc)/cm−1 3500-3100 (br), 3000-2800 (m), 1740 (s), 1610, 1580, 1520, 1330, 1280, 1220, 1130, 880; δH (300 MHz, CDCl3) 2.21 (2H, q, J6.5 Hz, CH2), 2.63 (2H, t, J6.5 Hz, CH2), 3.67 [6H, s, (OCH3)2), 3.81 (3H, s, OCH3), 3.88 (3H, s, OCH3), 3.93 (3H, s, OCH3), 4.16 (2H, t, J6.5 Hz, CH2), 5.40 (2H, s, CH2), 6.43 (1H, d, J12.9 Hz, olefinic CH), 6.47 (2H, s, H-2′″ and H-6′″), 6.48 (1H, d, J12.9 Hz, olefinic H), 6.81 (1H, d, J7.9 Hz, H-3″), 6.91-6.95 (2H, m, H-4″ and H-6″), 7.45 (1H, s, H-3′), 7.74 (1H, s, H-6′) ppm; δc (100 MHz, CDCl3) 24.0 (CH2), 30.1 (CH2), 55.9 (OCH3), 56.0 (OCH3), 56.2 (OCH3), 60.9 (OCH3), 68.0 (CH2), 68.4 (CH2), 105.9 (CH), 109.4 (CH), 109.7 (CH), 111.6 (CH), 115.6 (CH), 123.1 (CH), 129.2 (CH), 129.3 (CH), 129.7, 130.3, 132.7, 137.2, 138.8, 146.9, 147.3, 149.0, 152.9, 154.3, 173.5 (COOCH3) ppm; m/z (FAB) 583 [(M+), 100%].
  • Methyl 4-[2′-methoxy-4′-({2″-methoxy-5″-[(E)-2′″-3′″,4′″,5′″-trimethoxyphenyl)ethenyl]phenoxy}-methyl)-5′-nitrophenoxy]butanoate E-8
  • Figure US20070203217A1-20070830-C00035
  • To a suspension of methyl-4-[4′-(bromomethyl)-2′-methoxy-5′-nitrophenoxy] butanoate 7 (400 mg, 1.1 mmol) and trans-CA-4 (280 mg, 0.89 mmol) in anhydrous methanol (10 cm3) was added KtOBu (148 mg, 1.32 mmol). The resulting yellow mixture was stirred at r.t. for 30 mins after which time a yellow precipitate had formed. The precipitate was filtered and recrystallised from methanol furushing the desired compound E-8 as a pale yellow solid (367 mg, 73%). m.p. 142° C.; Found C, 65.5; H, 6.4; N, 2.4; C31H35NO11 requires C, 65.6; H, 6.2; N, 2.5%; Rf 0.44 (SiO2, hexane:EtOAc 1:1 v/v); νmax (KBr disc)/cm−1 3000-2800 (m), 1730 (s), 1610, 1580, 1520, 1320, 1280, 1220, 1130, 880; λmax (MeCN)/nm 219 (ε36612), 243 (ε33655) and 329 (ε41112); δH (300 MHz, CDCl3) 2.20 (2H, p, J6.8 Hz, CH2), 2.57 (2H, t, J6.8 Hz, CH2), 3.70 (3H, s, OCH3), 3.86 (3H, s, OCH3), 3.92 [6H, s, (OCH3)2], 3.93 (3H, s, OCH3), 3.97 (3H, s, OCH3), 4.41 (2H, t, J6.8 Hz, CH2), 5.61 (2H, s, CH2), 6.73 (2H, s, H-2′″ and H-6′″), 6.87 (1H, d, J16.2 Hz, olefinic H), 6.94 (1H, s, H-3″), 6.95 (1H, d, JH 16.2 Hz, olefinic CH), 7.11-7.14 (2H, m, H-4″ and H-6″), 7.55 (1H, s, H-3′), 7.77 (1H, s, H-6′) ppm; δc (100 MHz, CDCl3) 24.4 (CH2), 30.4 (CH2), 51.7 (OCH3), 56.1 [(OCH3)2], 56.2 (OCH3), 56.3 (OCH3), 61.0 (OCH3), 68.4 (2×CH2), 103.3 (CH), 109.3 (CH), 109.7 (CH), 112.0 (2×CH), 120.7 (CH), 127.2 (CH), 127.5 (CH), 129.9, 130.7, 133.1, 137.8, 138.8, 147.0, 147.8, 149.5, 153.4, 154.5, 173.3 (COOCH3) ppm; m/z (FAB) 597 [(M+), 25%].
  • 4-[2′-methoxy-4′-({2″-imethoxy-5″-[(E)-2′″-3′″,4′″,5′″-trimethoxyphenyl)ethenyl]phenoxy}methyl)-5′-nitrophenoxy]butanoic acid E-11
  • Figure US20070203217A1-20070830-C00036
  • In a foil wrapped flask a suspension of the methyl ester E-8 (250 mg, 0.42 mmol) in 1M aqueous NaOH (0.84 ml, 0.84 mmol) was prepared. The yellow suspension was then heated at reflux for 40 mins, after which time water (5 cm3) was added. The resulting orange solution was then acidified to pH 1 using conc. HCl and the subsequent yellow precipitate was filtered. The precipitate was purified by recrystallisation from ethanol, which provided the desired compound as a pale yellow solid (234 mg, 97%). m.p. 177° C.; Found C, 62.0; H, 5.8; N, 2.5; C30H33NO11 requires C, 61.7; H, 5.7; N, 2.4%; Rf 0.15 (SiO2, hexane:EtOAc 1:1 v/v); νmax (KBr disc)/cm−1 3500-3100 (br), 3000-2800 (m), 1710 (m), 1610, 1580, 1520, 1330, 1280, 1220, 1130; δH (300 MHz, CDCl3) 2.25 (2H, q, J6.7 Hz, CH2), 2.66 (2H, t, J6.7 Hz, CH2), 3.90 (3H, s, OCH3), 3.95 (3H, s, 2×OCH3), 3.96 (3H, s, OCH3), 4.00 (3H, s, OCH3), 4.19 (2H, t, J6.7 Hz, CH2), 5.64 (2H, s, CH2), 6.76 (2H, s, H-2′″ and H-6′″), 6.93-6.98 (3H, m), 7.14-7.16, (2H, m, H-4″ and H-6″), 7.59 (1H, s, H-3′), 7.82 (1H, s, H-6′) ppm; δc (100 MHz, CDCl3) 24.0 (CH2), 30.0 (CH2), 56.1 (2×OCH3), 56.2 (OCH3), 56.3 (OCH3), 61.0 (OCH3), 68.1 (CH2), 68.2 (CH2), 103.3 (2×CH), 109.3 (CH), 109.8 (CH), 112.0 (CH), 120.7 (CH), 127.2 (CH), 127.5 (CH), 130.1, 130.7, 133.1, 137.9 139.2, 146.8, 147.8, 149.5, 153.4, 154.8, 177.4 (COOH) ppm; m/z (FAB) 583 [(M+), 70%].
  • 4-[2′-methoxy-4′-({2″-methoxy-5″-[(E)-2′″-3′″,4′″,5′″-trimethoxyphenyl)ethenyl]phenoxy}methyl)-5′-nitrophenoxy]butanoic acid(potassium salt) E-12
  • Figure US20070203217A1-20070830-C00037
  • To a suspension of the butanoic acid E-11 (250 mg, 0.43 mmol) in methanol (3 cm3) was added KtOBu [0.43 cm3, 0.43 mmol, (1 M methanolic solution)]. The resulting clear yellow solution was stirred at r.t. for 5 minutes before being concentrated in vacuo to provide a pale brown solid (0.26 mg, 97%).
  • EXAMPLE 8 Photochemical Cleavage and Isomerisation Methyl 4-[2′-methoxy-4′-({2″-methoxy-5″-[(E)-2′″-3′″,4′″,5′″-trimnethoxyphenyl)ethenyl]phenoxy}methyl)-5′-nitrophenoxy]butanoate, E-8
  • In a 400 cm3 photochemical reaction vessel, N2 was bubbled through distilled benzene (300 cm3) for 30 mins. Following the degassing procedure, compound E-12 (300 mg, 0.5 mmol) was added to the benzene and allowed to dissolve. The colourless solution was then irradiated using a 400W medium pressure Hg lamp for a total of 20 mins. Aliquots were removed from the reaction vessel at specific time points throughout the 20 mins (0, 0.5, 1.0, 1.5, 2.0, 3.0, 4.0, 5.0, 7.5, 10.0, 12.5, 15.0, 17.5, 20.0 mins). Two aliquots were removed from the reaction vessel at each time point, one sample (0.5 cm3) was used for HPLC analysis (cyanopropyl column; mobile phase 95:5 hexane:IPA; flow rate 0.5 ml min−1; lamp 245 mn), whilst the other sample (10 cm3) was concentrated in vacuo and used to determine the IC50 at that time point. After irradiation for 20 mins the reaction mixture was yellow/orange in colour. The following compound retention times were observed:
    Trans-CA-4 23.35 mins
    Cis-CA-4 11.07 mins
    Coupled trans-CA-4 E-8 43.89 mins
    Coupled cis-CA-4 E-8 26.42 mins
  • EXAMPLE 9 Photochemical Isomerization of Combrestatatin A-4 and Derivatives
  • We detail the study of the trans to cis isomerisation of CA-4 herein. A solution of trains-CA-4 (200 mg, 0.63 mmol) in freshly distilled and degassed benzene (400 cm3), prepared by bubbling through Ar for 2 hours prior to use, was irradiated using a 400 W medium pressure Hg lamp for a total of 20 mins. Aliquots were removed at regular intervals for GC analysis (180-280° C. at 5° C. min−1; pressure program 195-225 kPa at 1.5 kPa min−1; on a SE54 column).
    Figure US20070203217A1-20070830-C00038
  • The results of GC analysis of the mixture are summarized in Table 3. Table 4 shows the retention times, by GC for the individual components of the reaction mixture.
    TABLE 3
    Time (mins) % trans-stilbene % cis-stilbene % phenanthrene
    0 100 0 0
    0.5 100 0 0
    1.5 100 0 0
    3.0 87 13 0
    5.0 69 31 0
    6.5 55 45 0
    8.0 53 47 0
    10.0 44 53 0
    12.5 32 68 0
    15.0 29 71 0
    17.5 22 74 4
    20.0 23 73 4
  • TABLE 4
    Component Retention time (mins)*
    trans-CA-4 20.32
    cis-CA-4 15.42
    CA-4 phenanthrene derivative 1 19.00

    *G.C. conditions; 180-280° C. @ 5° C. min−1, 195-225 kPa @ 1.5 kPa min−1, SE 54 column.
  • Table 5 shows the outcome of photoisomerisation of trans-CA-4 (99.7% E) and the corresponding IC50 values. The cytotoxicity (IC50 value) of the reaction mixture at each time point is shown in Table 3, showing an impressive increase in the cytotoxicity of the mixture over time.
  • The most exciting observation is that the IC50 at time zero is 4000 nM and after just half a minute it has decreased by more than 10 fold. This illustrates that the isomerization is rapid and also highlights just how potent cis-CA-4 is compared with trans-CA-4 since just 2.0% of cis-CA-4 present results in such a dramatic increase in cytotoxicity. The thousand-fold increase in activity obtained after six minutes, clearly shows that the process displays great potential.
    TABLE 5
    Time (mins) % trans % cis % phenanthrene IC50 (K562) (nM)
    0 99.7 0.3 0 4000
    0.5 98.0 2.0 0 310
    1.0 96.1 3.9 0 530
    1.5 93.7 6.3 0 50
    2.0 92.7 7.3 0 20
    3.0 74.3 25.7 0 12
    4.0 62.3 37.3 0 9
    5.0 52.1 47.9 0 10
    7.5 40.2 56.6 1.4 3
    10.0 34.0 63.1 2.9 6
    12.5 32.2 64.4 3.4 4
    15.0 31.7 64.6 3.7 1
    17.5 30.8 65.1 4.1 3
    20.0 30.6 65.7 3.7 2
  • Previously, the isomerisation study had been performed ex situ, i. e. in a photochemical reactor. However, it was necessary to illustrate the potential of the process as a real therapy by repeating the isomerizaton in the presence of the cancer cells i.e. in situ. Therefore, K562 cells were dosed with a known concentration of trans-CA-4, the resulting solutions were then irradiated using an ultra violet lamp consisting of 2×7 W ultra violet tubes for given lengths of time. Following irradiation, the cells were incubated in the normal way and the IC50 values were determined. A series of control experiments were performed simultaneously to verify that the results obtained (illustrated in Table 6) were in fact due to the isomerisation process occurring and not due to the effect of the ultraviolet radiation. Table 6 shows that the same pattern of results is obtained. However, the decrease in the IC50 value is now more rapid.
  • The experiment convincingly supports the results derived from the ex situ study and we are able to show that the trans to cis isomerisation is occurring in both circumstances, and more importantly is causing a rapid and significant increase in the cytotoxicity of the system. Due to the speed with which the process was occurring the experiment was repeated and monitored every 5 seconds in the first minute of irradiation. The results are shown in Table 7. It can be seen that after just 5 seconds there is a greater than 15 fold reduction in the IC50 and after a further 40 seconds a single nanomolar figure IC50 value is reached.
    TABLE 6
    Time/mins IC50 K562 nM
    0 1200
    1 3.6
    2 2.9
    3 1.7
    4 4.4
    5 2.7
    6 2.9
    7 1.9
    8 3.0
    9 2.5
    10 2.6
    12.5 2.4
    15.0 2.0
    17.5 1.7
    20.0 2.7
  • TABLE 7
    Time/secs IC50 K562 nM
    0 580
    5 35
    10 40
    15 20
    20 18
    25 17
    30 15
    35 11
    40 11
    45 9
    50 9
    55 9
    60 8
  • EXAMPLE 10 Synthesis of Trans-CA-4 Bearing a Photocleavable Group
  • We next demonstrate the potential use of light to trigger the release of CA-4 from a variety of non-toxic CA-4 derivatives. By way of example, two types of reaction were employed:
    • (1) Release of CA-4 from a trans-CA-4 derivative with E to Z isomerization.
    • (2) Release of CA-4 from a photolabile cis-CA-4 derivative.
  • As in some applications in the prior art, the insolubility of CA-4 in water results in clinical difficulties, it would be advantageous to utilize a photocleavable group to impart water solubility to the system.
  • We first considered the fate of a trans-CA-4 derivative (2) bearing a photocleavable water solubilising (PCWS) group (see the scheme below). When irradiated with ultraviolet light, two events can occur; (i) the PCWS group can be cleaved and (ii) trans to cis isomerisation can take place, (but perhaps not in that order). The overall effect of these transformations should be a dramatic increase in cytotoxicity.
    Figure US20070203217A1-20070830-C00039
  • It became apparent that whilst there are examples of the use of photocleavable groups in medical applications there are no examples of their use in this context. To demonstrate the validity of this approach we used an ortho-nitrobenzyl derivative as a prototypical photocleavable group. A vanillin based compound, developed by Holmes et al. (C. Holmes, J. Org. Chem., 1997, 62, 2370) as a solid phase linker, was used as a template for our photocleavable group. The synthetic strategy to the coupling agent 7 is outlined in the following scheme.
    Figure US20070203217A1-20070830-C00040
  • the desired product E-8 in a 73% yield, as illustrated below. E-8 precipitated out of solution as the reaction to place and was subsequently filtered and purified by recrystallisation. The coupling reaction was repeated using cis-CA-4 as the substrate-the reaction proceeded in a 69% yield.
    Figure US20070203217A1-20070830-C00041
  • As with the trans to cis photoinduced isomerisation study coupled trans-CA-4 was dissolved in benzene and the resulting solution was irradiated for a total of 20 minutes with aliquots removed at given time points to determine the isomeric ratios and the IC50 value. The results obtained are detailed in Table 8.
    TABLE 8
    % coupled trans-CA-4 % % IC50 K562
    Time/mins (E-8) trans-CA-4 cis-CA-4 (nM)
    0 100 >150000
    0.5 93 7 14580
    1.0 88 8 4 450
    1.5 80 12 8 90
    20. 72 19 9 59
    3.0 62 27 11 14
    4.0 61 31 8 12
    5.0 53 37 10 8
    7.5 42 45 13 4
    10.0 34 47 19 6
    12.5 20 35 45 2
    15.0 Unable to calculate* 2
    17.5 Unable to calculate* 4
    20.0 Unable to calculate* 3
  • The results show that the photocleavage of trans-CA-4 is the first event to occur, followed soon after by the isomerisation of trans-CA-4 to cis-CA-4. This process is accompanied by an even more dramatic increase in the cytotoxicity than was seen previously, this is largely because E-8 is much less cytotoxic than trans-CA-4, in fact it is 5 times less cytotoxic. This is highly beneficial if E-8 were to be used as a CA-4 prodrug. These results also highlight the speed with which the process occurs. After just 5 minutes exposure to ultra violet light the IC50 value has fallen to single nanomolar figures, with slightly more than 50% of the coupled starting material remaining and some 10% cis-CA-4 present.
  • Once again it was desirable to investigate the effect of the coupled trans-CA-4, E-8 upon irradiation in the presence of cancer cells. Therefore, K562 cells were dosed with a known concentration of coupled trans-CA-4 E-8 and subsequently exposed to ultra violet radiation for specific lengths of time and the IC50 value determined. As with the previous in situ experiment a series of control experiments were performed simultaneously to verify that any positive results obtained were due to the effect of ultra violet light on the drug candidate and not its effect on the cells. These consisted of i) exposing KS562 cells only (i.e. cells which were not dosed with any potential drug candidate) to ultra violet light, ii) preventing K562 cells dosed with drug candidate from being exposed to ultra violet light, and iii) preventing K562 cells only (i.e. cells which were not dosed with any potential drug candidate) from being exposed to ultra violet light. In all three control experiments no cytotoxic effect was observed. The results of the main experiment are illustrated in Table 9. It is apparent that the same general trend, with respect to the IC50 values is seen in both the ex situ and the in situ experiments.
    TABLE 9
    Time/Mins IC50 K562 (nm)
    0 >100000
    0.5 1540
    1 450
    1.5 250
    2 130
    3 40
    4 11
    5 9
    7.5 5
    10 2
    12.5 3
    15 3
    17.5 2
    20 2
  • We needed to demonstrate that the photo by-product, the nitrosobenzaldehyde, 10 did not possess any cytotoxicity and therefore was not contributing to the results obtained for the photoactivation of coupled trans-CA-4. The synthesis of the nitroso compound, 10 was achieved by exposing compound 9 to ultra violet light for 10 minutes and the desired compound was isolated from the resulting reaction mixture via column chromatography (see the scheme below). The compound 10 lacked significant cytotoxicity.
    Figure US20070203217A1-20070830-C00042
  • Having convincingly established the feasibility of prodrug photoactivation, the final objective was to render the coupled trans-CA-4 compound, E-8 water-soluble. A water soluble prodrug of CA-4 would clearly have clinical potential. Therefore, a water-soluble derivative was synthesised in an attempt to overcome this problem. The water-soluble derivative was prepared from the methyl ester, E-8 by a simple hydrolysis reaction using NaOH in ethanol. However, due to purification problems the carboxylic acid, E-11 was isolated and subsequently deprotonated with KtOBu, the water-soluble salt was isolated using ion-exchange chromatography.
    Figure US20070203217A1-20070830-C00043
  • The in situ study of the water-soluble derivative of frans-CA-4 was performed in the same manner as those described previously and the results can be seen in Table 10. The results obtained suggest that the water-soluble derivative, E-12 behaves in a similar fashion as the methyl ester derivative, E-8 when irradiated with ultra violet light in the presence of cancer cells. The IC50 values obtained are similar to those obtained for the in situ study of the methyl ester derivative, E-8.
    TABLE 10
    Time/Mins IC50 K562 (Nm)
    0 >50000
    0.5 1010
    1 410
    1.5 300
    2 160
    3 50
    4 58
    5 46
    7.5 22
    10 16
    12.5 12
    15 10
    17.5 9
    20 8
  • EXAMPLE 11 Further Biological Experiments
  • The effect of compounds 97-64H and 97-96 (quinone compound) was tested on H460 human lung xenograft grown subcutaneously by injecting equitoxic doses of the compounds (0.75×MTD). Twenty four hours following injection (i.p.) both compounds caused extensive damage to the tumours which was consistent with necrosis caused by destruction of vasculature.
  • The results from testing compounds of the invention in cell based assays are reported in Tables 12 to 27 below.
  • The references cited herein are incorporated by reference.
    TABLE II
    A2780 1C50 Tub Ass 1C50 Colchicine % HUVEC 1C50
    Compound P388 1C50 (μM) (μM) H460 1C50 (μM) K562 (μM) (μM) Inhibition (μM) Permeability
    Comb A-4 0.0026 0.00072 1.51 2.4 12 0.001 7.1, 6.9
    (1)
    97-64H 0.000489 0.00366 <1.25, 12.5 79, 70 .0024 5.8
    97-64L 0.171 0.113 >100 43 NT 1.0
    97-65 0.18 0.095 100 NT 0.35 3.4
    96-188 0.32 .081 NT NT
    96-167 0.0008 0.00037 9 3.8 0.04 5.5
    97-07 0.0023 0.00147 2.78 10 6.6, 12.3
    97-13H 0.0326 0.0179 .032-.035 >100 84.2 0.045 4.3, 3.2
    98-21 .000935 .00066 <1.25 1.4 0.0095 4.5
    97-96 0.57 0.19 .038 <1.25 82, 24 0.23, 0.22 2.2
    98-23 2.18 2.15 4.37 NT NT
    98-35H .00328 .00144 6.25 5 0.016 7.4
    98-29 0.45 0.38 >100 53 0.43 3.9
    99-03H 0.19 0.12 0.06 60
    00-82 .06 0.05 7.2 39.5
    00-105 0.02 >100 3.6
    17a 0.2
    17b 6
    19 0.04 1.8
    20 7 1.8
    23a 0.1
    23b 0.79
    27 9
    28 0.021 1.5
    31a 0.018 7.5
    32a 0.044 18
    45 0.12 1.8
    NT = NOT TESTED; ‘H’ COMPOUNDS ARE CIS, ‘L’ COMPOUNDS ARE TRANS. Stilbenes synthesised by either Wittig or 2-step synthesis routes.
    Quinone synthesised as in Section 9.
    Figure US20070203217A1-20070830-C00044
    97-64 R = OMe, X = F
    97-65 R = NMe2.HCl, X = H
    96-188 R = CHO, X = H
    98-21 R = Me,X = OH
    98-35 R = Me, X = F
    Figure US20070203217A1-20070830-C00045
    Figure US20070203217A1-20070830-C00046
    Figure US20070203217A1-20070830-C00047
    97-96 R = OMe
    98-23 R = Me
    Figure US20070203217A1-20070830-C00048
    Figure US20070203217A1-20070830-C00049
    Figure US20070203217A1-20070830-C00050
    Figure US20070203217A1-20070830-C00051
    Figure US20070203217A1-20070830-C00052
  • Figure US20070203217A1-20070830-C00053
    Figure US20070203217A1-20070830-C00054
    Figure US20070203217A1-20070830-C00055
    TABLE 12
    Biological activity of stilbenes with 3,4,5-trimethoxy substitution on the A
    ring
    Figure US20070203217A1-20070830-C00056
    Compound MTT (P388) MTT (K562) MA CD
    number R2 R1 Config IC50 (μM) IC50 (μM) IC50 (μM) IC50 (μM)
    CA4 (15) OH OMe Z 0.003 0.001 0.175 3
    32 H OMe Z ND 0.004 0.2 5.5
    90 F OMe Z ND 0.01 0.085 2.8
    93 Br OMe Z ND 0.001 0.4 10
    96 E ND 7.83 >10 >25
    101 OH H Z 0.3 0.14 >10 >25
    102 E 34 22 >10 >25
    103 Me Me Z 0.1 0.04 2 >25
    104 E 20 35.8 >10 >25
  • TABLE 13
    Growth inhibition studies using HUVEC
    HUVEC
    Drug IC50 (μM)
    CA4 (15) 0.0026
    90 0.004
    32 0.006
    93 0.067
  • TABLE 14
    Biological activity of stilbenes with 3,4,5-trimethyl substitution on the A
    ring
    Figure US20070203217A1-20070830-C00057
    Compound MTT (K562) MA CD
    number R IC50 (μM) IC50 (μM) IC50 (μM)
    CombretastatinA-4 (15) 0.001 0.175 3
    117 H 0.31 0.650 >25
    120 F 0.14 0.700 10
    133 OH 0.020 0.120 10
  • TABLE 15
    Growth inhibition studies using HUVECs
    HUVEC
    Drug IC50 (μM)
    117 >1
    120 0.46
    133 0.05
  • TABLE 16
    K562 cell cycle analysis
    % of cells with DNA
    % of cells content = 2n or >2n
    with DNA % of cells % of cells in S % of cells
    Drug content <2n in G0-G1 phase in G2-M
    117 15 3 10 87
    120 23 6 17 77
    133 6 3 6 91
  • TABLE 17
    Biological activity of stilbenes with 3,4-dimethyl substitution on the A-ring
    Figure US20070203217A1-20070830-C00058
    MTT MTT
    Compound (P388) (K562) MA CD
    number R R1 Config IC50 (μM) IC50 (μM) IC50 (μM) IC50 (μM)
    Combretastatin A-4 (15) Z 0.003 0.001 0.175 3
    137 H Me Z 2 3.4 >10 >25
    138 E 17 >50 >10 >25
    139 NO2 OMe Z 20 1.8 >10 >25
    140 E >50 24 >10 >25
    141 H OMe Z 4.9 1.6 >10 >25
    142 E 20.5 >50 >10 >25
    143 Br OMe Z 2.5 1.15 >10 >25
    144 E >50 >50 >10 >25
    146 OH OMe Z 0.4 0.07 3.09 12.5
    147 F OMe Z 1.7 0.09 6.57 >25
    148 E 36 2.5 >10 >25
    149 F Me Z 7.2 0.39 >10 >25
    150 E 40 >50 >10 >25
    151 NO2 Me Z 9.7 1.5 >10 >25
    152 E >50 >50 >10 >25
  • TABLE 18
    K562 cell cycle analysis
    % of cells with DNA
    % of cells content = 2n or >2n
    with DNA % of cells in % of cells in S % of cells
    Drug content <2n G0-G1 phase in G2-M
    Combretastatin 7 3 6 92
    A-4 (15)
    146 31 8 27 65
    147 28 10 33 57
  • TABLE 19
    Biological activity of stilbenes with 3,4,5-triethoxy substitution on the
    A-ring
    Figure US20070203217A1-20070830-C00059
    Compound MTT (K562) MA CD
    Combretastatin A-4 (15) Z 0.001 0.175 3
    247 Br Z 0.6 >10 >25
    248 E >50 >10 >25
    249 H Z 0.5 >10 >25
    250 E >50 >10 >25
    251 F Z 0.044 1.25 >25
    252 E >50 >10 >25
    253 OH Z 0.018 0.50 15.5
    254 E 0.2 >10 >25
    255 Br Z 0.6 >10 >25
    256 E >50 >10 >25
    257 Cl Z 0.45 >10 >25
    258 E 7 >10 >25
  • TABLE 20
    Growth inhibition studies using HUVECs
    HUVEC
    drug IC50 (μM)
    253 0.05
    251 0.19
  • TABLE 21
    K562 cell cycle analysis
    % of cells with DNA
    % of cells with content = 2n or >2n
    DNA content % of cells in S % of cells
    Drug <2n % of cells in G0-G1 phase in G2-M
    253 4 3 6 92
    251 24 7 18 75
  • TABLE 22
    Biological activity of stilbenes with substitution on the olefinic bond
    Figure US20070203217A1-20070830-C00060
    Com- MTT
    pound Con- (K562) MA CD
    number R1 R2 fig IC50 (μM) IC50 (μM) IC50 (μM)
    Combretastatin A-4 (15) Z 0.001 0.175 3
    208 Me OTBDMS Z 0.2 1.5 >25
    209 E 6 >10 >25
    210 Me OH Z 0.04 0.13 6
    211 E 0.7 >10 >25
    213 Me H Z 0.1 1.3 >25
    214 E 0.8 >10 >25
    217 Et OTBDMS Z 0.5 >10 >25
    218 E 3.4 >10 >25
    219 Et OH Z 0.12 0.13 >25
    220 E 4 >10 >25
    80 CO2H OH E >50 >10 >25
    81 CO2Me OH E >50 >10 >25
    82 CH2OH OH E >50 >10 >25
  • TABLE 23
    Growth inhibition studies using HUVECs
    HUVEC
    Drug IC50 (μM)
    210 0.09
    213 0.35
    219 0.22
  • TABLE 24
    K562 cell cycle analysis on double bond substituted analogues
    % of cells with DNA
    % of cells with content = 2n or >2n
    DNA content % of cells in S % of cells
    Drug <2n % of cells in G0-G1 phase in G2-M
    210 23 5 22 73
    213 23 5 21 73
    219 24 7 19 74
  • TABLE 25
    Biological activity of stilbenes with substitution on the double bond
    Figure US20070203217A1-20070830-C00061
    Compound MTT (P388) MTT (K562) MA CD
    number Config R R1 IC50 (μM) IC50 (μM) IC50 (μM) IC50 (μM)
    200 E H Me 7 2 9 >25
    210 Z Me H ND 0.04 0.13 6
    211 E Me H ND 0.7 >10 >25
  • TABLE 26
    Biological activity of monofluoro prodrug precursors
    Figure US20070203217A1-20070830-C00062
    Com-
    pound Con- MTT (K562) MA CD
    number R R1 fig IC50 (μM) IC50 (μM) IC50 (μM)
    Combretastatin A-4 (15) Z 0.001 0.175 3
    240 OTBDMS F Z 0.5 >10 >25
    241 E 10 >10 >25
    242 OH F Z 0.02 1.25 9
    243 E 5 >10 >25
    90 OMe F Z 0.01 0.085 2.8
    18 OH OH Z 0.04a 4-5 22

    aL1210 murine leukaemia cell line
  • TABLE 27
    K562 cell cycle analysis
    % of cells with DNA
    % of cells with content = 2n or >2n
    DNA content % of cells in S % of cells
    Drug <2n % of cells in G0-G1 phase in G2-M
    242 29 7 22 71
    90 16 5 16 79

Claims (33)

1. A compound represented by the structural formula:
Figure US20070203217A1-20070830-C00063
wherein:
the zigzag line indicates that the compound can be cis or trans;
Ra is selected from the groups consisting of:
Figure US20070203217A1-20070830-C00064
X is selected from hydroxyl, nitro, amino, aryl, heteroaryl, alkyl, alkoxy, CHO, COR, haloalkyl, NH2, NHR, NRR′, SR, CONH2, CONHR, CONHRR′, O-aryl, O-heteroaryl or O-ester;
R1, R2 and R3 are independently selected from hydrogen, alkyl, CHO, COR, alkoxy, hydroxyl, NH2, NHR, NRR′, SR, haloalkyl or halogen;
R4 and R5 are independently selected from hydrogen, alkyl, CH2NHCOR″ or CH2CONHR″; and,
R6, R7 and R8 are independently selected from alkyl or alkoxy;
wherein R and R′ are independently selected from C-1-10 alkyl groups and R″ is a C1-10 alkyl group, aryl group or heteroaryl group;
or a salt, an ester, a free acid or base or a hydrate thereof.
2. A compound according to claim 1 represented by the structural formula:
Figure US20070203217A1-20070830-C00065
wherein:
the zigzag line indicates that the compound can be cis or trans;
X is selected from hydroxyl, nitro, amino, aryl, heteroaryl, alkyl, alkoxy, CHO, COR, haloalkyl, NH2, NHR, NRR′, SR, CONH2, CONHR, CONHRR′, O-aryl, O-heteroaryl or O-ester;
R1 is selected from alkyl, CHO, alkoxy, NH2, NHR, NRR′, SR, CF3 or halogen;
R2 and R3 are independently selected from hydrogen, alkyl, alkoxy, hydroxyl, NH2, NHR, NRR′, SR, haloalkyl or halogen;
R4 and R5 are independently selected from hydrogen, alkyl, CH2NHCOR″ or CH2CONHR″; and,
R6, R7 and R8 are independently selected from alkyl or alkoxy;
wherein at least one of the substituents R4 and R5 is an alkyl group; and
wherein R and R′ are independently selected from C1-10 alkyl groups and R″ is a C1-10 alkyl group, aryl group or heteroaryl group;
or a salt, an ester, a free acid or base or a hydrate thereof.
3. The compound of claim 2 which is the cis or Z-isomer.
4. The compound of claim 2 which is the trans or E-isomer.
5. The compound of claim 2, wherein the alkyl group R4 and/or R5 is a methyl or ethyl group.
6. The compound of claim 2, wherein the compound is selected from one of compounds 208 to 220 or 80 to 82 having the formula defined in Table 22.
7. A compound according to claim 1 represented by the structural formula:
Figure US20070203217A1-20070830-C00066
wherein:
X is selected from hydroxyl, nitro, amino, aryl, heteroaryl, alkyl, alkoxy, CHO, COR, haloalkyl, NH2, NHR, NRR′, SR, CONH2, CONHR, CONHRR′, O-aryl, O-heteroaryl or O-ester;
R1 is selected from alkyl, CHO, alkoxy, NH2, NHR, NRR′, SR, CF3 or halogen;
R2 and R3 are independently selected from hydrogen, alkyl, alkoxy, hydroxyl, NH2, NHR, NRR′, SR, haloalkyl or halogen;
R4 and R5 are independently selected from hydrogen, alkyl, CH2NHCOR″ or CH2CONHR″; and,
wherein R6, R7 and R8 are independently selected from alkyl or alkoxy such that at least one of these substituents is an alkyl group; and
wherein R and R′ are independently selected from C1-10 alkyl groups and R″ is a C1-10 alkyl group, aryl group or heteroaryl group;
or a salt, an ester, a free acid or base or a hydrate thereof.
8. The compound of claim 7, wherein two of the R6, R7 and R8 groups are alkyl groups.
9. The compound of claim 7, wherein all three of the R6, R7 and R8 groups are alkyl groups.
10. The compound of claim 7, wherein the groups of the R6, R7 and R8 groups which are alkyl groups are methyl, ethyl or propyl groups.
11. The compound of claim 7, wherein the compound is selected from one of compounds 117, 120, 133 or 137 to 142 having the formula defined in Tables 14 or 17.
12. A compound represented by the structural formula:
Figure US20070203217A1-20070830-C00067
wherein:
R1 is selected from alkyl, alkoxy, NH2, NHR, NRR′, SR, CF3, CHO or halogen;
R2 and R3 are independently selected from hydrogen, alkyl, alkoxy, hydroxyl, NH2, NHR, NRR′, SR, haloalkyl or halogen;
R4 and R5 are independently selected from the hydrogen, alkyl, CH2NHCOR″ or CH2CONHR″; and,
R6, R7 and R8 are independently selected from hydrogen, alkyl or alkoxy; and,
wherein R and R are independently selected from substituted or unsubstituted C1-10 alkyl groups and R″ is a substituted or unsubstituted C1-10 alkyl group, aryl group or heteroaryl group;
or a salt thereof;
wherein X is a group represented by:
Figure US20070203217A1-20070830-C00068
wherein BOC represents a t-butoxycarbonyl group and the A group is a naturally occurring amino acid side chain.
13. The compound of claim 12, wherein the BOC amino acid ester comprises Phe, Ile, Gly, Trp, Met, Leu, Ala, His, Pro, D-Met, D-Trp, or Tyr.
14. The compound of claim 12, wherein the amino acid is Phe, the A group is —CH2Ph.
15. A compound according to claim 1 represented by the structural formula:
Figure US20070203217A1-20070830-C00069
wherein:
the zigzag line indicates that the compound can be cis or trans;
R1, R2 and R3 are independently selected from hydrogen, alkyl, CHO, COR, alkoxy, hydroxyl, NH2, NHR, NRR′, SR, haloalkyl or halogen;
R4 and R5 are independently selected from hydrogen, alkyl, CH2NHCOR″ or CH2CONHR″; and,
R6, R7 and R8 are independently selected from alkyl or alkoxy; and
wherein R and R′ are independently selected from substituted or unsubstituted C1-10 alkyl groups and R″ is a substituted or unsubstituted C1-10 alkyl group, aryl group or heteroaryl group; or
a salt, an ester, a free acid or base or a hydrate thereof.
16. The compound of claim 15, wherein the compound is compound 97-96 or 98-40, 98-23, 98-33 or 98-24 having the formula set out in Table 2.
17. A composition comprising the compound of claim 1 and a pharmaceutically acceptable carrier.
18. A composition comprising the compound of claim 2 and a pharmaceutically acceptable carrier.
19. A composition comprising the compound of claim 7 and a pharmaceutically acceptable carrier.
20. A composition comprising the compound of claim 12 and a pharmaceutically acceptable carrier.
21. A composition comprising the compound of claim 15 and a pharmaceutically acceptable carrier.
22. A method for the treatment of cancer or a condition characterized by abnormal proliferation of the vasculature in a patient in need of said treatment by administering a therapeutically effective amount of a compound of claim 1.
23. The method of claim 22, wherein said compound is administered for treatment of a condition characterized by abnormal proliferation of the vasculature that is selected from the group of diabetic retinopathy, psoriasis and endometriosis.
24. A method for the treatment of cancer or a condition characterized by abnormal proliferation of the vasculature in a patient in need of said treatment by administering a therapeutically effective amount of a compound of claim 2.
25. The method of claim 24, wherein said compound is administered for treatment of a condition characterized by abnormal proliferation of the vasculature that is selected from the group of diabetic retinopathy, psoriasis and endometriosis.
26. A method for treatment of cancer or a condition characterized by abnormal proliferation of the vasculature in a patient in need of said treatment by administering a therapeutically effective amount of a compound of claim 7.
27. The method of claim 26, wherein said compound is administered for treatment of a condition characterized by abnormal proliferation of the vasculature that is selected from the group of diabetic retinopathy, psoriasis and endometriosis.
28. A method for the treatment of cancer or a condition characterized by abnormal proliferation of the vasculature in a patient in need of said treatment by administering a therapeutically effective amount of a compound of claim 12.
29. The method of claim 28, wherein said compound is administered for treatment of a condition characterized by abnormal proliferation of the vasculature that is selected from the group of diabetic retinopathy, psoriasis and endometriosis.
30. A method for the treatment of cancer or a condition characterized by abnormal proliferation of the vasculature in a patient in need of said treatment by administering a therapeutically effective amount of a compound of claim 15.
31. The method of claim 30, wherein said compound is administered for treatment of a condition characterized by abnormal proliferation of the vasculature that is selected from the group of diabetic retinopathy, psoriasis and endometriosis.
32. The compound of claim 1, having the formula:
Figure US20070203217A1-20070830-C00070
33. The compound of claim 1, having the formula:
Figure US20070203217A1-20070830-C00071
US11/744,405 2000-12-21 2007-05-04 Substituted stilbenes and their reactions Abandoned US20070203217A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US11/744,405 US20070203217A1 (en) 2000-12-21 2007-05-04 Substituted stilbenes and their reactions
US13/588,331 US8853270B2 (en) 2000-12-21 2012-08-17 Substituted stilbenes and their reactions

Applications Claiming Priority (7)

Application Number Priority Date Filing Date Title
GB0031262.9 2000-12-21
GB0031262A GB0031262D0 (en) 2000-12-21 2000-12-21 Substituted stilbenes and their reactions
GB0100295A GB0100295D0 (en) 2001-01-05 2001-01-05 Substituted stilbenes and their reactions
GB0100295.5 2001-01-05
US10/451,213 US7220784B2 (en) 2000-12-21 2001-12-20 Substituted stilbenes and their reactions
PCT/GB2001/005702 WO2002050007A2 (en) 2000-12-21 2001-12-20 Substituted stilbenes, their reactions and anticancer activity
US11/744,405 US20070203217A1 (en) 2000-12-21 2007-05-04 Substituted stilbenes and their reactions

Related Parent Applications (3)

Application Number Title Priority Date Filing Date
PCT/GB2001/005702 Continuation WO2002050007A2 (en) 2000-12-21 2001-12-20 Substituted stilbenes, their reactions and anticancer activity
US10451213 Continuation 2001-12-20
US10/451,213 Continuation US7220784B2 (en) 2000-12-21 2001-12-20 Substituted stilbenes and their reactions

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US13/588,331 Continuation US8853270B2 (en) 2000-12-21 2012-08-17 Substituted stilbenes and their reactions

Publications (1)

Publication Number Publication Date
US20070203217A1 true US20070203217A1 (en) 2007-08-30

Family

ID=26245470

Family Applications (3)

Application Number Title Priority Date Filing Date
US10/451,213 Expired - Fee Related US7220784B2 (en) 2000-12-21 2001-12-20 Substituted stilbenes and their reactions
US11/744,405 Abandoned US20070203217A1 (en) 2000-12-21 2007-05-04 Substituted stilbenes and their reactions
US13/588,331 Expired - Fee Related US8853270B2 (en) 2000-12-21 2012-08-17 Substituted stilbenes and their reactions

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US10/451,213 Expired - Fee Related US7220784B2 (en) 2000-12-21 2001-12-20 Substituted stilbenes and their reactions

Family Applications After (1)

Application Number Title Priority Date Filing Date
US13/588,331 Expired - Fee Related US8853270B2 (en) 2000-12-21 2012-08-17 Substituted stilbenes and their reactions

Country Status (5)

Country Link
US (3) US7220784B2 (en)
EP (1) EP1351912A2 (en)
AU (1) AU2002216228A1 (en)
CA (1) CA2432640A1 (en)
WO (1) WO2002050007A2 (en)

Families Citing this family (44)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2432640A1 (en) * 2000-12-21 2002-06-27 Cancer Research Technology Limited Substituted stilbenes, their reactions and anticancer activity
CA2463902A1 (en) 2001-10-26 2003-05-01 Oxigene, Inc. Functionalized stilbene derivatives as improved vascular targeting agents
TWI317634B (en) 2001-12-13 2009-12-01 Nat Health Research Institutes Aroyl indoles compounds
US7632955B2 (en) 2001-12-13 2009-12-15 National Health Research Institutes Indole compounds
US7528165B2 (en) 2001-12-13 2009-05-05 National Health Research Institutes Indole compounds
WO2004078126A2 (en) 2003-02-28 2004-09-16 Oxigene, Inc. Compositions and methods with enhanced therapeutic activity
GB0306908D0 (en) * 2003-03-26 2003-04-30 Angiogene Pharm Ltd Bioreductively activated stilbene prodrugs
GB0306907D0 (en) * 2003-03-26 2003-04-30 Angiogene Pharm Ltd Boireductively-activated prodrugs
GB2403949A (en) * 2003-07-18 2005-01-19 Sigma Tau Ind Farmaceuti Combretastatin derivatives
EP1654210B1 (en) * 2003-08-14 2008-07-30 Thomas Szekeres Stilbene derivatives and their use in medicaments
US7223747B2 (en) * 2003-09-24 2007-05-29 Arizona Board Of Regents Acting For And On Behalf Of Arizona State University Halocombstatins and methods of synthesis thereof
HRP20040645A2 (en) * 2004-07-14 2007-01-31 Institut "Ruđer Bošković"
CN1723884A (en) * 2004-07-21 2006-01-25 中国人民解放军军事医学科学院放射医学研究所 Cis-1, the diphenyl ethylene derivatives that 2-replaces are used to prepare the purposes of the medicine of treatment or prevent diabetes
WO2006036743A2 (en) * 2004-09-24 2006-04-06 Arizona Board Of Regents, A Body Corporate Of The State Of Arizona Acting For And On Behalf Of Arizona State University Halocombstatins and methods of synthesis thereof
US7456289B2 (en) 2004-12-31 2008-11-25 National Health Research Institutes Anti-tumor compounds
US7759527B2 (en) * 2006-03-28 2010-07-20 Council Of Scientific & Industrial Research Microwave induced one pot process for the preparation of arylethenes
DE112007000790T5 (en) 2006-03-28 2009-04-23 Council Of Scientific & Industrial Research Single stage microwave-induced process for the preparation of substituted stilbenes and their analogues
CN101085743B (en) * 2006-06-06 2012-02-15 浙江大德药业集团有限公司 Fluorine-containing alkoxy combretastatin derivative, preparation method and use thereof
TW200819409A (en) * 2006-10-19 2008-05-01 Univ Taipei Medical Z-stilbenes derivatives and the pharmaceutical composition thereof
AU2008242626B2 (en) * 2007-04-20 2012-04-12 Acucela Inc. Styrenyl derivative compounds for treating ophthalmic diseases and disorders
WO2009059448A1 (en) * 2007-11-05 2009-05-14 Zhiquan Yong Styrene-acid derivative and use in manufacturing blood-vessel targeted-agent drugs
WO2009067706A1 (en) 2007-11-21 2009-05-28 Oxigene, Inc. Method for treating hematopoietic neoplasms
WO2009111543A2 (en) * 2008-03-05 2009-09-11 Edison Pharmaceuticals, Inc. Treatment of hearing and balance impairments with redox-active therapeutics
US8716532B2 (en) 2009-03-27 2014-05-06 Council Of Scientific And Industrial Research One pot multicomponent synthesis of some novel hydroxy stilbene derivatives with alpha, beta-carbonyl conjugation under microwave irradiation
JP2014527036A (en) 2011-06-27 2014-10-09 ザ ジャクソン ラボラトリー Methods and compositions for the treatment of cancer and autoimmune diseases
CN102863388B (en) * 2011-07-05 2015-04-29 南京圣和药业股份有限公司 Tumor targeted drug Combretastatin A4 derivatives
CN103889964B (en) * 2011-07-05 2016-03-02 南京圣和药业股份有限公司 Tumor-targeting drug Combretastatin A4 derivative
GB201113815D0 (en) * 2011-08-10 2011-09-28 Univ Salford The Multi-photon isomerisation of transcombretastatins
CN103030549B (en) * 2011-09-30 2016-09-14 中国科学院福建物质结构研究所 P-benzoquinone derivative and application thereof
PL220039B1 (en) * 2012-03-29 2015-08-31 Univ Medyczny Im Karola Marcinkowskiego W Poznaniu New derivatives of (Z)-1,2-diphenylethan
US9353150B2 (en) 2012-12-04 2016-05-31 Massachusetts Institute Of Technology Substituted pyrazino[1′,2′:1 ,5]pyrrolo[2,3-b]-indole-1,4-diones for cancer treatment
CA2905509A1 (en) 2013-03-15 2014-09-18 Memorial Sloan-Kettering Cancer Center Hsp90-targeted cardiac imaging and therapy
US20170051149A1 (en) 2014-04-29 2017-02-23 Ecole Normale Superieure De Lyon Azoaryls as reversibly modulatable tubulin inhibitors
TW201702218A (en) 2014-12-12 2017-01-16 美國杰克森實驗室 Compositions and methods relating to the treatment of cancer, autoimmune disease, and neurodegenerative disease
EP3337495A4 (en) 2015-08-18 2019-04-10 Mateon Therapeutics, Inc. Use of vdas to enhance immunomodulating therapies against tumors
US10918627B2 (en) 2016-05-11 2021-02-16 Massachusetts Institute Of Technology Convergent and enantioselective total synthesis of Communesin analogs
EP3571183B1 (en) 2017-01-20 2024-03-06 The Regents of the University of California 3-(phenyl)-2-(aminomethyl)-1-phenyl-2-propen-1-one derivatives as inhibitors of the n-terminal domain of the androgen receptor for treating prostate cancer
US11932650B2 (en) 2017-05-11 2024-03-19 Massachusetts Institute Of Technology Potent agelastatin derivatives as modulators for cancer invasion and metastasis
US10640508B2 (en) 2017-10-13 2020-05-05 Massachusetts Institute Of Technology Diazene directed modular synthesis of compounds with quaternary carbon centers
WO2020247054A1 (en) 2019-06-05 2020-12-10 Massachusetts Institute Of Technology Compounds, conjugates, and compositions of epipolythiodiketopiperazines and polythiodiketopiperazines and uses thereof
CN111875513B (en) * 2020-08-20 2022-12-30 湖南省人民医院 Resveratrol A ring N (CH) 3 ) 2 Base derivatives, preparation method and application thereof
CN112500278A (en) * 2020-12-24 2021-03-16 兰州大学 Design and preparation method of NQO1 activated Combretastatin prodrug
CN112759515B (en) * 2020-12-28 2023-06-13 广东药科大学 Novel FFA1 and PPAR alpha/gamma/delta quadruple agonist, preparation method thereof and application thereof as medicine
JP2024514770A (en) 2021-03-25 2024-04-03 セケレス、トーマス Compounds for use in the treatment and prevention of COVID-19

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4996237A (en) * 1987-01-06 1991-02-26 Arizona Board Of Regents Combretastatin A-4
US5430062A (en) * 1992-05-21 1995-07-04 Research Corporation Technologies, Inc. Stilbene derivatives as anticancer agents
US5561122A (en) * 1994-12-22 1996-10-01 Arizona Board Of Regents Acting On Behalf Of Arizona State University Combretastatin A-4 prodrug
US5569786A (en) * 1987-01-06 1996-10-29 Arizona Board Of Regents Acting On Behalf Of Arizona State University Isolation, structural elucidation and synthesis of novel antineoplastic substances denominated "combretastatins"
US7220784B2 (en) * 2000-12-21 2007-05-22 John Anthony Hadfield Substituted stilbenes and their reactions

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPS59189142A (en) 1983-04-12 1984-10-26 Ube Ind Ltd Electrically conductive thermoplastic resin composition
GB9106177D0 (en) * 1991-03-22 1991-05-08 Aston Molecules Ltd Substituted diphenylethylenes and analogues or derivatives thereof
TW325458B (en) 1993-09-08 1998-01-21 Ajinomoto Kk Stilbene derivatives and pharmaceutical compositions comprising the same for anti-cancer
CA2314238C (en) 1998-01-09 2008-09-02 Arizona Board Of Regents, A Body Corporate, Acting On Behalf Of Arizona State University Synthesis of combretastatin a-4 prodrugs and trans-isomers thereof
GB9903403D0 (en) 1999-02-16 1999-04-07 Angiogene Pharm Ltd Substituted stilbene compounds with vascular damaging activity
GB9918912D0 (en) * 1999-08-12 1999-10-13 Angiogene Pharm Ltd New stilbenes with vascular damaging activity

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4996237A (en) * 1987-01-06 1991-02-26 Arizona Board Of Regents Combretastatin A-4
US5569786A (en) * 1987-01-06 1996-10-29 Arizona Board Of Regents Acting On Behalf Of Arizona State University Isolation, structural elucidation and synthesis of novel antineoplastic substances denominated "combretastatins"
US5430062A (en) * 1992-05-21 1995-07-04 Research Corporation Technologies, Inc. Stilbene derivatives as anticancer agents
US5561122A (en) * 1994-12-22 1996-10-01 Arizona Board Of Regents Acting On Behalf Of Arizona State University Combretastatin A-4 prodrug
US7220784B2 (en) * 2000-12-21 2007-05-22 John Anthony Hadfield Substituted stilbenes and their reactions

Also Published As

Publication number Publication date
WO2002050007A2 (en) 2002-06-27
EP1351912A2 (en) 2003-10-15
WO2002050007A3 (en) 2002-10-17
US20130023663A1 (en) 2013-01-24
US20040152629A1 (en) 2004-08-05
AU2002216228A1 (en) 2002-07-01
US8853270B2 (en) 2014-10-07
CA2432640A1 (en) 2002-06-27
US7220784B2 (en) 2007-05-22

Similar Documents

Publication Publication Date Title
US8853270B2 (en) Substituted stilbenes and their reactions
RU2417216C2 (en) Fluoroalkoxy-combretastatin derivatives, use and synthesis methods thereof
JP4642226B2 (en) Prodrugs activated by hydroxylation
JP3908270B2 (en) Acylfulvene analogues and pharmaceutical compositions thereof
JP2004505888A (en) Tubulin binding ligands and corresponding prodrug structures
EP2297075A1 (en) Dihydro-iso-ca-4 and analogues: potent cytotoxics, inhibitors of tubulin polymerization
US20050014721A1 (en) Novel compounds and a novel process for their preparation
FR2896245A1 (en) NEW CHALCONE DERIVATIVES WITH ANTIMITOTIC ACTIVITY
EP2065358B1 (en) The prepartion and the use of ethoxy combretastatins and their prodrugs
US20050065213A1 (en) Combretastatin a-4 derivatives having antineoplastic activity
EP1351911B1 (en) Materials and methods for synthesizing stilbenes
EP3129368B1 (en) Cytotoxic compounds which are inhibitors of the polymerisation of tubulin
EP2142493B1 (en) Iso ca-4 and analogues thereof as potent cytotoxic agents inhibiting tubuline polymerisation
US8198302B2 (en) Compositions and methods with enhanced therapeutic activity
US7572778B2 (en) Fluorocombretastatin and derivatives thereof
US7183319B2 (en) Phenylethylamine derivatives and their use in the treatment of melanoma
Jelinek Discovery and development of dihydronaphthalene-based vascular disrupting agents and combretastatin-related analogs

Legal Events

Date Code Title Description
AS Assignment

Owner name: UNIVERSITEIT UTRECHT UU HOLDING B.V., NETHERLANDS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:VAN EDEN, WILLEM;VAN DER ZEE, RUURD;TLASKALOVA-HOGENOVA, HELENA;AND OTHERS;REEL/FRAME:022064/0570;SIGNING DATES FROM 20070821 TO 20070822

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION