WO2004046123A1 - Benzoxazole, benzthiazole and benzimidazole derivatives useful as heparanase inhibitors - Google Patents

Benzoxazole, benzthiazole and benzimidazole derivatives useful as heparanase inhibitors Download PDF

Info

Publication number
WO2004046123A1
WO2004046123A1 PCT/GB2003/004980 GB0304980W WO2004046123A1 WO 2004046123 A1 WO2004046123 A1 WO 2004046123A1 GB 0304980 W GB0304980 W GB 0304980W WO 2004046123 A1 WO2004046123 A1 WO 2004046123A1
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
compound
formula
hydrogen
halogen
Prior art date
Application number
PCT/GB2003/004980
Other languages
French (fr)
Inventor
Stephen Martin Courtney
Philip Andrew Hay
Original Assignee
Oxford Glycosciences (Uk) Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Oxford Glycosciences (Uk) Ltd filed Critical Oxford Glycosciences (Uk) Ltd
Priority to AU2003285498A priority Critical patent/AU2003285498A1/en
Publication of WO2004046123A1 publication Critical patent/WO2004046123A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D263/00Heterocyclic compounds containing 1,3-oxazole or hydrogenated 1,3-oxazole rings
    • C07D263/52Heterocyclic compounds containing 1,3-oxazole or hydrogenated 1,3-oxazole rings condensed with carbocyclic rings or ring systems
    • C07D263/54Benzoxazoles; Hydrogenated benzoxazoles
    • C07D263/56Benzoxazoles; Hydrogenated benzoxazoles with only hydrogen atoms, hydrocarbon or substituted hydrocarbon radicals, directly attached in position 2
    • C07D263/57Aryl or substituted aryl radicals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/04Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing three or more hetero rings

Definitions

  • the present invention relates to novel compounds useful as inhibitors of heparanase, methods for their synthesis, pharmaceutical compositions comprising the novel compounds and their use in medicine, in particular for the treatment of cancer.
  • the extracellular matrix is not only the structural surround for cells in a multicellular organism but also acts as a key modulator and mediator of their physiology, differentiation, organisation and repair.
  • Receptor ligands are stored, concentrated, processed and presented to the cell surface by components of the ECM, which include free and protein-bound heparan sulfate proteoglycans, free and protein-bound chondroitins, collagens, and a variety of cell-adhesive integrins, such as, fibronectin.
  • the ECM is in a constant flux of degradation and synthesis by neighbouring cells.
  • the ECM is also the principal barrier to tumour growth and metastasis. For a tumour cell to penetrate this barrier it must sufficiently degrade the ECM components so that there is ample space to traverse. The ECM must also be degraded in order to provide avenues for new blood vessel formation (angiogenesis) which are needed to supply the increased nutrient requirements of rapidly growing tumours.
  • angiogenesis new blood vessel formation
  • Carbohydrates represent a large fraction of the total mass of all ECM. Therefore, tumour cells secrete large quantities of carbohydrate degrading enzymes as they penetrate the ECM. In fact, there is good correlation between raised levels of carbohydrate processing enzymes, such as heparanases, secreted by tumour cells and their metastatic potential (e.g. Nlodavsky et al., (1994) Invasion Metastasis, 14:290- 302; (1999) Nature Medicine, 5:793-802).
  • Heparanases are enzymes that can degrade heparan sulfate as well as heparin and heparan sulfate proteoglycans.
  • the carbohydrate fragments generated by glycosidase action also promote the cancer phenotype since many are growth-stimulatory.
  • heparanase activity can release heparan sulfate fragments, which can increase the potency of a variety of growth factors, and can also elicit cell growth stimulation once bound by an appropriate cell surface receptor (e.g. Folkman and Shing (1992) Adv. Exp. Med. Biol., 313:355-64).
  • Inhibitors of ECM carbohydrate degradation are potent anticancer agents.
  • sulfated oligosaccharide heparanase inhibitors block tumour metastasis in some animal models (Nlodavsky et al., (1994) Invasion Metastasis, 14:290-302; Parish et al., (1999) Cancer Res., 59:3433-41).
  • heparanase activity results in the release of growth factors that can stimulate angiogenesis and promote tumour growth (Bashkin et al., (1989) Biochemistry, 28:1737-43).
  • Heparanase activity correlates with the ability of activated cells of the immune system to leave the circulation and elicit both inflammatory and autoimmune responses. Interaction of platelets, granulocytes, T and B lymphocytes, macrophage and mast cells with the subendothelial ECM is associated with degradation of heparan sulfate by heparanase activity (Nlodavsky et al., (1992) Invasion Metastasis, 12, 112-127). Heparanase inhibitors may be able to prevent or inhibit the progression of autoimmime and inflammatory diseases.
  • Heparinomimetic compounds are currently being developed as anticoagulant and antiproliferative agents for the control of thrombotic and proliferative disorders (Demir et al., Clin. Appl. Thromb. Hemost., 2001 Apr; 7(2): 131-40).
  • a secondary function of heparanase inhibitors may have a role in cardiovascular diseases including blood-clotting conditions, for example thromboembolic disease, arterial thrombosis and restenosis.
  • WO01/35967 discloses the use of heparanase inhibitors for the treatment or prevention of congestive heart failure e.g. primary cardiomyopathy. Associated conditions treated or prevented with the inhibitor are especially peripheral oedemas, pulmonary and hepatic congestion, dyspnoea, hydrothorax and ascites. Renal problems, e.g. nocturia can also be treated.
  • WO02/060374 discloses benzimidazole, benzoxazole and benzothiaziole derivatives as heparanase inhibitors.
  • the present invention provides a novel class of compounds, which can be used as inhibitors of heparanase. These compounds provide the opportunity for establishing new treatments for cancer, angiogenesis, inflammatory and autoimmune conditions and cardiovascular diseases.
  • the invention provides a compound of formula (I) or a pharmaceutically acceptable salt, ester or prodrug thereof:
  • R 1 , R 2 and R 3 are independently, hydrogen, halogen, CF 3 , OR 6 , NR 7 R 8 , NR 8 COR 10 , NR 8 SO 2 R 10 or C ⁇ _ 6 alkyl optionally substituted by hydroxy, C . 6 alkoxy or NR 7 R 8 ;
  • R 4 is NR 8 CONR 8 R 9 ;
  • X and Y are R 11 and the other is hydrogen or halogen; or X and Y together with the carbon atoms to which they are attached form a fused six-membered aromatic ring; Z is NR 8 , O or S;
  • R 6 is hydrogen or C ⁇ _ 6 alkyl, C 3 . 6 alkenyl or C 3 . 6 alkynyl any of which can optionally be substituted by hydroxy, C . 6 alkoxy or NR 7 R 8 ;
  • R 7 is hydrogen or C ⁇ _ 6 alkyl or C 3 . 6 alkenyl either of which is optionally substituted by d- ⁇ alkoxy or a 5- or 6-membered heterocyclic ring containing up to three heteroatoms selected from NR 8 , S and O;
  • R 8 is hydrogen or C ⁇ _ 6 alkyl; or the groups R 7 and R 8 may together with the nitrogen to which they are attached form a 5- or 6- membered ring which optionally contains up to two further heteroatom selected from NR 8 , S and O;
  • R 9 is Ci-io alkyl or C 3 - ⁇ 0 alkenyl wherein a -CH 2 - group other than that adjacent to the N may be replaced by -O- and wherein the alkyl or alkenyl is substituted by one or more carboxylic acid or tetrazole groups; or in R 4 the groups R 8 and R 9 may together with the nitrogen to which they are attached form a 5- or 6-membered ring, which is substituted with one or more carboxylic acid or tetrazole groups;
  • R 10 is C ⁇ . 6 alkyl
  • R 11 is hydrogen, halogen, C ⁇ _ 6 alkyl, OR 6 or phenyl optionally substituted by one or more substituents selected from halogen, . 6 alkyl, CF 3 , OCF 3 , OR 6 , CN and methylenedioxo; or a 5- to 10- membered heteroaryl group containing up to three heteroatoms selected from O, N and S, which heteroaryl group may optionally be substituted by one or more substituents selected from C ⁇ - 6 alkyl, - ⁇ alkoxy and halogen.
  • R 1 , R 2 and R 3 are independently, hydrogen, halogen, CF 3 , OR 6 , NR 7 R 8 , NR 8 COR 10 , NR 8 SO 2 R 10 or - ⁇ alkyl optionally substituted by hydroxy or C ⁇ - 6 alkoxy.
  • R 1 , R 2 and R 3 are independently, hydrogen, halogen, OR 6 , NR 7 R 8 , or C ⁇ _ 6 alkyl optionally substituted by hydroxy or C ⁇ - 6 alkoxy.
  • Z is O.
  • R 6 is hydrogen or C ⁇ . 6 alkyl or C 3 . 6 alkenyl either of which can optionally be substituted by hydroxy or C ⁇ . 6 alkoxy.
  • R s is preferably:
  • At least one of X and Y is other than hydrogen.
  • X is preferably R 11 .
  • R 11 is phenyl optionally substituted by one or more substituents selected from halogen, C ⁇ - 6 alkyl, CF 3 , OCF 3 , OR 6 , CN and methylenedioxo; or a 5- to 10-membered heteroaryl group containing up to three heteroatoms selected from O, N and S which heteroaryl group may optionally be substituted by one or more substituents selected from C . 6 alkyl, . 6 alkoxy and halogen.
  • R 1 is hydrogen, OR 6 or NR 7 R 8 .
  • R 2 is hydrogen
  • R 3 is hydrogen, halogen or OR 6 .
  • the ring may be, for example, morpholine, piperazine or N- methyl piperazine.
  • R ⁇ is a 5- to 10-membered heteroaryl group containing up to three heteroatoms selected from O, N and S, the group may be, for example, benzofuran or benzothiophene.
  • the configuration of the R groups is preferably,
  • alkyl and alkylene as used herein whether on its own or as part of a larger group e.g. "alkoxy” includes both straight and branched chain radicals.
  • alkyl also includes those radicals wherein one or more hydrogen atoms are replaced by fluorine.
  • alkenyl alkenylene
  • alkynyl alkynylene
  • heteroaryl as used herein means a 5- to 10-membered, substituted or unsubstituted, mono- or bicyclic aromatic ring containing up to three heteroatoms selected from oxygen, nitrogen and sulfur.
  • heterofuran e.g. 2-benzofuran
  • benzothiophene e.g. 2-benzothiophene
  • benzoxazole e.g. 2-benzoxazole
  • benzothiazole e.g. 2-benzothiazole
  • quinoline isoquinoline
  • pyridine pyrimidine
  • pyrazine oxadiazole
  • imidazole tetrazole
  • furan and thiophene e.g. 2-benzothiophene
  • Specific compounds of the invention that may be mentioned include those provided in the examples.
  • a preferred list of specific compounds of the invention include those compounds provided in Examples 2, 9, 14, 26, 27, 28, 29, 30, 31, 34, 35 and 36.
  • example compounds a) are esters of example compounds b).
  • the compounds of the invention preferably have a molecular weight of less than 800, more preferably less than 600.
  • Suitable pharmaceutically acceptable salts of the compounds include those derived from inorganic and organic bases.
  • suitable inorganic bases include the hydroxides, carbonates, and bicarbonates of ammonia, lithium, sodium, calcium, potassium, aluminium, iron, magnesium, zinc and the like. Salts can also be formed with suitable organic bases.
  • Such organic bases are well known in the art and may include amino acids such as arginine and lysine, mono-, di-, or trihydroxyalkylamines such as mono-, di-, and triethanolamine, choline, mono-, di-, and trialkylamines, such as methylamine, dimethylamine, and trimethylamine, guanidine; N-methylglucosamine; N-methylpiperazine; morpholine; ethylenediamine; N-benzylphenethylamine; tris(hydroxymethyl) aminomethane; meglumine; and the like.
  • amino acids such as arginine and lysine, mono-, di-, or trihydroxyalkylamines such as mono-, di-, and triethanolamine, choline, mono-, di-, and trialkylamines, such as methylamine, dimethylamine, and trimethylamine, guanidine; N-methylglucosamine; N-methylpiperazine; morpholine; ethylenediamine; N
  • Salts may be prepared in a conventional manner using methods well known in the art, for example by treatment of a solution of the compound of formula (I) with a solution of the base, for example, potassium or sodium hydroxide, or potassium or sodium hydrogen carbonate.
  • a solution of the base for example, potassium or sodium hydroxide, or potassium or sodium hydrogen carbonate.
  • the invention also includes prodrugs of the aforementioned compounds.
  • a prodrug is commonly described as an inactive or protected derivative of an active ingredient or a drug, which is converted to the active ingredient or drug in the body.
  • Examples of prodrugs include pharmaceutically acceptable esters, including C ⁇ -C 6 alkyl esters and pharmaceutically acceptable amides, including secondary - alkylamides.
  • Some of the compounds of this invention may be crystallised or recrystallised from solvents such as aqueous and organic solvents. In such cases solvates may be formed.
  • This invention includes within its scope stoichiometric solvates including hydrates as well as compounds containing variable amounts of water that may be produced by processes such as lyophilisation.
  • Certain of the compounds of formula (I) may exist in the form of optical isomers, e.g. diastereoisomers and mixtures of isomers in all ratios, e.g. racemic mixtures.
  • the invention includes all such forms, in particular the pure isomeric forms.
  • the different isomeric forms may be separated or resolved one from the other by conventional methods, or any given isomer may be obtained by conventional synthetic methods or by stereospecific or asymmetric syntheses.
  • the alkene can be presented as a cis or trans isomer or a mixture thereof.
  • an isomeric form of a compound of the invention When an isomeric form of a compound of the invention is provided substantially free of other isomers, it will preferably contain less than 5% w/w, more preferably less than 2% w/w and especially less than 1% w/w of the other isomers.
  • the compounds of formula (I) are intended for use in pharmaceutical compositions it will readily be understood that they are each preferably provided in substantially pure form, for example at least 60% pure, more suitably at least 75% pure and preferably at least 85%, especially at least 98% pure (% are on a weight for weight basis). Impure preparations of the compounds may be used for preparing the more pure forms used in the pharmaceutical compositions; these less pure preparations of the compounds should contain at least 1%, more suitably at least 5% and preferably at least 10% of a compound of the formula (I) or pharmaceutically acceptable derivative thereof.
  • the compounds of formula (I) can be prepared by art-recognized procedures from known or commercially available starting materials. If the starting materials are unavailable from a commercial source, their synthesis is described herein, or they can be prepared by procedures known in the art.
  • the invention also provides a process for preparing a compound of formula (I), from a compound of formula (II): dD wherein R x is NO 2 , NHR 8 , or NCO and R 1 , R 2 , R 3 , R 5 . and R 8 are as defined for formula (I), by the processes described below.
  • a compound of formula (II) where R x is NH 2 may be prepared from a corresponding compound where R x is NO 2 by methods well known to those skilled in the art, for example, hydrogenation with palladium on a charcoal catalyst or treatment with Zn and acetic acid.
  • a compound of formula (II) wherein R x is NH 2 may be converted to a compound of formula (H) wherein R x is NCO by reaction with, for example, triphosgene.
  • a compound of formula (II) wherein R x is NH 2 may be converted to another compound of formula (II) wherein R x is NHR 8 and R 8 is -e alkyl, by an alkylation or reductive amination reaction using methods well known to those skilled in the art.
  • the invention provides a process for preparing a compound of formula (I), comprising: treating a compound of formula (II) wherein R x is NHR 8 , with a compound of formula (-TJ):
  • a suitable solvent for example THF.
  • An ester compound of formula (I) can be converted to a free acid compound of formula (I) by hydrolysis, a method well known to those skilled in the art.
  • R A is as defined for formula (IH), e.g. by stirring at room temperature or with heating in a suitable solvent.
  • Compounds of formula (111) and (TV) may be available through the usual commercial sources. They and derivatives thereof miay also be prepared by methods well known to those skilled in the art.
  • this may be achieved via oxidative cyclisation of a Schiff base, derived from the condensation of the 2-aminophenol or 2-aminothiophenol and aldehydes, using various oxidants such as PhI(OAc) 2 , Pb(OAc) 4 or DDQ..
  • R B in compound (V) is CHO, heating in acetonitrile followed by oxidation using for example O 2 /FeCl 3 (cat.) in acetonitrile.
  • the compounds of formula (II), wherein X or Y is halogen, can be modified to give a corresponding set of compounds of formula (H) wherein X or Y is phenyl or a 5- to 10-membered heteroaryl group optionally substituted by one or more substituents as defined in formula (I).
  • the modification may be achieved by a coupling reaction with compounds of formula (X):
  • R c is phenyl or a 5- to 10-membered heteroaryl group optionally substituted by one or more substituents as defined for formula (I), using an appropriate catalyst for example tetrakis (triphenylphosphine) palladium.
  • compounds of formula (II) where R x is NO 2 , R 3 is halogen at a position ortho or para to the R x group and R 1 , R 2 and R 5 are as defined for formula (I) may be converted to a corresponding subset of compounds of formula (II) where R 3 is OR 6 or NR 7 R 8 , by reaction with an alcohol or amine via a nucleophilic aromatic substitution.
  • R 3 is NR 8 COR 10 or NR 8 SO 2 R 10
  • R 3 is NHR 8 by reaction with the appropriate carboxylic acid/chloride or sulfonyl chloride, i.e. R 10 CO 2 H/(R 10 CO 2 C1) or R 10 SO 2 C1 wherein R 10 is as defined for formula (I).
  • labile functional groups in the intermediate compounds e.g. hydroxy, carboxy and amino groups
  • the protecting groups may be removed at any stage in the synthesis of the compounds of formula (I) or may be present on the final compound of formula (I).
  • a comprehensive discussion of the ways in which various labile functional groups groups may be protected and methods for cleaving the resulting protected derivatives is given in for example Protective Groups in Organic Chemistry, T.W. Greene and P.G.M. Wuts, (Wiley- Interscience, New York, 2nd edition, 1991).
  • the compounds of formula (I) may be prepared singly or as compound libraries comprising at least 2, for example 5 to 1,000 compounds, and more preferably 10 to 100 compounds of formula (I).
  • Libraries of compounds of formula (I) may be prepared by multiple parallel synthesis using either solution phase or solid phase chemistry, by procedures known to those skilled in the art.
  • a compound library comprising at least 2 compounds of formula (I) or pharmaceutically acceptable salts, esters and prodrugs thereof.
  • any novel intermediate compounds as described herein also fall within the scope of the present invention, e.g. the compounds of fomula (H).
  • the invention provides a compound of formula
  • R x is NO 2 , NHR 8 , or NCO
  • R 5 is wherein one of X and Y is R 11 and the other is hydrogen or halogen; or X and Y together with the carbon atoms to which they are attached form a fused six-membered aromatic ring;
  • R 11 is phenyl optionally, and preferably, substituted by one or more substituents selected from halogen, C ⁇ - 6 alkyl, CF 3 , OCF 3 , OR 6 , CN and methylenedioxo; or a 5- to 10-membered heteroaryl group containing up to three heteroatoms selected from O, N and S, which heteroaryl group may optionally be substituted by one or more substituents selected from C ⁇ _ 6 alkyl, . 6 alkoxy and halogen; and
  • R 1 , R 2 , R 3 and R 8 are as defined for formula (I).
  • Preferred intermediate compounds of the invention include Intermediate compounds 1 to 31 described in the Examples, in particular Intermediate compounds 2, 9, 14b), 14c), 26a), 26b), 27a), 27b), 28a)-31a) and 28b)-31b).
  • the invention also provides a compound of formula (I) when prepared by any of the above mentioned methods.
  • the pharmaceutically effective compounds of formula (I) and pharmaceutically acceptable salts, esters and prodrugs thereof may be administered in conventional dosage forms prepared by combining a compound of formula (I) ("active ingredient") with standard pharmaceutical carriers or excipients according to conventional procedures well known in the art. These procedures may involve mixing, granulating and compressing or dissolving the ingredients as appropriate to the desired preparation.
  • a pharmaceutical composition comprising a compound of formula (I), or a pharmaceutically acceptable salt, ester or prodr ⁇ g thereof, together with one or more pharmaceutically acceptable carriers or excipients.
  • compositions of the invention may be formulated for administration by any route, and include those in a form adapted for oral, topical or parenteral administration to mammals including humans.
  • compositions may be adapted for administration by any appropriate route, for example by the oral (including buccal or sublingual), rectal, nasal, topical (including buccal, sublingual or transdermal), vaginal or parenteral (including subcutaneous, intramuscular, intravenous or mtradermal) route.
  • Such formulations may be prepared by any method known in the art of pharmacy, for example by bringing into association the active ingredient with the carrier(s) or excipient(s).
  • compositions adapted for oral administration may be presented as discrete units such as capsules or tablets; powders or granules; solutions or suspensions in aqueous or non-aqueous liquids; edible foams or whips; or oil-in-water liquid emulsions or water-in-oil liquid emulsions.
  • compositions adapted for transdermal administration may be presented as discrete patches intended to remain in intimate contact with the epidermis of the recipient for a prolonged period of time.
  • the active ingredient may be delivered from the patch by iontophoresis as generally described in Pharmaceutical Research, 3(6), 318, (1986).
  • compositions adapted for topical administration may be formulated as ointments, creams, suspensions, lotions, powders, solutions, pastes, gels, impregnated dressings, sprays, aerosols or oils and may contain appropriate conventional additives such as preservatives, solvents to assist drug penetration and emollients in ointments and creams.
  • the formulations are preferably applied as a topical ointment or cream.
  • the active ingredient may be employed with either a paraffinic or a water-miscible ointment base.
  • the active ingredient may be formulated in a cream with an oil-in-water cream base or a water-in-oil base.
  • compositions adapted for topical administration to the eye include eye drops wherein the active ingredient is dissolved or suspended in a suitable carrier, especially an aqueous solvent.
  • compositions adapted for topical administration in the mouth include lozenges, pastilles and mouth washes.
  • compositions adapted for rectal administration may be presented as suppositories or enemas.
  • compositions adapted for nasal administration wherein the carrier is a solid include a coarse powder having a particle size for example in the range 20 to 500 microns.
  • Suitable formulations wherein the carrier is a liquid, for administration as a nasal spray or as nasal drops, include aqueous or oil solutions of the active ingredient.
  • Fine particle dusts or mists which may be generated by means of various types of metered dose pressurised aerosols, nebulizers or insufflators.
  • compositions adapted for vaginal administration may be presented as pessaries, tampons, creams, gels, pastes, foams or spray formulations.
  • compositions adapted for parenteral administration include aqueous and non-aqueous sterile injection solutions which may contain anti-oxidants, buffers, bacteriostats and solutes which render the formulation isotonic with the blood of the intended recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents.
  • the formulations may be presented in unit-dose or multi-dose containers, for example sealed ampoules and vials, and may be stored in a freeze- dried (lyophilized) condition requiring only the addition of the sterile liquid carrier, for example water for injections, immediately prior to use.
  • Extemporaneous injection solutions and suspensions may be prepared from sterile powders, granules and tablets.
  • formulations may also include other agents conventional in the art having regard to the type of formulation in question, for example those suitable for oral administration may include flavouring agents.
  • the pharmaceutical formulations according to the invention are preferably adapted for oral administration.
  • the formulations may also contain compatible conventional carriers, such as cream or ointment bases and ethanol or oleyl alcohol for lotions.
  • suitable conventional carriers such as cream or ointment bases and ethanol or oleyl alcohol for lotions.
  • Such carriers may be present as from about 1% up to about 98% of the formulation. More usually they will form up to about 80% of the formulation.
  • Tablets and capsules for oral administration may be in unit dose presentation form, and may contain conventional excipients such as binding agents, for example syrup, acacia, gelatin, sorbitol, tragacanth, or polyvinylpyrrolidone; fillers, for example lactose, sugar, maize-starch, calcium phosphate, sorbitol or glycine; tabletting lubricants, for example magnesium stearate, talc, polyethylene glycol or silica; disintegrants, for example potato starch; or acceptable wetting agents such as sodium lauryl s ⁇ lphate.
  • the tablets may be coated according to methods well known in normal pharmaceutical practice.
  • Oral liquid preparations may be in the form of, for example, aqueous or oily suspensions, solutions, emulsions, syrups or elixirs, or may be presented as a dry product for reconstitution with water or other suitable vehicle before use.
  • Such liquid preparations may contain conventional additives, such as suspending agents, for example sorbitol, methyl cellulose, glucose syrup, gelatin, hydroxyethyl cellulose, carboxymethyl cellulose, aluminium stearate gel or hydrogenated edible fats, emulsifying agents, for example lecithin, sorbitan monooleate, or acacia; non-aqueous vehicles (which may include edible oils), for example almond oil, oily esters such as glycerine, propylene glycol, or ethyl alcohol; preservatives, for example methyl or propyl p-hydroxybenzoate or sorbic acid, and, if desired, conventional flavouring or colouring agents.
  • suspending agents for example sorbitol, methyl cellulose, glucose syrup, gelatin, hydroxyethyl cellulose, carboxymethyl cellulose, aluminium stearate gel or hydrogenated edible fats, emulsifying agents, for example lecithin, sorbitan monooleate, or
  • Suppositories will contain conventional suppository bases, e.g. cocoa-butter or other glyceride.
  • fluid unit dosage forms are prepared utilizing the active ingredient and a sterile vehicle, water being preferred.
  • the active ingredient depending on the vehicle and concentration used, can be either suspended or dissolved in the vehicle.
  • the active ingredient can be dissolved in water for injection and filter sterilised before filling into a suitable vial or ampoule and sealing.
  • agents such as a local anaesthetic, preservative and buffering agents can be dissolved in the vehicle.
  • the composition can be frozen after filling into the vial and the water removed under vacuum.
  • the dry lyophilized powder is then sealed in the vial and an accompanying vial of water for injection may be supplied to reconstitute the liquid prior to use.
  • Parenteral suspensions are prepared in substantially the same manner except that the active ingredient is suspended in the vehicle instead of being dissolved and sterilization cannot be accomplished by filtration.
  • the active ingredient can be sterilised by exposure to ethylene oxide before suspending in the sterile vehicle.
  • a surfactant or wetting agent is included in the composition to facilitate uniform distribution of the compound.
  • compositions may contain from 0.1% by weight, preferably from 10-60% by weight, of the active ingredient, depending on the method of administration.
  • compositions may be presented in unit dose forms containing a predetermined amount of active ingredient per dose.
  • a unit may contain for example lOOmg/kg to lmg/kg depending on the condition being treated, the route of administration and the age, weight and condition of the patient.
  • Preferred unit dosage formulations are those containing a daily dose or sub-dose, as herein above recited, or an appropriate fraction thereof, of the active ingredient.
  • the optimal quantity and spacing of individual dosages of a formula (I) compound will be determined by the nature and extent of the condition being treated, the form, route and site of administration, and the particular mammal being treated, and that such optimums can be determined by conventional techniques. It will also be appreciated by one of skill in the art that the optimal course of treatment, i.e., the number of doses of the compound of formula (I) given per day for a defined number of days, can be ascertained by those skilled in the art using conventional course of treatment determination tests.
  • the compounds of the present invention are useful in that they are capable of inhibiting heparanase.
  • the compounds can be used in the treatment of cancer.
  • the compounds of the present invention can also be used in combination with one or more additional treatments or therapeutic compounds for cancer.
  • treatments include, surgery and radiation therapy.
  • therapeutic compounds include but are not limited to cisplatin, cyclophosphamide, methotrexate, 5-fluorouracil, paclitaxel, docetaxel, vincristine, vinblastine, vinorelbine, doxorubicin, tamoxifen, toremifene, megestrol acetate, anastrozole, goserelin, anti-HER2 monoclonal antibody, capecitabine and raloxifene hydrochloride.
  • the compounds of the present invention can also be used in the treatment of angiogenesis and angiogenesis dependent diseases, which include angiogenesis associated with the growth of solid tumours and retinopathy.
  • the compounds of the present invention can also be used in combination with one or more additional treatments or therapeutic compounds for angiogenesis.
  • additional treatments or therapeutic compounds for angiogenesis include, but are not limited to, recombinant platelet-derived growth factor-BB (RegranexTM).
  • the compounds of the present invention can also be used in the treatment of inflammatory conditions, including, but not limited to, rheumatoid arthritis, inflammatory bowel disease, and wound healing.
  • the compounds of the present invention can also be used in the treatment of autoimmune diseases, such as, but not limited to, multiple sclerosis.
  • the compounds of the present invention can also be used in the treatment of cardiovascular diseases, such as, but not limited to, blood clotting conditions, for example thromboembolic disease, arterial thrombosis and restenosis.
  • cardiovascular diseases such as, but not limited to, blood clotting conditions, for example thromboembolic disease, arterial thrombosis and restenosis.
  • treating is meant either prophylactic or therapeutic therapy.
  • cancer or 'carcinoma' is a malignant new growth that arises from epithelium, found in skin or, more commonly, the lining of body organs. Carcinomas tend to infiltrate into adjacent tissues and spread (metastasise) to distant organs, for example to bone, liver, lung or the brain.
  • cancer includes both metastatic tumour cells and tissue and examples include, but are not limited to, melanoma, mesothelioma, lymphoma, leukaemia, fibrosarcoma, rhabdomyosarcoma, mastocytoma and the following tissue carcinomas: colorectal, colon, prostate, lung, breast, pancreatic, intestinal, renal, gastric, bladder, ovarian, uterine, cervical, hepatic and stomach.
  • tissue carcinomas colorectal, colon, prostate, lung, breast, pancreatic, intestinal, renal, gastric, bladder, ovarian, uterine, cervical, hepatic and stomach.
  • the present invention provides:
  • (ix) a method for the treatment of inflammatory diseases, such as but not limited to rheumatoid arthritis, inflammatory bowel disease, and wound healing which comprises the step of administering to a patient an effective amount of a compound of formula (I).
  • inflammatory diseases such as but not limited to rheumatoid arthritis, inflammatory bowel disease, and wound healing
  • autoimmune diseases such as but not limited to multiple sclerosis
  • cardiovascular diseases such as but not limited to blood clotting conditions, for example thromboembolic disease, arterial thrombosis and restenosis which comprises the step of administering to a patient an effective amount of a compound of formula (I).
  • Example 32 4-[[2-Methoxy-5-[(5-phenyl)-benzoxazol-2-yl]phenylamino]carbonylamino]butanoic acid a) 4-[[2-Methoxy-5-[(5-phenyl)-benzoxazol-2-yl]phenylamino]carbonylamino]butanoic acid ethyl ester
  • Example 33 4-[[4-N-n-propyIamino-5-[(5-phenyl)-benzoxazol-2-yl]phenylamino]carbonylamino] butanoic acid a) 4-[[4-N-n-propylamino-5-[(5-phenyl)-benzoxazol-2-yl]phenylaniino]carbonylamino] butanoic acid ethyl ester
  • Triphosgene (89mg, 0.3mmol) was added to a solution of 2-(3-amino-4-methoxyphenyl)-5- (benzofuran-2-yl)benzoxazole, Intermediate 27b), (250mg, 0.7mmol) in DCM (2ml). After 30 min the aminoester (0.7mmol) and DIPEA (122 ⁇ L, 0.7mmol) were added and the reaction vessel sealed and heated (Microwave) to 110°C for 10 min. The reaction mixture was diluted with DCM (20ml) and washed with 2M HCl (20ml) and then water (20ml). The organic layer was dried over sodium sulfate and concentrated to give the product.
  • the following compounds were prepared by adding a solution of 2-fluoro-5-nitrobenzoyl chloride (lg, 5.0mmol) in THF (5ml) to a solution of the corresponding 2-aminophenol (5.0mmol). After stirring overnight the reactions were filtered and the solid dried under vacuum. The crude product (2.5mmol) was added to a solution of p-toluenesulfonic acid (1.04g, 5.5mmol) in toluene (10ml) and the mixture heated to reflux for 6 h. The cooled reaction mixture was neutralized with saturated NaHCO 3 , the slurry filtered and the solid dried under vacuum.
  • Heparanase assay The assay is based upon the use of the specific binding of basic fibroblast growth factor (bFGF) to heparan sulfate. Heparan sulfate can be detected via binding of bFGF using a horse radish peroxidase-conjugated bFGF antibody. Following cleavage of high molecular weight heparan sulfate by heparanase, the smaller material generated will no longer adhere to the surface of a 96 well plate and hence heparanase activity can be followed as a reduction in bFGF binding.
  • bFGF basic fibroblast growth factor
  • Nunc Maxisorp 96-well plates are coated for 16h at RT with 100 ⁇ ]/well 0.04mg/ml heparan sulfate in PBS. The wells are then aspirated and blocked for lh with 200 ⁇ l ⁇ vell 1% BSA-PBS. Following five washes with 0.01% BSA, 0.05% Tween20 PBS (wash buffer), 100 ⁇ l of recombinant human basic FGF (90ng/ml in 0.1% BSA/PBS) is added per well and the plate is incubated at room temperature for lh.
  • test compound in 10% DMSO
  • human heparanase in lOOmM Sodium acetate, 5mM CaCl 2 , pH 5.5 are added to each well and the plate incubated for 2h at 37°C.
  • the wells are washed again with wash buffer and lOO ⁇ l of bFGF antibody-horse radish peroxidase conjugate added.
  • the plate is incubated at room temperature for lh and washed again five times with wash buffer.
  • lOO ⁇ l of TMB peroxidase substrate is added and the colour allowed to develop for 10 min.
  • the reaction is stopped with 50 ⁇ l 1M H 2 SO 4 and the colour read at 450nm on a plate reader.

Abstract

Compounds of formula (I): wherein R1, R2 and R3 are independently, hydrogen, halogen, CF3, OR6, NR7 R8, NR8COR10, NR8SO2R10 or C 1-6 alkyl optionally substituted by hydroxy, C 1-6 alkoxy or NR7R8; R4 is NR8CONR8R9; and R5 formula (Ia) is methods for their synthesis, pharmaceutical compositions comprising them and their use in medicine, in particular for the treatment of cancer.

Description

BENZOXAZOLE , BENZTHIAZOLE AND BENZIMIDAZOLE DERIVATIVES USEFUL AS HEPARANASE . INHIBITORS
The present invention relates to novel compounds useful as inhibitors of heparanase, methods for their synthesis, pharmaceutical compositions comprising the novel compounds and their use in medicine, in particular for the treatment of cancer.
The extracellular matrix (ECM) is not only the structural surround for cells in a multicellular organism but also acts as a key modulator and mediator of their physiology, differentiation, organisation and repair. Receptor ligands are stored, concentrated, processed and presented to the cell surface by components of the ECM, which include free and protein-bound heparan sulfate proteoglycans, free and protein-bound chondroitins, collagens, and a variety of cell-adhesive integrins, such as, fibronectin. As such, the ECM is in a constant flux of degradation and synthesis by neighbouring cells.
The ECM is also the principal barrier to tumour growth and metastasis. For a tumour cell to penetrate this barrier it must sufficiently degrade the ECM components so that there is ample space to traverse. The ECM must also be degraded in order to provide avenues for new blood vessel formation (angiogenesis) which are needed to supply the increased nutrient requirements of rapidly growing tumours.
A broad spectrum of degradative enzymes are secreted by tumor cells to break down the ECM's complex composition. However, recent studies have demonstrated that inhibiting even just one ECM degrading enzyme appears to provide significant benefit in treating cancer. For example, inhibitors of certain proteases that degrade ECM protein component have been studied in preclinical and clinical trials as anticancer agents.
Carbohydrates represent a large fraction of the total mass of all ECM. Therefore, tumour cells secrete large quantities of carbohydrate degrading enzymes as they penetrate the ECM. In fact, there is good correlation between raised levels of carbohydrate processing enzymes, such as heparanases, secreted by tumour cells and their metastatic potential (e.g. Nlodavsky et al., (1994) Invasion Metastasis, 14:290- 302; (1999) Nature Medicine, 5:793-802). Heparanases, are enzymes that can degrade heparan sulfate as well as heparin and heparan sulfate proteoglycans.
The carbohydrate fragments generated by glycosidase action also promote the cancer phenotype since many are growth-stimulatory. For example, heparanase activity can release heparan sulfate fragments, which can increase the potency of a variety of growth factors, and can also elicit cell growth stimulation once bound by an appropriate cell surface receptor (e.g. Folkman and Shing (1992) Adv. Exp. Med. Biol., 313:355-64).
Inhibitors of ECM carbohydrate degradation are potent anticancer agents. For example, sulfated oligosaccharide heparanase inhibitors block tumour metastasis in some animal models (Nlodavsky et al., (1994) Invasion Metastasis, 14:290-302; Parish et al., (1999) Cancer Res., 59:3433-41). Furthermore, heparanase activity results in the release of growth factors that can stimulate angiogenesis and promote tumour growth (Bashkin et al., (1989) Biochemistry, 28:1737-43).
Heparanase activity correlates with the ability of activated cells of the immune system to leave the circulation and elicit both inflammatory and autoimmune responses. Interaction of platelets, granulocytes, T and B lymphocytes, macrophage and mast cells with the subendothelial ECM is associated with degradation of heparan sulfate by heparanase activity (Nlodavsky et al., (1992) Invasion Metastasis, 12, 112-127). Heparanase inhibitors may be able to prevent or inhibit the progression of autoimmime and inflammatory diseases.
Heparinomimetic compounds are currently being developed as anticoagulant and antiproliferative agents for the control of thrombotic and proliferative disorders (Demir et al., Clin. Appl. Thromb. Hemost., 2001 Apr; 7(2): 131-40). Thus, a secondary function of heparanase inhibitors may have a role in cardiovascular diseases including blood-clotting conditions, for example thromboembolic disease, arterial thrombosis and restenosis.
WO01/35967 discloses the use of heparanase inhibitors for the treatment or prevention of congestive heart failure e.g. primary cardiomyopathy. Associated conditions treated or prevented with the inhibitor are especially peripheral oedemas, pulmonary and hepatic congestion, dyspnoea, hydrothorax and ascites. Renal problems, e.g. nocturia can also be treated.
WO02/060374 discloses benzimidazole, benzoxazole and benzothiaziole derivatives as heparanase inhibitors.
The present invention provides a novel class of compounds, which can be used as inhibitors of heparanase. These compounds provide the opportunity for establishing new treatments for cancer, angiogenesis, inflammatory and autoimmune conditions and cardiovascular diseases.
The invention provides a compound of formula (I) or a pharmaceutically acceptable salt, ester or prodrug thereof:
Figure imgf000003_0001
(I) wherein
R1, R2 and R3 are independently, hydrogen, halogen, CF3, OR6, NR7R8, NR8COR10, NR8SO2R10 or Cι_6 alkyl optionally substituted by hydroxy, C .6 alkoxy or NR7R8; R4 is NR8CONR8R9;
R5 is
Figure imgf000003_0002
wherein one of X and Y is R11 and the other is hydrogen or halogen; or X and Y together with the carbon atoms to which they are attached form a fused six-membered aromatic ring; Z is NR8, O or S;
R6 is hydrogen or Cι_6 alkyl, C3.6 alkenyl or C3.6 alkynyl any of which can optionally be substituted by hydroxy, C .6 alkoxy or NR7R8;
R7 is hydrogen or Cι_6 alkyl or C3.6 alkenyl either of which is optionally substituted by d-β alkoxy or a 5- or 6-membered heterocyclic ring containing up to three heteroatoms selected from NR8, S and O;
R8 is hydrogen or Cι_6 alkyl; or the groups R7 and R8 may together with the nitrogen to which they are attached form a 5- or 6- membered ring which optionally contains up to two further heteroatom selected from NR8, S and O;
R9 is Ci-io alkyl or C30 alkenyl wherein a -CH2- group other than that adjacent to the N may be replaced by -O- and wherein the alkyl or alkenyl is substituted by one or more carboxylic acid or tetrazole groups; or in R4 the groups R8 and R9 may together with the nitrogen to which they are attached form a 5- or 6-membered ring, which is substituted with one or more carboxylic acid or tetrazole groups;
R10 is Cι.6 alkyl; and
R11 is hydrogen, halogen, Cι_6 alkyl, OR6 or phenyl optionally substituted by one or more substituents selected from halogen, .6 alkyl, CF3, OCF3, OR6, CN and methylenedioxo; or a 5- to 10- membered heteroaryl group containing up to three heteroatoms selected from O, N and S, which heteroaryl group may optionally be substituted by one or more substituents selected from Cι-6 alkyl, -β alkoxy and halogen.
Preferably R1, R2 and R3 are independently, hydrogen, halogen, CF3, OR6, NR7R8, NR8COR10, NR8SO2R10 or -β alkyl optionally substituted by hydroxy or Cι-6 alkoxy.
More preferably R1, R2 and R3 are independently, hydrogen, halogen, OR6, NR7R8, or Cι_6 alkyl optionally substituted by hydroxy or Cι-6 alkoxy.
Preferably Z is O.
Preferably R6 is hydrogen or Cχ.6 alkyl or C3.6 alkenyl either of which can optionally be substituted by hydroxy or Cι.6 alkoxy.
Rs is preferably:
Figure imgf000004_0001
Preferably at least one of X and Y is other than hydrogen.
X is preferably R11.
Preferably R11 is phenyl optionally substituted by one or more substituents selected from halogen, Cι-6 alkyl, CF3, OCF3, OR6, CN and methylenedioxo; or a 5- to 10-membered heteroaryl group containing up to three heteroatoms selected from O, N and S which heteroaryl group may optionally be substituted by one or more substituents selected from C .6 alkyl, .6 alkoxy and halogen.
More preferably R1 is hydrogen, OR6 or NR7R8.
More preferably R2 is hydrogen.
More preferably R3 is hydrogen, halogen or OR6.
In the group NR7R8, when the R7 and R8 substituents, together with the nitrogen to which they are attached form a 5- or 6-membered ring, the ring may be, for example, morpholine, piperazine or N- methyl piperazine.
When Rπ is a 5- to 10-membered heteroaryl group containing up to three heteroatoms selected from O, N and S, the group may be, for example, benzofuran or benzothiophene.
The configuration of the R groups is preferably,
Figure imgf000004_0002
The term "alkyl" and "alkylene" as used herein whether on its own or as part of a larger group e.g. "alkoxy" includes both straight and branched chain radicals. The term alkyl also includes those radicals wherein one or more hydrogen atoms are replaced by fluorine. The terms "alkenyl", "alkenylene", "alkynyl" and "alkynylene" should be interpreted accordingly.
The term "heteroaryl" as used herein means a 5- to 10-membered, substituted or unsubstituted, mono- or bicyclic aromatic ring containing up to three heteroatoms selected from oxygen, nitrogen and sulfur. Examples include benzofuran e.g. 2-benzofuran, benzothiophene e.g. 2-benzothiophene, benzoxazole e.g. 2-benzoxazole, benzothiazole e.g. 2-benzothiazole, quinoline, isoquinoline, pyridine, pyrimidine, pyrazine, oxadiazole, imidazole, tetrazole, furan and thiophene.
Specific compounds of the invention that may be mentioned include those provided in the examples. A preferred list of specific compounds of the invention include those compounds provided in Examples 2, 9, 14, 26, 27, 28, 29, 30, 31, 34, 35 and 36. For clarity, example compounds a) are esters of example compounds b).
The compounds of the invention preferably have a molecular weight of less than 800, more preferably less than 600.
Suitable pharmaceutically acceptable salts of the compounds include those derived from inorganic and organic bases. Examples of suitable inorganic bases include the hydroxides, carbonates, and bicarbonates of ammonia, lithium, sodium, calcium, potassium, aluminium, iron, magnesium, zinc and the like. Salts can also be formed with suitable organic bases. Such organic bases are well known in the art and may include amino acids such as arginine and lysine, mono-, di-, or trihydroxyalkylamines such as mono-, di-, and triethanolamine, choline, mono-, di-, and trialkylamines, such as methylamine, dimethylamine, and trimethylamine, guanidine; N-methylglucosamine; N-methylpiperazine; morpholine; ethylenediamine; N-benzylphenethylamine; tris(hydroxymethyl) aminomethane; meglumine; and the like.
Salts may be prepared in a conventional manner using methods well known in the art, for example by treatment of a solution of the compound of formula (I) with a solution of the base, for example, potassium or sodium hydroxide, or potassium or sodium hydrogen carbonate.
The invention also includes prodrugs of the aforementioned compounds. A prodrug is commonly described as an inactive or protected derivative of an active ingredient or a drug, which is converted to the active ingredient or drug in the body. Examples of prodrugs include pharmaceutically acceptable esters, including Cι-C6 alkyl esters and pharmaceutically acceptable amides, including secondary - alkylamides.
Some of the compounds of this invention may be crystallised or recrystallised from solvents such as aqueous and organic solvents. In such cases solvates may be formed. This invention includes within its scope stoichiometric solvates including hydrates as well as compounds containing variable amounts of water that may be produced by processes such as lyophilisation.
Certain of the compounds of formula (I) may exist in the form of optical isomers, e.g. diastereoisomers and mixtures of isomers in all ratios, e.g. racemic mixtures. The invention includes all such forms, in particular the pure isomeric forms. The different isomeric forms may be separated or resolved one from the other by conventional methods, or any given isomer may be obtained by conventional synthetic methods or by stereospecific or asymmetric syntheses. Where a compound contains an alkene moiety, the alkene can be presented as a cis or trans isomer or a mixture thereof. When an isomeric form of a compound of the invention is provided substantially free of other isomers, it will preferably contain less than 5% w/w, more preferably less than 2% w/w and especially less than 1% w/w of the other isomers.
Since the compounds of formula (I) are intended for use in pharmaceutical compositions it will readily be understood that they are each preferably provided in substantially pure form, for example at least 60% pure, more suitably at least 75% pure and preferably at least 85%, especially at least 98% pure (% are on a weight for weight basis). Impure preparations of the compounds may be used for preparing the more pure forms used in the pharmaceutical compositions; these less pure preparations of the compounds should contain at least 1%, more suitably at least 5% and preferably at least 10% of a compound of the formula (I) or pharmaceutically acceptable derivative thereof.
The compounds of formula (I) can be prepared by art-recognized procedures from known or commercially available starting materials. If the starting materials are unavailable from a commercial source, their synthesis is described herein, or they can be prepared by procedures known in the art.
The invention also provides a process for preparing a compound of formula (I), from a compound of formula (II):
Figure imgf000006_0001
dD wherein Rx is NO2, NHR8, or NCO and R1, R2, R3, R5 .and R8 are as defined for formula (I), by the processes described below.
A compound of formula (II) where Rx is NH2 may be prepared from a corresponding compound where Rx is NO2 by methods well known to those skilled in the art, for example, hydrogenation with palladium on a charcoal catalyst or treatment with Zn and acetic acid. A compound of formula (II) wherein Rx is NH2 may be converted to a compound of formula (H) wherein Rx is NCO by reaction with, for example, triphosgene. A compound of formula (II) wherein Rx is NH2 may be converted to another compound of formula (II) wherein Rx is NHR8 and R8 is -e alkyl, by an alkylation or reductive amination reaction using methods well known to those skilled in the art.
The invention provides a process for preparing a compound of formula (I), comprising: treating a compound of formula (II) wherein Rxis NHR8, with a compound of formula (-TJ):
R^— N==1=0 (HI) wherein RA is CWo alkyl or C30 alkenyl wherein a -CH2- group other than that adjacent to the N may be replaced by -O- and wherein the alkyl or alkenyl is substituted with one or more carboxylate ester or CN groups, e.g. by stirring at room temperature or with heating in a suitable solvent, for example THF. When RAis substituted by CN a further reaction with for example, sodium azide and ammonium chloride in a suitable solvent, e.g. DMF, converts the CN group to a tetrazole group.
An ester compound of formula (I) can be converted to a free acid compound of formula (I) by hydrolysis, a method well known to those skilled in the art.
Alternatively, a compound of formula (II) wherein Rxis NCO, is treated with a compound of formula (IV):
R— NH2
(IN) wherein RA is as defined for formula (IH), e.g. by stirring at room temperature or with heating in a suitable solvent. Compounds of formula (111) and (TV) may be available through the usual commercial sources. They and derivatives thereof miay also be prepared by methods well known to those skilled in the art.
The compounds of formula (II) wherein Rx is ΝO2, R1, R2, R3 and R5 are as defined for formula (I) and Z is O or S, may be prepared by treatment of a compound of formula (V):
Figure imgf000006_0002
(V) wherein RR11,, RR22 aanndd RR33 aarree aass ddeefifinneedd ffoorr ffcormula (I) and RB is CO2H or CHO, with a compound of formula (VI):
Figure imgf000007_0001
(VI) wherein X and Y are as defined for formula (I) and Z is S or O, by e.g. either (i) heating in a condensation/cyclisation reaction, using for example polyphosphoric acid, or (ii) by firstly coupling a compound of formula (V) to a compound of formula (VI) via either an ester/ thioester or amide formation reaction using methods well known to those of skill in the art followed by direct heating or heating with an acidic media with a suitable solvent to effect cyclisation, for example, p-toluenesulfonic acid in toluene. Alternatively, this may be achieved via oxidative cyclisation of a Schiff base, derived from the condensation of the 2-aminophenol or 2-aminothiophenol and aldehydes, using various oxidants such as PhI(OAc)2, Pb(OAc)4 or DDQ..
Compounds of formulae (V) and (VI) may be available through the usual commercial sources. They and derivatives thereof may also be prepared by methods well known to those skilled in the art.
The compounds of formula (II) wherein Rx is NO2, R1, R2, R3 and R5 are as defined for formula (I) and Z is NR8, may be prepared by treatment of a compound of formula (V) with a compound of formula (NH):
Figure imgf000007_0002
(NIT) wherein X, Y and R8 are as defined for formula (I), in e.g. the following reactions:
(i) if RB in compound (V) is CO2H, heating in a condensation/cyclisation reaction using for example polyphosphoric acid; or
(ii) if RB in compound (V) is CHO, heating in acetonitrile followed by oxidation using for example O2/FeCl3 (cat.) in acetonitrile.
Compounds of formula (NIT) may be prepared by reduction of compounds, of formula (NDI):
Figure imgf000007_0003
(vm) wherein X, Y and R8 are as defined for formula (I) by methods well known to those skilled in the art, for example, hydrogenation with palladium on a charcoal catalyst or treatment with Zn and acetic acid.
Compounds of formula (NDI) may be prepared by nitration of compounds of formula (IX):
Figure imgf000007_0004
(IX) wherein X, Y and R8 are as defined for formula (I), by methods well known to those skilled in the art, for example using fuming nitric acid and sulfuric acid or fuming nitric acid and tin (IN) chloride. Compounds of formula (IX) may be available through the usual commercial sources. They and derivatives thereof may also be prepared by methods well known to those skilled in the art.
The compounds of formula (II), wherein X or Y is halogen, can be modified to give a corresponding set of compounds of formula (H) wherein X or Y is phenyl or a 5- to 10-membered heteroaryl group optionally substituted by one or more substituents as defined in formula (I). The modification may be achieved by a coupling reaction with compounds of formula (X):
Rc B(OH)2
(X) where Rc is phenyl or a 5- to 10-membered heteroaryl group optionally substituted by one or more substituents as defined for formula (I), using an appropriate catalyst for example tetrakis (triphenylphosphine) palladium.
Additionally, compounds of formula (II) where Rx is NO2, R3 is halogen at a position ortho or para to the Rx group and R1, R2 and R5 are as defined for formula (I) may be converted to a corresponding subset of compounds of formula (II) where R3 is OR6 or NR7R8, by reaction with an alcohol or amine via a nucleophilic aromatic substitution.
Compounds of formula (IT) where R3 is NR8COR10 or NR8SO2R10 may be prepared from corresponding compounds of formula (II) where R3 is NHR8 by reaction with the appropriate carboxylic acid/chloride or sulfonyl chloride, i.e. R10CO2H/(R10CO2C1) or R10SO2C1 wherein R10 is as defined for formula (I).
During the synthesis of the compounds of formula (I), labile functional groups in the intermediate compounds, e.g. hydroxy, carboxy and amino groups, may be protected. The protecting groups may be removed at any stage in the synthesis of the compounds of formula (I) or may be present on the final compound of formula (I). A comprehensive discussion of the ways in which various labile functional groups groups may be protected and methods for cleaving the resulting protected derivatives is given in for example Protective Groups in Organic Chemistry, T.W. Greene and P.G.M. Wuts, (Wiley- Interscience, New York, 2nd edition, 1991).
Further details for the preparation of compounds of formula (I) are found in the examples.
The compounds of formula (I) may be prepared singly or as compound libraries comprising at least 2, for example 5 to 1,000 compounds, and more preferably 10 to 100 compounds of formula (I). Libraries of compounds of formula (I) may be prepared by multiple parallel synthesis using either solution phase or solid phase chemistry, by procedures known to those skilled in the art.
Thus according to a further aspect of the invention there is provided a compound library comprising at least 2 compounds of formula (I) or pharmaceutically acceptable salts, esters and prodrugs thereof.
Any novel intermediate compounds as described herein also fall within the scope of the present invention, e.g. the compounds of fomula (H). In particular, the invention provides a compound of formula
Figure imgf000008_0001
(Ha) wherein Rx is NO2, NHR8, or NCO; R5 is
Figure imgf000009_0001
wherein one of X and Y is R11 and the other is hydrogen or halogen; or X and Y together with the carbon atoms to which they are attached form a fused six-membered aromatic ring;
R11 is phenyl optionally, and preferably, substituted by one or more substituents selected from halogen, Cι-6 alkyl, CF3, OCF3, OR6, CN and methylenedioxo; or a 5- to 10-membered heteroaryl group containing up to three heteroatoms selected from O, N and S, which heteroaryl group may optionally be substituted by one or more substituents selected from Cι_6 alkyl, .6 alkoxy and halogen; and
Z, R1, R2, R3 and R8 are as defined for formula (I).
Preferred intermediate compounds of the invention include Intermediate compounds 1 to 31 described in the Examples, in particular Intermediate compounds 2, 9, 14b), 14c), 26a), 26b), 27a), 27b), 28a)-31a) and 28b)-31b).
The invention also provides a compound of formula (I) when prepared by any of the above mentioned methods.
The pharmaceutically effective compounds of formula (I) and pharmaceutically acceptable salts, esters and prodrugs thereof, may be administered in conventional dosage forms prepared by combining a compound of formula (I) ("active ingredient") with standard pharmaceutical carriers or excipients according to conventional procedures well known in the art. These procedures may involve mixing, granulating and compressing or dissolving the ingredients as appropriate to the desired preparation.
According to a further aspect of the invention there is provided a pharmaceutical composition comprising a compound of formula (I), or a pharmaceutically acceptable salt, ester or prodrύg thereof, together with one or more pharmaceutically acceptable carriers or excipients.
The pharmaceutical compositions of the invention may be formulated for administration by any route, and include those in a form adapted for oral, topical or parenteral administration to mammals including humans.
Pharmaceutical formulations may be adapted for administration by any appropriate route, for example by the oral (including buccal or sublingual), rectal, nasal, topical (including buccal, sublingual or transdermal), vaginal or parenteral (including subcutaneous, intramuscular, intravenous or mtradermal) route. Such formulations may be prepared by any method known in the art of pharmacy, for example by bringing into association the active ingredient with the carrier(s) or excipient(s).
Pharmaceutical formulations adapted for oral administration may be presented as discrete units such as capsules or tablets; powders or granules; solutions or suspensions in aqueous or non-aqueous liquids; edible foams or whips; or oil-in-water liquid emulsions or water-in-oil liquid emulsions.
Pharmaceutical formulations adapted for transdermal administration may be presented as discrete patches intended to remain in intimate contact with the epidermis of the recipient for a prolonged period of time. For example, the active ingredient may be delivered from the patch by iontophoresis as generally described in Pharmaceutical Research, 3(6), 318, (1986).
Pharmaceutical formulations adapted for topical administration may be formulated as ointments, creams, suspensions, lotions, powders, solutions, pastes, gels, impregnated dressings, sprays, aerosols or oils and may contain appropriate conventional additives such as preservatives, solvents to assist drug penetration and emollients in ointments and creams.
For applications to the eye or other external tissues, for example the mouth and skin, the formulations are preferably applied as a topical ointment or cream. When formulated in an ointment, the active ingredient may be employed with either a paraffinic or a water-miscible ointment base. Alternatively, the active ingredient may be formulated in a cream with an oil-in-water cream base or a water-in-oil base.
Pharmaceutical formulations adapted for topical administration to the eye include eye drops wherein the active ingredient is dissolved or suspended in a suitable carrier, especially an aqueous solvent.
Pharmaceutical formulations adapted for topical administration in the mouth include lozenges, pastilles and mouth washes.
Pharmaceutical formulations adapted for rectal administration may be presented as suppositories or enemas.
Pharmaceutical formulations adapted for nasal administration wherein the carrier is a solid include a coarse powder having a particle size for example in the range 20 to 500 microns. Suitable formulations wherein the carrier is a liquid, for administration as a nasal spray or as nasal drops, include aqueous or oil solutions of the active ingredient.
Pharmaceutical formulations adapted for administration by inhalation include fine particle dusts or mists which may be generated by means of various types of metered dose pressurised aerosols, nebulizers or insufflators.
Pharmaceutical formulations adapted for vaginal administration may be presented as pessaries, tampons, creams, gels, pastes, foams or spray formulations.
Pharmaceutical formulations adapted for parenteral administration include aqueous and non-aqueous sterile injection solutions which may contain anti-oxidants, buffers, bacteriostats and solutes which render the formulation isotonic with the blood of the intended recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents. The formulations may be presented in unit-dose or multi-dose containers, for example sealed ampoules and vials, and may be stored in a freeze- dried (lyophilized) condition requiring only the addition of the sterile liquid carrier, for example water for injections, immediately prior to use. Extemporaneous injection solutions and suspensions may be prepared from sterile powders, granules and tablets.
It should be understood that in addition to the ingredients particularly mentioned above, the formulations may also include other agents conventional in the art having regard to the type of formulation in question, for example those suitable for oral administration may include flavouring agents.
The pharmaceutical formulations according to the invention are preferably adapted for oral administration.
The formulations may also contain compatible conventional carriers, such as cream or ointment bases and ethanol or oleyl alcohol for lotions. Such carriers may be present as from about 1% up to about 98% of the formulation. More usually they will form up to about 80% of the formulation.
Tablets and capsules for oral administration may be in unit dose presentation form, and may contain conventional excipients such as binding agents, for example syrup, acacia, gelatin, sorbitol, tragacanth, or polyvinylpyrrolidone; fillers, for example lactose, sugar, maize-starch, calcium phosphate, sorbitol or glycine; tabletting lubricants, for example magnesium stearate, talc, polyethylene glycol or silica; disintegrants, for example potato starch; or acceptable wetting agents such as sodium lauryl sμlphate. The tablets may be coated according to methods well known in normal pharmaceutical practice.
Oral liquid preparations may be in the form of, for example, aqueous or oily suspensions, solutions, emulsions, syrups or elixirs, or may be presented as a dry product for reconstitution with water or other suitable vehicle before use. Such liquid preparations may contain conventional additives, such as suspending agents, for example sorbitol, methyl cellulose, glucose syrup, gelatin, hydroxyethyl cellulose, carboxymethyl cellulose, aluminium stearate gel or hydrogenated edible fats, emulsifying agents, for example lecithin, sorbitan monooleate, or acacia; non-aqueous vehicles (which may include edible oils), for example almond oil, oily esters such as glycerine, propylene glycol, or ethyl alcohol; preservatives, for example methyl or propyl p-hydroxybenzoate or sorbic acid, and, if desired, conventional flavouring or colouring agents.
Suppositories will contain conventional suppository bases, e.g. cocoa-butter or other glyceride.
For parenteral administration, fluid unit dosage forms are prepared utilizing the active ingredient and a sterile vehicle, water being preferred. The active ingredient, depending on the vehicle and concentration used, can be either suspended or dissolved in the vehicle. In preparing solutions the active ingredient can be dissolved in water for injection and filter sterilised before filling into a suitable vial or ampoule and sealing.
Advantageously, agents such as a local anaesthetic, preservative and buffering agents can be dissolved in the vehicle. To enhance the stability, the composition can be frozen after filling into the vial and the water removed under vacuum. The dry lyophilized powder is then sealed in the vial and an accompanying vial of water for injection may be supplied to reconstitute the liquid prior to use. Parenteral suspensions are prepared in substantially the same manner except that the active ingredient is suspended in the vehicle instead of being dissolved and sterilization cannot be accomplished by filtration. The active ingredient can be sterilised by exposure to ethylene oxide before suspending in the sterile vehicle. Advantageously, a surfactant or wetting agent is included in the composition to facilitate uniform distribution of the compound.
The compositions may contain from 0.1% by weight, preferably from 10-60% by weight, of the active ingredient, depending on the method of administration.
Pharmaceutical formulations may be presented in unit dose forms containing a predetermined amount of active ingredient per dose. Such a unit may contain for example lOOmg/kg to lmg/kg depending on the condition being treated, the route of administration and the age, weight and condition of the patient. Preferred unit dosage formulations are those containing a daily dose or sub-dose, as herein above recited, or an appropriate fraction thereof, of the active ingredient.
It will be recognized by one of skill in the art that the optimal quantity and spacing of individual dosages of a formula (I) compound will be determined by the nature and extent of the condition being treated, the form, route and site of administration, and the particular mammal being treated, and that such optimums can be determined by conventional techniques. It will also be appreciated by one of skill in the art that the optimal course of treatment, i.e., the number of doses of the compound of formula (I) given per day for a defined number of days, can be ascertained by those skilled in the art using conventional course of treatment determination tests.
The compounds of the present invention are useful in that they are capable of inhibiting heparanase. Thus, the compounds can be used in the treatment of cancer.
The compounds of the present invention can also be used in combination with one or more additional treatments or therapeutic compounds for cancer. Examples of such treatments include, surgery and radiation therapy. Examples of therapeutic compounds include but are not limited to cisplatin, cyclophosphamide, methotrexate, 5-fluorouracil, paclitaxel, docetaxel, vincristine, vinblastine, vinorelbine, doxorubicin, tamoxifen, toremifene, megestrol acetate, anastrozole, goserelin, anti-HER2 monoclonal antibody, capecitabine and raloxifene hydrochloride.
The compounds of the present invention can also be used in the treatment of angiogenesis and angiogenesis dependent diseases, which include angiogenesis associated with the growth of solid tumours and retinopathy. The compounds of the present invention can also be used in combination with one or more additional treatments or therapeutic compounds for angiogenesis. Examples of such other therapeutic compounds include, but are not limited to, recombinant platelet-derived growth factor-BB (Regranex™).
The compounds of the present invention can also be used in the treatment of inflammatory conditions, including, but not limited to, rheumatoid arthritis, inflammatory bowel disease, and wound healing.
The compounds of the present invention can also be used in the treatment of autoimmune diseases, such as, but not limited to, multiple sclerosis.
The compounds of the present invention can also be used in the treatment of cardiovascular diseases, such as, but not limited to, blood clotting conditions, for example thromboembolic disease, arterial thrombosis and restenosis.
By the term "treating" is meant either prophylactic or therapeutic therapy.
The term cancer or 'carcinoma' is a malignant new growth that arises from epithelium, found in skin or, more commonly, the lining of body organs. Carcinomas tend to infiltrate into adjacent tissues and spread (metastasise) to distant organs, for example to bone, liver, lung or the brain. Herein, cancer includes both metastatic tumour cells and tissue and examples include, but are not limited to, melanoma, mesothelioma, lymphoma, leukaemia, fibrosarcoma, rhabdomyosarcoma, mastocytoma and the following tissue carcinomas: colorectal, colon, prostate, lung, breast, pancreatic, intestinal, renal, gastric, bladder, ovarian, uterine, cervical, hepatic and stomach.
In additional aspects, therefore, the present invention provides:
(i) the use of a compound of formula (I) as an inhibitor of the enzyme heparanase.
(ii) the use of a compound of formula (I) in the manufacture of a medicament for the treatment of cancer.
(iii) the use of a compound of formula (I) in the manufacture of a medicament for the treatment of angiogenesis and angiogenesis dependent diseases which include angiogenesis associated with the growth of solid tumours and retinopathy.
(iv) the use of a compound of formula (I) in the manufacture of a medicament for the treatment of inflammatory conditions such as but not limited to rheumatoid arthritis, inflammatory bowel disease, and wound healing.
(v) the use of a compound of formula (I) in the manufacture of a medicament for the treatment of autoimmune diseases such as but not limited to multiple sclerosis.
(vi) the use of a compound of formula (I) in the manufacture of a medicament for the treatment of cardiovascular diseases such as but not limited to blood clotting conditions, for example thromboembolic disease, arterial thrombosis and restenosis.
(vii) a method for the treatment of cancer, which comprises the step of administering to a patient an effective amount of a compound of formula (I).
(viii) a method for the treatment of angiogenesis and angiogenesis dependent diseases, which include angiogenesis associated with the growth of solid tumours and retinopathy, which comprises the step of administering to a patient an effective amount of a compound of formula (I).
(ix) a method for the treatment of inflammatory diseases, such as but not limited to rheumatoid arthritis, inflammatory bowel disease, and wound healing which comprises the step of administering to a patient an effective amount of a compound of formula (I).
(x) a method for the treatment of autoimmune diseases, such as but not limited to multiple sclerosis, which comprises the step of administering to a patient an effective amount of a compound of formula (I). (xi) a method for the treatment of cardiovascular diseases, such as but not limited to blood clotting conditions, for example thromboembolic disease, arterial thrombosis and restenosis which comprises the step of administering to a patient an effective amount of a compound of formula (I).
All publications, including, but not limited to, patents and patent applications cited in this specification, are herein incorporated by reference as if each individual publication were specifically and individually indicated to be incorporated by reference herein as though fully set forth.
The invention will now be described by reference to the following examples, which are merely illustrative and are not to be construed as a limitation of the scope of the present invention.
Examples Examples la)-31a)
The following compounds were prepared by adding ethyl 6-isocyanatohexanoate (36μL, 0.2mmol) to a solution of the corresponding aminobenzoxazole, Intermediates 1-9, 10c)-14c), 15b)-26b), 27e) and 28b)-31b) (0.2mmol) in THF (2ml). After stirring overnight at 40°C the reaction was concentrated under reduced pressure to give the product.
Figure imgf000013_0001
Figure imgf000013_0002
Figure imgf000014_0001
Figure imgf000015_0001
Figure imgf000016_0001
Figure imgf000017_0002
Examples lb)-31b)
The following compounds were prepared by addition of a solution of LiOH (24mg l.Ommol) in water (0.5ml) to a solution of the corresponding ethyl ester, Examples la)-31a), (0.2mmol) in THF/MeOH (1:1, 1.5ml). After stirring for 6 h the reaction was acidified with 2M HCl and the resulting precipitate filtered and dried under vacuum.
Figure imgf000017_0001
Figure imgf000017_0003
Figure imgf000018_0001
Figure imgf000019_0001
Figure imgf000020_0001
Figure imgf000021_0001
Figure imgf000022_0004
Example 32: 4-[[2-Methoxy-5-[(5-phenyl)-benzoxazol-2-yl]phenylamino]carbonylamino]butanoic acid a) 4-[[2-Methoxy-5-[(5-phenyl)-benzoxazol-2-yl]phenylamino]carbonylamino]butanoic acid ethyl ester
Figure imgf000022_0001
Ethyl-4-isocyanatobutyrate (46μL, 0.16mmol) was added to a solution of 2-(3-amino-4- methoxyphenyl)-5-phenylbenzoxazole, Intermediate 9, (50mg, O.lόmmol) in THF at 40°C. After stirring overnight the reaction was concentrated to give the subtitle compound. MS (APCI-)«z/z: 472.1. b) 4-[[2-Methoxy-5-[(5-phenyl)-benzoxazol-2-yl]phenylamino]carbonylamino]butanoic acid
Figure imgf000022_0002
Prepared by the addition of a solution of LiOH (19mg, 0.8mmol) in water to a solution of 4-[[2- methoxy-5-[(5-phenyl)-benzoxazol-2-yl]phenylamino]carbonylamino]butanoic acid ethyl ester, Example 32a), (73mg, 0.16mmol) in THF/MeOH (1:1, 1.5ml). After stirring for 6 h the reaction was acidified with 2M HCl and the resulting title product precipitate filtered and dried under vacuum (44mg, 62%). H NMR (DMSO) δ 1.68 (2H, m), 2.29 (2H, t), 3.14 (2H, q), 3.96 (3H, s), 7.02 (IH, bt), 7.20 (IH, d), 7.38 (IH, t), 7.50 (2H, m), 7.65-7.85 (5H, m), 8.01 (IH, d), 8.13 (IH, s), 9.08 (IH, d). MS (APCl-)m/z: 444.0. Example 33: 4-[[4-N-n-propyIamino-5-[(5-phenyl)-benzoxazol-2-yl]phenylamino]carbonylamino] butanoic acid a) 4-[[4-N-n-propylamino-5-[(5-phenyl)-benzoxazol-2-yl]phenylaniino]carbonylamino] butanoic acid ethyl ester
Figure imgf000022_0003
Ethyl-4-isocyanatobutyrate (15μl, O.lmmol) was added to a solution of 2-(3-amino-6-N-«- propylaminophenyl)-5-phenylbenzoxazole, Intermediate 14c), (35mg, O.lmmol) in THF (2ml). After stirring overnight at 40°C the reaction was concentrated under reduced pressure to give the subtitle compound (49mg, 99%). !H NMR (DMSO) δ 1.04 (3H, t), 1.19 (3H, t), 1.65-1.75 (4H, m), 2.33 (2H, t), 3.11 (2H, q), 3.27 (2H, q), 4.07 (2H, q), 6.11 (IH, bt), 6.83 (IH, d), 7.32 (IH, dd), 7.40 (IH, d), 7.49 (2H, m), 7.68-7.78 (4H, m), 7.86 (IH, d), 7.99-8.03 (2H, m), 8.25-8.28 (2H, m). b) 4-[[4-N-ra-propylamino-5-[(5-phenyl)-benzoxazol-2-yl]phenylamino]carbonylamino] butanoic acid
Figure imgf000023_0001
Prepared by the method of Examples lb)-31b) from 4-[[4-N-n-propylamino-5-[(5-phenyl)- benzoxazol-2-yl]phenylamino]carbonylamino] butanoic acid ethyl ester, Example 33a), (47mg, O.lmmol) the title compound was obtained. W NMR (DMSO) δ 1.04 (3H, t), 1.70 (4H, m), 2.25 (2H, t), 3.10 (2H, q), 3.27 (2H, q), 6.11 (IH, bt), 6.82 (IH, d), 7.29-7.41 (2H, m), 7.49 (2H, t), 7.68-7.77 (3H, m), 7.86 (IH, d), 7.99-8.03 (2H, m), 8.25-8.28 (2H, m). MS (APCI- n/z: 471.2. Examples 34a)-36a)
Triphosgene (89mg, 0.3mmol) was added to a solution of 2-(3-amino-4-methoxyphenyl)-5- (benzofuran-2-yl)benzoxazole, Intermediate 27b), (250mg, 0.7mmol) in DCM (2ml). After 30 min the aminoester (0.7mmol) and DIPEA (122μL, 0.7mmol) were added and the reaction vessel sealed and heated (Microwave) to 110°C for 10 min. The reaction mixture was diluted with DCM (20ml) and washed with 2M HCl (20ml) and then water (20ml). The organic layer was dried over sodium sulfate and concentrated to give the product.
Figure imgf000023_0002
Figure imgf000023_0003
Figure imgf000024_0002
Examples 34b)-36b)
The following compounds were prepared by addition of a solution of LiOH (24mg l.Ommol) in water (0.5ml) to a solution of the corresponding ethyl ester, Examples 34a)-36a), (0.2mmol) in THF MeOH (1:1, 1.5ml). After stirring for 6 h the reaction was acidified with 2M HCl and the resulting precipitate filtered and dried under vacuum.
Figure imgf000024_0001
Figure imgf000024_0003
Preparation of Intermediates Intermediates 1-9
The following intermediates were prepared by mixing the corresponding aminobenzoic acid (3.0mmol) and corresponding aminophenol (3.0mmol) in polyphosphoric acid (5ml) and heating the mixture to 200°C for 4 h. The reaction mixture was slowly poured into ice water (100ml) and the resulting mixture basified with solid sodium hydroxide. At pH5-6 the precipitate was filtered, washed with water and dried under vacuum.
Figure imgf000025_0001
Intermediates 10a)-14a)
The following compounds were prepared by adding a solution of 2-fluoro-5-nitrobenzoyl chloride (lg, 5.0mmol) in THF (5ml) to a solution of the corresponding 2-aminophenol (5.0mmol). After stirring overnight the reactions were filtered and the solid dried under vacuum. The crude product (2.5mmol) was added to a solution of p-toluenesulfonic acid (1.04g, 5.5mmol) in toluene (10ml) and the mixture heated to reflux for 6 h. The cooled reaction mixture was neutralized with saturated NaHCO3, the slurry filtered and the solid dried under vacuum.
Figure imgf000026_0002
Intermediates 10b)-14b)
The following intermediates were prepared by adding propylamine (492μL, ό.Ommol) to a solution of the corresponding fluorobenzoxazole, Intermediates 10a)-14a), (2.0mmol) in THF (10ml). After stirring overnight the reaction was concentrated and the solid triturated with methanol. The resulting product was filtered and dried under vacuum.
Figure imgf000026_0001
Figure imgf000027_0004
Intermediate 37 : 2-(2-Fluoro-5-nitrophenyl)-5-bromobenzoxazole
Figure imgf000027_0001
A solution of 2-fluoro-5-nitrobenzoyl chloride (4.42g, 21.7mmol) in THF (75ml) was slowly added to a solution of 4-bromo-2-aminophenol (4.08g, 21.7mmol) in THF (75ml). After stirring overnight the reaction was filtered and the solid dried under vacuum. The crude product was added to a solution of p-toluenesulfonic acid (8.66g, (45.6mmol) in toluene (200ml) and the mixture heated to reflux with a Dean-Stark trap fitted for 6 h. The cooled reaction mixture was neutralized with saturated NaHCO3, the slurry filtered and the solid dried under vacuum. Η NMR (DMSO) δ 7.72 (IH, dd), 7.87 (IH, t), 7.93 (IH, d), 8.23 (IH, d), 8.57-8.63 (IH, m), 8.97-9.00 (IH, m). Intermediate 38 : 2-(2-Methoxy-5-nitrophenyl)-5-bromobenzoxazole
Figure imgf000027_0002
Sodium methoxide (80mg, 1.5mmol) in MeOH was added to a suspension of 2-(2-fluoro-5- nitrophenyl)-5 -bromobenzoxazole (Intermediate 37) (500mg, 1.5mmol) in MeOH (2ml). The mixture was heated to 85°C for 4 h. The reaction was quenched with ice water and extracted with EtOAc. The organic layer was washed with brine, dried over sodium sulfate and concentrated to give the title compound, 360mg (78%). *H NMR (DMSO) δ 4.09 (3H, s), 7.52 (IH, d), 7.63 (IH, dd), 7.83 (IH, d),
8.12 (IH, d), 8.48 (IH, dd), 8.84 (IH, d).
Intermediate 39: 2-(2-N-Propylamine-5-nitrophenyl)-5-bromobenzoxazoIe
Figure imgf000027_0003
Propylamine (1.95ml, 23.7mmol) was added to a solution of 2-(2-fluoro-5-nitrophenyl)-5- bromobenzoxazole (Intermediate 37) (2.0g, 5.9mmol) in THF (50ml). After stirring for 4 h the reaction was concentrated and the residue triturated with methanol. The precipitate was filtered and dried under vacuum to give the title compound. !H NMR (DMSO) δ 1.04 (3H, t), 1.74 (2H, m), 3.44 (2H, m), 7.05 (IH, d), 7.63 (IH, dd), 7.84 (IH, d), 8.03 (IH, d), 8.21 (IH, dd), 8.77 (IH, d), 9.08 (IH, t). Intermediate 40: 2-(3-Nitro-4-methoxyphenyl)-5-bromobenzoxazole
Figure imgf000028_0001
a) 4-Methoxy-3-nitro-benzoyl chloride
Oxalyl chloride (15.8ml, 180mmol) was added dropwise with stirring to a solution of 4-methoxy- 3-nitrobenzoic acid (7.00g, 36.00mmol) in THF containing lOμL DMF. After 1 h the solvent was removed under reduced pressure. The product was used directly in the next step. b) 2-(4-Methoxy-3-nitrophenyl)-carbonylamino-4-bromophenol
A solution of 4-methoxy-3-nitro-benzoyl chloride (7.12g, 33.0mmol) in THF (50 ml) was added dropwise with stirring to a solution of 4-bromo-2-aminophenol (6.20g, 33.0mmol) in THF (50ml) containing triethylamine (6.82ml, 66.0mmol). After addition was complete the reaction was stirred at room temperature overnight. The reaction mixture was concentrated to approximately half the original volume and the precipitate collected by filtration. The solid was washed with methanol and ether and dried under vacuum to give the subtitle compound as a brown solid (5.24g, 42%). H NMR (DMSO) δ 4.05 (3H, s), 6.68 (IH, dd), 6.68 (IH, dd), 6.96 (2H, m), 7.55 (IH, d), 8.36 (IH, dd), 8.58 (IH, d). c) 2-(3-Nitro-4-methoxyphenyl)-5-bromobenzoxazole
A suspension of 2-(4-methoxy-3-nitrophenyl)-carbonylamino-4-bromophenol (5.24g, 14.2mmol) and toluenesulfonic acid (5.36g, 31.2mmol) in toluene (100ml) was heated to reflux overnight. The cooled reaction mixture was washed with saturated sodium hydrogen carbonate solution (care foaming) and the organic layer separated. The aqueous layer was extracted with ethyl acetate (2x50ml) and the combined organic layers dried over sodium sulfate and the solvent removed under reduced pressure. The residue was triturated with ether, filtered and dried under vacuum to give the title compound as a pale pink solid (4.5 lg, 93%). XH NMR (DMSO) δ 4.05 (3H, s), 7.61 (2H, m), 7.79 (IH, d), 8.05 (IH, d), 8.42 (IH, dd), 8.61 (IH, d). MS m/z 349.0 (M+H)+. Intermediates 15a)-26a) and 28a)-31a)
The following intermediates were prepared by adding tetrakis(triphenylphosphine)palladium(0) (0.06 mmol) to a degassed solution of the corresponding bromobenzoxazole, Intermediates 38, 39 and 40c), (1.1 mmol) in 1,4-dioxane (18 ml) at room temperature under argon. The solution was further degassed and 2M aqueous sodium carbonate (1.0 ml, 2.3 mmol) and the boronic acid (1.7 mmol) were added. The resulting suspension was heated at reflux for 16 h. On cooling to room temperature the mixture was diluted with DCM (10 ml), washed with water (10 ml), brine (10 ml) and dried (Na2SO ). The solvent was removed under reduced pressure and the residue triturated with methanol. The solid was collected by filtration and dried under vacuum to give the product.
Figure imgf000028_0002
Figure imgf000029_0001
Figure imgf000030_0001
Intermediates 10c)-14c), 15b)-26b) and 28b)-31b)
The following intermediates were prepared by adding zinc dust (523mg, 8.0mmol) to a stirred solution of the corresponding nitrobenzoxazole, Intermediates 10b)-26b) and 28b)-31b), (0.8mmol) in acetic acid/ethanol (1:1, 10ml). After stirring overnight the reactions were concentrated, the residue dissolved in EtOAc (50ml) and washed with saturated NaHCO3. The organic layer was separated, dried over sodium sulfate and concentrated under reduced pressure to give the product.
Figure imgf000031_0001
Figure imgf000032_0001
Figure imgf000033_0001
Intermediate 27: 2-(3-Amino-4-methoxyphenyI)-5-(benzofuran-2-yl)benzoxazole
Figure imgf000034_0001
a) 2-(3-Nitro-4-methoxyphenyl)-5-(benzofuran-2-yl)benzoxazole
2-(3-Nitro-4-methoxyphenyl)-5-bromobenzoxazole, Intermediate 40c), (200mg, 0.57mmol) was suspended in degassed ethylene glycol dimethyl ether (DME, 10ml). Tetrakis
(triphenylphosphine)palladium (0) (33mg, 0.03mmol), 2M sodium carbonate (0.5ml) and benzofuran-2- boronic acid (137mg (0.85mmol) were added and the reaction was further degassed. The reaction was heated to reflux for 16 h. The cooled reaction mixture was diluted with water (10ml) and extracted with dichloromethane (2xl0ml). The combined organic extracts were dried over sodium sulfate, filtered and the solvent removed under reduced pressure. The residue was triturated with methanol (5ml) and filtered. The solid was dried under vacuum to give the subtitle compound (50mg, 15%). *H NMR (DMSO) δ 4.04 (3H, s), 7.25-7.36 (2H, m), 7.54 (IH, s), 7.59-7.69 (3H, m), 7.92 (IH, d), 8.01 (IH, dd), 8.29 (IH, d), 8.44 (IH, dd), 8.63 (IH, d). b) 2-(3-Amino-4-methoxyphenyl)-5-(benzofuran-2-yl)benzoxazole
A suspension of 2-(3-nitro-4-methoxyphenyl)-5-(benzofuran-2-yl)benzoxazole (50mg, 0.13mmol) in dioxane (10ml) was placed under an atmosphere of argon. Palladium on carbon (10%) (lOmg) was added and the reaction purged with hydrogen and stirred at room temperature for 36 h. The reaction was filtered through a bed of celite and the filtrate concentrated to give the title compound (40mg, 86%). MS m/z 357.1 (M+H)+.
Biological Data
Heparanase assay: The assay is based upon the use of the specific binding of basic fibroblast growth factor (bFGF) to heparan sulfate. Heparan sulfate can be detected via binding of bFGF using a horse radish peroxidase-conjugated bFGF antibody. Following cleavage of high molecular weight heparan sulfate by heparanase, the smaller material generated will no longer adhere to the surface of a 96 well plate and hence heparanase activity can be followed as a reduction in bFGF binding.
Nunc Maxisorp 96-well plates are coated for 16h at RT with 100μ]/well 0.04mg/ml heparan sulfate in PBS. The wells are then aspirated and blocked for lh with 200μlΛvell 1% BSA-PBS. Following five washes with 0.01% BSA, 0.05% Tween20 PBS (wash buffer), 100μl of recombinant human basic FGF (90ng/ml in 0.1% BSA/PBS) is added per well and the plate is incubated at room temperature for lh.
After a further five washes with the wash buffer, 10μl of test compound (in 10% DMSO) and 90μl of human heparanase in lOOmM Sodium acetate, 5mM CaCl2, pH 5.5 are added to each well and the plate incubated for 2h at 37°C. The wells are washed again with wash buffer and lOOμl of bFGF antibody-horse radish peroxidase conjugate added. The plate is incubated at room temperature for lh and washed again five times with wash buffer. lOOμl of TMB peroxidase substrate is added and the colour allowed to develop for 10 min. The reaction is stopped with 50μl 1M H2SO4and the colour read at 450nm on a plate reader.
The following table gives the heparanase inhibitory activity of represenatative compounds of the invention.
Figure imgf000035_0001

Claims

CLAIMS:
1. A compound of formula (I) or a pharmaceutically acceptable salt, ester or prodrug thereof:
Figure imgf000036_0001
(I) wherein
R1, R2 and R3 are independently, hydrogen, halogen, CF3, OR6, NR7R8, NR8COR10, NR8SO2R10 or Q.6 alkyl optionally substituted by hydroxy, Q.6 alkoxy or NR7R8;
R4 is NR8CONR8R9; R5 is
Figure imgf000036_0002
wherein one of X and Y is R11 and the other is hydrogen or halogen; or X and Y together with the carbon atoms to which they are attached form a fused six-membered aromatic ring; Z is NR8, O or S;
R6 is hydrogen or Q.6 alkyl, C3.6 alkenyl or C3.6 alkynyl any of which can optionally be substituted by hydroxy, Cι.6 alkoxy or NR7R8;
R7 is hydrogen or Q.6 alkyl or C3.6 alkenyl either of which is optionally substituted by Q.6 alkoxy or a 5- or 6-membered heterocyclic ring containing up to three heteroatoms selected from NR8, S and O;
R8 is hydrogen or Q.6 alkyl; or the groups R7 and R8 may together with the nitrogen to which they are attached form a 5- or 6- membered ring which optionally contains up to two further heteroatom selected from NR8, S and O;
R9 is Cι_ιo alkyl or C30 alkenyl wherein a -CH2- group other than that adjacent to the N may be replaced by -O- and wherein the alkyl or alkenyl is substituted by one or more carboxylic acid or tetrazole groups; or in R4 the groups R8 and R9 may together with the nitrogen to which they are attached form a 5- or 6-membered ring, which is substituted with one or more carboxylic acid or tetrazole groups;
R10 is .6 alkyl; and
R11 is hydrogen, halogen, Q-6 alkyl, OR6 or phenyl optionally substituted by one or more substituents selected from halogen, Q.6 alkyl, CF3, OCF3, OR6, CN and methylenedioxo; or a 5- to 10- membered heteroaryl group containing up to three heteroatoms selected from O, N and S, which heteroaryl group may optionally be substituted by one or more substituents selected from Q.β alkyl, -6 alkoxy and halogen.
2. A compound according to claim 1 where R1, R2 and R3 are independently, hydrogen, halogen, OR6, NR7R8, or Q.6 alkyl optionally substituted by hydroxy or Q_6 alkoxy.
3. A compound according to any one of the preceding claims where Z is O.
4. A compound according to any one of the preceding claims where R6 is hydrogen or Q.6 alkyl, C3.6 alkenyl or .6 alkynyl any of which can optionally be substituted by hydroxy or Q.β alkoxy.
5. A compound according to any one of the preceding claims where Ru is phenyl optionally substituted by one or more substituents selected from halogen, Q.6 alkyl, CF3, OCF3, OR6, CN and methylenedioxo; or a 5- to 10-membered heteroaryl group containing up to three heteroatoms selected from O, N and S, which heteroaryl group may optionally be substituted by one or more substituents selected from Q.6 alkyl, Q.6 alkoxy and halogen.
6. A compound according to any one of the preceding claims where R1 is hydrogen, OR6 or NR7R8.
7. A compound according to any one of the preceding claims where R2 is hydrogen.
8. A compound according to any one of the preceding claims where R3 is hydrogen, halogen or OR6
9. A compound according to any one of the preceding claims where the configuration of the R groups is:
Figure imgf000037_0001
10. A compound of formula (I) as described in any one of Examples 1 to 36 or a pharmaceutically acceptable salt, ester or prodrug thereof.
11. A compound as defined in any one of claims 1 to 10 for use in medicine.
12. A process for the preparation of a compound as defined in any one of claims 1 to 10 which comprises: a) treating a compound of formula (H):
R2
R fc R 3:
(II) wherein Rxis NHR8, with a compound of formula (HI):
R^L_Nχχ__0
(HI) wherein RA is Cwo alkyl or Q_10 alkenyl wherein a -CH2- group other than that adjacent to the N may be replaced by -O- and wherein the alkyl or alkenyl is substituted with one or more carboxylate ester or CN groups; and when RA is substituted by CN, conversion of the CN to a tetrazole group; or b) treating a compound of formula (II) wherein Rx is NCO, with a compound of formula (IV):
R — NHa (IV) wherein RA is as defined for formula (HI).
13. A pharmaceutical formulation comprising a compound according to any one of claims 1 to 10 together with a pharmaceutically acceptable carrier or excipient.
14. The use of a compound as defined in any one of claims 1 to 10 in the manufacture of a medicament for the inhibition of heparanase.
15. The use of a compound as defined in any one of claims 1 to 10 in the manufacture of a medicament for the treatment of a subject with cancer.
16. The use of a compound as defined in any one of claims 1 to 10, in the manufacture of a medicament for the treatment of a disease selected from angiogenesis or an angiogenesis dependent disease, an inflammatory disease, an autoimmune disease and a cardiovascular disease.
17. A compound of formula (II).
Figure imgf000038_0001
(Ha) wherein Rx is NO2, NHR8, or NCO;
R5 is
Figure imgf000038_0002
wherein one of X and Y is R11 and the other is hydrogen or halogen; or X and Y together with the carbon atoms to which they are attached form a fused six-membered aromatic ring;
Rπ is phenyl optionally substituted by one or more substituents selected from halogen, Q_6 alkyl, CF3, OCF3, OR6, CN and methylenedioxo; or a 5- to 10-membered heteroaryl group containing up to three heteroatoms selected from O, N and S, which heteroaryl group may optionally be substituted by one or more substituents selected from Q_6 alkyl, Q.6 alkoxy and halogen; and
Z, R1, R2, R3 and R8 are as defined in claim 1.
PCT/GB2003/004980 2002-11-16 2003-11-17 Benzoxazole, benzthiazole and benzimidazole derivatives useful as heparanase inhibitors WO2004046123A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU2003285498A AU2003285498A1 (en) 2002-11-16 2003-11-17 Benzoxazole, benzthiazole and benzimidazole derivatives useful as heparanase inhibitors

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
GB0226822.5 2002-11-16
GB0226822A GB0226822D0 (en) 2002-11-16 2002-11-16 Novel compounds

Publications (1)

Publication Number Publication Date
WO2004046123A1 true WO2004046123A1 (en) 2004-06-03

Family

ID=9948008

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/GB2003/004980 WO2004046123A1 (en) 2002-11-16 2003-11-17 Benzoxazole, benzthiazole and benzimidazole derivatives useful as heparanase inhibitors

Country Status (3)

Country Link
AU (1) AU2003285498A1 (en)
GB (1) GB0226822D0 (en)
WO (1) WO2004046123A1 (en)

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7214696B2 (en) 2002-12-19 2007-05-08 The Scripps Research Institute Compositions and methods for stabilizing transthyretin and inhibiting transthyretin misfolding
US7868033B2 (en) 2004-05-20 2011-01-11 Foldrx Pharmaceuticals, Inc. Compounds, compositions and methods for stabilizing transthyretin and inhibiting transthyretin misfolding
US9249112B2 (en) 2011-09-16 2016-02-02 Pfizer Inc. Solid forms of a transthyretin dissociation inhibitor
CN106536478A (en) * 2014-05-22 2017-03-22 悉尼大学 Omega-3 analogues
CN108299413A (en) * 2018-01-22 2018-07-20 复旦大学 A method of no metal catalytic prepares 2- (aminocarbonyl phenyl) benzoxazoles and its derivative
EP3381906A1 (en) 2017-03-27 2018-10-03 Leadiant Biosciences SA Compounds for use as heparanase inhibitors
EP3381907A1 (en) 2017-03-27 2018-10-03 Leadiant Biosciences SA Symmetrical 2-aminophenyl-benzazolyl-5-acetate compounds and their use as anti-heparanase

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002060374A2 (en) * 2001-01-29 2002-08-08 Insight Strategy And Marketing Ltd Benz-1,3-azole derivatives and their uses as heparanase inhibitors
WO2003074516A1 (en) * 2002-03-06 2003-09-12 Oxford Glycosciences (Uk) Ltd Phthalimide carboxylic acid derivatives

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002060374A2 (en) * 2001-01-29 2002-08-08 Insight Strategy And Marketing Ltd Benz-1,3-azole derivatives and their uses as heparanase inhibitors
WO2003074516A1 (en) * 2002-03-06 2003-09-12 Oxford Glycosciences (Uk) Ltd Phthalimide carboxylic acid derivatives

Non-Patent Citations (10)

* Cited by examiner, † Cited by third party
Title
BULL.FAC.SCI.ASSIUT UNIV., vol. 4, - 1975, pages 133 - 138 *
CAN.J.CHEM., vol. 48, 1970, pages 2227 - 2233 *
DATABASE CROSSFIRE BEILSTEIN [online] Beilstein Institut zur Förderung der Chemischen Wissenschaften, Frankfurt am Main, DE; XP002271206, Database accession no. BRN: 297724 *
DATABASE CROSSFIRE BEILSTEIN [online] Beilstein Institut zur Förderung der Chemischen Wissenschaften, Frankfurt am Main, DE; XP002271207, Database accession no. BRN: 1009131 *
DATABASE CROSSFIRE BEILSTEIN [online] Beilstein Institut zur Förderung der Chemischen Wissenschaften, Frankfurt am Main, DE; XP002271208, Database accession no. BRN: 1009327 *
DATABASE CROSSFIRE BEILSTEIN [online] Beilstein Institut zur Förderung der Chemischen Wissenschaften, Frankfurt am Main, DE; XP002271209, Database accession no. BRN: 531928 *
DATABASE CROSSFIRE BEILSTEIN [online] Beilstein Institut zur Förderung der Chemischen Wissenschaften, Frankfurt am Main, DE; XP002271210, Database accession no. BRN: 1009179 *
INDIAN J. CHEM., vol. 12, 1974, pages 48 - 50 *
J. AMER. CHEM. SOC., vol. 57, 1935, pages 2121 - 2123 *
PROC.-INDIAN ACAD.SCI.SECT.A, vol. 61, 1965, pages 139 - 143 *

Cited By (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8653119B2 (en) 2002-12-19 2014-02-18 The Scripps Research Institute Methods for treating transthyretin amyloid diseases
US7214695B2 (en) 2002-12-19 2007-05-08 The Scripps Research Institute Compositions and methods for stabilizing transthyretin and inhibiting transthyretin misfolding
US7560488B2 (en) 2002-12-19 2009-07-14 The Scripps Research Institute Methods for treating transthyretin amyloid diseases
US7214696B2 (en) 2002-12-19 2007-05-08 The Scripps Research Institute Compositions and methods for stabilizing transthyretin and inhibiting transthyretin misfolding
US8168663B2 (en) 2002-12-19 2012-05-01 The Scripps Research Institute Pharmaceutically acceptable salt of 6-carboxy-2-(3,5 dichlorophenyl)-benzoxazole, and a pharmaceutical composition comprising the salt thereof
US7868033B2 (en) 2004-05-20 2011-01-11 Foldrx Pharmaceuticals, Inc. Compounds, compositions and methods for stabilizing transthyretin and inhibiting transthyretin misfolding
US8338459B2 (en) 2004-05-20 2012-12-25 Foldrx Pharmaceuticals, Inc. Compounds, compositions and methods for stabilizing transthyretin and inhibiting transthyretin misfolding
US9249112B2 (en) 2011-09-16 2016-02-02 Pfizer Inc. Solid forms of a transthyretin dissociation inhibitor
CN106536478A (en) * 2014-05-22 2017-03-22 悉尼大学 Omega-3 analogues
EP3381906A1 (en) 2017-03-27 2018-10-03 Leadiant Biosciences SA Compounds for use as heparanase inhibitors
EP3381907A1 (en) 2017-03-27 2018-10-03 Leadiant Biosciences SA Symmetrical 2-aminophenyl-benzazolyl-5-acetate compounds and their use as anti-heparanase
WO2018177857A1 (en) 2017-03-27 2018-10-04 Leadiant Biosciences Sa In Liquidazione Symmetrical 2-aminophenyl-benzazolyl-5-acetate compounds and their use as anti-heparanase
WO2018177865A1 (en) 2017-03-27 2018-10-04 Leadiant Biosciences Sa In Liquidazione Compounds for use as heparanase inhibitors
CN108299413A (en) * 2018-01-22 2018-07-20 复旦大学 A method of no metal catalytic prepares 2- (aminocarbonyl phenyl) benzoxazoles and its derivative

Also Published As

Publication number Publication date
AU2003285498A1 (en) 2004-06-15
GB0226822D0 (en) 2002-12-24

Similar Documents

Publication Publication Date Title
EP3733659B1 (en) Aromatic vinyl or aromatic ethyl derivative, preparation method therefor, intermediate, pharmaceutical composition, and application
EP2951153B1 (en) Selective hdac3 inhibitors
CA2714181C (en) Ampk modulators
JP6117430B2 (en) Novel compounds as selective histone deacetylase inhibitors and pharmaceutical compositions containing the same
EP2021330B1 (en) Benzimidazole modulators of vr1
JP7175888B2 (en) Piperazine Derivatives as Selective HDAC1,2 Inhibitors
JP2010520162A (en) Thiadiazole derivatives that are stearoyl-CoA desaturase inhibitors
KR20100071048A (en) Oxadiazole- and oxazole-substituted benzimidazole- and indole-derivatives as dgat1 inhibitors
JP2007527904A (en) Pyrrolopyridine-2-carboxylic acid hydrazide compounds as glycogen phosphorylase inhibitors
WO2003053915A2 (en) Compounds for the treatment of inflammatory disorders
CA3201443A1 (en) Tetrahydroquinoline derivative and medicinal use thereof
WO2020192650A1 (en) Preparation method for amide compound and application thereof in field of medicine
US7326727B2 (en) Furanthiazole derivatives as heparanase inhibitors
RU2675375C1 (en) New heterocyclic compound, preparation method therefor, pharmaceutical composition thereof
JP2022502497A (en) Inhibitors of HDAC1 and 2
WO2004046123A1 (en) Benzoxazole, benzthiazole and benzimidazole derivatives useful as heparanase inhibitors
MXPA06000538A (en) Novel compounds.
TW202239756A (en) 1,3,4-oxadiazole thiocarbonyl compounds as histone deacetylase 6 inhibitor, and pharmaceutical composition comprising the same
JP2018087173A (en) Anti-malignant brain tumor therapeutic agent
CA3136725A1 (en) Inhibitors of cd40-cd154 binding
TW202035406A (en) Heterocyclic compound as CDK-HDAC dual pathway inhibitor
CN117247358A (en) Benzodiazepine compound with HDAC6 inhibition activity and preparation method and application thereof
WO2019141095A1 (en) Amidine and guanidine derivative, preparation method therefor and medical use thereof
TW202321231A (en) Small molecule urea derivatives as sting antagonists
CN114401935A (en) Ion channel antagonists/blockers and uses thereof

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): BW GH GM KE LS MW MZ SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IT LU MC NL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
32PN Ep: public notification in the ep bulletin as address of the adressee cannot be established

Free format text: NOTING OF LOSS OF RIGHTS PURSUANT TO RULE 69(1) EPC (EPO FORM 1205A DATED 01.08.2005)

122 Ep: pct application non-entry in european phase
NENP Non-entry into the national phase

Ref country code: JP

WWW Wipo information: withdrawn in national office

Country of ref document: JP