WO2005011661A1 - Pharmaceutical agents - Google Patents

Pharmaceutical agents Download PDF

Info

Publication number
WO2005011661A1
WO2005011661A1 PCT/GB2004/003155 GB2004003155W WO2005011661A1 WO 2005011661 A1 WO2005011661 A1 WO 2005011661A1 GB 2004003155 W GB2004003155 W GB 2004003155W WO 2005011661 A1 WO2005011661 A1 WO 2005011661A1
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
alkoxy
hydrogen
compound
amino
Prior art date
Application number
PCT/GB2004/003155
Other languages
French (fr)
Inventor
Charles Marson
Original Assignee
University College London
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 University College London filed Critical University College London
Publication of WO2005011661A1 publication Critical patent/WO2005011661A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/195Carboxylic acids, e.g. valproic acid having an amino group
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/095Sulfur, selenium, or tellurium compounds, e.g. thiols
    • A61K31/10Sulfides; Sulfoxides; Sulfones
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/16Amides, e.g. hydroxamic acids
    • A61K31/18Sulfonamides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/192Carboxylic acids, e.g. valproic acid having aromatic groups, e.g. sulindac, 2-aryl-propionic acids, ethacrynic acid 
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/21Esters, e.g. nitroglycerine, selenocyanates
    • A61K31/215Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids
    • A61K31/216Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids of acids having aromatic rings, e.g. benactizyne, clofibrate

Definitions

  • HDAC histone deacetylase
  • Inhibitors of the enzyme histone deacetylase (HDAC) are a new and promising class of therapeutic agents.
  • HDAC inhibitors are especially promising for cancer therapy since they are able to regulate transcription and induce apoptosis or differentiation in cancer cells and are effective in animal xenograft models.
  • Histones in the nucleus of the cell are complex proteins integrally associated with DNA.
  • Histone deacetylase is acknowledged to be a critical regulator of chromatin structure and gene regulation, and HDAC inhibitors induce hyperacetylation in chromatin that leads to activation of specific genes. Conversely, deacetylation of histones results in repression of transcription.
  • HDAC histone deacetylases
  • HDAC histone deacetylases
  • acetyltransf erases Malignancies can arise by aberrant histone acetylation.
  • Fundamental nuclear processes including DNA replication, transcription and repair are influenced by chromatin structure and the binding of regulatory proteins to DNA. These processes can be modulated by altering the extent of acetylation of the ⁇ -amino groups of highly conserved lysine residues in the N-terminal tails that protrude from the histone octamer in the nucleus, thereby changing nucleosome conformation and regulating gene expression.
  • HDAC inhibitors show promise in countering a variety of cancers including breast and prostate cancer, as well as haematological disorders including leukaemia.
  • HDAC inhibitors are also of potential benefit in the treatment of Huntingdon's disease and possibly other neurological disorders especially Alzheimer's disease, and probably in a wide range of disorders, especially those of genetic and/or metabolic origin. Consequently, potent and metabolically stable HDAC inhibitors of good pharmacological profiles would possess important advantages over current HDAC inhibitors that have usually proved inadequate in clinical trials.
  • WO 02/085400 describes the treatment of diseases associated with aberrant silencing of gene expression, such as cancer, by administration of a HDAC inhibitor and a DNA methylation inhibitor.
  • HDAC inhibitors A new class of compounds that are HDAC inhibitors has now been prepared in which a saturated, partly saturated or unsaturated alkyl chain is a key feature of the inhibitor and to which is attached a metal-binding terminal group, the other end of the chain typically being linked to an aromatic or heteroaromatic system which is usually of an extended and substituted nature.
  • HDAC inhibitors can possess superior metabolic stability, lower toxicity or higher potency than previously described and related HDAC inhibitors. Accordingly, the present invention provides a compound of formula (I):
  • R 1 to R 5 each independently represent hydrogen, - Q alkyl, C,-C 10 alkenyl, - C 10 alkynyl, - Q alkoxy, C ⁇ -C ⁇ 0 thioalkoxy, hydroxyl, - o hydroxyalkyl, halo, - C 10 haloalkyl, amino, C t -C w alkylamino, di(C 1 -C 10 alkyl)amino, amido, nitro, cyano, (C r C 10 alkyl)carbonyloxy, ( - Q alkoxy )carbonyl, (CrC 10 alkyl)carbonyl, (C ⁇ -C w alkyl)thiocarbonyl, ( - o alkyl)sulfonylamino, aminos
  • R 17 and R 18 each independently represents hydrogen, unsubstituted or substituted - o alkyl, an unsaturated hydrocarbon chain of up to ten carbon atoms comprising one or more carbon-carbon double and/or triple bonds, or C 10 aryl, a 5- to 10-membered heterocyclic group, - o alkoxy, C ⁇ -C w thioalkoxy, hydroxyl, halo, cyano, nitro, amino, amido, (C r C 10 alkyl)carbonyloxy, ( - o alkoxy )carbonyl, (Cj-C 10 alkyl)carbonyl, ( -C JO alkyl)thiocarbonyl, (C ⁇ -C 10 alkyl)sulfonylamino, aminosulfonyl, C,-C 10 alkylsulfinyl, C ⁇ -C w alkyls
  • halo is typically chlorine, fluorine, bromine or iodine and is preferably chlorine or fluorine.
  • a - ,, haloalkyl group is typically a said - ,, alkyl group, for example a - alkyl group or - alkyl group, substituted by one or more said halo atoms. Typically, it is substituted by 1, 2 or 3 said halogen atoms.
  • Preferred haloalkyl groups include perhaloalkyl groups such as -CX 3 wherein X is a said halogen atom. Particularly preferred haloalkyl groups are -CF 3 and -CC1 3 .
  • a substituted - o alkyl group is typically a said - o alkyl group, for example a - alkyl group or a -C 4 alkyl group, substituted by one or more, for example from one to three, atoms o other groups such as hydroxy, halo, amino, C r C 4 alkoxy, - alkylthio, - alkylamino and di(Cj-C 4 alkyl)amino.
  • the substituted -C JO alkyl group may be a said C C 10 haloalkyl group.
  • Other suitable substituted - C 10 alkyl groups include - Q hydroxyalkyl.
  • An unsaturated hydrocarbon chain of up to ten carbon atoms comprising one or more carbon-carbon double or triple bonds is typically a said C 2 -C 10 alkenyl or C 2 -C 10 alkynyl group.
  • a C 6 or C 10 aryl group or moiety is typically a phenyl or naphthyl group or moiety.
  • the group or moiety may be substituted by one or more, for example from one to three, atoms or groups such as hydroxy, halo, cyano, amido, nitro, amino, C r C 4 alkyl, C r C 4 alkoxy, C r C 4 alkylthio, C r C 4 alkylamino and di(C C 4 alkyl)amino.
  • a 5- to 10-membered heterocyclic group may be a heteroaryl group. It may therefore be a 5- to 10-membered aromatic, i.e. fully unsaturated, ring such as a 5- or 6- membered ring, containing at least one heteroatom, for example one, two, three or four heteroatoms, selected from O, S and N.
  • C 5 -C 10 carbocyclic ring in which one or more, for example one, two, three or four, of the carbon atoms is replaced by a heteroatom selected from O, S and N.
  • suitable such heterocyclyl groups include piperidinyl, piperazinyl, mo ⁇ holinyl, pyrrolidinyl, tetrahydrofuranyl, imidazolidinyl, thiazolidinyl, 1,4-dioxanyl and 1,3-dioxolanyl.
  • the invention also provides the use of a compound of formula (I) for the manufacture of a medicament for use in treating a disorder mediated by histone deacetylase.
  • R 1 , R 2 and R 3 are each independently selected from hydrogen, - alkyl, C ⁇ -C 6 alkoxy, amino, - alkylamino, di(C r C 6 alkyl)amino, halo, - haloalkyl, (C ⁇ -C 6 alkoxy )carbonyl or - alkyl substituted by amino, C r C 6 alkoxy, C r C 6 alkylamino or di (C j - alkyl)amino.
  • R 1 , R 2 and R 3 are most preferably selected from hydrogen, methyl, ethyl, methoxy, ethoxy, dimethylamino, chloro, fluoro, trifluoromethyl, difluoromethyl, fluoromethyl, methoxymethyl, ethoxymethyl, aminomethyl, methylaminomethyl or dimethylaminomethyl.
  • one or two of R 1 , R 2 and R 3 is hydrogen.
  • the others or other of R 1 , R 2 and R 3 may be located at any position on the benzene ring.
  • the other is preferably located at the 4-position on the benzene ring.
  • R 4 and R 5 are each independently selected from hydrogen, - alkyl, C,-C 6 alkoxy, halo, C r C 6 haloalkyl or C C 6 alkyl substituted by amino, C,-C 6 alkoxy, - alkylamino or di(C C 6 alkyl)amino.
  • R 4 and R 5 may therefore be selected from hydrogen, C C 4 alkyl, C,-C 4 alkoxy, chloro, fluoro, C,-C 4 alkyl substituted by one, two or three chlorine or fluorine atoms, or - alkyl substituted by amino, C ⁇ or C 2 alkoxy, C or alkylamino or di( or alkyl)amino.
  • R 4 and R 5 are most preferably selected from hydrogen, methyl, ethyl, methoxy, ethoxy, chloro, fluoro, trifluoromethyl, difluoromethyl, fluoromethyl, methoxymethyl, ethoxymethyl, aminomethyl, methylaminomethyl or dimethylaminomethyl.
  • R 4 and R 5 is hydrogen.
  • the other may be located at any position on the benzene ring.
  • R 6 represents hydrogen, amino, - alkylamino, di(C C 6 alkyl)amino, halo such as chlorine or fluorine, C r C 6 alkyl, - alkoxy, - haloalkyl or - alkyl substituted by amino, C C 6 alkoxy, - alkylamino or di(C C 6 alkyl)amino.
  • halo such as chlorine or fluorine
  • C r C 6 alkyl - alkoxy, - haloalkyl or - alkyl substituted by amino, C C 6 alkoxy, - alkylamino or di(C C 6 alkyl)amino.
  • R 6 may therefore be selected from hydrogen, halo such as chlorine or fluorine, - alkyl, - alkoxy, -Q alkyl substituted by one, two or three halo atoms, or -Q alkyl substituted by amino, or C 2 alkoxy, or C 2 alkylamino or di( or C 2 alkyl)amino.
  • R 6 may therefore be hydrogen, methyl, ethyl, methoxy, ethoxy, fluoro, trifluoromethyl, difluoromethyl, fluoromethyl, dimethylamino, methoxymethyl, ethoxymethyl, aminomethyl, methylaminomethyl or dimethylaminomethyl.
  • R 7 represents hydrogen, chloro, fluoro, - alkoxy, - alkoxy, C C 4 alkylthio, amino, C t -C 4 alkylamino or d ⁇ - alkyl)amino.
  • R 7 and if present R 8 each represent hydrogen.
  • R u , R 12 and R 13 each independently represents hydrogen, C x -C 6 alkyl, C 6 or C 10 aryl or a 5 to 10-membered heterocylic group and p is an integer of from 1 to 4.
  • R 11 , R 12 and R 13 are as defined above.
  • R", R 12 and R 13 are independently selected from hydrogen, methyl or ethyl.
  • R 1 represents hydrogen, C C 4 alkyl, C C 4 alkoxy or halo; R 6 and R 18 are each independently selected from hydrogen, C t -C 4 alkyl or -Q alkoxy; X represents -OR 14 or -NHOH wherein R 14 is hydrogen or C r C 4 alkyl; Z represents C or SO; and m is 1, 2 or 3; and pharmaceutically acceptable salts thereof. More preferably R 1 represents hydrogen, C C 4 alkyl, C C 4 alkoxy or halo; R 6 and R 18 are each independently selected from hydrogen, C t -C 4 alkyl or -Q alkoxy; X represents -OR 14 or -NHOH wherein R 14 is hydrogen or C r C 4 alkyl; Z represents C or SO; and m is 1, 2 or 3; and pharmaceutically acceptable salts thereof. More preferably R 1 represents hydrogen, C C 4 alkyl, C C 4 alkoxy or halo; R 6 and R 18 are each independently selected from hydrogen, C t -
  • R 1 represents hydrogen, C C 4 alkyl, - alkoxy or halo
  • R 6 and R 18 are each independently selected from hydrogen, - alkyl or C r C 4 alkoxy
  • X represents -OR 14 or -NHOH wherein R 14 is hydrogen or C,-C 4 alkyl
  • Z represents C or SO
  • n is 1, 2 or 3
  • R 1 represents hydrogen, Cj-C 4 alkyl, - alkoxy or halo
  • R 6 is hydrogen and especially methyl.
  • R 1 to R 5 each independently represent hydrogen, C 2 -C 10 alkyl, C 2 -C 10 alkenyl, C 2 -C 10 alkynyl, - o alkoxy, - o thioalkoxy, hydroxyl, - o hydroxyalkyl, halo, C j -C 10 haloalkyl, amino, -C JQ alkylamino, alkyl)amino, amido, nitro, cyano, (C ⁇ -C 10 alkyl)carbonyloxy, ( - o alkoxy )carbonyl, ( - o alkyl)carbonyl, ( -C IQ alkyl)thiocarbonyl, (Cj-C 10 alkyl)sulfonylamino, aminosulfonyl, ( - o alkyl
  • a pharmaceutically acceptable salt is a salt with a pharmaceutically acceptable acid or base.
  • Pharmaceutically acceptable acids include both inorganic acids such as hydrochloric, sulphuric, phosphoric, diphosphoric, hydrobromic or nitric acid and organic acids such as citric, fumaric, maleic, malic, ascorbic, succinic, tartaric, benzoic, acetic, methanesulphonic, ethanesulphonic, benzenesulphonic or p-toluenesulphonic acid.
  • Pharmaceutically acceptable bases include alkali metal (e.g. sodium or potassium), alkali earth metal (e.g.
  • a primary amine salt can be the cyclohexylammonium salt
  • a suitable secondary amine salt may be the piperidine salt
  • a tertiary amine salt may be the triethylamine salt.
  • Tautomers of compounds of formula (I) and (VI) also form part of the invention.
  • the compounds of the invention can contain one or more chiral centre.
  • preferred compounds of the invention are optically active isomers.
  • preferred compounds of formula (I) containing only one chiral centre include an R enantiomer in substantially pure form, an S enantiomer in substantially pure form and enantiomeric mixtures which contain an excess of the R enantiomer or an excess of the S enantiomer.
  • the compounds of formula (I) and (VI) and their salts may be prepared by adaptation of conventional procedures. For example, Scheme 1 below illustrates one way in which compounds of the invention may be prepared.
  • R 1 to R 5 , R 7 and W are as defined above.
  • R 7 represents hydrogen.
  • Compound (A) can be obtained by a variety of methods, depending on the nature of W.
  • W represents a single bond, one of the following will typically be used: Suzuki coupling (of a boronic acid or ester with an aryl halide or triflate), Ullman coupling (copper-catalysed coupling of aryl halides or related compounds), Gomberg reaction (arylation of diazonium salts) or other metal-catalysed aryl-aryl coupling of aryl halides, triflates or related compounds.
  • compound (A) may be prepared from an arylamine of formula Ar-NHR 11 and an aromatic aldehyde or ketone.
  • R n is OH
  • W in the resulting compound of formula (A) will exist as the amide group of formula -N(R n )C(O)- or -C(O)NR n -, rather than as the corresponding hydroxyimine, due to keto-enol tautomerism.
  • compound (A) may be prepared by reduction of the corresponding imine or amide using hydrogen and a catalyst or a metal hydride system, or by addition of an organometallic reagent to the imine preceded and followed by suitable protection and deprotection respectively of the ketone group present in (A).
  • a preferred method of preparing such compounds (A) is by reaction of a suitable amine with a suitable aldehyde or ketone under reductive conditions, using reagents such as sodium cyanoborohydride or sodium triacetoxyborohydride.
  • compound (A) When W represents -NR u -CO- or -CO-NR 11 -, compound (A) may be prepared by reaction of ArCOQ (wherein Q is a leaving group, typically CI) with an arylamine in the presence of a base.
  • ArCOQ wherein Q is a leaving group, typically CI
  • W represents CO Friedel-Crafts arylation or metal-catalysed coupling involving ArCOQ compounds are preferred (wherein Q is a leaving group, typically CI).
  • W represents -SO 2 -NR ⁇ - or -R ⁇ N-SO 2 - compound (A) may be prepared by sulfonylation (typically using ArSO 2 Cl) of an arylamine in the presence of a base.
  • compound (A) When W represents -[C(R n )R 12 ] p -, compound (A) may be prepared by a Friedel-Crafts procedure (especially when R 11 and R 12 are both hydrogen) followed by reduction (hydrogen and a catalyst) or N 2 H 4 -KOH (Wolf-Kishner) or related procedures. For an alkyl chain in W, a Wittig reaction followed by reduction, typically using hydrogen and a catalyst, is the preferred method. When W represents -NR 11 -, compound (A) may be prepared by catalytic amination of an aryl halide, typically with a palladium-based catalyst, although displacement of an aryl halide or triflate by an arylamine or metal salt may also be used.
  • compound (A) When W represents O, compound (A) may be prepared by etherification of an aryl halide or triflate with a phenol, or more preferably its metal salt, typically in the presence of a metal catalyst such as a copper, palladium or nickel derivative or the metal itself.
  • a metal catalyst such as a copper, palladium or nickel derivative or the metal itself.
  • compound (A) When W represents S, compound (A) may be prepared by displacement of an aryl halide or triflate, with a thiophenol. This may be in the presence of a metal catalyst or more preferably a metal salt of the thiophenol. Alternatively, the metal salt of the thiophenol may also be reacted with a diazonium salt.
  • compound (A) When W represents SO, compound (A) may be prepared by oxidation of the corresponding compound (A) where W represents S with hydrogen peroxide or NaIO 4 .
  • W represents SO 2 compound (A) may be prepared by oxidation with H 2 O 2 or NaIO 4 of the corresponding compound (A) where W represents S with a peracid, typically /n-chloroperoxybenzoic acid, or by oxidation of the corresponding compound (A) where W represents SO with NaIO 4 .
  • compound (A) may be prepared by a Williamson ether-type synthesis involving a phenol or more preferably its metal salt with an alkyl halide, triflate or related displaceable group, i.e. ArC(R u )(R 12 )Q, wherein Q is a leaving group, typically halogen or triflate, preferably Br.
  • R 17 is preferably H, CH 3 or CH 2 CH 3 . More preferably, R 17 is H or Me. Typically, R 17 is H. The aromatic ring and the -C(O)R 17 moiety are typically trans with respect to each other in the resulting compound (B). Compound (B) is then reacted with compound (C) to form compound (D).
  • R 17 and R 18 are independently selected from H, CH 3 and CH 2 CH 3 . More preferably, they are H or CH 3 and typically both denote H.
  • R 17 and R 18 are H.
  • the other may be CH 3 but typically both R 17 and R 18 are H.
  • the group R in compound (C) is hydrogen or - o alkyl.
  • R is H, CH 3 or CH 2 CH 3 .
  • reaction of the acid group to form the corresponding methyl or ethyl ester is typically carried out before reaction with (B).
  • a reduction is carried out to convert compound (D) to compound (E).
  • One, two or more of the carbon-carbon double bonds in the chain of compound (D), for example all such bonds, are thus reduced to single bonds.
  • a metal catalyst such as Pd or palladium-on-carbon
  • suitable reagents are (a) Mg/ROH wherein R is C r C 10 alkyl or (b) R 3 SiH/H + .
  • the partial reduction can be carried out selectively.
  • a magnesium or related metal together with an alcohol, typically methanol or ethanol, can be used.
  • a trialkylsilane or related agent together with an acid, can be used.
  • the desired degree of saturation may alternatively be inco ⁇ orated from the Wittig reagent as appropriate.
  • compound (G) is desired, compound (D) is reacted with reagent (F).
  • compound (H) is formed by reaction of compound (E) with reagent (F).
  • a carboxylic acid (D) or (E) can be reacted with (i) oxalyl chloride, (ii) (CF 3 CO) 2 O and pyridine, and then (iii) water.
  • the preferred preparation method includes reduction of (B) to the corresponding alcohol using hydrogen and a metal catalyst or using a metal-hydride system.
  • Cyclopropanation is then effected with an iodoorganozinc reagent.
  • a chiral co-reagent such as a dialkyl tartrate or dialkyl tartramide is added, and subsequent oxidation (back to the aldehyde) to form the derivative of (B) that has the structure ArCR 7 (CH 2 )CR 6 CHO.
  • the cyclopropyl ring may alternatively be introduced by cyclopropanation of more unsaturated systems, such as compound (D), by a variety of reagents including CH 2 I 2 with a zinc-copper couple.
  • the product may then be reacted with compound (C) to give a nitro ester that is reduced, preferably with Fe and aqueous ammonia, to convert the -NO 2 group into an -NH 2 group, that amine then being reacted with an acid chloride or sulfonyl chloride to give a compound of formula (D) or a compound of formula (E).
  • R 7 and R 8 together represent O (i.e.
  • Schemes 2 to 5 shown below illustrate syntheses for producing certain aspects of the invention, for example when R 1 , R 2 , R 3 , R 4 and R 5 are selected from H, - o alkyl and - o alkoxy.
  • R 17 and R 18 are independently defined as above.
  • R 17 and R 18 are independently H, CH 3 or CH 2 CH 3 .
  • they are H.
  • one or R 17 and R 18 is H and the other is CH 3 . More preferably, both are H.
  • other substituents are defined as above.
  • the group R of (5) is typically hydroxy or - o alkoxy such as, in particular, -OCH 3 or -OCH 2 CH 3 .
  • Reduction of the nitro group of (6) to the amino derivative (7) can be carried out by many reagents, but iron and aqueous ammonia mixtures are shown to be particularly effective and convenient.
  • N-Sulfonylation and N-acylation of (7) affords the corresponding respective sulfonamides and esters (8).
  • Such sulfonylations are typically carried out with an arenesulfonyl chloride and base, and the corresponding acylations with an acid chloride and a base.
  • Many bases may possibly be employed. However, use of pyridine or a related base or of an aliphatic tertiary amine is preferred. In some cases, alkali metal hydroxides may suffice.
  • Scheme 3 shows a route to carboxylic acids (17) and their ester derivatives, and also to the hydroxamic acid derivatives (18).
  • the unsaturated aldehydes (4) undergo a Wittig or related process to afford the trienic carbonyl compounds (17).
  • a related process includes Horner-Wadsworth-Emmons or other modifications, as well as use of compounds other than phosphorus or arsenic ylid (10) (e.g. simple or complex enolates, or enolate equivalents of an appropriate carbonyl compound, such as enamino esters, enamino ketones or metal complexes derived from ⁇ -halo esters and related compounds).
  • the trienic carbonyl compound (17) so obtained can then be converted into other derivatives, but preferably the hydroxamic acids (18).
  • the phosphorus or arsenic ylid may have a variety of substituents (e.g. trialkyl or unsymmetrical alkyl and/or aryl substituents) and is not intended to be limited to Ph 3 P- or Ph 3 As- as shown in the Scheme.
  • the group R of the phosphorus or arsenic ylid (10) and of compound (17) is hydroxy or C r C 10 alkoxy.
  • R is C C 10 alkoxy.
  • it is -OCH 3 or -OCH 2 CH 3 .
  • R should denote hydroxy in compound (17), this may be achieved by hydrolysis of the corresponding ester compound (17) in which R is - o alkoxy. If R denotes hydroxy in compound (17), the reaction of (17) to (18) may require activation of the acid group of compound (17).
  • ester derivatives (22) and (25) can be converted, typically by aqueous hydroxylamine, but also by hydroxylamine derivatives with or without bases, respectively into the hydroxamic acids (23) and (26).
  • the group R in compounds (21) and (24) is typically hydroxy or C r C 10 alkoxy.
  • R is C,-C 10 alkoxy and especially is -OCH 3 or -OCH 2 CH 3 .
  • R is OH
  • the reaction of (22) to (23) and (25) to (26) may require activation of the acid group of compound (22) or (25) respectively.
  • Preferred reaction procedures are set out in Schemes 6 to 9 shown below.
  • enolates simple or complex enolates, or enolate equivalents of an appropriate carbonyl compound, such as enamino esters, enamino ketones or metal complexes derived from ⁇ -halo esters and related compounds).
  • the group R of reagent (5) is typically hydroxy or Cj-Cy, alkoxy such as, in particular, -OCH 3 or - OCH 2 CH 3 .
  • Reduction of the nitro group of (6) to the amino derivative (7) can be carried out be many reagents, but iron and aqueous ammonia mixtures are shown to be particularly effective and convenient.
  • N-Sulfonylation and N-acylation of (7) affords the corresponding respective sulfonamides and esters (8) in which X is C or SO.
  • Such sulfonylations are typically carried out with an arenesulfonyl chloride and base, and the corresponding acylations with an acid chloride and a base.
  • Many bases may possibly be employed. However, use of pyridine or a related base or of an aliphatic tertiary amine is preferred. In some cases, alkali metal hydroxides may suffice.
  • Scheme 7 is similar to Scheme 6.
  • Scheme 7 provides a route to carboxylic acids (17) and their ester derivatives, and also their hydroxamic acid derivatives (18).
  • An aldol or related condensation typically initiated by base but acid and other catalysts or reagents are feasible, is followed by dehydration to give the unsaturated aldehydes (4) which undergo a Wittig or related process to afford the dienic carbonyl compounds (6).
  • a related process includes Horner-Wadsworth-Emmons or other modifications, as well as use of compounds other than the phosphorus ylid (13) (e.g.
  • R in the phosphorus ylid (13) is typically hydroxy or - Q alkoxy.
  • R is - o alkoxy and especially -OCH 3 or - OCH 2 CH 3 .
  • Reduction of the nitro group of compound (6) to the amino derivative (15) can be carried out be many reagents, but iron and aqueous ammonia mixtures are particularly effective and convenient.
  • N-Sulfonylation and N-acylation of (15) affords the corresponding respective sulfonamides and esters (16) in which X is C or SO.
  • Such sulfonylations are typically carried out with an arenesulfonyl chloride and base, and the corresponding acylations with an acid chloride and a base.
  • Many bases may possibly be employed. However, use of pyridine or a related base or of an aliphatic tertiary amine is preferred although in some cases alkali metal hydroxides may suffice.
  • Scheme 8 provides an alternative route to carboxylic acids (17) and their ester derivatives, and also their hydroxamic acid derivatives (18).
  • the group X denotes C or SO.
  • a Wittig or related process affords the trienic compounds (17).
  • a related process includes Homer- Wadsworth-Emmons or other modifications, and also use of analogous arsenic ylides as well as use of compounds other than (10) and (11) (e.g. simple or complex enolates, or enolate equivalents of an appropriate carbonyl compound, such as enamino esters, enamino ketones or metal complexes derived from ⁇ -halo esters and related compounds).
  • the group R in reagents (10) and (11) is typically hydroxy or C x -C 0 alkoxy and especially -OCH 3 or - OCH 2 CH 3 .
  • the trienic compound (17) can be converted into other compounds of formula (I) according to the invention but especially to the hydroxamic acids (18).
  • the phosphorus or arsenic ylid (10) may have a variety of substituents (e.g. trialkyl or unsymmetrical alkyl and/or aryl substituents) and is not intended to be limited to Ph 3 P- or Ph 3 As- as shown.
  • substituents e.g. trialkyl or unsymmetrical alkyl and/or aryl substituents
  • R should denote hydroxy in compound (17) this may be achieved by hydrolysis of the corresponding ester compound (17) in which R is -C K , alkoxy.
  • reaction of (17) to (18) may require activation of the acid group of compound (17).
  • a substituted or unsubstituted 4-nitrophenylpropenal can be reacted with (10) to give the corresponding 4-nitrophenylhepta-2,4,6-trienoic acid ester that is reduced to give the corresponding 4-amino compound that is then treated with an aromatic sulphonyl chloride or aromatic chloride to give the corresponding compound (17).
  • the carbonyl compound (22) so obtained can then be converted into other derivatives, but especially the hydroxamic acids (23).
  • the phosphorus or arsenic ylid may have a variety of substituents (e.g. trialkyl or unsymmetrical alkyl and/or aryl substituents) and is not intended to be limited to Ph 3 P- or Ph 3 As- as shown.
  • the carbonyl compounds (22) can also be reduced, typically with hydrogen and a catalysts (e.g. Pd, Ni, Co, etc.) or by hydrosilylation or hydride-acid systems and other processes to give the carboxylic acids (25) and their derivatives.
  • ester derivatives (22) and (25) can be converted, typically by aqueous hydroxylamine, but also by hydroxylamine derivatives with or without bases, respectively into the hydroxamic acids (23) and (26).
  • the group R in reagent (21) and in compound (24) is hydroxy or - o alkoxy.
  • R is C ⁇ -C 10 alkoxy and especially -OCH 3 or - OCH 2 CH 3 .
  • Scheme 10 shows how an intermediate (7) bearing an amino group can be converted into compounds of the invention where W is -C(R U )(R 12 )-NR 13 -.
  • R 11 and R 12 are both hydrogen, although other reactants could readily be chosen in order to prepare compounds having other R 11 and R 12 groups.
  • the reactant R 10 CHO is shown in parentheses because it need not be used if it is desired that R 10 be hydrogen.
  • a preferred example obtained using procedures in Scheme 10 include the reaction of an amine (7) with a benzaldehyde in the presence of formaldehyde and sodium cyanoborohydride to give the corresponding tertiary amine whose ester group is subsequently converted into the corresponding hydroxamic acid.
  • hydroxylamine is shown as the means of obtaining a hydroxamic acid. While this is the preferred reagent, especially in the form of an aqueous 50% solution, and in its action upon methyl or ethyl esters that are to undergo hydroxamation, this protocol is not intended to exclude other variants.
  • a hydroxylamine salt especially hydroxylamine hydrochloride (or hydrates thereof) in combination with a base or alkali, especially sodium hydroxide or potassium hydroxide, and commonly followed by filtration to give a "salt-free" solution of hydrazine (often in an alcoholic solvent) is an effective procedure.
  • carboxylic acids e.g.
  • the compounds of the invention are inhibitors of histone deacetylase (HDAC). They may therefore be used to treat a HDAC-mediated disorder.
  • a therapeutically effective amount of a compound of the invention is administered to a subject, typically a human being, having such a disorder. The condition of the subject can thus be improved. Symptoms associated with the disorder may be ameliorated.
  • Compounds of the invention may also be inhibitors of DNA methyl transferase.
  • HDAC-mediated disorders that may be treated according to the invention include cancer such as breast cancer, colon cancer, colorectal cancer, esophageal cancer, glioma, leukemia, lung small and non-small cell cancers, neuroblastoma, prostate cancer, thoracic cancer, melanoma, ovarian cancer, cervical cancer and renal cancer; cardiac hypertrophy; hematological disorders such as haemoglobinopathies, thalessemia and sickle cell anemia; and genetic-related metabolic disorders such as cystic fibrosis, peroxisome biogenesis disorders and adrenoleukodystrophy.
  • HDAC inhibitors have also been proposed for stimulating hematopoietic cells ex vivo, ameliorating protozoal parasitic infection, accelerating wound healing and protecting hair follicles.
  • a compound of the invention may be used in combination with another chemotherapeutic or antineoplastic agent in the treatment of a cancer.
  • chemotherapeutic or antineoplastic agents examples include mitoxantrone; Vinca alkaloids such as vincristine and vinblastine; anthracycline antibiotics such as daunorubicin and doxorubicin; alkylating agents such as chlorambucil and melphalan; taxanes such as paclitaxel; antifolates such as methotrexate and tomudex; epipodophyllotoxins such as etoposide; camptothecins such as irinotecan and its active metabolite SN-38 and DNA methylation inhibitors such as the DNA methylation inhibitors disclosed in WO 02/085400.
  • products which contain a compound of the invention and another chemotherapeutic or antineoplastic agent as a combined preparation for simultaneous, separate or sequential use in creating a cancer.
  • the compound of the invention and the other agent may be administered together or, if separately, in any order as determined by a physician.
  • the present compounds can be administered in a variety of dosage forms, for example orally such as in the form of tablets, capsules, sugar- or film-coated tablets, liquid solutions or suspensions or parenterally, for example intramuscularly, intravenously or subcutaneously.
  • the present compounds may therefore be given by injection or infusion.
  • the dosage depends on a variety of factors including the age, weight and condition of the patient and the route of administration.
  • the dosage for a particular patient will be determined by a physician.
  • the dosage adopted for each route of administration when a compound of the invention is administered to adult humans is 0.001 to 500 mg/kg, most commonly in the range of 0.01 to 100 mg/kg, body weight, for instance 0.01 to 50 mg/kg.
  • Such a dosage may be given, for example, from 1 to 5 times daily by bolus infusion, infusion over several hours and/or repeated administration.
  • the dosage and timing of administration of, for example, another chemotherapeutic or antineoplastic agent which may be given to a cancer patient with a compound of the invention will similarly be dependent on a variety of factors and will be determined by a physician.
  • a compound of any of the formulae (I) to (VII) described above or a pharmaceutically acceptable salt thereof is formulated for use as a pharmaceutical composition also comprising a pharmaceutically acceptable carrier or diluent.
  • the compositions are typically prepared following conventional methods and are administered in a pharmaceutically suitable form.
  • Preferred pharmaceutical compositions are sterile and pyrogen-free.
  • the pharmaceutical compositions provided by the invention typically contain a compound of the invention which is a substantially pure optical isomer.
  • Compositions suitable for oral administration may, if required, contain a colouring or flavouring agent.
  • a capsule or tablet comprises from 5 to 500 mg, preferably 10 to 500 mg, more preferably 15 to 100 mg, of a compound of formula (I) or a pharmaceutically acceptable salt thereof.
  • Solid oral forms of the pharmaceutical compositions of the invention may contain, together with the active compound, diluents, e.g. lactose, dextrose, saccharose, cellulose, corn starch or potato starch; lubricants, e.g. silica, talc, stearic acid, magnesium or calcium stearate, and/or polyethylene glycols; binding agents; e.g. starches, arabic gums, gelatin, methylcellulose, carboxymethylcellulose or polyvinyl pyrrolidone; disaggregating agents, e.g.
  • diluents e.g. lactose, dextrose, saccharose, cellulose, corn starch or potato starch
  • lubricants e.g. silica, talc, stearic acid, magnesium or calcium stearate, and/or polyethylene glycols
  • binding agents e.g. starches, arabic gums, gelatin, methylcellulose, carboxymethylcellulose or
  • Liquid dispersions for oral administration may be syrups, emulsions and suspensions.
  • the syrups may contain as carriers, for example, saccharose or saccharose with glycerine and/or mannitol and/or sorbitol.
  • Suspensions and emulsions may contain as carrier, for example a natural gum, agar, sodium alginate, pectin, methylcellulose, carboxymethylcellulose, or polyvinyl alcohol.
  • the suspension or solutions for intramuscular injections may contain, together with the active compound, a pharmaceutically acceptable carrier, e.g. sterile water, olive oil, ethyl oleate, glycols, e.g. propylene glycol, and if desired, a suitable amount of lidocaine hydrochloride.
  • Solutions for injection or infusion may contain as carrier, for example, sterile water or preferably they may be in the form of sterile, aqueous, isotonic saline solutions. The following Examples illustrate the invention.
  • Example 1 (2E.4E)-5-(4-benzenesulfonylaminophenyl)-4-methylpenta-2,4-dienoic acid ethyl ester (8a) To a stirred solution of (2E,4E)-5-(4-aminophenyl)-4-methylpenta-2,4-dienoic acid ethyl ester (Preparation Example 7; 7b) (0.35 g, 1.52 mmol) in pyridine (8.0 mL) at 25 °C was added benzenesulfonyl chloride (0.39 mL, 3.04 mmol) by means of a stainless steel cannula.
  • Example 2 (2E t 4£)-5-(4-(4-chlorobenzenesulfonylamino)phenyl)penta-2.4-dienoic acid ethyl ester
  • 8b A solution of (2E,4E)-5-(4-aminophenyl)penta-2,4-dienoic acid ethyl ester (Preparation Example 6, 7a) (0.32 g, 1.47 mmol) andp-chlorobenzenesulfonyl chloride (0.405 g, 1.91 mmol) in pyridine (3 mL) was heated at reflux for 6 h, then removed to ambient temperature and stirred for 12 h.
  • Example 5 (2£.4 ⁇ )-5-(4-(4-chlorobenzenesulfonylamino)phenyl)-4-methylpenta- 2.4-dienoic acid (8e)
  • Example 7 (2E.4E)-5-(4-(4-methoxybenzenesulfonylamino)phenyl)-2.4- dimethylpenta-2,4-dienoic acid ethyl ester (8g)
  • Example 9 (2--?.4-g)-5-(4-(4-methoxybenzenesulfonylamino)phenyl)-4- methylpenta-2.4-dienoic acid (8i)
  • Example 10 (2ig.4E)-5-(4-(4-chlorobenzoylamino)phenyl) -4-methylpenta-2.4- dienoic acid ethyl ester (8k)
  • j-Chlorobenzoyl chloride was prepared by a modification of a procedure described in: J. P. Dickie, M. E. Loomans, T. M. Farley and F. M. Strong, J. Med. Chem., 1968, 6, 424.
  • p-Chlorobenzoic acid (0.78 g, 4.98 mmol) was dissolved in toluene (20 mL) and one drop of DMF added.
  • 1,2-Phenylene phosphorochloridite (0.56 g, 3.21 mmol) in dry toluene (15 mL) was then added to the mixture, and the reaction heated at reflux for 2.5 h. A solution of j-chlorobenzoyl chloride was then added, and the mixture heated at reflux for a further 24 h. The solvent was evaporated and the residue dissolved in dichloromethane (50 mL). The solution was washed with 2 M hydrochloric acid (3 x 40 mL), then water (2 x 40 mL) and lastly saturated aqueous sodium hydrogen carbonate (3 x 40 mL). The organic layer was dried (MgSO 4 ), filtered and evaporated. The residue was recrystallised from propan-2-ol to give the title compound (0.73 g, 45%) as white plates, mp 169-170 °C.
  • Example 11 (2E,4E)-5-(4-(4-methoxybenzoylamino)phenyl)-4-methylpenta-2.4- dienoic acid ethyl ester (81) (2E,4E)-5-(4-Aminophenyl)-4-methylpenta-2,4-dienoic acid ethyl ester (Preparation Example 7; 7b) (1.00 g, 4.33 mmol) was dissolved in dry toluene (13 mL) and triethylamine (1.26 mL, 8.96 mmol).
  • o-Phenylene phosphorochloridite (0.98 g, 5.62 mmol) in dry toluene (6.2 mL) was then added to the mixture, and the reaction heated at reflux for 2.5 h.
  • p-Chlorobenzoic acid (0.37 g, 2.37 mmol) was then added, and the mixture heated at reflux for a further 24 h.
  • the solvent was evaporated and the residue purified by flash chromatography on silica gel (80 g) (1:4 ethyl acetate: 60-80 °C petroleum ether) to give a residue was recrystallised from propan-2-ol to give the title compound (0.07 g, 5%) as yellow platelets, mp 121-122 °C.
  • Example 12 (2i ⁇ 1 4i t ?)-5-(4-benzenesulfonylaminophenyl)-4-methylpenta-2.4- dienoic acid hydroxyamide (9a)
  • hydroxylamine hydrochloride (0.56 g, 8.06 mmol)
  • methanol 4.0 mL
  • potassium hydroxide 0.45 g, 8.04 mmol
  • methanol 4.0 mL
  • Example 13 (2£.4E)-5-(4-chlorobenzenesulfonylaminophenyl)-4-methylpenta- 2.4-dienoic acid hydroxyamide (9b)
  • Example 14 (2E.4E)-5-(4-methoxybenzenesulfonylaminophenyl)-4-methylpenta- 2.4-dienoic acid hydroxyamide (9c)
  • (2E,4E)-5-(4-(4-methoxybenzenesulfonylamino)phenyl)-4- methylpenta-2,4-dienoic acid ethyl ester (Example 8, 8f) (0.70 g, 1.74 mmol) in THF (10 mL) cooled to 0 °C was added dropwise a 50% aqueous solution of hydroxylamine (0.52 g, 0.78 mmol) and potassium hydroxide (0.31 g, 5.54 mmol) in methanol (3.0 mL) at 0 °C over 30 min.
  • Example 15 (2£.4E)-5-(4-methoxybenzenesulfonylaminophenyl)penta-2,4-dienoic acid hydroxyamide (9d)
  • Example 16 (2E,4ig.6E)-7-(4-(4-methoxybenzenesulfonylamino)phenyl)-6- methvIhepta-2.4.6-trienoic acid ethyl ester (17h)
  • Example 17 (2£ t 4£.6£)-7-(4-(4-chlorobenzoylamino)phenyl)hepta-2.4.6-trienoic acid methyl ester (17s) 4-Chloro-N-(4-formylphenyl)benzamide (Preparation Example 1; 2g) (0.27 g, 1.05 mmol), [(2£,4E)-6-methoxy-6-oxo-2,4-hexadienyl]triphenylphosphonium bromide (0.49 g, 1.05 mmol) and potassium carbonate (0.72 g, 5.22 mmol) were added to THF (20 mL). The mixture was stirred at 40 °C for 72 h.
  • Example 18 5-r4-(4-methoxybenzenesulfonylamino)phenyll-4-methylpentanoic acid ethyl ester (25c)
  • Ethyl 5-(4-aminophenyl)-4-methylpentanoate (Preparation Example 11, 25a) (0.65 g, 2.76 mmol) was dissolved in pyridine (4.0 mL), and 4-methoxybenzenesulfonyl chloride (0.57 g, 2.76 mmol) was added to the stirred mixture. The mixture was stirred for an additional 16 h at 25 °C. The pyridine was removed under a high vacuum and the residue was dissolved in dichloromethane (50 mL).
  • Example 20 5-r4-(4-chlorobenzenesulfonylamino)phenyll-4-methylpentanoic acid hydroxyamide
  • 26b Ethyl 5-[4-(4-chlorobenzenesulfonylamino)phenyl]-4-methylpentanoate (Preparation Example 12, 25b) (0.63 g, 1.53 mmol) was dissolved in methanol (5.0 mL) at 0 °C. The solution was stirred and 50% aqueous hydroxylamine (0.91 g, 13.8 mmol) was added dropwise over 15 min, and then aqueous potassium hydroxide (1.3 mL, 1.6 M) in methanol added to the mixture in one batch.
  • Example 21 5-r4-(4-chlorobenzoylamino)phenyll-4-methylpentanoic acid hydroxyamide
  • Example 22 (2E.4E.6E)-7(4-(p-Chlorobenzenesulphonylamino)-6-methylhepta- 2.4.6-trienoic acid ethyl ester
  • (2E,4E,6E)-7(p-aminophenyl)-6-methylhepta-2,4,6-trienoic acid ethyl ester (0.37 g, 1.44 mmol) and -chlorobenzenesulphonylchloride (0.365 g, 2.88 mmol) in pyridine (5 mL) was heated at reflux for 24 h. Stirring was then continued for an additional 48 h at room temperature.
  • Histone deacetylase inhibitory activity was measured as described by Vigushin et al., Clin. Cancer Res., 7, 971-976 (2001) based on methods published by Taunton et al., Science, 272, 408-411 (1995) and Emiliani et al, Proc. Natl. Acad. Sci. U.S ⁇ ., 95, 2795-2800 (1998). Briefly, the assay begins by incubating histone deacetylase enzymes contained in a nuclear extract from the HeLa human cervical adenocarcinoma cell line with a compound of the invention followed by addition of a radiolabeled substrate.
  • the substrate was a synthetic peptide corresponding to histone H4 (amino acids 14-21) that had been chemically acetylated on lysine residues with sodium [ 3 H]acetate according to the method published by Taunton et al, Science 272, 408-411 (1995). Released [ 3 H] acetic acid (a measure of histone deacetylase activity) was then extracted with ethyl acetate and quantified in a scintillation counter. The concentration of a compound of the invention that inhibits histone deacetylase activity by 50% (i.e. IC 50 ) was then determined by repeating the assay with a range of different concentrations of compound. Each assay was performed in duplicate with control samples in triplicate for accuracy.
  • HeLa cell nuclear extract HeLa cell nuclear extract was prepared according to the method of Dignam et al, Nucleic Acids Res., 11, 1475-1489 (1983). HeLa human cervical adenocarcinoma cells were grown at 37°C in DMEM medium containing 5% foetal calf serum to a concentration of 5x10 s cells per ml prior to harvesting. Cells were then harvested by centrifugation for 10 minutes at 2,000 rpm in a Sorvall HG4L rotor. The cell pellet was resuspended in 5 volumes of cold phosphate buffered saline, collected by centrifugation at 4°C and all subsequent manipulations were performed at 4°C.
  • Cells were suspended in 5 packed cell volumes of lOmM HEPES (pH 7.9 at 4°C), 1.5 mM MgCl 2 , lOmM KC1 and 0.5 mM DDT and allowed to equilibrate for 10 minutes.
  • the cells were pelleted by centrifugation as above, resuspended in 2 packed cell volumes of the same buffer and then lysed by 10 strokes of a glass Dounce homogenizer. The homogenate was centrifuged as before and the pellet was then centrifuged for 20 minutes at 25,000 x g in a Sorvall SS34 rotor to remove residual cytoplasmic material, yielding crude nuclei.
  • the supernatant was dialysed against 50 volumes at 20 mM HEPES (pH 7.9), 20% (v/v) glycerol, 100 mM KC1, 0.2 mM EDTA, 0.5 mM PMSF and 0.5 mM DTT for 5 hours and the dialysate was then centrifuged at 25,000 x g for 20 minutes (Sorvall SS34 rotor).
  • the supernatant designated the nuclear extract, was snap frozen in liquid nitrogen and then stored at -80°C.
  • the protein concentration measured by Bradford assay was 10 mg/ml and 50 mg of protein was obtained from 10 9 cells.
  • GAKRHRKV was synthesised in an automated peptide synthesiser (ABI 433; Applied Biosystems, Cheshire, UK), purified by reverse phase high performance liquid chromatography (HPLC), and lyophilised.
  • the peptide was >95% pure by reverse phase HPLC, mass spectrometry and capillary electrophoresis. All subsequent steps were performed in a fume hood.
  • the supernatant containing [ 3 H]acetate-labelled histone H4 peptide was purified by gel filtration on Sephadex G-25 (PD10 column; Amersham Biosciences UK Limited, Buckinghamshire, UK). After equilibrating with 10 column volumes of TEN, the supernatant was loaded onto the column and then eluted with TEN. 0.5 ml fractions were collected and the radioactivity in each was quantified by liquid scintillation counting. After the column void volume, the [ 3 H]acetate-labelled histone H4 peptide elutes first followed by free unincorporated label ([ 3 H]acetic acid). Eluates containing the purified radiolabeled peptide are pooled, divided into aliquots and stored at -70°C until use.
  • Histone deacetylase assay Histone deacetylase inhibition by compounds of the invention was assayed as described in Vigushin et al, Clin Cancer Res. 7, 971-976 (2001) based on methods published by Taunton et al, Science, 272, 408-411 (1995) and Emiliani et al. Proc. Natl. Acad. Sci. U.S ⁇ ., 95, 2795-2800 (1998).
  • the substrate was a synthetic peptide corresponding to amino acids 14-21 of histone H4 that had been chemically acetylated on lysine residues with sodium [ 3 H]acetate as described by Taunton et al, Science 272, 408-411 (1995).
  • a stock solution in dimethylsulfoxide (DMSO) was prepared for each compound of the invention to be tested and trichostatin A as a positive control.
  • Stock solutions were diluted in DMSO to give a range of lOOx working solutions.
  • the assay was performed in a final reaction volume of 200 ⁇ l.
  • reaction mixture was incubated for 60 minutes at room temperature. Fifty ⁇ l of a quenching solution [1 M HC1/0.16 M acetic acid] was then added to stop the reaction. The released [ 3 H]acetate in each assay reaction was extracted into 600 ⁇ l ethyl acetate. After mixing by vortex, the organic and aqueous phases were separated by centrifugation (14,000 x g for 1 minute at room temperature).
  • Duplicate 200 ⁇ l aliquots of the upper organic phase were transferred into separate scintillation vials each containing 5 ml scintillant (Hionic Fluor; Canberra Harwell Ltd., Didcot, UK) and the radioactivity in each measured by ⁇ -scintillation counting.
  • An initial assay was performed to established the range of activity of each compound of the invention. The assay was then repeated using four log dilutions in range according to the expected potency for each test compound.
  • the concentration of each compound of the invention that inhibited histone deacetylase enzyme activity by 50% was determined graphically in each case using non-linear regression analysis to fit inhibition data to the appropriate dose-response curve (GraphPad Prism Version 3.0; GraphPad Software Inc., San Diego, CA). Each test compound was assayed in duplicate whilst positive and negative control samples were assayed in triplicate. Test compounds of the invention were found to be potent histone deacetylase inhibitors, some having IC 50 values in the low nanomolar concentration range (e.g. two test compounds had IC S0 values of 49 nM and 74 nM).

Abstract

Use of compounds of formula (I) in the manufacture of a medicament for use in treating a disorder mediated by histone deacetylase: wherein the symbol ---- represents a single bond or a double bond or the symbol ---- R6 and R8 together represent cyclopropyl and R1 to R8 W, X and Y are as defined herein; and pharmaceutically acceptable salts thereof. Also disclosed are compounds for such uses. The compounds are useful in the treatment of cancers. They may be utilised in combination therapies with DNA methylation inhibitors and other anti cancer agents.

Description

PHARMACEUTICAL AGENTS The present invention inhibitors of histone deacetylase and the use of such compounds in medicine. Inhibitors of the enzyme histone deacetylase (HDAC) are a new and promising class of therapeutic agents. HDAC inhibitors are especially promising for cancer therapy since they are able to regulate transcription and induce apoptosis or differentiation in cancer cells and are effective in animal xenograft models. Histones in the nucleus of the cell are complex proteins integrally associated with DNA. Histone deacetylase is acknowledged to be a critical regulator of chromatin structure and gene regulation, and HDAC inhibitors induce hyperacetylation in chromatin that leads to activation of specific genes. Conversely, deacetylation of histones results in repression of transcription. There is increasing evidence that cell proteins involved in the regulation of proliferation and differentiation act by recruiting histone deacetylases (HDAC) and also histone ace tyltransf erases. Malignancies can arise by aberrant histone acetylation. Fundamental nuclear processes including DNA replication, transcription and repair are influenced by chromatin structure and the binding of regulatory proteins to DNA. These processes can be modulated by altering the extent of acetylation of the ω-amino groups of highly conserved lysine residues in the N-terminal tails that protrude from the histone octamer in the nucleus, thereby changing nucleosome conformation and regulating gene expression. Accordingly, HDAC inhibitors could be used in therapy to relieve gene repression and to reinstate the program of cell differentiation and apoptosis, a form of "transcription therapy". There is intense interest in finding new inhibitors of histone deacetylase (HDAC), especially because most of the early compounds showed severe limitations. Thus, the natural product trichostatin A is thought to mimic such acetylated lysine residues, and its potent inhibition of HDAC is consistent with the binding of its hydroxamic acid unit to a zinc atom in the catalytic pocket of the enzyme. However the high HDAC inhibitory activity of trichostatin A is offset by rapid metabolism to many products, resulting in a short clinical half -life that makes it ineffective for therapy. Sodium phenylbutyrate, a feeble inhibitor of histone deacetylase, was used (together with retinoic acid) to induce complete remission of multi-resistant acute promyelocytic leukaemia (APML) in a patient unbeatable by conventional therapy. Depsipeptide is a potent HDAC inhibitor but exhibited significant cardiotoxicity in clinical trials. Further HDAC inhibitors are disclosed in WO 02/076941. HDAC inhibitors show promise in countering a variety of cancers including breast and prostate cancer, as well as haematological disorders including leukaemia. HDAC inhibitors are also of potential benefit in the treatment of Huntingdon's disease and possibly other neurological disorders especially Alzheimer's disease, and probably in a wide range of disorders, especially those of genetic and/or metabolic origin. Consequently, potent and metabolically stable HDAC inhibitors of good pharmacological profiles would possess important advantages over current HDAC inhibitors that have usually proved inadequate in clinical trials. WO 02/085400 describes the treatment of diseases associated with aberrant silencing of gene expression, such as cancer, by administration of a HDAC inhibitor and a DNA methylation inhibitor. A new class of compounds that are HDAC inhibitors has now been prepared in which a saturated, partly saturated or unsaturated alkyl chain is a key feature of the inhibitor and to which is attached a metal-binding terminal group, the other end of the chain typically being linked to an aromatic or heteroaromatic system which is usually of an extended and substituted nature. These HDAC inhibitors can possess superior metabolic stability, lower toxicity or higher potency than previously described and related HDAC inhibitors. Accordingly, the present invention provides a compound of formula (I):
Figure imgf000003_0001
wherein: the symbol :===, represents a single bond or a double bond or the symbol zzz, R6 and R8 together represent cyclopropyl; R1 to R5 each independently represent hydrogen, - Q alkyl, C,-C10 alkenyl, - C10alkynyl, - Q alkoxy, Cα-Cι0 thioalkoxy, hydroxyl, - o hydroxyalkyl, halo, - C10 haloalkyl, amino, Ct-Cw alkylamino, di(C1-C10 alkyl)amino, amido, nitro, cyano, (CrC10alkyl)carbonyloxy, ( - Q alkoxy )carbonyl, (CrC10 alkyl)carbonyl, (C^-Cw alkyl)thiocarbonyl, ( - o alkyl)sulfonylamino, aminosulfonyl, ( -Cj,, alkyl)sulfinyl, (Cj-Cno alkyl)sulfonyl or C -Cw alkyl substituted by amino, - Q alkoxy,
Figure imgf000004_0001
alkylamino or di - o alkyl)amino; R6 represents hydrogen, -CK, alkyl, substituted Cj-C10 alkyl, an unsaturated hydrocarbon chain of up to ten carbon atoms comprising one or more carbon-carbon double or triple bond, C6 or C10 aryl, a 5- to 10-membered heterocyclic group, -Cy, alkoxy, CJ-CJO thioalkoxy, hydroxyl, halo, cyano, nitro, amino, - o alkylamino, alkyl)amino, amido, ( - o alkyl)carbonyloxy, (Cι-C10 alkoxy )carbonyl, ( - Cw alkyl)carbonyl, ( -Cjo alkyl)thiocarbonyl, ( - o alkyl)sulfonylamino, aminosulfonyl, (Cχ-C10 alkyl)sulfinyl, (Cι-C10 alkyl)sulfonyl, a saturated or unsaturated C3-C12 hydrocarbon chain interrupted by O, S, NR, CO, C=NR, N(R)SO2, SO2N(R), N(R)C(O)O, OC(O)N(R), N(R)C(O)N(R), OC(O), C(O)O, OSO2, SO2O or OC(O)O where: (a) R independently represents hydrogen, - o alkyl, C2-C10 alkenyl, C2-Cω alkynyl, C^C^ alkoxy, -Ciohydroxyalkyl, hydroxyl or Cj- ,, haloalkyl, and (b) the saturated or unsaturated hydrocarbon chain is optionally substituted with CrC10 alkyl, C2-C10 alkenyl, -CJO alkynyl, CrC10 alkoxy, hydroxyl, Cλ-Cw hydroxyalkyl, halo, - o haloalkyl, amino, ( - o alkyl)carbonyloxy, (CJ- Q alkoxy )carbonyl, (Cι-C10 alkyl)carbonyl, (Cr C10 alkyl)sulfonylamino, aminosulfonyl or
Figure imgf000004_0002
when the symbol ™ represents a single bond, R7 and R8 each independently represents hydrogen, halo,
Figure imgf000004_0003
alkyl, C6 or C10 aryl, a 5- to 10-membered heterocyclic group, OR9, SR9 or NR9R10 wherein R9 and R10 each independently represent hydrogen or Cj- alkyl or one of R9 and R10 is H and the other is -CO^- alkyl) or R7 and R8 together represent =O, =CH2 or =CHR9 wherein R9 is as defined above; - when the symbol z represents a double bond, R7 represents hydrogen, halo, -CJO alkyl, C6 or C10 aryl, a 5- to 10-membered heterocyclic group, OR9, SR9 or NR9R10 wherein R9 and R10 are as defined above and R8 is absent; W represents a single bond, -C(Rπ)=N-, -N=C(Rπ)-, -C(Rπ)(R12)-NR13-, -NR13-C(Rπ)(R12)-, -CO-NR11-, -NRn-CO-, -SO2-NRπ-, -NRu-SO2-, -C(Ru)(R12)-O-, -O-C(Ru)(R12)-, -C(Rπ)(R12)-S-, -S-C(RU)(R12)-, -CO-, -NR11-, -SO-, -SO2-, O, S or -[C(Rπ)R12]p- wherein R", R12 and R13 each independently represents hydrogen, - alkyl, C6 or Cι0 aryl or a 5 to 10-membered heterocylic group and p is an integer of from 1 to 4; X represents -OR14, -SR14, -NR14OR15,-NR14NR15R16' -CF3, -CF2H or CH2F wherein R14, R15 and R16 each independently represents hydrogen or - alkyl; and Y represents
Figure imgf000005_0001
wherein m is an integer from 1 to 4; n is an integer from 1 to 8; and R17 and R18 each independently represents hydrogen, unsubstituted or substituted - o alkyl, an unsaturated hydrocarbon chain of up to ten carbon atoms comprising one or more carbon-carbon double and/or triple bonds, or C10 aryl, a 5- to 10-membered heterocyclic group, - o alkoxy, Cλ-Cw thioalkoxy, hydroxyl, halo, cyano, nitro, amino, amido, (CrC10 alkyl)carbonyloxy, ( - o alkoxy )carbonyl, (Cj-C10 alkyl)carbonyl, ( -CJO alkyl)thiocarbonyl, (Cι-C10 alkyl)sulfonylamino, aminosulfonyl, C,-C10 alkylsulfinyl, C^-Cw alkylsulfonyl, or a saturated or unsaturated C3-C12 hydrocarbon chain interrupted by O, S, NR, CO, C(NR), N(R)SO2, SO2N(R), N(R)C(O)O, OC(O)N(R), N(R)C(O)N(R), OC(O), C(O)O, OSO2, SO2O or OC(O)O where R is as defined above and the saturated or unsaturated hydrocarbon chain is optionally substituted as defined above; and pharmaceutically acceptable salts thereof. As used herein, a C -Cw alkyl group or moiety is a linear or branched alkyl group or moiety containing from 1 to 10 carbon atoms, such as a - alkyl group or moiety or a C1-C4 alkyl group or moiety, for example methyl, ethyl, n-propyl, i-propyl, n-butyl, i-butyl and t-butyl. A C2-C10 alkenyl group is typically a C2- alkenyl group such as a C3-C6 alkenyl group, for example C3 or C4 alkenyl and in particular allyl. A C2-C10 alkynyl group is typically a C2-C6 alkynyl group such as a C3-C6 alkynyl group, for example C3 or C4 alkynyl and in particular propargyl. A -CJO alkoxy group or moiety is a linear or branched alkoxy group or moiety containing from 1 to 10 carbon atoms, such as a CrC6 alkoxy group or moiety or a C C4 alkoxy group or moiety. The -Q alkoxy group may be methoxy, ethoxy, n- propoxy, i-propoxy, n-butoxy or i-butoxy, preferably methoxy or ethoxy. As used herein, halo is typically chlorine, fluorine, bromine or iodine and is preferably chlorine or fluorine. A - ,, haloalkyl group is typically a said - ,, alkyl group, for example a - alkyl group or - alkyl group, substituted by one or more said halo atoms. Typically, it is substituted by 1, 2 or 3 said halogen atoms. Preferred haloalkyl groups include perhaloalkyl groups such as -CX3 wherein X is a said halogen atom. Particularly preferred haloalkyl groups are -CF3 and -CC13. A substituted - o alkyl group is typically a said - o alkyl group, for example a - alkyl group or a -C4 alkyl group, substituted by one or more, for example from one to three, atoms o other groups such as hydroxy, halo, amino, CrC4 alkoxy, - alkylthio, - alkylamino and di(Cj-C4 alkyl)amino. The substituted -CJO alkyl group may be a said C C10 haloalkyl group. Other suitable substituted - C10 alkyl groups include - Q hydroxyalkyl. An unsaturated hydrocarbon chain of up to ten carbon atoms comprising one or more carbon-carbon double or triple bonds is typically a said C2-C10 alkenyl or C2-C10 alkynyl group. A C6 or C10 aryl group or moiety is typically a phenyl or naphthyl group or moiety. The group or moiety may be substituted by one or more, for example from one to three, atoms or groups such as hydroxy, halo, cyano, amido, nitro, amino, CrC4 alkyl, CrC4 alkoxy, CrC4 alkylthio, CrC4 alkylamino and di(C C4 alkyl)amino. A 5- to 10-membered heterocyclic group may be a heteroaryl group. It may therefore be a 5- to 10-membered aromatic, i.e. fully unsaturated, ring such as a 5- or 6- membered ring, containing at least one heteroatom, for example one, two, three or four heteroatoms, selected from O, S and N. Examples include pyridyl, pyrazinyl, pyrimidinyl, pyridazinyl, furanyl, thienyl, imidazolyl, pyrazolidinyl, pyrrolyl, oxadiazolyl, oxazyl, isoxazyl, thiadiazolyl, thiazolyl, 1,2,3-triazolyl, 1,2,4-triazolyl, tetrazolyl and pyrazolyl groups. Alternatively, the 5- to 10-membered heterocyclic group is a non-aromatic, i.e. saturated or partially unsaturated, C5-C10 carbocyclic ring in which one or more, for example one, two, three or four, of the carbon atoms is replaced by a heteroatom selected from O, S and N. Examples of suitable such heterocyclyl groups include piperidinyl, piperazinyl, moφholinyl, pyrrolidinyl, tetrahydrofuranyl, imidazolidinyl, thiazolidinyl, 1,4-dioxanyl and 1,3-dioxolanyl. The invention also provides the use of a compound of formula (I) for the manufacture of a medicament for use in treating a disorder mediated by histone deacetylase. In this embodiment, preferably one or two of R1, R2 and R3 is/are hydrogen and the other one or two of R1, R2 and R3 is/are hydrogen. Preferably R1, R2 and R3 are each independently selected from hydrogen, - alkyl, Cα-C6 alkoxy, amino, - alkylamino, di(CrC6 alkyl)amino, halo, - haloalkyl, (Cα-C6 alkoxy )carbonyl or - alkyl substituted by amino, CrC6 alkoxy, CrC6 alkylamino or di (Cj- alkyl)amino. R1, R2 and R3 may therefore be selected from hydrogen, - alkyl, - alkoxy, amino, -Q alkylamino, d^ -Q alkyl)amino, chloro, fluoro, C,-C4 alkyl substituted by one, two or three chlorine or fluorine atoms, ( -Q alkoxy)carbonyl or -C4 alkyl substituted by amino, Cx or C2 alkoxy, or alkylamino or di( or C2 alkyl)amino. R1, R2 and R3 are most preferably selected from hydrogen, methyl, ethyl, methoxy, ethoxy, dimethylamino, chloro, fluoro, trifluoromethyl, difluoromethyl, fluoromethyl, methoxymethyl, ethoxymethyl, aminomethyl, methylaminomethyl or dimethylaminomethyl. Generally, one or two of R1, R2and R3 is hydrogen. The others or other of R1, R2 and R3 may be located at any position on the benzene ring. When two of R1, R2 and R3 are hydrogen, the other is preferably located at the 4-position on the benzene ring. Preferably R4 and R5 are each independently selected from hydrogen, - alkyl, C,-C6 alkoxy, halo, CrC6 haloalkyl or C C6 alkyl substituted by amino, C,-C6 alkoxy, - alkylamino or di(C C6 alkyl)amino. R4 and R5 may therefore be selected from hydrogen, C C4 alkyl, C,-C4 alkoxy, chloro, fluoro, C,-C4 alkyl substituted by one, two or three chlorine or fluorine atoms, or - alkyl substituted by amino, Cλ or C2 alkoxy, C or alkylamino or di( or alkyl)amino. R4 and R5 are most preferably selected from hydrogen, methyl, ethyl, methoxy, ethoxy, chloro, fluoro, trifluoromethyl, difluoromethyl, fluoromethyl, methoxymethyl, ethoxymethyl, aminomethyl, methylaminomethyl or dimethylaminomethyl. Generally, one or both of R4 and R5 is hydrogen. When one R4 and R5 is hydrogen, the other may be located at any position on the benzene ring. Preferably, R6 represents hydrogen, amino, - alkylamino, di(C C6 alkyl)amino, halo such as chlorine or fluorine, CrC6 alkyl, - alkoxy, - haloalkyl or - alkyl substituted by amino, C C6 alkoxy, - alkylamino or di(C C6 alkyl)amino. R6 may therefore be selected from hydrogen, halo such as chlorine or fluorine, - alkyl, - alkoxy, -Q alkyl substituted by one, two or three halo atoms, or -Q alkyl substituted by amino, or C2 alkoxy, or C2 alkylamino or di( or C2 alkyl)amino. R6 may therefore be hydrogen, methyl, ethyl, methoxy, ethoxy, fluoro, trifluoromethyl, difluoromethyl, fluoromethyl, dimethylamino, methoxymethyl, ethoxymethyl, aminomethyl, methylaminomethyl or dimethylaminomethyl. Most preferably, R6 is - alkyl or - alkyl and especially methyl. When the symbol ^^ represents a single bond, R7 and R8 preferably each independently represent hydrogen, halo such as chloro or fluoro, hydroxy, - alkoxy, CrC4 alkylthio, amino, - alkylamino or di(Cι-C4 alkyl)amino. When the symbol ™ represents a double bond, R7 is preferably hydrogen, halo, - o alkyl, C6 or C10 aryl, pyrazinyl, pyrimidinyl, pyridazinyl, furanyl, thienyl, imidazolyl, pyrazolidinyl, oxadiazolyl, oxazyl, isoxazyl, thiadiazolyl, thiazolyl, 1,2,3-triazolyl, tetrazolyl, pyrazolyl, OR9, SR9 or NR9R10 wherein R9 and R10 are as defined above.
More preferably R7 represents hydrogen, chloro, fluoro, - alkoxy, - alkoxy, C C4 alkylthio, amino, Ct-C4 alkylamino or d^ - alkyl)amino. Typically, R7 and if present R8 each represent hydrogen. Preferred W groups include single bond, -C(Rπ)=N-, -N=C(Rn)-, -C(RU)(R12)- NR13-, -NR13-C(Rn)(R12)-, -CO-NR11-, -NRπ-CO-, -SO2-NRu-, -NRu-SO2-,
-C(Ru)(R12)-O-, -O-C(Rn)(R12)-, -C(Rπ)(R12)-S-, -S-C(RU)(R12)-, -CO-, -NR11-, -SO-, -SO2-, S or -[C(Rn)R12]p- wherein Ru, R12 and R13 each independently represents hydrogen, Cx-C6 alkyl, C6 or C10 aryl or a 5 to 10-membered heterocylic group and p is an integer of from 1 to 4. More preferably W is -C(Rπ)=N-, -N=C(Rn)-, -C(RU)(R12)- NR13-, -NR13-C(Rn)(R12)-, -CO-NR11-, -NRn-CO-, -SO2-NRπ-, -NRu-SO2-,
-C(Ru)(R12)-O-, -O-C(Rπ)(R12)-, -C(Rn)(R12)-S-, -S-C(Rn)(R12)-, -CO-, -NR11-, -SO-, -SO2-, S or -[C(R")R12]p- wherein R11, R12 and R13 are as defined above. Preferably, R", R12 and R13 are independently selected from hydrogen, methyl or ethyl. Consequently, W preferably represents a single bond, -CH=N-, -N=CH-, -CONH-, -NHCO-, -SO2NH-, NHSO2-, -OCH2-, -CH2O-, -CH2S- or -SCH2-, or the equivalent groups where one or more of the hydrogen atoms are replaced with methyl or ethyl. Most preferably R11, R12 and R13 are hydrogen. Particularly preferred are compounds of formula (I) in which W represents -CH=N-, -CONH-, -SO2NH-, -CH2O or -CH2S-. The double bonds are typically in the trans-configuration. Preferably X represents -CF3, -OR5 or -NHOR5 wherein R5 is hydrogen or - C6 alkyl such as methyl or ethyl. Preferably Y represents:
Figure imgf000009_0001
wherein m is 1, 2 or 3; n is 1, 2 or 3; and R18 is hydrogen, - alkyl or 0,- alkoxy. A preferred class of compounds according to the invention have the formula (II):
Figure imgf000009_0002
wherein R1, R2, R6, R18, W and X are as defined above and m is 1, 2 or 3; and pharmaceutically acceptable salts thereof. More especially, R1 and R2 each independently represent hydrogen, - alkyl, Cx-C6 alkoxy, halo or (C,-C6 alkoxy )carbonyl; R6 and R18 are each independently selected from hydrogen, - alkyl or Cj-C4 alkoxy; W represents a single bond, -CH=N-, -N=CH-, -CONH-, -NHCO-, -SO2NH-, -NHSO2-, -OCH2-, -CH2O-, -CH2S- or -SCH2-; and X represents -NHOH, -CF3 or -OR14 wherein R14 is hydrogen or -C4 alkyl. More preferred are compounds having the formula (III):
Figure imgf000010_0001
wherein R1 represents hydrogen, C C4 alkyl, C C4 alkoxy or halo; R6 and R18 are each independently selected from hydrogen, Ct-C4 alkyl or -Q alkoxy; X represents -OR14 or -NHOH wherein R14 is hydrogen or CrC4 alkyl; Z represents C or SO; and m is 1, 2 or 3; and pharmaceutically acceptable salts thereof. More preferably R1 represents
C C4 alkyl, -Q alkoxy or halo. Most preferably R6 is hydrogen and especially methyl. Preferred compounds of formula (III) are: (2E,4E)-5-(4-(4-chlorobenzenesulfonylamino)phenyl)penta-2,4-dienoic acid ethyl ester;
(2E,4E)-5 -(4-(4-methoxybenzenesulf onylamino)pheny l)penta-2,4-dienoic acid ethyl ester;
(2E,4E)-5-(4-(4-chlorobenzenesulfonylamino)phenyl)-4-methylpenta-2,4-dienoic acid ethyl ester; (2£,4E)-5-(4-(4-chlorobenzenesulfonylamino)phenyl)-4-methylpenta-2,4-dienoic acid;
(2E,4E)-5-(4-(4-methoxybenzenesulfonylamino)phenyl)-4-methylpenta-2,4-dienoic acid ethyl ester;
(2E,4E)-5-(4-(4-methoxybenzenesulfonylamino)phenyl)-2,4-dimethylpenta-2,4-dienoic acid ethyl ester; (2E,4E)-5-(4-(4-methoxybenzenesulfonylamino)phenyl)-4-methylpenta-2,4-dienoic acid;
(2E,4E)-5-(4-(4-chlorobenzenesulfonylamino)phenyl)-4-methylpenta-2,4-dienoic acid ethyl ester; (2E,4E)-5-(4-methoxybenzenesulfonylaminophenyl)-4-methylpenta-2,4-dienoic acid hydroxamide;
(2£,4E,6E)-7-(4-(4-methoxybenzenesulfonylamino)phenyl)-6-methylhepta-2,4,6- trienoic acid ethyl ester; and
(2E,4E,6E)-7-(4-(4-chlorobenzoylamino)phenyl)hepta-2,4,6-trienoic acid methyl ester. Another preferred class of compounds according to the invention have the formula (IV):
Figure imgf000011_0001
wherein R1, R2, R6, R18, W and X are as defined above, the symbol represents a single bond or a double bond and n is 1, 2 or 3; and pharmaceutically acceptable salts thereof. More especially, R1 and R2 each independently represent hydrogen, - alkyl, - alkoxy, halo or (C C6 alkoxy)carbonyl; R6 and R18 are each independently selected from hydrogen, CrC4 alkyl or - alkoxy; W represents a single bond, - CH=N-, -N=CH-, -CONH-, -NHCO-, -SO2NH-, -NHSO2-. -OCH2-, -CH2O-, -CH2S- or -SCH2-; and X represents -NHOH, -CF3 or -OR14 wherein R14 is hydrogen or C C4 alkyl. More preferred are compounds which have the formula (V):
Figure imgf000011_0002
wherein the symbol represents a single bond or a double bond; R1 represents hydrogen, C C4 alkyl, - alkoxy or halo; R6 and R18 are each independently selected from hydrogen, - alkyl or CrC4 alkoxy; X represents -OR14 or -NHOH wherein R14 is hydrogen or C,-C4 alkyl; Z represents C or SO; and n is 1, 2 or 3; and pharmaceutically acceptable salts thereof. More preferably R1 represents hydrogen, Cj-C4 alkyl, - alkoxy or halo. Preferably R6 is hydrogen and especially methyl. Preferred compounds of formula (V) are: 5-[4-(4-methoxybenzenesulfonylamino)phenyl]-4-methylpentanoic acid ethyl ester; ethyl 5 - [4-(4-chlorobenzoy lamino)pheny 1] -4-me thy lpentanoate ; ethyl 5 - [4-(4-chlorobenzoy lamino)phenyl] -4-methy lpentanoate ; 5-[4-(4-chlorobenzenesulfonylamino)phenyl]-4-methylpentanoic acid hydroxyamide; and 5-[4-(4-chlorobenzoylamino)phenyl]-4-methylpentanoic acid hydroxyamide. The invention also provides compounds of formula (VI):
Figure imgf000012_0001
wherein the symbol ™ represents a single bond or a double bond or the symbol zz z, 6 and R8 together represent cyclopropyl; R1 to R5 each independently represent hydrogen, C2-C10 alkyl, C2-C10 alkenyl, C2-C10 alkynyl, - o alkoxy, - o thioalkoxy, hydroxyl, - o hydroxyalkyl, halo, Cj-C10 haloalkyl, amino, -CJQ alkylamino,
Figure imgf000012_0002
alkyl)amino, amido, nitro, cyano, (Cι-C10 alkyl)carbonyloxy, ( - o alkoxy )carbonyl, ( - o alkyl)carbonyl, ( -CIQ alkyl)thiocarbonyl, (Cj-C10 alkyl)sulfonylamino, aminosulfonyl, ( - o alkyl)sulfinyl, (CJ-CK, alkyl)sulfonyl or -CK, alkyl substituted by amino, -Cn, alkoxy, Cj- ,, alkylamino or di(Cι-C10 alkyl)amino; - R6 represents hydrogen, CrC10 alkyl, substituted - Q alkyl, an unsaturated hydrocarbon chain of up to ten carbon atoms comprising one or more carbon-carbon double or triple bond, C6 or C10 aryl, a 5- to 10-membered heterocyclic group, - Q alkoxy, -CJO thioalkoxy, hydroxyl, halo, cyano, nitro, amino, - o alkylamino, di(Cι-C10 alkyl)amino, amido, (Cj- o alkyl)carbonyloxy, (CJ-CJO alkoxy )carbonyl, (C^CJO alkyl)carbonyl, (CrC10 alkyl)thiocarbonyl, (Cj-C10 alkyl)sulfonylamino, aminosulfonyl, (C -Cw alkyl)sulfinyl, (C -Cw alkyl)sulfonyl, a saturated or unsaturated C3-C12 hydrocarbon chain interrupted by O, S, NR, CO, C=NR, N(R)SO2, SO2N(R), N(R)C(O)O, OC(O)N(R), N(R)C(O)N(R), OC(O), C(O)O, OSO2, SO2O or OC(O)O where: (a) R independently represents hydrogen, CrC10 alkyl, C2-C10 alkenyl, C2-C10 alkynyl, - ,, alkoxy, -CJQ hydroxyalkyl, hydroxyl or C C10 haloalkyl, and (b) the saturated or unsaturated hydrocarbon chain is optionally substituted with Cj- o alkyl, C o alkenyl, C2-C]0 alkynyl, CrC10 alkoxy, hydroxyl, Cχ-Cl0 hydroxyalkyl, halo, -C^ haloalkyl, amino, (CrC10 alkyl)carbonyloxy, ( - o alkoxy )carbonyl, (Cj-C10 alkyl)carbonyl, (Cj- o alkyl)sulfonylamino, aminosulfonyl or -CK, alkylsulfonyl, when the symbol ™ represents a single bond, R7 and R8 each independently represents hydrogen, halo, -C,,, alkyl, C6 or C10 aryl, a 5- to 10-membered heterocyclic group, OR9, SR9 or NR9R10 wherein R9 and R10 each independently represent hydrogen or - alkyl or one of R9 and R10 is H and the other is -CO(CrC6 alkyl). or R7 and R8 together represent =O, =CH2 or =CHR9 wherein R9 is as defined above; when the symbol ^^ represents a double bond, R7 represents hydrogen, halo, Cj- o alkyl, C6 or Cι0 aryl, a 5- to 10-membered heterocyclic group, OR9, SR9 or NR9R10 wherein R9 and R10 are as defined above and R8 is absent; - W represents a single bond, -C(Rπ)=N-, -N=C(RU)-, -C(RU)(R12)-NR13-,
-NR13-C(RU)(R12)-, -CO-NR11-, -NRπ-CO-, -SO2-NRπ-, -NRπ-SO2-, -C(Ru)(R12)-O-, -O-C(Rn)(R12)-, -C(Rn)(R12)-S-, -S-C(Rn)(R12)-, -CO-, -NR11-, -SO-, -SO2-, S or -[C(Rπ)R12]p- wherein R11, R12 and R13 each independently represents hydrogen, - alkyl, C6 or C10 aryl or a 5- to 10- membered heterocyclic group alkyl and p is an integer of from 1 to 4; X represents -OR14, -SR14, -NR14OR15,-NR14NR15R16, -CF3, -CF2H or CH2F wherein R14, R1S and R16 each independently represents hydrogen or CrC6 alkyl; and Y represents
Figure imgf000013_0001
wherein m is an integer from 1 to 4; n is an integer from 1 to 8; and R17 and R18 each independently represents hydrogen, unsubstituted or substituted - Q alkyl, an unsaturated hydrocarbon chain of up to ten carbon atoms comprising one or more carbon-carbon double and/or triple bonds, C6 or C10 aryl, a 5- to 10-membered heterocyclic group, - Q alkoxy, C C10 thioalkoxy, hydroxyl, halo, cyano, nitro, amino, amido, ( - ,, alkyl)carbonyloxy, ( - o alkoxy )carbonyl, ( - o alkyl)carbonyl, ( - o alkyl)thiocarbonyl, (CJ-CJQ alkyl)sulfonylamino, aminosulfonyl, Cj-Cjo alkylsulfinyl, - o alkylsulfonyl, or a saturated or unsaturated C3-C12 hydrocarbon chain interrupted by O, S, NR, CO, C(NR), N(R)SO2, SO2N(R), N(R)C(O)O, OC(O)N(R), N(R)C(O)N(R), OC(O), C(O)O, OSO2, SO2O or OC(O)O where R is as defined above and the saturated or unsaturated hydrocarbon chain is optionally substituted as defined above; and pharmaceutically acceptable salts thereof. Preferred features of the compounds of formula (VI) are as previously defined for the compounds of formula (I). In particular, it is preferred that R1, R2 and R3 are selected from hydrogen, - alkyl, - alkoxy, amino, - alkylamino, di(Cι-C6 alkyl)amino, halo, C C6 haloalkyl, (Ct- alkoxy )carbonyl or - alkyl substituted by amino, - alkoxy, - alkylamino or di(Cj-C6 alkyl) amino. More preferably one or two of R1, R2 and R3 is hydrogen. Preferred compounds of formula (VI) are those of formula (VII):
Figure imgf000014_0001
wherein R1 represents hydrogen, - alkyl, CrC4 alkoxy or halo; R6 and R18 are each independently selected from hydrogen, - alkyl or C:-C4 alkoxy; X represents -OR14 or -NHOH wherein R14 is hydrogen or - alkyl; Z represents C or SO; and m is 1, 2 or 3; and pharmaceutically acceptable salts thereof. More preferably R6 is methyl. The preferred compounds of formula (VI) include:
(2E,4E)-5-(4-benzenesulfonylaminophenyl)-4-methylpenta-2,4-dienoic acid ethyl ester; (2E,4E)-5-(4-benzenesulfonylaminophenyl)-4-methylpenta-2,4-dienoic acid; and (2E,4E)-5-(4-benzenesulfonylaminophenyl)-4-methylpenta-2,4-dienoic acid hydroxamide. The compounds of formula (VI) can also be used in the manufacture of medicaments for use in the treatment of a disorder mediated by histone deacetylase. As used herein, a pharmaceutically acceptable salt is a salt with a pharmaceutically acceptable acid or base. Pharmaceutically acceptable acids include both inorganic acids such as hydrochloric, sulphuric, phosphoric, diphosphoric, hydrobromic or nitric acid and organic acids such as citric, fumaric, maleic, malic, ascorbic, succinic, tartaric, benzoic, acetic, methanesulphonic, ethanesulphonic, benzenesulphonic or p-toluenesulphonic acid. Pharmaceutically acceptable bases include alkali metal (e.g. sodium or potassium), alkali earth metal (e.g. calcium or magnesium), zinc and iron hydroxides and organic bases such as - alkyl amines, aralkyl amines or heterocyclic amines. An example of a primary amine salt can be the cyclohexylammonium salt, a suitable secondary amine salt may be the piperidine salt and a tertiary amine salt may be the triethylamine salt. Tautomers of compounds of formula (I) and (VI) also form part of the invention. Also, the compounds of the invention can contain one or more chiral centre. For the avoidance of doubt, the chemical structures depicted herein are intended to embrace all stereoisomers of the compounds shown, including racemic and non-racemic mixtures and pure enantiomers and/or diastereoisomers. Preferred compounds of the invention are optically active isomers. Thus, for example, preferred compounds of formula (I) containing only one chiral centre include an R enantiomer in substantially pure form, an S enantiomer in substantially pure form and enantiomeric mixtures which contain an excess of the R enantiomer or an excess of the S enantiomer. The compounds of formula (I) and (VI) and their salts may be prepared by adaptation of conventional procedures. For example, Scheme 1 below illustrates one way in which compounds of the invention may be prepared. The compounds thus prepared can then be converted as desired into pharmaceutically acceptable salts by conventional routes. In all Schemes and in the Examples, the benzene ring and the chain incoφorating the group Y which are either side of the symbol shown in formula (I) are in the trans configuration with respect to each other when the symbol represents a carbon-carbon double bond. Compounds in which the symbol represents a carbon-carbon double bond therefore are isolated as the trα/is-isomer, trαns-trα/w-isomer, trans-trans-trans-isomei, etc. Scheme 1
Figure imgf000016_0001
In the starting material (A), R1 to R5, R7 and W are as defined above. Typically R7 represents hydrogen. Compound (A) can be obtained by a variety of methods, depending on the nature of W. Thus: When W represents a single bond, one of the following will typically be used: Suzuki coupling (of a boronic acid or ester with an aryl halide or triflate), Ullman coupling (copper-catalysed coupling of aryl halides or related compounds), Gomberg reaction (arylation of diazonium salts) or other metal-catalysed aryl-aryl coupling of aryl halides, triflates or related compounds. When W represents an imine of formula -N=C(RU)- or -C(Rπ)=N- , compound (A) may be prepared from an arylamine of formula Ar-NHR11 and an aromatic aldehyde or ketone. When Rn is OH, W in the resulting compound of formula (A) will exist as the amide group of formula -N(Rn)C(O)- or -C(O)NRn-, rather than as the corresponding hydroxyimine, due to keto-enol tautomerism. When W represents -C(RU)(R12)-NR13- or -NR13-C(Rn)(R12)-, compound (A) may be prepared by reduction of the corresponding imine or amide using hydrogen and a catalyst or a metal hydride system, or by addition of an organometallic reagent to the imine preceded and followed by suitable protection and deprotection respectively of the ketone group present in (A). A preferred method of preparing such compounds (A) is by reaction of a suitable amine with a suitable aldehyde or ketone under reductive conditions, using reagents such as sodium cyanoborohydride or sodium triacetoxyborohydride. When W represents -NRu-CO- or -CO-NR11-, compound (A) may be prepared by reaction of ArCOQ (wherein Q is a leaving group, typically CI) with an arylamine in the presence of a base. When W represents CO, Friedel-Crafts arylation or metal-catalysed coupling involving ArCOQ compounds are preferred (wherein Q is a leaving group, typically CI). - When W represents -SO2-NRπ- or -RπN-SO2-, compound (A) may be prepared by sulfonylation (typically using ArSO2Cl) of an arylamine in the presence of a base. When W represents -[C(Rn)R12]p-, compound (A) may be prepared by a Friedel-Crafts procedure (especially when R11 and R12 are both hydrogen) followed by reduction (hydrogen and a catalyst) or N2H4-KOH (Wolf-Kishner) or related procedures. For an alkyl chain in W, a Wittig reaction followed by reduction, typically using hydrogen and a catalyst, is the preferred method. When W represents -NR11-, compound (A) may be prepared by catalytic amination of an aryl halide, typically with a palladium-based catalyst, although displacement of an aryl halide or triflate by an arylamine or metal salt may also be used. When W represents O, compound (A) may be prepared by etherification of an aryl halide or triflate with a phenol, or more preferably its metal salt, typically in the presence of a metal catalyst such as a copper, palladium or nickel derivative or the metal itself. - When W represents S, compound (A) may be prepared by displacement of an aryl halide or triflate, with a thiophenol. This may be in the presence of a metal catalyst or more preferably a metal salt of the thiophenol. Alternatively, the metal salt of the thiophenol may also be reacted with a diazonium salt. When W represents SO, compound (A) may be prepared by oxidation of the corresponding compound (A) where W represents S with hydrogen peroxide or NaIO4. When W represents SO2, compound (A) may be prepared by oxidation with H2O2 or NaIO4 of the corresponding compound (A) where W represents S with a peracid, typically /n-chloroperoxybenzoic acid, or by oxidation of the corresponding compound (A) where W represents SO with NaIO4. When W represents -O-C(Rn)(R12)- or -C(Ru)(R12)-O-, compound (A) may be prepared by a Williamson ether-type synthesis involving a phenol or more preferably its metal salt with an alkyl halide, triflate or related displaceable group, i.e. ArC(Ru)(R12)Q, wherein Q is a leaving group, typically halogen or triflate, preferably Br. When W represents -S-C(RU)(R12)- or -C(RU)(R12)-S-, compound (A) may be prepared by reaction of a thiophenol or more preferably its metal salt with an alkyl halide [ArC(Rn)(R12)Q (wherein Q is a leaving group, typically halogen or triflate, preferably Br]. > In Scheme 1, compound (A) is reacted with either an aldehyde R6CH2CR17O, typically in the presence of an acidic or basic catalyst, or a Wittig or related reagent such as PH3P=C(R6)CR17O. R17 is preferably H, CH3 or CH2CH3. More preferably, R17 is H or Me. Typically, R17 is H. The aromatic ring and the -C(O)R17 moiety are typically trans with respect to each other in the resulting compound (B). Compound (B) is then reacted with compound (C) to form compound (D). (C) is typically either a Wittig or related reagent of type Ph3P=C(R18)(CR17=CR18)qCO2R wherein q = 0, 1, 2 or 3 and R17 and R18 are each independently as defined above. Preferably R17 and R18 are independently selected from H, CH3 and CH2CH3. More preferably, they are H or CH3 and typically both denote H. Within any (CR17=CR18) moiety, it is preferred that one of R17 and R18 is H. The other may be CH3 but typically both R17 and R18 are H. The group R in compound (C) is hydrogen or - o alkyl. Preferably R is H, CH3 or CH2CH3. When R is H, reaction of the acid group to form the corresponding methyl or ethyl ester is typically carried out before reaction with (B). A reduction is carried out to convert compound (D) to compound (E). One, two or more of the carbon-carbon double bonds in the chain of compound (D), for example all such bonds, are thus reduced to single bonds. Where saturation of all such double bonds is desired, excess hydrogen and a metal catalyst such as Pd or palladium-on-carbon may be used. Where partial saturation is desired, suitable reagents are (a) Mg/ROH wherein R is CrC10 alkyl or (b) R3SiH/H+. The partial reduction can be carried out selectively. Thus, where the double bond adjacent to the -COOR group of compound (D) is to be hydrogenated, a magnesium or related metal, together with an alcohol, typically methanol or ethanol, can be used. Where at least one of the double bonds that is not adjacent to the -COOR group of compound (D) is to be hydrogenated, a trialkylsilane or related agent, together with an acid, can be used. The desired degree of saturation may alternatively be incoφorated from the Wittig reagent as appropriate. Where the compound (G) is desired, compound (D) is reacted with reagent (F). In an analogous fashion, compound (H) is formed by reaction of compound (E) with reagent (F). The nature of reagent (F) is dependent on the nature of the desired group X of compound (G) or (H). Suitable methods for forming the group X as are follows. For X = NHOH, this is typically achieved by reaction of the methyl or ethyl ester of (D) or (E) with aqueous hydroxylamine. A compound (G) or (H) in which X = OH can be obtained by alkaline hydrolysis of an ester (D) or (E) with sodium or potassium hydroxide (or other alkali or related metal hydroxide) followed by acidification. When it is wished to form a compound (G) or (H) in which X = CF3, a carboxylic acid (D) or (E) can be reacted with (i) oxalyl chloride, (ii) (CF3CO)2O and pyridine, and then (iii) water. A compound (G) or (H) in which X= CH2F is typically obtained by alkaline hydrolysis of an ester (D) or (E) with sodium or potassium hydroxide (or other alkali or related metal hydroxide) followed by acidification and treatment of the carboxylic acid so obtained with (i) ClCO2i-Bu and N-methyl-moφholine, (ii) CH2N2 and Et3N and (iii) HBr and AcOH to give the corresponding bromomethyl ketone. That ketone is then converted into the corresponding fluoromethyl ketone using KF and 18-crown-6 in acetonitrile. When it is wished to form a compound (G) or (H) in which X = CHF2, this is typically achieved by alkaline hydrolysis of an ester (D) or (E) with sodium or potassium hydroxide (or other alkali or related metal hydroxide) followed by acidification and treatment of the carboxylic acid so obtained with EtOC(O)CHF2. A compound (G) or (H) in which X = SR14 can be prepared by reaction of an ester (D) or (E) with a metal salt of a thiol having a SR14 group. A compound (G) or (H) in which X = NR14OR15 is obtainable by reaction of an ester (D) or (E) with HNR14OR15. A compound (G) or (H) in which X = -NR14NR15R16 can be formed by reaction of an ester (D) or (E) with NHR14NR15R16. Alternatively such a compound can be obtained by reaction of the corresponding carboxylic acid (obtainable from the ester as described above for X=OH) with an activating reagent such as iso-butyl chloroformate or DCC, followed by the addition of HNR14NR15R16. For compounds containing a cyclopropyl ring, the preferred preparation method includes reduction of (B) to the corresponding alcohol using hydrogen and a metal catalyst or using a metal-hydride system. Cyclopropanation is then effected with an iodoorganozinc reagent. Where an enantio-enriched product is required, a chiral co-reagent such as a dialkyl tartrate or dialkyl tartramide is added, and subsequent oxidation (back to the aldehyde) to form the derivative of (B) that has the structure ArCR7(CH2)CR6CHO. This can then be reacted further, as described above. The cyclopropyl ring may alternatively be introduced by cyclopropanation of more unsaturated systems, such as compound (D), by a variety of reagents including CH2I2 with a zinc-copper couple. In a variation of Scheme 1, compound (A) or compound (B) may be reacted with a Wittig reagent of the type Ph3P=C(R')(CH2)nCOOR where R' is typically H, R is typically CH3 or CH2CH3 and n is as defined above. In this way, compound (E) and like saturated ester derivatives can be prepared. When W represents -CONR11- or -S(O)NRu-, the Ar group in Ar-W-C6H2(R4)(R5)CHO can attached later in another variation of Scheme 1. Thus, a 7-nitrobenzaldehyde replaces compound (A) and is reacted with R6CH2CR17O or Ph3P=C(R6)CR17O. The product may then be reacted with compound (C) to give a nitro ester that is reduced, preferably with Fe and aqueous ammonia, to convert the -NO2 group into an -NH2 group, that amine then being reacted with an acid chloride or sulfonyl chloride to give a compound of formula (D) or a compound of formula (E). When R7 and R8 together represent =O (i.e. a ketone group) in a compound of formula (I), a variant procedure typically involves the addition of an organometallic reagent such as XZn(CH2)nCO2R wherein X is halogen, typically Br or I, to compound (A) where R7=H to give the corresponding alcohol. That alcohol is then oxidised (e.g. with MnO2 or chromium (IV) reagent or oxygen or air and a metal catalyst, or other oxidising reagent) to give a compound of formula (I) (typically where Y is a saturated chain). This ketone of formula (I) may be converted into the alkene using Ph3P=CHR9 or a related reagent. Schemes 2 to 5 shown below illustrate syntheses for producing certain aspects of the invention, for example when R1, R2, R3, R4 and R5 are selected from H, - o alkyl and - o alkoxy. Where they appear in Schemes 2 to 5, R17 and R18 are independently defined as above. Preferably, R17 and R18 are independently H, CH3 or CH2CH3. Typically, they are H. Within any (CR17=CR18) moiety, it is preferred that one or R17 and R18 is H and the other is CH3. More preferably, both are H. Unless stated otherwise, other substituents are defined as above.
Scheme 2
Figure imgf000021_0001
In Scheme 2, a route is shown to carboxylic acids (8) and their ester derivatives, and also their hydroxamic acid derivatives (9). An aldol or related condensation, typically initiated by base though acid and other catalysts or reagents are feasible, is followed by dehydration to give the unsaturated aldehydes (4) which undergo a Wittig or related process to afford the dienic carbonyl compounds (6). Here, a related process includes Horner-Wadsworth-Emmons or other modifications, as well as compounds other than (5) (e.g. simple or complex enolates, or enolate equivalents of an appropriate carbonyl compound, such as enamino esters, enamino ketones or metal complexes derived from α-halo esters and related compounds). The group R of (5) is typically hydroxy or - o alkoxy such as, in particular, -OCH3 or -OCH2CH3. Reduction of the nitro group of (6) to the amino derivative (7) can be carried out by many reagents, but iron and aqueous ammonia mixtures are shown to be particularly effective and convenient. N-Sulfonylation and N-acylation of (7) affords the corresponding respective sulfonamides and esters (8). Such sulfonylations are typically carried out with an arenesulfonyl chloride and base, and the corresponding acylations with an acid chloride and a base. Many bases may possibly be employed. However, use of pyridine or a related base or of an aliphatic tertiary amine is preferred. In some cases, alkali metal hydroxides may suffice.
Scheme 3
Figure imgf000022_0001
Scheme 3 shows a route to carboxylic acids (17) and their ester derivatives, and also to the hydroxamic acid derivatives (18). The unsaturated aldehydes (4) undergo a Wittig or related process to afford the trienic carbonyl compounds (17). Here, a related process includes Horner-Wadsworth-Emmons or other modifications, as well as use of compounds other than phosphorus or arsenic ylid (10) (e.g. simple or complex enolates, or enolate equivalents of an appropriate carbonyl compound, such as enamino esters, enamino ketones or metal complexes derived from α-halo esters and related compounds). The trienic carbonyl compound (17) so obtained can then be converted into other derivatives, but preferably the hydroxamic acids (18). The phosphorus or arsenic ylid may have a variety of substituents (e.g. trialkyl or unsymmetrical alkyl and/or aryl substituents) and is not intended to be limited to Ph3P- or Ph3As- as shown in the Scheme. The group R of the phosphorus or arsenic ylid (10) and of compound (17) is hydroxy or CrC10 alkoxy. Preferably, R is C C10 alkoxy. Typically, it is -OCH3 or -OCH2CH3. When it is desired that R should denote hydroxy in compound (17), this may be achieved by hydrolysis of the corresponding ester compound (17) in which R is - o alkoxy. If R denotes hydroxy in compound (17), the reaction of (17) to (18) may require activation of the acid group of compound (17).
Scheme 4
Figure imgf000023_0001
Figure imgf000023_0002
In Scheme 4, a route is shown to carboxylic acids and their ester derivatives (28) and also their hydroxamic acid derivatives (29) via a Horner-Wadsworth-Emmons reaction. The phosphorus ylid reagent provides the group Y shown in formula (I). In this case, the ylid provides a -CR17=CR18-CR17=CR18- group. The group R in Scheme 4 is generally hydroxy or Cj- o alkoxy.
Figure imgf000024_0001
x o £ o z
Figure imgf000024_0002
U-) In Scheme 5 are shown routes to carboxylic acids (22) and (25) and their ester derivatives, and also their hydroxamic acid derivatives (23) and (26). A Wittig or related process affords the carbonyl compounds (22). Here, a related process includes Horner-Wadsworth-Emmons or other modifications, and also the analogous arsenic ylids, as well as compounds other than the phosphorus or arsenic ylid (21) (e.g. simple or complex enolates, or enolate equivalents of an appropriate carbonyl compound, such as enamino esters, enamino ketones or metal complexes derived from α-halo esters and related compounds). The carbonyl compound (22) so obtained can then be converted into other derivatives, but especially the hydroxamic acids (23). The phosphorus or arsenic ylid may have a variety of substituents (e.g. trialkyl or unsymmetrical alkyl and/or aryl substituents) and is not intended to be limited to Ph3P- or Ph3As- as shown in the Scheme. The carbonyl compounds (22) can also be reduced, typically with hydrogen and a catalyst(s) (e.g. Pd, Ni, Co etc.) or by hydrosilylation or hydride-acid systems and other processes to give the carboxylic acids (25) and their derivatives. In particular, the ester derivatives (22) and (25) can be converted, typically by aqueous hydroxylamine, but also by hydroxylamine derivatives with or without bases, respectively into the hydroxamic acids (23) and (26). The group R in compounds (21) and (24) is typically hydroxy or CrC10 alkoxy. Preferably, R is C,-C10 alkoxy and especially is -OCH3 or -OCH2CH3. When R=OH is desired in compound (22), this may be achieved by standard methods of hydrolysis of the corresponding ester compound (22) formed when R is - o alkoxy in compound (21). If R is OH, the reaction of (22) to (23) and (25) to (26) may require activation of the acid group of compound (22) or (25) respectively. Preferred reaction procedures are set out in Schemes 6 to 9 shown below. In
Schemes 6 to 9, the symbols shown in the formulae are as defined above unless otherwise indicated. Scheme 6
Figure imgf000026_0001
Figure imgf000026_0002
aOB
Figure imgf000026_0003
Figure imgf000026_0004
Figure imgf000026_0005
In Scheme 6 is shown a route to carboxylic acids (8) and their ester derivatives, and also their hydroxamic acid derivatives (9). An aldol or related condensation, typically initiated by base though acid and other catalysts or reagents being feasible, is followed by dehydration to give the unsaturated aldehydes (4) which undergo a Wittig or related process to afford the dienic carbonyl compounds (6). Here, a related process includes Horner-Wadsworth-Emmons or other modifications, as well as compounds other than (5) (e.g. simple or complex enolates, or enolate equivalents of an appropriate carbonyl compound, such as enamino esters, enamino ketones or metal complexes derived from α-halo esters and related compounds). The group R of reagent (5) is typically hydroxy or Cj-Cy, alkoxy such as, in particular, -OCH3 or - OCH2CH3. Reduction of the nitro group of (6) to the amino derivative (7) can be carried out be many reagents, but iron and aqueous ammonia mixtures are shown to be particularly effective and convenient. N-Sulfonylation and N-acylation of (7) affords the corresponding respective sulfonamides and esters (8) in which X is C or SO. Such sulfonylations are typically carried out with an arenesulfonyl chloride and base, and the corresponding acylations with an acid chloride and a base. Many bases may possibly be employed. However, use of pyridine or a related base or of an aliphatic tertiary amine is preferred. In some cases, alkali metal hydroxides may suffice.
Scheme 7
,N
Figure imgf000027_0001
Figure imgf000027_0002
Scheme 7 is similar to Scheme 6. Scheme 7 provides a route to carboxylic acids (17) and their ester derivatives, and also their hydroxamic acid derivatives (18). An aldol or related condensation, typically initiated by base but acid and other catalysts or reagents are feasible, is followed by dehydration to give the unsaturated aldehydes (4) which undergo a Wittig or related process to afford the dienic carbonyl compounds (6). Here, a related process includes Horner-Wadsworth-Emmons or other modifications, as well as use of compounds other than the phosphorus ylid (13) (e.g. simple or complex enolates, or enolate equivalents of an appropriate carbonyl compound, such as enamino esters, enamino ketones or metal complexes derived from α-halo esters and related compounds). The group R in the phosphorus ylid (13) is typically hydroxy or - Q alkoxy. Preferably, R is - o alkoxy and especially -OCH3 or - OCH2CH3. Reduction of the nitro group of compound (6) to the amino derivative (15) can be carried out be many reagents, but iron and aqueous ammonia mixtures are particularly effective and convenient. N-Sulfonylation and N-acylation of (15) affords the corresponding respective sulfonamides and esters (16) in which X is C or SO. Such sulfonylations are typically carried out with an arenesulfonyl chloride and base, and the corresponding acylations with an acid chloride and a base. Many bases may possibly be employed. However, use of pyridine or a related base or of an aliphatic tertiary amine is preferred although in some cases alkali metal hydroxides may suffice.
Scheme 8
Figure imgf000028_0001
Scheme 8 provides an alternative route to carboxylic acids (17) and their ester derivatives, and also their hydroxamic acid derivatives (18). The group X denotes C or SO. A Wittig or related process affords the trienic compounds (17). Here, a related process includes Homer- Wadsworth-Emmons or other modifications, and also use of analogous arsenic ylides as well as use of compounds other than (10) and (11) (e.g. simple or complex enolates, or enolate equivalents of an appropriate carbonyl compound, such as enamino esters, enamino ketones or metal complexes derived from α-halo esters and related compounds). The group R in reagents (10) and (11) is typically hydroxy or Cx-C 0 alkoxy and especially -OCH3 or - OCH2CH3. The trienic compound (17) can be converted into other compounds of formula (I) according to the invention but especially to the hydroxamic acids (18). The phosphorus or arsenic ylid (10) may have a variety of substituents (e.g. trialkyl or unsymmetrical alkyl and/or aryl substituents) and is not intended to be limited to Ph3P- or Ph3As- as shown. When it is desired that R should denote hydroxy in compound (17), this may be achieved by hydrolysis of the corresponding ester compound (17) in which R is -CK, alkoxy. If R denotes hydroxy in compound (17), the reaction of (17) to (18) may require activation of the acid group of compound (17). Optionally (see Scheme 8) a substituted or unsubstituted 4-nitrophenylpropenal can be reacted with (10) to give the corresponding 4-nitrophenylhepta-2,4,6-trienoic acid ester that is reduced to give the corresponding 4-amino compound that is then treated with an aromatic sulphonyl chloride or aromatic chloride to give the corresponding compound (17).
Figure imgf000030_0001
a X o o £ a z z
Figure imgf000030_0002
Figure imgf000030_0003
lO In Scheme 9 is shown routes to carboxylic acids (22) and (25) and their ester derivatives, and also their hydroxamic acid derivatives (23) and (26). A Wittig or related process affords the carbonyl compounds (22). Here, a related process includes Horner-Wadsworth-Emmons or other modifications, and also use of the analogous arsenic ylides as well as use of compounds other than (21) (e.g. simple or complex enolates, or enolate equivalents of an appropriate carbonyl compound, such as enamino esters, enamino ketones or metal complexes derived from α-halo esters and related compounds). The carbonyl compound (22) so obtained can then be converted into other derivatives, but especially the hydroxamic acids (23). The phosphorus or arsenic ylid may have a variety of substituents (e.g. trialkyl or unsymmetrical alkyl and/or aryl substituents) and is not intended to be limited to Ph3P- or Ph3As- as shown. The carbonyl compounds (22) can also be reduced, typically with hydrogen and a catalysts (e.g. Pd, Ni, Co, etc.) or by hydrosilylation or hydride-acid systems and other processes to give the carboxylic acids (25) and their derivatives. In particular, the ester derivatives (22) and (25) can be converted, typically by aqueous hydroxylamine, but also by hydroxylamine derivatives with or without bases, respectively into the hydroxamic acids (23) and (26). The group R in reagent (21) and in compound (24) is hydroxy or - o alkoxy. Preferably, R is Cι-C10 alkoxy and especially -OCH3 or - OCH2CH3. When it is desired that R should denote hydroxy in compound (17), this may be achieved by hydrolysis of the corresponding ester compound (17) in which R is Cj- o alkoxy. When R=OH is desired in compound (22) or (25), this may be preferably achieved by standard methods of ester hydrolysis of the corresponding ester compound (22) or (25) formed in the preferred aspect when R is Cj- o alkoxy. If R is OH, the reaction of (22) to (23) and of (25) to (26) may require activation of the acid group.
Scheme 10
Figure imgf000032_0001
Scheme 10 shows how an intermediate (7) bearing an amino group can be converted into compounds of the invention where W is -C(RU)(R12)-NR13-. In this particular scheme R11 and R12 are both hydrogen, although other reactants could readily be chosen in order to prepare compounds having other R11 and R12 groups. The reactant R10CHO is shown in parentheses because it need not be used if it is desired that R10 be hydrogen. A preferred example obtained using procedures in Scheme 10 include the reaction of an amine (7) with a benzaldehyde in the presence of formaldehyde and sodium cyanoborohydride to give the corresponding tertiary amine whose ester group is subsequently converted into the corresponding hydroxamic acid. In the Schemes, hydroxylamine is shown as the means of obtaining a hydroxamic acid. While this is the preferred reagent, especially in the form of an aqueous 50% solution, and in its action upon methyl or ethyl esters that are to undergo hydroxamation, this protocol is not intended to exclude other variants. For example, the use of a hydroxylamine salt, especially hydroxylamine hydrochloride (or hydrates thereof) in combination with a base or alkali, especially sodium hydroxide or potassium hydroxide, and commonly followed by filtration to give a "salt-free" solution of hydrazine (often in an alcoholic solvent) is an effective procedure. Also effective is the activation of carboxylic acids (e.g. by chloroformates such as iso-butyl chloroformate or ethyl chloroformate) and subsequent treatment with hydroxylamine or its equivalent in the form of derivatives with or without additional reagents such as a base. Thus, any known method of hydroxamation may be considered. The compounds of the invention are inhibitors of histone deacetylase (HDAC). They may therefore be used to treat a HDAC-mediated disorder. A therapeutically effective amount of a compound of the invention is administered to a subject, typically a human being, having such a disorder. The condition of the subject can thus be improved. Symptoms associated with the disorder may be ameliorated. Compounds of the invention may also be inhibitors of DNA methyl transferase. They may therefore be used to treat diseases and disorders for which inhibition of methyl transferase is relevant. The compounds may target a variety of cancers, including prostate, colon and esophageal cancers. HDAC-mediated disorders that may be treated according to the invention include cancer such as breast cancer, colon cancer, colorectal cancer, esophageal cancer, glioma, leukemia, lung small and non-small cell cancers, neuroblastoma, prostate cancer, thoracic cancer, melanoma, ovarian cancer, cervical cancer and renal cancer; cardiac hypertrophy; hematological disorders such as haemoglobinopathies, thalessemia and sickle cell anemia; and genetic-related metabolic disorders such as cystic fibrosis, peroxisome biogenesis disorders and adrenoleukodystrophy. HDAC inhibitors have also been proposed for stimulating hematopoietic cells ex vivo, ameliorating protozoal parasitic infection, accelerating wound healing and protecting hair follicles. A compound of the invention may be used in combination with another chemotherapeutic or antineoplastic agent in the treatment of a cancer. Examples of such other chemotherapeutic or antineoplastic agents include mitoxantrone; Vinca alkaloids such as vincristine and vinblastine; anthracycline antibiotics such as daunorubicin and doxorubicin; alkylating agents such as chlorambucil and melphalan; taxanes such as paclitaxel; antifolates such as methotrexate and tomudex; epipodophyllotoxins such as etoposide; camptothecins such as irinotecan and its active metabolite SN-38 and DNA methylation inhibitors such as the DNA methylation inhibitors disclosed in WO 02/085400. According to the invention, therefore, products are provided which contain a compound of the invention and another chemotherapeutic or antineoplastic agent as a combined preparation for simultaneous, separate or sequential use in creating a cancer. The compound of the invention and the other agent may be administered together or, if separately, in any order as determined by a physician. The present compounds can be administered in a variety of dosage forms, for example orally such as in the form of tablets, capsules, sugar- or film-coated tablets, liquid solutions or suspensions or parenterally, for example intramuscularly, intravenously or subcutaneously. The present compounds may therefore be given by injection or infusion. The dosage depends on a variety of factors including the age, weight and condition of the patient and the route of administration. The dosage for a particular patient will be determined by a physician. Typically, however, the dosage adopted for each route of administration when a compound of the invention is administered to adult humans is 0.001 to 500 mg/kg, most commonly in the range of 0.01 to 100 mg/kg, body weight, for instance 0.01 to 50 mg/kg. Such a dosage may be given, for example, from 1 to 5 times daily by bolus infusion, infusion over several hours and/or repeated administration. The dosage and timing of administration of, for example, another chemotherapeutic or antineoplastic agent which may be given to a cancer patient with a compound of the invention will similarly be dependent on a variety of factors and will be determined by a physician. A compound of any of the formulae (I) to (VII) described above or a pharmaceutically acceptable salt thereof is formulated for use as a pharmaceutical composition also comprising a pharmaceutically acceptable carrier or diluent. The compositions are typically prepared following conventional methods and are administered in a pharmaceutically suitable form. Preferred pharmaceutical compositions are sterile and pyrogen-free. Further, the pharmaceutical compositions provided by the invention typically contain a compound of the invention which is a substantially pure optical isomer. Compositions suitable for oral administration may, if required, contain a colouring or flavouring agent. Typically, a capsule or tablet comprises from 5 to 500 mg, preferably 10 to 500 mg, more preferably 15 to 100 mg, of a compound of formula (I) or a pharmaceutically acceptable salt thereof. Solid oral forms of the pharmaceutical compositions of the invention may contain, together with the active compound, diluents, e.g. lactose, dextrose, saccharose, cellulose, corn starch or potato starch; lubricants, e.g. silica, talc, stearic acid, magnesium or calcium stearate, and/or polyethylene glycols; binding agents; e.g. starches, arabic gums, gelatin, methylcellulose, carboxymethylcellulose or polyvinyl pyrrolidone; disaggregating agents, e.g. starch, alginic acid, alginates or sodium starch glycolate; effervescing mixtures; dyestuffs; sweeteners; wetting agents, such as lecithin, polysorbates, laurylsulphates; and, in general, non toxic and pharmacologically inactive substances used in pharmaceutical formulations. Such pharmaceutical preparations may be manufactured in known manner, for example, by means of mixing, granulating, tableting, sugar coating, or film coating processes. Liquid dispersions for oral administration may be syrups, emulsions and suspensions. The syrups may contain as carriers, for example, saccharose or saccharose with glycerine and/or mannitol and/or sorbitol. Suspensions and emulsions may contain as carrier, for example a natural gum, agar, sodium alginate, pectin, methylcellulose, carboxymethylcellulose, or polyvinyl alcohol. The suspension or solutions for intramuscular injections may contain, together with the active compound, a pharmaceutically acceptable carrier, e.g. sterile water, olive oil, ethyl oleate, glycols, e.g. propylene glycol, and if desired, a suitable amount of lidocaine hydrochloride. Solutions for injection or infusion may contain as carrier, for example, sterile water or preferably they may be in the form of sterile, aqueous, isotonic saline solutions. The following Examples illustrate the invention. Several Preparation Examples are also provided. Starting materials were purchased from Avocado or Aldrich and used as supplied, unless otherwise stated. The following compounds were prepared by literature methods: - (triphenyl-γs-phosphanylidene)acetic acid ethyl ester (5b) (J. J. Cappon, J. Boart,
G. A. M. Walle, J. Lugtenburg, Reel. Trav. Chim. Pays-Bas, 1991, 5, 158); (l-ethoxycarbonylethyl)triphenylphosphonium bromide (I. Shimizu, K. Hayashi, N. Ide and M. Oshima, Tetrahedron, 1991, 47, 2991); (E)-4-(triphenyl-γ4-arsanylidene)but-2-enoic acid ethyl ester bromide (Y. Huang, Y. Shen, J. Zhang and S. Zhang, Synthesis, 1985, 57); (E -2-methyl-3-(4-nitrophenyl)propenal (4b) (H. Hirata, H. Nakata, K. Yamada, K. Okuhara and T. Naito, Tetrahedron, 1961, 14, 252); (E -2-methyl-3-(4-dimethylaminophenyl)propenal (4i) (V. Sunjic, M. Majeric and Z. Hamersak, Croat. Chem. Ada, 1996, 69, 643); (2E,4E)-2,4-dimethyl-5-(p-nitrophenyl)penta-2,4-dienal (14a) (E. Suzuki and S. Inoue, J. Chem. Soc, Perkin Trans. 1, 1976, 404); - (ethoxycarbonylmethylene)triphenylphosphorane (R. W. Lang and H.-J.
Hansen, Org. Synth., 1994, 62, 203); 2-(4-nitrophenyl)-l,3-dioxane (S. Fukuzawa, T. Tsuchimoto and T. Hiyama, J. Org. Chem., 1997, 62, 151); 4-(l,3-dioxan-2-yl)phenylamine (M. C. Cesa, j. E. Rinz and T. T. Kopp, U. S. Pat., 1998, 4,868,061; and [(2E,4E)-6-methoxy-6-oxo-2,4-hexadienyl]triphenylphosphonium bromide (E. Vedejs and j. P. Bershas, Tetrahedron Lett., 1975, 16, 1359).
Preparation Example 1; 4-chloro-N-(4-formylphenyl)benzamide (2g) 2-(4-aminophenyl)-[l,3] dioxane (1.37 g, 7.62 mmol) was dissolved in pyridine
(15 mL) and 4-chlorobenzoyl chloride (1.74 g, 9.94 mmol) was then added dropwise to the stirred solution over 10 min and the mixture subsequently stirred for at 20 °C 5 h. The mixture was then added to dichloromethane (100 mL) and water (100 mL), and the whole gently shaken. The organic layer was separated and washed with 2 M hydrochloric acid (4 x 50 mL), then the organic layer was poured into a mixture of methanol (100 mL) and water (100 mL). More methanol was added until the solution became homogenous. A catalytic amount of p-toluenesulphonic acid was then added to the stirred solution followed by dropwise addition of 10 M hydrochloric acid (15 mL). The mixture was subsequently stirred at 20 °C 24 h. The solvents were evaporated and the residue suspended in water (100 mL) prior to extraction with dichloromethane (3 x 50 mL). The organic layers were combined, washed with saturated aqueous sodium hydrogen carbonate (3 x 50 mL), and then with water (50 mL). The organic layer was dried (Na^O^, filtered, and evaporated to give the title compound (0.97 g, 49%) as a fine pale yellow powder, mp 183-185 °C.
Preparation Example 2: N-(4-formylphenyl)-4-rnethoxybenzamide (2h 2-(4-aminophenyl)-[l,3] dioxane (1.00 g, 5.58 mmol) was dissolved in pyridine (11 mL) and 4-methoxybenzoyl chloride (1.05 g, 6.10 mmol) was then added dropwise to the stirred solution over 10 min and the mixture subsequently heated at reflux for 14 h. The solvent was evaporated and the residue dissolved in dichloromethane (50 mL) to give a solution that was washed with 2 M hydrochloric acid (3 x 40 mL). The organic layer was then separated and poured into water (100 mL). Methanol was then added until the solution became homogenous. A catalytic amount of 7-toluenesulfonic acid was then added to the stirred solution and the mixture was subsequently stirred at 25 °C 24 h. The solvents were evaporated and the residue suspended in water (100 mL) prior to extraction with dichloromethane (3 x 50 mL). The organic layers were combined, washed with saturated aqueous sodium hydrogen carbonate (3 x 50 mL), and then with water (50 mL). The organic layer was dried (Na^O , filtered, and evaporated to give the title compound (0.45 g, 32%) as a fine pale yellow powder, mp 142-144 °C.
Preparation Example 3: (2jg.4£)-4-methyl-5-(4-nitrophenyl)penta-2.4-dienoic acid ethyl ester (6c) To a stirred solution of (E)-2-methyl-3-(4-nitrophenyl)propenal (1.00 g, 5.24 mmol) in toluene (6.0 mL) was added (triphenyl-γ5-phosphanylidene)acetic acid ethyl ester (5b) (2.76 g, 7.93 mmol) at 25 °C. The mixture was heated to 40 °C and stirring was continued at 40 °C for 4 h. The mixture was then evaporated and the residue purified by flash chromatography on silica gel (1:9 ethyl acetate: 60-80 °C petroleum ether) to give the title compound (1.26 g, 92%) as crystalline yellow platelets, mp 124-125 °C IR (nujol) vmax 1712 (COOEt), 1625 (C=C), 1519 (NO2) cm-1; Η NMR (300 MHz, CDC13) δH 8.09 (2H, d, J=8Hz, ArH), 7.35 (3H, dd, 7=8 and 13 Hz, ArH and CH=CH-CHO), 6.73 (1H, s, Ph-CH), 5.99 (1H, d, J=13 Hz, CH-CHO), 4.12 (2H, q, J=7Hz, CH2O), 1.94 (3H, s, =CCH3), 1.18 (3H, t, J=7Hz, CH3).
Preparation Example 4: (2E.4E)-2.4-dimethyl-5-(4-nitrophenyl)penta-2.4-dienoic acid ethyl ester (6e) To a cooled suspension of (l-ethoxycarbonylethyl)triphenylphosphonium bromide (1.0 g, 2.25 mmol) in dry THF (10 mL) was added dropwise /i-butyllithium (1.6 M in hexanes, 1.4 mL, 2.3 mmol) at 0 °C. Stirring was continued at 0 °C for 20 min, then 10 min at 20 °C. To the above solution was added (E)-2-methyl-3-(4-nitrophenyl)propenal (4b) (0.36 g, 1.88 mmol) in portions over 5 min. The mixture was then stirred at 20 °C for 16 h. The mixture was then evaporated and the residue purified by flash chromatography on silica gel (1:4 ethyl acetate: 60-80 °C petroleum ether) to give a residue that was recrystallised from methanol to give the title compound (0.38 g, 73%) as yellow platelets, mp 80-81 °C; IR (nujol) vmax 1697 (COOEt), 1606 and 1589 (C=C), 1508 (NO2) cm 1; Η NMR (300 MHz, CDC13) δH 8.16 (2H, d, 7=9.0 Hz, ArH), 7.37 (2H, d, 7=9.0 Hz, ArH), 7.19 (IH, s, CH=CH-COO), 6.54 (IH, s, Ph-C/T), 4.14 (2H, q, 7=7 Hz, CH2O), 2.03 (6H, s, =CCH3), 1.29 (3H, t, 7=7 Hz, CH5CH2O); 13C NMR (75 MHz, CDC13) δc 168.91 (COO), 144.13, 142.29, 138,48, 131.75, 130.16, 129.18, 123.97 (ArC=C and C=C), 61.34 (CH2O), 18.97, 14.74, 14.68 (CH3). Elemental analysis: C15H17NO4. Calc. (%) C: 65.45, H: 6.18, N: 5.09. Found (%) C: 64.93, H: 6.15, 4.88.
Preparation Example 5: (2JE,.4£)-5-(4-Nitrophenyl)penta-2.4-dienoic acid ethyl ester (6a) To a suspension of 3-ethoxycarbonylallylidenenetriphenylarsonium bromide (1.40 g, 2.81 mmol) in dry THF (12 mL) was added dropwise n-butyllithium (2.5 M in hexanes, 1.1 mL, 2.81 mmol) at 0 °C. Stirring was continued at 0 °C for 20 min, then 10 min at 20 °C. To the above solution was added p-nitrobenzaldehyde (0.385 g, 2.55 mmol) in portions over 5 min. The mixture was then stirred at 20 °C for 16 h. The mixture was evaporated and the residue purified by flash chromatography on silica gel (1:4 ethyl acetate: 60-80 °C petroleum ether) to give a residue that was recrystallised from methanol to give the tide compound (0.51 g, 81%) as yellow prisms, mp 114-115 °C. IR (nujol) vmax 1710 (COO), 1625 (C=C), 1512 (ArC=C) cm 1; δH Η NMR (300 MHz, CDC13) δc 8.03 (2H, d, 7=9 Hz, ArH), 7.40 (2H, d, 7=9 Hz, ArH), 7.26 (IH, dd, 7= 10 and 15 Hz, C77=CH-COO), 6.86-6.78 (2H, m, ?h-CH=CH), 5.82 (IH, d, 7=15 Hz, CH-COO), 4.02 (2H, q, 7=7 Hz, CH2O), 1.11 (3H, t, 7=7 Hz, CH3). 13C NMR (75 MHz, CDC13) δc 166.84 (COO), 143.39, 142.68, 137.52, 130.81, 127.96, 127.96, 124.63, 124.53 (ArC=C and CH=CH), 60.98 (CH2O), 14.64 (CH3). Elemental analysis: C13H13NO4. Calc. (%): C: 63.15, H: 5.26, N: 5.66. Found (%): C: 63.11, H: 5.33, N: 5.64.
Preparation Example 6: (2E.4E)-5-(4-aminophenyl)penta-2.4-dienoic acid ethyl ester (7a) To a stirred solution of iron(II) sulfate heptahydrate (6.88 g, 24.7 mmol) in water (26 mL) and 0.880 concentrated aqueous ammonia (13 mL) at 60 °C was added a solution of (2E,4E)-5-(4-nitrophenyl)penta-2,4-dienoic acid ethyl ester (Preparation Example 5, 6a) (0.51 g, 2.06 mmol) in ethanol (20 mL). The mixture was then heated to 60 °C for 10 min. The solution was allowed to cool, extracted with dichloromethane (2 x 30 mL). The combined organic layers were dried (MgSO4), filtered and evaporated. The residue was recrystallised from methanol - diethyl ether - 60-80 °C petroleum ether to give the title compound (0.37 g, 83%) as orange needles, mp=91-92 °C. IR (nujol) vmax3398 and 3332 (NH2), 1697 (COO), 1622 (C=C), 1512 (ArC=C) cm 1; Η NMR (300 MHz, CDC13) δH 7.25 (IH, dd, 7=10 and 15 Hz, CH=CH-COO), 7.09 (2H, d, 7=8 Hz, ArH), 6.60 (IH, d, 7=15 Hz, CH-COO), 6.55-6.44 (3H, m, ArH and Ph-CH=CH), 5.71 (IH, d,7=15 Hz, Ph-CH), 4.03 (2H, q, 7=7 Hz, CH2O), 3.62 (2H, br s, NH2), 1.13 (3H, t, 7=7Hz, CH3); 13C NMR (75 MHz, CDC13) δc 167.79 (COO), 147.93, 145.84, 141.21, 129.75, 127.01, 123.00, 119.42, 115.42 (ArC=C and CH=CH), 60.50 (CH2O), 14.74 (CH3). Elemental analysis: C13H15NO2. Calc. (%): C: 71.88, H: 6.91, N: 6.45. Found (%): C: 71.97, H: 6.90, N: 6.36.
Preparation Example 7: (2Jg.4£)-5-(4-aminophenyl)-4-methylpenta-2.4-dienoic acid ethyl ester (7b) To a stirred solution of iron(II) sulfate heptahydrate (9.71 g, 34.9 mmol) in water (40 mL) and 0.880 concentrated aqueous ammonia (18 mL) at 40 °C was added a solution of (2E,4E)-4-methyl-5-(4-nitrophenyl)penta-2,4-dienoic acid ethyl ester (Preparation Example 3; 6c) (0.760 g, 2.91 mmol) in ethanol (25 mL). The mixture was then heated to 60 °C for 10 min. The solution was allowed to cool, extracted with dichloromethane (3 x 40 mL). The combined organic layers were dried (MgSO4), filtered and evaporated to give the title compound (0.62 g, 92%) as yellow prisms, mp 107-108 °C. IR (film) vmax 1720 (COOEt), 1530 (C=C) cm 1; Η NMR (300 MHz, CDC13) δH 7.38 (IH, d, 7=15Hz, CH=CH-CHO), 7.11 (2H, d, 7=8Hz, ArH), 6.62 (IH, s, Ph-CH), 6.60 (2Η, d, 7= 8Hz, ArH), 5.80 (IH, d, 7=15Hz, CH-CHO), 4.09 (2H, q, 7=7 Hz, CH2O), 3.73 (2H, br s, NH2), 1.94 (3H, s, =CCH3), 1.24 (3H, t, 7=7 Hz, CH3); 13C NMR (75 MHz, CDC13) δc 168.04 (C=O), 151.16, 147.03, 143.57, 137.79, 136.19, 131.58, 123.91, 115.02 (ArC=C and C=C), 60.51 (CH2O), 14.66, 14.18 (CH3). Elemental analysis: C14H NO2. Calc (%) C: 72.70, H: 7.40, N: 6.05. Found (%) C: 72.65, H: 7.45, N: 6.00.
Preparation Example 8: (2E.4E)-5-(4-aminophenyl)-2.4-dimethylpenta-2.4-dienoic acid ethyl ester (7c) To a stirred solution of iron(II) sulfate heptahydrate (3.0 g, 10.8 mmol) in water (10 mL) and 0.880 concentrated aqueous ammonia (6 mL) at 60 °C was added a solution of (2E,4E)-5-(4-nitrophenyl)-2,4-dimethylpenta-2,4-dienoic acid ethyl ester (Preparation Example 4; 6e) (0.25 g, 0.09 mmol) in ethanol (10 mL). The mixture was then heated to 60 °C for 15 min. The solution was allowed to cool, extracted with dichloromethane (3 x 30 mL). The combined organic layers were dried (MgSO4), filtered and evaporated. The mixture was then evaporated and the residue purified by flash chromatography on silica gel (1:4 ethyl acetate: 60-80 °C petroleum ether) to give the title compound as an oil.
Example 1: (2E.4E)-5-(4-benzenesulfonylaminophenyl)-4-methylpenta-2,4-dienoic acid ethyl ester (8a) To a stirred solution of (2E,4E)-5-(4-aminophenyl)-4-methylpenta-2,4-dienoic acid ethyl ester (Preparation Example 7; 7b) (0.35 g, 1.52 mmol) in pyridine (8.0 mL) at 25 °C was added benzenesulfonyl chloride (0.39 mL, 3.04 mmol) by means of a stainless steel cannula. The stirred mixture was then heated and maintained at 90 °C for 2 h. Crushed ice (2.5 g) was added cautiously and the resulting suspension was filtered through a sintered glass funnel. The solid was dried in air and recrystallised from ethanol to give the title compound (0.41 g, 73%) as dark yellow platelets, mp 134-136 °C.
Example 2: (2Et4£)-5-(4-(4-chlorobenzenesulfonylamino)phenyl)penta-2.4-dienoic acid ethyl ester (8b) A solution of (2E,4E)-5-(4-aminophenyl)penta-2,4-dienoic acid ethyl ester (Preparation Example 6, 7a) (0.32 g, 1.47 mmol) andp-chlorobenzenesulfonyl chloride (0.405 g, 1.91 mmol) in pyridine (3 mL) was heated at reflux for 6 h, then removed to ambient temperature and stirred for 12 h. Evaporation gave a residue that was dissolved in dichloromethane (30 mL) and the solution washed with 1.2 M hydrochloric acid (20 mL) then with saturated aqueous sodium chloride (20 mL). The organic layer was dried (MgSO4), filtered and evaporated. The residue was recrystallised from methanol to give the title compound (0.42 g, 73%) as orange prisms, mp 191-192 °C. IR (nujol) vmax 3238 (NH), 1683 (C=O), 1625 (C=C), 1512 (ArC=C) cm 1; Η NMR (300 MHz, CDC13) δH 7.92 (2H. d, 7=8 Hz, ArH), 7.41-7.28 (5H, m, ArH and CH=CH-C=O), 6.99 (2H, d, 7=9 Hz, ArH), 6.70 (2H, m, Ph-CH and Ph-CH=CH), 5.91 (1Η, d, 7=15 Ηz, CΗ-C=O), 4.15 (2H, q, 7=7 Hz, CH2O), 1.21 (3H, t, 7=7 Hz, CH3); 13CNMR (75 MHz, CDC13) δc 166.04 (C=O), 143.24, 138.80, 137.93, 136.50, 135.60, 132.51, 128.45, 127.64, 127.27, 125.35, 120.57 (ArC=C and C=C), 59.41 (CH2O), 13.29 (CH3). Elemental analysis: C19H18ClNO4S. Calc. (%) C: 58.23, H: 4.59, N: 3.57. Found (%) C: 57.85, H: 4.64, N: 3.49.
Example 3: (2E.4£')-5-(4-(4-methoχybenzenesulfonylamino)phenyl)penta-2.4- dienoic acid ethyl ester (8c) A solution of (2E,4E)-5-(4-aminophenyl)penta-2,4-dienoic acid ethyl ester
(Preparation Example 6, 7a) (0.35 g, 1.61 mmol) and ?-methoxybenzenesulfonyl chloride (0.50 g, 2.41 mmol) in pyridine (3 mL) was heated at reflux for 24 h. The pyridine was removed under reduced pressure and the residue was dissolved in dichloromethane (30 mL). The solution was washed with 1.2 M hydrochloric acid (20 mL) then with water (20 mL). The organic layer was dried (MgSO4), filtered and evaporated. The residue was recrystallised from methanol to give the title compound (0.56 g, 90%) as the hemi-hydrate, a pale yellow solid, mp 181-182 °C. Η NMR (300 MHz, CDC13) δ„ 7.66 (2H, d, 7=8 Hz, ArH), 7.32 (IH, dd, 7= 15 and 10 Hz, CH=CH- C=O), 7.28 (2H. d, 7=8 Hz, ArH), 7.01 (2H, d, 7=9Hz, ArH), 6.81 (2H, d, 7=8 Hz, ArH), 6.70 (2H, m, Ph-CH and Ph-CH=CH), 5.87 (1Η, d, 7=15 Ηz, CΗ-C=O), 4.06 (2H, q, 7=7 Hz, CH2O), 3.76 (3H, s, CH3O), 1.20 (3H, t, 7=7 Hz, CH3); 13CNMR (75 MHz, CDC13) δc 167.53 (C=O), 144.84, 139.67, 137.76, 133.16, 130.85, 129.83, 128.59, 126.27, 121.61, 121.34, 114.69, 113.43 (ArC=C and C=C), 60.80 (CH2O), 55.98 (CH3O), 14.70 (CH3). Elemental analysis: C20H21NO5S, l/2H2O. Calc. (%) C: 60.60, H: 5.55, N: 3.53. Found (%) C: 60.94, H: 5.51, N: 3.57.
Example 4: (2E.4E)-5-(4-(4-chlorobenzenesulfonylamino)phenyl)-4-methylpenta- 2.4-dienoic acid ethyl ester (8d) A stirred solution of (2E,4E)-5-(4-aminophenyl)-4-methylpenta-2,4-dienoic acid ethyl ester (Preparation Example 7; 7b) (0.30 g, 1.29 mmol) and /?-chlorobenzenesulfonyl chloride (0.548 g, 2.58 mmol) in pyridine (8 mL) was heated at reflux for 2 h. Dichloromethane (20 mL) was then added to the solution which was then washed with 1.2 M hydrochloric acid (3 x 10 mL) then with water (10 mL). The organic layer was dried (MgSO4), filtered and evaporated. The residue was purified by flash chromatography on silica gel (1:4 ethyl acetate: 60-80 °C petroleum ether) to give a solid which was recrystallised from methanol to give the title compound (0.23 g, 44%) as yellow needles, mp 156-157 °C. IR (nujol) vmax3180 (NH), 1683 (COO), 1625 (C=C), 1508 (ArC=C) cm 1; Η NMR (300 MHz, CDC13) δH 7.85 (2H, d, 7=9 Hz, ArH), 7.58 (IH, d, 7=15 Hz, CH=CH-CHO), 7.53 (IH, s, NH), 7.52 (2H, d, 7= 8 Hz, ArH), 7.36 (2H, d, 7=9 Hz, ArH), 7.28 (2H, d, 7= 8 Hz, ArH), 6.83 (IH, s, Ph-CH), 6.07 (1Η, d, 7=15 Ηz, CH-CΗO), 4.35 (2Η, q,7=7Hz, CH2O), 2.10 (3H, s, =CCH3), 1.44 (3H, t, J=7 Hz, CH3CH2O); 13C NMR (75 MHz, CDC13) δc 167.77 (COO), 149.90, 140.12, 138.00, 137.91, 135.92, 134.65, 134.48, 131.02, 129.81, 129.05, 121.57, 118.15 (ArC=C and C=C), 60.79 (CH2O), 14.69, 14.11 (CH3). Elemental analysis: C20H20NC1O4S. Required (%) C: 59.18, H: 4.93, N: 3.45. Found (%): C: 58.73, H: 4.63 , N: 3.27 .
Example 5: (2£.4^)-5-(4-(4-chlorobenzenesulfonylamino)phenyl)-4-methylpenta- 2.4-dienoic acid (8e) A stirred solution of (2E,4E)-5-(4-(4-chlorobenzenesulfonylamino)phenyl-4- methylpenta-2,4-dienoic acid ethyl ester (Example 4; 8d) (0.30 g, 0.73 mmol) and lithium hydroxide (0.154 g, 3.65 mmol) in THF (10 mL) and water (10 mL) was heated at 70 °C for 16 h. The mixture was extracted with diethyl ether (10 mL), acidified to pH 2 with 1.2 M hydrochloric acid and then extracted with ethyl acetate (2 x 20 mL). The organic layer was dried (MgSO4), filtered and evaporated. The residue was recrystallised from ethyl acetate to give the title compound (0.25 g, 91%) as a yellow solid, mp 218-220 °C. mp=218-220 °C; IR (nujol) vmax Η NMR (300 MHz, DMSO-<26); δH 7.97 (2H, d, 7=8 Hz, ArH), 7.81 (2H, d, 7=8 Hz, ArH), 7.55-7.48 (M, 3H, ArH and CH=CH-C=O), 7.32 (2H, d,7=8 Hz, ArH), 6.76 (IH, s, Ph-CH), 6.08 (IH, d, 7=15 Hz, CH-C=O), 2.14 (3H, s, CH3); 13C NMR (75 MHz, DMSO-rf6) δc 168.04 (C=O), 149.50, 138.75, 138.25, 137.84, 137.27, 133.61, 132.67, 130.89, 129.84, 128.93, 120.09, 118.44 (ArC=C and C=C), 13.92 (CH3). Elemental analysis: C18H16NClO4S. Calc. ( '%) C: 57.21, H: 4.23, N: 3.70. Found (%) C: 56.78, H: 4.53, N: 3.58.
Example 6: (2-g.4JE)-5-(4-(4-methoxybenzenesulfonylamino)phenyl)-4- methylpenta-2.4-dienoic acid ethyl ester (8f) A stirred solution of (2E,4E)-5-(4-aminophenyl)-4-methylpenta-2,4-dienoic acid ethyl ester (Preparation Example 7; 7b) (0.30 g, 1.29 mmol) and ?-methoxybenzenesulfonyl chloride (0.535 g, 2.58 mmol) in pyridine (8 mL) was heated at reflux for 2 h. Dichloromethane (20 mL) was then added and the solution washed with 1.2 M hydrochloric acid (3 x 10 mL) then with water (10 mL). The organic layer was dried (MgSO4), filtered and evaporated. The residue was purified by flash chromatography on silica gel (3:7 ethyl acetate: 60-80 °C petroleum ether) to give a solid which was recrystallised from methanol to give the title compound (0.43 g, 83%) as yellow prisms, mp 131-132 °C. mp=131-132 °C, IR (nujol) vmax3199 (NH), 1683 (COO), 1625 (C=C), 1508 (ArC=C) cm 1; Η NMR (300 MHz, CDC13) δH 7.98 (2H, d, 7=9 Hz, ArH), 7.58 (IH, d, 7=15.5 Hz, CH=CH-CHO), 7.51 (IH, s, NH), 7.45 (2H, d, 7= 8 Hz, ArH), 7.34 (2H, d, 7=9 Hz, ArH), 7.14 (2H, d, 7= 8 Hz, ArH), 6.94 (IH, s, Ph- CH), 6.22 (1Η, d,7=16 Ηz, CH-CΗO), 4.40 (2Η, q, 7=7Hz, CH2O), 4.03 (3H, s, CH3O), 2.21 (3H, s, =CCH3), 1.56 (3H, t, J=7 Hz, CH3CH2O); "CNMR (75 MHz, CDC13) δc 167.77 (COO), 163.29, 150.02, 138.19, 136.65, 134.31, 133.82, 131.10, 130.91, 129.82, 121.06, 117.91, 114.67 (ArC=C and C=C), 60.74 (CH3O), 55.95 (CH2O), 14.70, 14.10 (CH3). Elemental analysis:
Figure imgf000043_0001
l/8H2O. Required (%) C: 62.84, H: 5.73, N: 3.49. Found (%): C: 62.49, H: 5.76, N: 3.47.
Example 7: (2E.4E)-5-(4-(4-methoxybenzenesulfonylamino)phenyl)-2.4- dimethylpenta-2,4-dienoic acid ethyl ester (8g) A stirred solution of (2E,4E)-5-(4-aminophenyl)-2,4-dimethylpenta-2,4-dienoic acid ethyl ester (Preparation Example 8; 7c) (0.15 g, 0.61 mmol) and 7-methoxybenzenesulfonyl chloride (0.253 g, 1.22 mmol) in pyridine (5 mL) was heated at 90 °C for 4 h. The pyridine was removed under reduced pressure and the residue dissolved in dichloromethane (20 mL). The solution was washed with 1.2 M hydrochloric acid (2 x 10 mL) then with water (10 mL). The organic layer was dried (MgSO4), filtered and evaporated. The residue was purified by flash chromatography on silica gel (3:7 ethyl acetate: 60-80 °C petroleum ether) to give the title compound (0.23 g, 91%) as an oil, a mixture of diastereoisomers.
Example 8: (2£.4 )-5-(4-benzenesulfonylaminophenyl)-4-methylpenta-2.4-dienoic acid (8h) To a stirred solution of (2E,4£)-5-(4-benzenesulfonylaminophenyl)-4- methylpenta-2,4-dienoic acid ethyl ester (Example 1, 8a) (0.40 g, 1.08 mmol) in THF (5.0 mL) and water (5.0 mL) was added lithium hydroxide (0.15 g, 6.25 mmol). The stirred mixture was then heated and maintained at 80 °C for 5 h. The mixture was allowed to cool, transferred to a separating runnel and extracted with diethyl ether (3 x 10 mL). The aqueous layer was then acidified to pH 2 with 1M hydrochloric acid (approx. 2.5 mL), extracted with diethyl ether (3 x 10 mL). The combined organic layers were dried (MgSO4), filtered through a fluted filter paper and evaporated to give the title compound (0.31 g, 84%) as bright orange platelets, mp 162-163 °C.
Example 9; (2--?.4-g)-5-(4-(4-methoxybenzenesulfonylamino)phenyl)-4- methylpenta-2.4-dienoic acid (8i) A stirred suspension of (2E,4E)-5-(4-(4-methoxybenzenesulfonylamino)phenyl- 4-methylpenta-2,4-dienoic acid ethyl ester (Example 6; 8f) (0.15 g, 0.37 mmol) and lithium hydroxide (0.045 g, 1.88 mmol) in THF (2 mL) and water (2 mL) was heated at 70 °C for 3.5 h. The mixture was extracted with diethyl ether (20 mL), acidified to pH 2 with 1.2 M hydrochloric acid and then extracted with dichloromethane (3 x 15 mL). The combined organic layers were dried (MgSO4), filtered and evaporated to give the title compound (0.10 g, 73%) as a white solid, mp 215 °C.
Example 10: (2ig.4E)-5-(4-(4-chlorobenzoylamino)phenyl) -4-methylpenta-2.4- dienoic acid ethyl ester (8k) j-Chlorobenzoyl chloride was prepared by a modification of a procedure described in: J. P. Dickie, M. E. Loomans, T. M. Farley and F. M. Strong, J. Med. Chem., 1968, 6, 424. p-Chlorobenzoic acid (0.78 g, 4.98 mmol) was dissolved in toluene (20 mL) and one drop of DMF added. This mixture was cooled at -30 °C, and oxalyl chloride (0.43 mL, 5.01 mmol) added dropwise over 10 min. The mixture was allowed to warm to 20 °C, the solvent evaporated and the residue redissolved in dry toluene (10 mL) to give a solution of /?-chlorobenzoyl chloride. (2E,4E)-5-(4-Aminophenyl)-4-methylpenta-2,4-dienoic acid ethyl ester (Preparation Example 7; 7b) (1.00 g, 4.32 mmol) was dissolved in dry toluene (26 mL) and triethylamine (0.42 mL, 3.03 mmol). 1,2-Phenylene phosphorochloridite (0.56 g, 3.21 mmol) in dry toluene (15 mL) was then added to the mixture, and the reaction heated at reflux for 2.5 h. A solution of j-chlorobenzoyl chloride was then added, and the mixture heated at reflux for a further 24 h. The solvent was evaporated and the residue dissolved in dichloromethane (50 mL). The solution was washed with 2 M hydrochloric acid (3 x 40 mL), then water (2 x 40 mL) and lastly saturated aqueous sodium hydrogen carbonate (3 x 40 mL). The organic layer was dried (MgSO4), filtered and evaporated. The residue was recrystallised from propan-2-ol to give the title compound (0.73 g, 45%) as white plates, mp 169-170 °C.
Example 11: (2E,4E)-5-(4-(4-methoxybenzoylamino)phenyl)-4-methylpenta-2.4- dienoic acid ethyl ester (81) (2E,4E)-5-(4-Aminophenyl)-4-methylpenta-2,4-dienoic acid ethyl ester (Preparation Example 7; 7b) (1.00 g, 4.33 mmol) was dissolved in dry toluene (13 mL) and triethylamine (1.26 mL, 8.96 mmol). o-Phenylene phosphorochloridite (0.98 g, 5.62 mmol) in dry toluene (6.2 mL) was then added to the mixture, and the reaction heated at reflux for 2.5 h. p-Chlorobenzoic acid (0.37 g, 2.37 mmol) was then added, and the mixture heated at reflux for a further 24 h. The solvent was evaporated and the residue purified by flash chromatography on silica gel (80 g) (1:4 ethyl acetate: 60-80 °C petroleum ether) to give a residue was recrystallised from propan-2-ol to give the title compound (0.07 g, 5%) as yellow platelets, mp 121-122 °C.
Example 12: (2i^14it?)-5-(4-benzenesulfonylaminophenyl)-4-methylpenta-2.4- dienoic acid hydroxyamide (9a) To a stirred solution of hydroxylamine hydrochloride (0.56 g, 8.06 mmol) in methanol (4.0 mL) at 40 °C was added a solution of potassium hydroxide (0.45 g, 8.04 mmol) in methanol (4.0 mL). The solution was cooled to 0 °C and the suspension filtered through a glass funnel. To the stirred filtrate was added
(2E,4E)-5-(4-benzenesulfonylaminophenyl)-4-methylpenta-2,4-dienoic acid ethyl ester (Example 1, 8a) (0.30 g, 0.81 mmol) at 25 °C. To the stirred mixture at 25 °C was added potassium hydroxide (0.20 g, 3.57 mmol) in portions over 30 min. The mixture was then stirred at 25 °C for a further 4 h. Water (4.0 mL) was added and the mixture was acidified to pH 6 with 0.5 M hydrochloric acid, and extracted with ethyl acetate (2 x 5 mL). The combined organic layers were dried (MgSO4), filtered through a fluted filter paper and evaporated to give a solid which was recrystallised from benzene to give the title compound (0.20 g, 69%) as bright orange crystals, mp 137-139 °C.
Example 13: (2£.4E)-5-(4-chlorobenzenesulfonylaminophenyl)-4-methylpenta- 2.4-dienoic acid hydroxyamide (9b) To a suspension of (2£,4E)-5-(4-4-chlorobenzenesulfonylamino)phenyl)-4- methylpenta-2,4-dienoic acid ethyl ester (Example 4; 8d) (0.45 g, 1.1 mmol) in THF (6 mL) cooled to 0 °C was added dropwise a 50% aqueous solution of hydroxylamine (0.66 g, 20.0 mmol) and potassium hydroxide (0.197 g, 3.52 mmol) in methanol (2.0 mL) at 0 °C over 30 min. Stirring was continued for a further 30 min at 0 °C, then at 20 °C for 4 days. Water (5.0 mL) was then added and the mixture was acidified to pH 6 with 1.2 M hydrochloric acid, and extracted with ethyl acetate (2 x 20 mL). The combined organic layers were dried (MgSO4), filtered and evaporated to give a solid which was recrystallised from ethyl acetate to give the title compound (0.24 g, 44%) as a white powder, mp 177-178 °C (decomp.); IR (nujol) vraax 3436 (OH), 3217 (NH), 1664 (C=O), 1631 (C=C), 1502 (ArC=C) cm 1; Η NMR (300 MHz, DMSO-rf6) δH 8.92 (IH, br s, NH), 7.79 (2H, d, 7=9 Hz, ArH), 7.61 (2H, d, 7=9 Hz, ArH), 7.36 (IH, s, NHSO2), 7.29-7.21 (3H, m, ArH, CH=CH-COO), 7.13 (2H, d, 7=9 Hz, ArH), 6.75 (IH, s, Ph-CH), 5.90 (1Η, d, 7=15 Ηz, CΗ-COO), 1.94 (3Η, s, CH3); 13C NMR (75 MHz, DMSO-rf6) δc 163.46 (C=O), 143.97, 138.75, 138.24, 136.92, 135.97, 133.71, 133.02, 130.71, 129.84, 128.94, 120.17, 118.47 (ArC=C and C=C), 14.06 (CH3). Elemental analysis: C18H N2ClO4S. Calc (%) C: 55.03, H: 4.33, N: 7.13. Found (%) C: 54.85, H: 4.61, N: 6.89.
Example 14: (2E.4E)-5-(4-methoxybenzenesulfonylaminophenyl)-4-methylpenta- 2.4-dienoic acid hydroxyamide (9c) To a suspension of (2E,4E)-5-(4-(4-methoxybenzenesulfonylamino)phenyl)-4- methylpenta-2,4-dienoic acid ethyl ester (Example 8, 8f) (0.70 g, 1.74 mmol) in THF (10 mL) cooled to 0 °C was added dropwise a 50% aqueous solution of hydroxylamine (0.52 g, 0.78 mmol) and potassium hydroxide (0.31 g, 5.54 mmol) in methanol (3.0 mL) at 0 °C over 30 min. Stirring was continued for a further 30 min at 0 °C, then at 20 °C for 48 h. Water (10 mL) was then added and the mixture was acidified to pH 6 with 1.2 M hydrochloric acid, and extracted with ethyl acetate (2 x 20 mL). The combined organic layers were dried (MgSO4), filtered and evaporated to give a solid which was recrystallised from ethyl acetate to give the title compound (0.40 g, 59%) as a yellow powder, mp 171-172 °C (decomp.); IR (nujol) vmax3456 (OH), 3359 (NH), 1664 (C=O), 1649 (C=C), 1508 (ArC=C) cm 1; Η NMR (300 MHz, DMSO-rf6) δH 8.89 (IH, br s, NH), 7.69 (2H, d, 7=9 Hz, ArH), 7.10 (IH, d, 7=15 Hz, CH=CH-C=O), 7.05 (4H, m, ArH), 6.72 (IH, s, Ph-CH), 5.99 (1Η, d, 7=15 Ηz, CΗ-C=O), 3.76 (3H, s, CH3O), 1.92 (3H, s, CH3); 13C NMR (75 MHz, DMSO- /6) δc 162.85 (C=O), 144.01, 137.56, 136.09, 133.45, 132.46, 131.58, 130.62, 129.23, 119.61, 118.31, 114.78 (ArC=C and C=C), 55.99 (CH3O), 14.06 (CH3). Elemental analysis: Cι9H20N2O5S, l/4H2O. Calc (%) C: 58.1, H: 5.22, N: 7.13. Found (%) C: 58.33, H: 5.30, N: 6.89.
Example 15: (2£.4E)-5-(4-methoxybenzenesulfonylaminophenyl)penta-2,4-dienoic acid hydroxyamide (9d) To a suspension of (2£,4E)-5-(4-(4-methoxybenzenesulfonylamino)phenyl) penta-2,4-dienoic acid ethyl ester (Example 3, 8c) (0.47 g, 1.21 mmol) in THF (10 mL) cooled to 0 °C was added dropwise a 50% aqueous solution of hydroxylamine (0.72 g, 10.9 mmol) and potassium hydroxide (0.23 g, 4.11 mmol) in methanol (5.0 mL) at 0 °C over 30 min. Stirring was continued for a further 30 min at 0 °C, then at 20 °C for 48 h. Water (10 mL) was then added and the mixture was acidified to pH 6 with 1.2 M hydrochloric acid, and extracted with ethyl acetate (2 x 20 mL). The combined organic layers were dried (MgSO4), filtered and evaporated to give a solid which was recrystallised from ethyl acetate to give the title compound (0.28 g, 62%) as a pale yellow powder, mp 181-182 °C (decomp.); Η NMR (300 MHz, DMSO-d6) δ„ 7.90 (2H, d, 7=9 Hz, ArH), 7.59 (2H, d, 7=8 Hz, ArH), 7.33 (IH, dd, 7=15 and 10 Hz, CH=CH-C=O), 7.25 (4H, m, ArH), 7.18 (2H, m, Ph-CH and Ph-CΗ=CH), 6.14 (1Η, d, 7=15 Ηz, CΗ-C=O), 3.98 (3H, s, CH3O); 13C NMR (75 MHz, DMSO-d6) δc 162.78
(C=O), 138.83, 137.47, 132.02, 131.69, 129.21, 128.18, 126.33, 122.05, 120.01, 114.74 (ArC=C and C=C), 55.97 (CH3O). Elemental analysis: C18H17N2O5SK,l/2H2O. Calc (%) C: 51.2, H: 4.27, N: 6.63. Found (%) C: 50.84, H: 4.83, N: 6.73. Preparation Example 9: (2E.4ig.6E)-6-methyl-7-(4-nitrophenyl)hepta-2,4.6-trienoic acid ethyl ester To a suspension of 3-ethoxycarbonylallylidenenetriphenylarsonium bromide (1.0 g, 2.0 mmol) in dry THF (10 mL) was added dropwise n-butyllithium (2.5 M in hexanes, 1.1 mL, 2.75 mmol) at 0 °C. Stirring was continued at 0 °C for 20 min, then 10 min at 20 °C. To the above solution was added (E)-2-methyl-3-(4- -nitrophenyl) propenal (4b) (0.318 g, 1.66 mmol) in portions over 5 min. The mixture was then stirred at 20 °C for 16 h. The mixture was evaporated and the residue purified by flash chromatography on silica gel (1:4 ethyl acetate: 60-80 °C petroleum ether) to give the title compound (0.28 g, 59%) as yellow prisms, mp 143-144 °C. IR (nujol) vmax 1699 (COOEt), 1620 (C=C), 1504 (NO2) cm"1, Η NMR (300 MHz, CDC13) δH 8.06 (2H, d, 7= 9.0 Hz, ArH), 7.40 (2H, d, 7=9.0 Hz, ArH), 7.28 (IH, dd, 7= 11 and 15 Hz, CH=CH- CHO), 6.71 (IH, d, 7=15 Hz, CH-CHO), 6.62 (IH, s, Ph-CH), 6.45 (1Η, dd, 7= 11 and 15 Ηz, =C(CΗ3)-CΗ=CH), 5.91 (1Η, d, 7=15 Ηz, =C(CΗ3)-CH), 4.18 (2Η, q, 7=7 Hz, OCH2), 2.02 (3H, s, =CCH3), 1.24 (3H, t, 7=7 Hz, CH3CH2O); 13C NMR (75 MHz, CDC13) δc 165 (COO), 144.87, 144.47, 144.21, 139.13, 133.13, 130.26, 128.28, 123.98, 122.52 (ArC=C and C=C), 60.79 (CH2O), 14.69, 14.45 (CH3). Elemental analysis: C16H17NO4. Calc. (%) C: 66.89, H: 5.92, N: 4.87. Found (%) C: 66.59, H: 5.82, N: 4.74.
Preparation Example 10: (2E.4E.6E)-7-(4-aminophenyl)-6-methylhepta-2.4.6- trienoic acid ethyl ester To a stirred solution of iron(II) sulfate heptahydrate (4.64 g, 16.7 mmol) in water (10 mL) and 0.880 concentrated aqueous ammonia (10 mL) at 60 °C was added a solution of (2E,4E,6E)-6-methyl-7-(4-nitrophenyl)hepta-2,4,6-trienoic acid ethyl ester (Preparation Example 9) (0.40 g, 1.39 mmol) in ethanol (10 mL). The mixture was then heated to 60 °C for 15 min. The solution was allowed to cool, extracted with dichloromethane (2 x 20 mL). The combined organic layers were dried (MgSO4), filtered and evaporated. The residue was recrystallised from methanol to give the title compound (0.32 g, 90%) as orange needles, mp 123-124 °C. Η NMR (300 MHz,
CDC13) δH 7.35 (IH, dd,7=ll and 15 Hz, CH=CH-COO), 7.15 (2H, d, 7= 8 Hz, ArH), 6.67 (IH, d, 7=15 Hz, CH-COO), 6.64 (2H, d, 7=8 Hz, ArH), 6.60 (IH, s, Ph-CH), 6.31 (IH, dd, 7=ll and 15 Hz, CH-CH=CH-COO), 5.84 (IH, d, 7=15 Hz, Ph-CH=C(CH3)- CH), 4.14 (2Η, q, 7=7 Hz, CH2O), 3.67 (2H, br s, NH2), 2.00 (3H, s, CH3), 1.28 (3H, t, 7=7 Hz, CH3); 13C NMR (75 MHz, CDC13) δc 167.78 (COO), 147.32, 146.33, 145.76, 136.73, 132.86, 131.29, 127.98, 124.75, 119.88, 115.13 (ArC=C and CH=CH), 60.54 (CH2O), 14.75 and 14.29 (CH3). Elemental analysis: C16H19NO2, l/16H2O. Calc. (%) C: 74.70, H: 7.39, N: 5.44. Found (%) C: 74.38, H: 7.40, N: 5.42.
Example 16: (2E,4ig.6E)-7-(4-(4-methoxybenzenesulfonylamino)phenyl)-6- methvIhepta-2.4.6-trienoic acid ethyl ester (17h) A stirred solution of (2E,4£,6£)-7-(4-aminophenyl)-6-methylhepta-2,4,6- trienoic acid ethyl ester (Preparation Example 10) (0.55 g, 2.14 mmol) and -methoxybenzenesulfonyl chloride (0.531 g, 2.57 mmol) in pyridine (5 mL) was heated at reflux for 12 h. The pyridine was removed under reduced pressure and the residue dissolved in dichloromethane (20 mL). The solution was washed with 1.2 M hydrochloric acid (2 x 10 mL) then with water (10 mL). The organic layer was dried (MgSO4), filtered and evaporated. The residue was purified by flash chromatography on silica gel (3:7 ethyl acetate: 60-80 °C petroleum ether) to give the title compound (0.66 g, 72%) as an oil, a mixture of diastereoisomers.
Example 17: (2£t4£.6£)-7-(4-(4-chlorobenzoylamino)phenyl)hepta-2.4.6-trienoic acid methyl ester (17s) 4-Chloro-N-(4-formylphenyl)benzamide (Preparation Example 1; 2g) (0.27 g, 1.05 mmol), [(2£,4E)-6-methoxy-6-oxo-2,4-hexadienyl]triphenylphosphonium bromide (0.49 g, 1.05 mmol) and potassium carbonate (0.72 g, 5.22 mmol) were added to THF (20 mL). The mixture was stirred at 40 °C for 72 h. The solvent was evaporated and the residue was suspended in ethyl acetate (50 mL), sonicated for 10 min, and stirred for 1 h at 20 °C. The mixture was then filtered through a thin pad of silica, which was subsequently washed with ethyl acetate 2 x 10 mL. The combined filtrates were evaporated, and the residue recrystallised from isopropanol to give the title compound (0.12 g, 31 %) as a yellow prisms, mp 169-170 °C.
Preparation Example 11: ethyl 5-(4-aminophenyl)-4-methylpentanoate (25a) Ethyl 4-methyl-5-(4-nitrophenyl)penta-2,4-dienoate (Preparation Example 3, 6c) (1.80 g, 6.89 mmol) was dissolved in ethanol (50 mL), and a catalytic amount of 10% palladium-on-carbon was added cautiously. Nitrogen gas was passed over the mixture. A gentle vacuum was then applied for a short time. Nitrogen gas was then slowly admitted and the gas above the mixture then evacuated. Hydrogen was then carefully admitted until atmospheric pressure reached. The mixture was stirred under hydrogen (1 arm) for 2 h. The hydrogen gas was then evacuated and replaced by nitrogen. The mixture was filtered through Celite® and the solvent evaporated to give the tide compound (1.51 g, 97%) as a clear oil. Η NMR (300 MHz) δH 6.93 (d, 2H, 2,6-Ar), 6.60 (d, 2H, 3,5-Ar), 4.08 (q, 2H, CO-OCH2-CH3), 3.48(s, broad, 2H, NH2), 2.52 (q, IH, Ar-CH2-CHMe-), 2.30 (m, 3H, Ar-CH2-CHMe- and -CH2CO2Et), 1.69 (m, 2H, - CHMe-CH2-CH2-CO2Et), 1.47 (m, IH, Ar-CH2-CHMe-CH2-), 1.24 (t, 3H, -COO-CH2- CH3), 0.84 (d, 3H, -CH2-CHMe-CH2-); 13C NMR (75 MHz, CDC13) δc 173.97 (CI), 144.24 (4-Ar), 130.97 (1-Ar), 129.91 (3,5-Ar), 115.11 (2,6-Ar), 60.20 (O-CH2-CH3), 42.50 (C5), 34.78 (C4), 32.28 (C3), 31.59 (C2), 18.99 (C6), 14.25 (O-CH2-CH3).
Example 18: 5-r4-(4-methoxybenzenesulfonylamino)phenyll-4-methylpentanoic acid ethyl ester (25c) Ethyl 5-(4-aminophenyl)-4-methylpentanoate (Preparation Example 11, 25a) (0.65 g, 2.76 mmol) was dissolved in pyridine (4.0 mL), and 4-methoxybenzenesulfonyl chloride (0.57 g, 2.76 mmol) was added to the stirred mixture. The mixture was stirred for an additional 16 h at 25 °C. The pyridine was removed under a high vacuum and the residue was dissolved in dichloromethane (50 mL). This solution was washed with 2M hydrochloric acid (3 x 20 mL), then with water (20 mL) and lastly with saturated aqueous sodium hydrogen carbonate (20 mL). The organic layer was dried (MgSO4), filtered and evaporated to give a residue that was purified by flash chromatography (gradient elution from pure 40-60 °C petroleum ether to 40:60: ethyl acetate: 40-60 °C petroleum ether with 5% increase in ethyl acetate for every 200 mL) to give the title compound (0.78 g, 70%) as a yellow oil.
Example 19: Ethyl 5-r4-(4-chlorobenzoylamino)phenyll-4-methylpentanoate (25d) Ethyl 5-(4-aminophenyl)-4-methylpentanoate (Preparation Example 11, 25a)
(0.54 g, 2.29 mmol) was dissolved in pyridine (4.0 mL), and 4-chlorobenzoyl chloride (0.52 g, 2.97 mmol) was added to the stirred mixture. The mixture was stirred for an additional 16 h at 25 °C. The pyridine was removed under a high vacuum and the residue was dissolved in dichloromethane (50 mL). This solution was washed with 2M hydrochloric acid (3 x 20 mL), then with water (20 mL) and lastly with saturated aqueous sodium hydrogen carbonate (20 mL). The organic layer was dried (MgSO4), filtered and evaporated. The pink residue was recrystallised from propan-2-ol to give the title compound (0.48 g, 56%) as shiny white platelets, mp 114-115 °C. Η (300 MHz) δH 8.06(s, IH, CO-NH-Ar), 7.76 (d, 2H, 2,6-Ar), 7.51 (d, 2H, 3,5-Ar), 7.40 (d, 2H, 3,5-Ar), 7.10 (d, 2H, 2,6-Ar), 4.10 (q, 2H, O-CH2-CH3), 2.61 (q, IH, Ar-CH2- CHMe-), 2.34 (m, 3H, Ar-CH2-CHMe- and -CH2CO2Et), 1.72 (m, 2H, -CHMe-CH2- CH2-CO2Et), 1.46 (m, IH, Ar-CH2-CHMe-CH2-), 1.24 (t, 3H, -COO-CH2-CH3), 0.85 (d, 3H, -CH2-CHMe-CH2-); 13C NMR (75 MHz, CDC13) δc 173.93 (CI), 164.76
(CONH), 137.93 (4-Ar), 137.50 (4-Ar), 135.65 (1-Ar), 133.45 (1-Ar), 129.67 (2,6-Ar), 128.91 (2,6-Ar), 128.63 (3,5-Ar), 120.39 (3,5-Ar), 60.29 (O-CH2-CH3), 42.79 (C5), 34.61 (C4), 32.21 (C3), 31.58 (C2), 18.97 (C6), 14.25 (O-CH2-CH3).
Preparation Example 12: ethyl 5-f4-(4-chlorobenzenesulfonylamino)phenyll- 4-methylpentanoate (25b) Ethyl 5-(4-aminophenyl)-4-methylpentanoate (Preparation Example 11, 25a) (0.52 g, 2.21 mmol) was dissolved in pyridine (2.0 mL), and a solution of 4-chlorobenzenesulfonyl chloride (0.61 g, 2.89 mmol) in pyridine (2.0 mL) was added to the stirred mixture. The mixture was stirred for an additional 16 h at 25 °C. The pyridine was removed under a high vacuum and the residue was dissolved in dichloromethane (50 mL). This solution was washed with 2 M hydrochloric acid (3 x 20 mL), then with water (20 mL) and lastly with saturated aqueous sodium hydrogen carbonate (20 mL). The organic layer was dried (MgSO4), filtered and evaporated. The orange oil was purified by flash chromatography on 70 g of silica gel (gradient elution from pure 60-80 °C petroleum ether to 40:60: ethyl acetate: 60-80 °C petroleum ether). Evaporation of the intermediate fractions afforded the title compound (0.52 g, 57%) as an light yellow oil. Η (300 MHz) δH 7.66 (d, 2H, 2,6-Ar), 7.37 (2, 2H, 3,5-Ar), 6.99 (dd, 4H, 2,6-Ar), 5.29 (s, IH, SO2NH), 4.10 (q, 2H, O-CH2-CH3), 2.56 (q, IH, Ar-CH2- CHMe-), 2.31(m, 3H, Ar-CH2-CHMe- and -CH2CO2Et), 1.66 (m, 2H, -CHMe-CH2-
CH2-CO2Et), 1.45 (m, IH, Ar-CH2-CHMe-CH2-), 1.23(t, 3H, -COO-CH2-CH3), 0.80 (d, 3H, -CH2-CHMe-CH2-); 13C NMR (75 MHz, CDC13) δc 173.85 (CI), 139.42 (4-Ar), 138.87 (1-Ar), 137.59 (4-Ar), 133.81 (1-Ar), 130.05 (2,6-Ar), 129.24 (2,6-Ar), 128.70 (3,5), 60.33 (O-CH2-CH3), 42.69 (C5), 34.47 (C4), 32.13 (C3), 31.51 (C2), 18.93 (C6), 14.24 (O-CH2-CH3).
Example 20: 5-r4-(4-chlorobenzenesulfonylamino)phenyll-4-methylpentanoic acid hydroxyamide (26b) Ethyl 5-[4-(4-chlorobenzenesulfonylamino)phenyl]-4-methylpentanoate (Preparation Example 12, 25b) (0.63 g, 1.53 mmol) was dissolved in methanol (5.0 mL) at 0 °C. The solution was stirred and 50% aqueous hydroxylamine (0.91 g, 13.8 mmol) was added dropwise over 15 min, and then aqueous potassium hydroxide (1.3 mL, 1.6 M) in methanol added to the mixture in one batch. The stirred mixture was allowed to warm up to 25° C over 14 h. Water (10 mL) was then added, and dilute hydrochloric acid added until pH 6 was attained. The mixture was extracted with 85:15 ethyl acetate: methanol (5 x 30 mL) and the combined organic layers dried (MgSO4), filtered and evaporated to give the title compound (0.32 g, 52%) as an oil.
Example 21: 5-r4-(4-chlorobenzoylamino)phenyll-4-methylpentanoic acid hydroxyamide (26d) Ethyl 5-[4-(4-chlorobenzoylamino)phenyl]-4-methylpentanoate (Example 19; 25d) (0.52 g, 1.39 mmol) was dissolved in methanol (5.0 mL) at 0 °C. The solution was stirred and 50% aqueous hydroxylamine (0.84 g, 12.7 mmol) was added dropwise over 15 min, and then aqueous potassium hydroxide (1.3 mL, 1.6 M),in methanol added to the mixture in one batch. The stirred mixture was allowed to warm up to 25° C over 14 h. Water (10 mL) was then added, and dilute hydrochloric acid added until pH 6 was attained. The mixture was extracted with 85:15 ethyl acetate: methanol (5 x 30 mL) and the combined organic layers dried (MgSO4), filtered and evaporated. The residue was recrystallised from methanol/hexane, to give the title compound (0.21 g, 41%) as a fine white powder, mp 185-186 °C.
Example 22: (2E.4E.6E)-7(4-(p-Chlorobenzenesulphonylamino)-6-methylhepta- 2.4.6-trienoic acid ethyl ester A solution of (2E,4E,6E)-7(p-aminophenyl)-6-methylhepta-2,4,6-trienoic acid ethyl ester (0.37 g, 1.44 mmol) and -chlorobenzenesulphonylchloride (0.365 g, 2.88 mmol) in pyridine (5 mL) was heated at reflux for 24 h. Stirring was then continued for an additional 48 h at room temperature. After the removal of the pyridine under vacuum, the residue was dissolved in dichloromethane (30 mL), washed with 1.2 M hydrochloric acid, and the organic layer dried over anhydrous MgSO4, filtered and evaporated. The residue was chromatographed on silica gel column using 1:4 ethyl acetate:40-60 °C petroleum ether to give the title compound (0.40 g, 64%) as a yellow oil; Η NMR (300 MHz, CDC13) δH 7.94 (2H, d, 7=8 Hz, ArH), 7.59 (3H, m, ArH and CH=CH-C=O), 7.41 (2H, d, 7=8 Hz, ArH), 7.29 (2H, d, 7=8 Hz, ArH), 6.86 (IH, d, 7=15 Hz, C(CH3)-CH=), 6.84 (IH, s, Ph-CH), 6.70 (IH, dd,7=15 and 11 Hz, C(CH3)- CH=CH), 6.11 (IH, d, 7=15 Hz, CH-C=O), 4.39 (2H, t, 7=7 Hz, CH2O), 2.08 (3H, s, CH3), 1.47 (3H, s, CH3); 13C NMR (75 MHz, CDC13) δc 167.86 (C=O), 146.29, 145.40, 140.03, 137.91, 135.80, 135.58, 134.97, 134.84, 130.86, 129.80, 129.08, 126.36, ' 121.58, 121.00 (ArC=C and C=C), 60.83 (CH2O), 14.72, 14.26 (CH3).
Determination of Histone Deacetylase Inhibition Activity Histone deacetylase inhibitory activity was measured as described by Vigushin et al., Clin. Cancer Res., 7, 971-976 (2001) based on methods published by Taunton et al., Science, 272, 408-411 (1995) and Emiliani et al, Proc. Natl. Acad. Sci. U.SΛ., 95, 2795-2800 (1998). Briefly, the assay begins by incubating histone deacetylase enzymes contained in a nuclear extract from the HeLa human cervical adenocarcinoma cell line with a compound of the invention followed by addition of a radiolabeled substrate. The substrate was a synthetic peptide corresponding to histone H4 (amino acids 14-21) that had been chemically acetylated on lysine residues with sodium [3H]acetate according to the method published by Taunton et al, Science 272, 408-411 (1995). Released [3H] acetic acid (a measure of histone deacetylase activity) was then extracted with ethyl acetate and quantified in a scintillation counter. The concentration of a compound of the invention that inhibits histone deacetylase activity by 50% (i.e. IC50) was then determined by repeating the assay with a range of different concentrations of compound. Each assay was performed in duplicate with control samples in triplicate for accuracy.
HeLa cell nuclear extract HeLa cell nuclear extract was prepared according to the method of Dignam et al, Nucleic Acids Res., 11, 1475-1489 (1983). HeLa human cervical adenocarcinoma cells were grown at 37°C in DMEM medium containing 5% foetal calf serum to a concentration of 5x10s cells per ml prior to harvesting. Cells were then harvested by centrifugation for 10 minutes at 2,000 rpm in a Sorvall HG4L rotor. The cell pellet was resuspended in 5 volumes of cold phosphate buffered saline, collected by centrifugation at 4°C and all subsequent manipulations were performed at 4°C. Cells were suspended in 5 packed cell volumes of lOmM HEPES (pH 7.9 at 4°C), 1.5 mM MgCl2, lOmM KC1 and 0.5 mM DDT and allowed to equilibrate for 10 minutes. The cells were pelleted by centrifugation as above, resuspended in 2 packed cell volumes of the same buffer and then lysed by 10 strokes of a glass Dounce homogenizer. The homogenate was centrifuged as before and the pellet was then centrifuged for 20 minutes at 25,000 x g in a Sorvall SS34 rotor to remove residual cytoplasmic material, yielding crude nuclei. Crude nuclei from 109 cells were resuspended in 3 ml of 20 mM HEPES (pH 7.9), 25% (v/v) glycerol, 420 mM NaCl, 1.5 mM MgCl2, 0.2 mM EDTA, 0.5 mM phenylmethylsulfonyl fluoride (PMSF) and 0.5 mM DTT with a glass Dounce homogenizer (10 strokes). After stirring gently with a magnetic stirrer for 30 minutes, the suspension was centrifuged for 30 minutes at 25,000 x g (Sorvall SS34 rotor). The supernatant was dialysed against 50 volumes at 20 mM HEPES (pH 7.9), 20% (v/v) glycerol, 100 mM KC1, 0.2 mM EDTA, 0.5 mM PMSF and 0.5 mM DTT for 5 hours and the dialysate was then centrifuged at 25,000 x g for 20 minutes (Sorvall SS34 rotor). The supernatant, designated the nuclear extract, was snap frozen in liquid nitrogen and then stored at -80°C. The protein concentration measured by Bradford assay was 10 mg/ml and 50 mg of protein was obtained from 109 cells.
Acetylation of histone H4 peptide with [3H]acetate A peptide corresponding to amino-terminal residues 14-21 of histone H4
(GAKRHRKV) was synthesised in an automated peptide synthesiser (ABI 433; Applied Biosystems, Cheshire, UK), purified by reverse phase high performance liquid chromatography (HPLC), and lyophilised. The peptide was >95% pure by reverse phase HPLC, mass spectrometry and capillary electrophoresis. All subsequent steps were performed in a fume hood. To 1 mg of the lyophilised peptide in a 2 ml screw top amber glass vial was added 500 μl (12.5 mCi) of sodium [3H]acetate (9.9 Ci/mmol, 25 mCi/ml in ethanol; ICN Pharmaceuticals, Basingstoke, UK). Then 20 μl of a freshly prepared solution of 0.24 M benzotriazol-l-yloxytris(dimethylamino)phosphonium hexafluorophosphate (BOP reagent; Sigma-Aldrich, Gillingham, UK) and 0.2 M triethylamine in acetonitrile was added, the reaction vial was capped, and the labelling mixture incubated on a rotating platform overnight at room temperature. The mixture was then concentrated to dryness under reduced pressure and the residue dissolved in 1 ml TEN buffer (50 mM Tris (pH 8.0), 150 mM NaCl, 5 mM EDTA). After centrifugation at 14,000 x g for 15 seconds at room temperature, the supernatant containing [3H]acetate-labelled histone H4 peptide was purified by gel filtration on Sephadex G-25 (PD10 column; Amersham Biosciences UK Limited, Buckinghamshire, UK). After equilibrating with 10 column volumes of TEN, the supernatant was loaded onto the column and then eluted with TEN. 0.5 ml fractions were collected and the radioactivity in each was quantified by liquid scintillation counting. After the column void volume, the [3H]acetate-labelled histone H4 peptide elutes first followed by free unincorporated label ([3H]acetic acid). Eluates containing the purified radiolabeled peptide are pooled, divided into aliquots and stored at -70°C until use.
Histone deacetylase assay Histone deacetylase inhibition by compounds of the invention was assayed as described in Vigushin et al, Clin Cancer Res. 7, 971-976 (2001) based on methods published by Taunton et al, Science, 272, 408-411 (1995) and Emiliani et al. Proc. Natl. Acad. Sci. U.SΛ., 95, 2795-2800 (1998). HeLa cell nuclear extract prepared according to Dignam et al, Nucleic Acids. Res. 11, 1475-1489 (1983) was used as a source of histone deacetylase enzymes. The substrate was a synthetic peptide corresponding to amino acids 14-21 of histone H4 that had been chemically acetylated on lysine residues with sodium [3H]acetate as described by Taunton et al, Science 272, 408-411 (1995). A stock solution in dimethylsulfoxide (DMSO) was prepared for each compound of the invention to be tested and trichostatin A as a positive control. Stock solutions were diluted in DMSO to give a range of lOOx working solutions. The assay was performed in a final reaction volume of 200 μl. To each tube was added 40 μl of 5x HDAC buffer (50 mM Tris (pH 8.0), 750 mM NaCl, 50% (v/v) glycerol, 1 mM PMSF], 4 μl (40 μg total protein) HeLa cell nuclear extract, 2 μl lOOx inhibitor in DMSO or 2 μl DMSO as a negative control, and water to a total of 199 μl. After mixing by vortex and brief centrifugation (14,000 x g for 5 seconds at room temperature), the reaction mixture was incubated for 30 minutes at room temperature. The assay was then initiated by addition of 1 μl (37 kBq) of [3H]acetate-labelled histone H4 peptide substrate. After brief vortex and centrifugation as above, the reaction mixture was incubated for 60 minutes at room temperature. Fifty μl of a quenching solution [1 M HC1/0.16 M acetic acid] was then added to stop the reaction. The released [3H]acetate in each assay reaction was extracted into 600 μl ethyl acetate. After mixing by vortex, the organic and aqueous phases were separated by centrifugation (14,000 x g for 1 minute at room temperature). Duplicate 200 μl aliquots of the upper organic phase were transferred into separate scintillation vials each containing 5 ml scintillant (Hionic Fluor; Canberra Harwell Ltd., Didcot, UK) and the radioactivity in each measured by β -scintillation counting. An initial assay was performed to established the range of activity of each compound of the invention. The assay was then repeated using four log dilutions in range according to the expected potency for each test compound. The concentration of each compound of the invention that inhibited histone deacetylase enzyme activity by 50% (IC50) was determined graphically in each case using non-linear regression analysis to fit inhibition data to the appropriate dose-response curve (GraphPad Prism Version 3.0; GraphPad Software Inc., San Diego, CA). Each test compound was assayed in duplicate whilst positive and negative control samples were assayed in triplicate. Test compounds of the invention were found to be potent histone deacetylase inhibitors, some having IC50 values in the low nanomolar concentration range (e.g. two test compounds had ICS0 values of 49 nM and 74 nM).
Figure imgf000056_0001

Claims

1. Use of a compound of formula (I) for the manufacture of a medicament for use in treating a disorder mediated by histone deacetylase:
Figure imgf000057_0001
wherein: the Symbol __^ represents a single bond or a double bond or the symbol ^ , R6 and R8 together represent cyclopropyl; R1 to R5 each independently represent hydrogen, -CJQ alkyl, C2-C10 alkenyl, C2-C10 alkynyl, C C10 alkoxy, - o thioalkoxy, hydroxyl, C C10 hydroxyalkyl, halo, -CJO haloalkyl, amino, C -Cw alkylamino, di(CrC10 alkyl)amino, amido, nitro, cyano, (Cj-C^alky^carbonyloxy, ( -Cn, alkoxy )carbonyl, (CrC10 alkyl)carbonyl, ( - o alkyl)thiocarbonyl, ( - o alkyl)sulfonylamino, aminosulfonyl, (CrC10 alkyl)sulfinyl, (CJ-CJO alkyl)sulfonyl or -CJO alkyl substituted by amino, Cι-C10 alkoxy, -CK, alkylamino or di(C1-C10 alkyl)amino, and wherein one or two of R1, R2 and R3 is/are hydrogen and the other one or two of R1, R2 and R3 is/are other than hydrogen; R6 represents hydrogen, - o alkyl, substituted Cι-C10 alkyl, an unsaturated hydrocarbon chain of up to ten carbon atoms comprising one or more carbon-carbon double or triple bond, C6 or C]0aryl, a 5- to 10-membered heterocyclic group, CrC10 alkoxy, C^-Cw thioalkoxy, hydroxyl, halo, cyano, nitro, amino, - ,, alkylamino, d^ - o alkyl)amino, amido, ( - oalky carbonyloxy, ( - o alkoxy )carbonyl, alkyl)carbonyl, (C,-C10 alkyl)thiocarbonyl, (CJ-CJO alkyl)sulfonylamino, aminosulfonyl, (Cι-C10 alkyl)sulfinyl, ( -CJO alkyl)sulfonyl, a saturated or unsaturated C3-C12 hydrocarbon chain interrupted by O, S, NR, CO, C=NR, N(R)SO2, SO2N(R), N(R)C(O)O, OC(O)N(R), N(R)C(O)N(R), OC(O), C(O)O, OSO2, SO2O or OC(O)O where: (a) R independently represents hydrogen, C C10 alkyl, C2-C10 alkenyl, C2-C10 alkynyl, -Cm alkoxy, - o hydroxyalkyl, hydroxyl or - o haloalkyl, and (b) the saturated or unsaturated hydrocarbon chain is optionally substituted with C C10 alkyl, C2-C10 alkenyl, C2-C10 alkynyl, - o alkoxy, hydroxyl, CJ- Q hydroxyalkyl, halo, - o haloalkyl, amino, ( - Q alkyl)carbonyloxy, ( - o alkoxy )carbonyl, (CJ-CJ,, alkyl)carbonyl, (Cj-C^ alkyl)sulfonylamino, aminosulfonyl or - o alkylsulfonyl, when the symbol — represents a single bond, R7 and R8 each independently represents hydrogen, halo, - o alkyl, C6 or C10 aryl, a 5- to 10-membered heterocyclic group, OR9, SR9 or NR9R10 wherein R9 and R10 each independently represent hydrogen or C,-C6 alkyl or one of R9 and R10 is H and the other is -CO(Cx-C6 alkyl). or R7 and R8 together represent =O, =CH2 or =CHR9 wherein R9 is as defined above; - when the symbol ^^ represents a double bond, R7 represents hydrogen, halo,
CrC10 alkyl, or C10 aryl, a 5- to 10-membered heterocyclic group, OR9, SR9 or NR9R10 wherein R9 and R10 are as defined above and R8 is absent; W represents a single bond, -C(RU)=N-, -N=C(Rπ)-, -C(Rπ)(R12)-NR13-, -NR13-C(Rπ)(R12)-, -CO-NR11-, -NRπ-CO-, -SO2-NRn-, -NRπ-SO2-, -C(Rn)(R12)-O-, -O-C(Rn)(R12)-, -C(Rn)(R12)-S-, -S-C(RU)(R12)-, -CO-, -NR11-,
-SO-, -SO2-, S or -[C(Rπ)R1 ]p- wherein R", R12 and R13 each independently represents hydrogen, - alkyl, C6 or C]0 aryl or a 5- to 10- membered heterocyclic group alkyl and p is an integer of from 1 to 4; X represents -OR14, -SR14, -NR14OR15,-NR14NR15R16' -CF3, -CF2H or CH2F wherein R14, R1S and R16 each independendy represents hydrogen or - alkyl; and Y represents
Figure imgf000058_0001
wherein m is an integer from 1 to 4; n is an integer from 1 to 8; and R17 and R18 each independently represents hydrogen, unsubstituted or substituted Cj- o alkyl, an unsaturated hydrocarbon chain of up to ten carbon atoms comprising one or more carbon-carbon double and/or triple bonds, C6 or C10 aryl, a 5- to 10-membered heterocyclic group, Cι-C10 alkoxy, - ,, thioalkoxy, hydroxyl, halo, cyano, nitro, amino, amido, (Cι-C10 alkyl)carbonyloxy,
Figure imgf000059_0001
alkoxy)carbonyl, ( - o alkyl)carbonyl, ( -Cu, alkyl)thiocarbonyl, ( -CH, alkyl)sulfonylamino, aminosulfonyl, 0,-CJO alkylsulfinyl, -CK, alkylsulfonyl, or a saturated or unsaturated C3-C12 hydrocarbon chain interrupted by O, S, NR, CO, C(NR), N(R)SO2, SO2N(R), N(R)C(O)O, OC(O)N(R), N(R)C(O)N(R), OC(O), C(O)O, OSO2, SO2O or OC(O)O where R is as defined above and the saturated or unsaturated hydrocarbon chain is optionally substituted as defined above; and pharmaceutically acceptable salts thereof.
2. Use according to claim 1, wherein R1, R2 and R3 are selected from hydrogen, C C5 alkyl, CrC6 alkoxy, amino, Cx-C6 alkylamino, di - alkyl)amino, halo, - haloalkyl, (C C6 alkoxy )carbonyl or - alkyl substituted by amino, -C6 alkoxy, -C6 alkylamino or di - alkyl) amino.
3. Use according to claim 1 or claim 2, wherein R4 and R5 are selected from hydrogen, - alkyl, - alkoxy, halo, - haloalkyl or -C6 alkyl substituted by amino, - alkoxy, C,-C6 alkylamino or di(Cj-C6 alkyl) amino.
4. Use according to any one of the preceding claims, wherein either or both of R4 and Rs is hydrogen.
5. Use according to any one of the preceding claims, wherein R11, R12 and R13 are each independently selected from hydrogen, methyl or ethyl.
6. Use according to any one of the preceding claims, wherein W represents -CH=N-, -CONH-, -SO2NH-, -CH2O- or -CH2S-.
7. Use according to any one of the preceding claims, wherein X represents -CF3, -OR5 or -NHOR5 wherein R5 is hydrogen or C,-C6 alkyl.
8. Use according to any one of the preceding claims, wherein Y represents:
Figure imgf000060_0001
wherein m is 1, 2 or 3; n is 1, 2 or 3; and R18 is hydrogen, - alkyl or - alkoxy
9. Use according to claim 1, wherein the compound is of formula (II):
Figure imgf000060_0002
wherein R1, R2, R6, R18, W and X are as defined above and m is 1, 2 or 3; and pharmaceutically acceptable salts thereof.
10. Use according to claim 9, wherein R1 and R2 independently represent hydrogen, Cj- alkyl, CrC6 alkoxy, halo or (C C6 alkoxy )carbonyl; R6 and R18 are each independendy selected from hydrogen, -Q alkyl or -Q, alkoxy; W represents a single bond, -CH=N-, N=CH-, -CONH-, -NHCO-, -SO2NH-, -NHSO2-, -OCH2-, -CH2O-, -CH2S- or -SCH2-; and X represents-NHOH, -CF3 or -OR14 wherein R14 is hydrogen or -Q alkyl.
11. Use according to claim 9, wherein the compound is of formula (III):
Figure imgf000060_0003
wherein R1 represents -Q alkyl, Ct-C4 alkoxy or halo; R6 and R18 are each independently selected from hydrogen, -C4 alkyl or -Q alkoxy; X represents -OR14 or -NHOH wherein R14 is hydrogen or -C4 alkyl; Z represents C or SO; and m is 1, 2 or 3; and pharmaceutically acceptable salts thereof.
12. Use according to claim 11, wherein R6 is methyl.
13. Use according to claim 11, wherein the compound is selected from: (2E,4E)-5-(4-(4-chlorobenzenesulfonylamino)phenyl)penta-2,4-dienoic acid ethyl ester; (2E,4E)-5-(4-(4-methoxybenzenesulfonylamino)phenyl)penta-2,4-dienoic acid ethyl ester; (2£,4E)-5-(4-(4-chlorobenzenesulfonylamino)phenyl)-4-methylpenta-2,4-dienoic acid ethyl ester;
(2E,4E)-5 -(4-(4-chlorobenzenesulf ony lamino)pheny l)-4-methy lpenta-2,4-dienoic acid ;
(2E,4E)-5-(4-(4-methoxybenzenesulfonylamino)phenyl)-4-methylpenta-2,4-dienoic acid ethyl ester; (2E,4E)-5-(4-(4-methoxybenzenesulfonylamino)phenyl)-2,4-dimethylpenta-2,4-dienoic acid ethyl ester;
(2E,4E)-5-(4-(4-methoxybenzenesulfonylamino)phenyl)-4-methylpenta-2,4-dienoic acid;
(2E,4E)-5r(4-(4-chlorobenzenesulfonylamino)phenyl)-4-methylpenta-2,4-dienoic acid ethyl ester;
(2E,4E)-5-(4-methoxybenzenesulfonylaminophenyl)-4-methylpenta-2,4-dienoic acid hydroxamide;
(2E,4E,6E)-7-(4-(4-methoxybenzenesulfonylamino)phenyl)-6-methylhepta-2,4,6- trienoic acid ethyl ester; and (2E,4E,6E)-7-(4-(4-chlorobenzoylamino)phenyl)hepta-2,4,6-trienoic acid methyl ester.
14. Use according to claim 1, wherein the compound is of formula (IV):
Figure imgf000062_0001
wherein R1, R2, R6, R18, W and X as defined in claim 1, the symbol represents a single bond or a double bond and n is 1, 2 or 3; and pharmaceutically acceptable salts thereof.
15. Use according to claim 14, wherein R1 and R2 each independently represent hydrogen, - alkyl, C,-C6 alkoxy, halo or (CrC6 alkoxy )carbonyl; R6 and R18 are each independently selected from hydrogen, CrC4 alkyl or C C4 alkoxy; W represents a single bond, -CH=N-, -N=CH-, CONH-, -NHCO-, -SO2NH-, -NHSO2-, -OCH2-, -CH2O-, -CH2S- or -SCH2-; and X represents -NHOH, CF3 or -OR14 wherein R14 is hydrogen or C,-C4 alkyl.
16. Use according to claim 14, wherein the compound is of formula (V):
Figure imgf000062_0002
wherein the symbol represents a single bond or a double bond; R1 represents -C4 alkyl, C,-C4 alkoxy or halo; R6 and R18 are each independently selected from hydrogen, - alkyl or - alkoxy; X represents -OR14 or -NHOH wherein R14 is hydrogen or C C4 alkyl; Z represents C or SO; and n is 1, 2 or 3; and pharmaceutically acceptable salts thereof.
17. Use according to claim 16, wherein R6 is methyl.
18. Use according to claim 16, wherein the compound is selected from: 5-[4-(4-methoxybenzenesulfonylamino)phenyl]-4-methylpentanoic acid ethyl ester; ethyl 5-[4-(4-chlorobenzenesulfonylamino)phenyl]-4-methylpentanoate; ethyl 5- [4-(4-chlorobenzoy lamino)phenyl] -4-me thy lpentanoate ;
5-[4-(4-chlorobenzenesulfonylamino)phenyl]-4-methylpentanoic acid hydroxyamide; and
5-[4-(4-chlorobenzoylamino)phenyl]-4-methylpentanoic acid hydroxyamide.
19. Use of a compound of formula (VI) for the manufacture of a medicament for use in treating a disorder mediated by histone deacetylase:
Figure imgf000063_0001
wherein the symbol ™ represents a single bond or a double bond or the symbol ^ , R6 and R8 together represent cyclopropyl; R1 to R5 each independently represent hydrogen, -CK, alkyl, C2-C10 alkenyl, C2-C10 alkynyl, - o alkoxy, - o thioalkoxy, hydroxyl,
Figure imgf000063_0002
halo, - o haloalkyl, amino, -CK, alkylamino, d^ - o alkyl)amino, amido, nitro, cyano, ( - o alkyl)carbonyloxy, (CrC10 alkoxy )carbonyl, ( - o alkyl)carbonyl, (Cι-C10 alkyl)thiocarbonyl, (CrC10 alkyl)sulfonylamino, aminosulfonyl, ( - o alkyl)sulfinyl, (Cx-C10 alkyl)sulfonyl or
Figure imgf000063_0003
alkyl substituted by amino, - Q alkoxy, - o alkylamino or di(C1-C10 alkyl)amino; R6 represents hydrogen, C,-C10 alkyl, substituted CrC10 alkyl, an unsaturated hydrocarbon chain of up to ten carbon atoms comprising one or more carbon-carbon double or triple bond, C6 or C10aryl, a 5- to 10-membered heterocyclic group, -C,,, alkoxy, - o thioalkoxy, hydroxyl, halo, cyano, nitro, amino, -CK, alkylamino, di(C1-C10 alkyl)amino, amido, ( - Q alkyl)carbonyloxy, ( - o alkoxy )carbonyl, ( -CJO alkyl)carbonyl, (C1-C10 alkyl)thiocarbonyl, (Cj-C,,, alkyl)sulfonylamino, aminosulfonyl, (CJ-CJO alkyl)sulfinyl, (C C10 alkyl)sulfonyl, a saturated or unsaturated C3-C12 hydrocarbon chain interrupted by O, S, NR, CO, C=NR, N(R)SO2, SO2N(R), N(R)C(O)O, OC(O)N(R), N(R)C(O)N(R), OC(O), C(O)O, OSO2, SO2O or OC(O)O where: (a) R independently represents hydrogen, C C10 alkyl, C2-C10 alkenyl, C2-C10 alkynyl, - o alkoxy, CrC10 hydroxyalkyl, hydroxyl or Cγ-Cw haloalkyl, and (b) the saturated or unsaturated hydrocarbon chain is optionally substituted with -Cjo alkyl, C2-C10 alkenyl, C>-C10 alkynyl, - ,, alkoxy, hydroxyl, - o hydroxyalkyl, halo, - o haloalkyl, amino, (CrC10 alkyl)carbonyloxy, ( -CK, alkoxy )carbonyl, (Cj- o alkyl)carbonyl, (Ci-Cjo alkyl)sulfonylamino, aminosulfonyl or - o alkylsulfonyl, when the symbol ™ represents a single bond, R7 and R8 each independently represents hydrogen, halo, Cj-C10 alkyl, C6 or C10 aryl, a 5- to 10-membered heterocyclic group, OR9, SR9 or NR9R10 wherein R9 and R10 each independently represent hydrogen or - alkyl or one of R9 and R10 is H and the other is -CO(CrC6 alkyl). or R7 and R8 together represent =O, =CH2 or =CHR9 wherein R9 is as defined above; when the symbol z zz represents a double bond, R7 represents hydrogen, halo, Cj- o alkyl, C6 or Cw aryl, a 5- to 10-membered heterocyclic group, OR9, SR9 or NR9R10 wherein R9 and R10 are as defined above and R8 is absent; - W represents a single bond, -C(Rn)=N-, -N=C(Rn)-, -C(Rπ)(R12)-NR13-,
-NR13-C(RU)(R12)-, -CO-NR11-, -NRπ-CO-, -SO2-NRu-, -NR"-SO2-, -C(Ru)(R12)-O-, -O-C(Rn)(R12)-, -C(RU)(R12)-S-, -S-C(RU)(R12)-, -CO-, -NR11-, -SO-, -SO2-, S or -[C(Rn)R12]p- wherein R11, R12 and R13 each independently represents hydrogen, - alkyl, C6 or C10 aryl or a 5- to 10- membered heterocyclic group alkyl and p is an integer of from 1 to 4; X represents -OR14, -SR14, -NR14OR15,-NR14NR15R16' -CF3, -CF2H or CH2F wherein R14, R15 and R16 each independendy represents hydrogen or - alkyl; and Y represents
Figure imgf000064_0001
wherein m is an integer from 1 to 4; n is an integer from 1 to 8; and R17 and R18 each independently represents hydrogen, unsubstituted or substituted -CJO alkyl, an unsaturated hydrocarbon chain of up'to ten carbon atoms comprising one or more carbon-carbon double and/or triple bonds, C6 or C10 aryl, a 5- to 10-membered heterocyclic group, -CH, alkoxy, -CJO thioalkoxy, hydroxyl, halo, cyano, nitro, amino, amido, ( - o alkyl)carbonyloxy, ( -Cj,, alkoxy )carbonyl, (Cj- o alkyl)carbonyl, ( - Q alkyl)thiocarbonyl, (Cj-C10 alkyl)sulfonylamino, aminosulfonyl, CJ-CJO alkylsulfinyl, - o alkylsulfonyl, or a saturated or unsaturated C3-C12 hydrocarbon chain interrupted by O, S, NR, CO, C(NR), N(R)SO2, SO2N(R), N(R)C(O)O, OC(O)N(R), N(R)C(O)N(R), OC(O), C(O)O, OSO2, SO2O or OC(O)O where R is as defined above and the saturated or unsaturated hydrocarbon chain is optionally substituted as defined above; and pharmaceutically acceptable salts thereof.
20. Use according to claim 19 wherein R1, R2 and R3 are selected from hydrogen, - alkyl, - alkoxy, amino, - alkylamino, di - alkyl)amino, halo, - haloalkyl, ( - alkoxy)carbonyl or - alkyl substituted by amino, Cj-C6 alkoxy, CrC6 alkylamino or di -Cg alkyl) amino.
21. Use according to claim 19 or claim 20 wherein one or two of R1, R2 and R3 is hydrogen.
22. Use according to any one of claims 19 to 21 and wherein the compound is additionally as defined in any one of claims 3 to 7, 9 or 10.
23. Use according to claim 19 wherein the compound is of formula (VII):
Figure imgf000065_0001
wherein R1 represents hydrogen, Ca-C4 alkyl, - alkoxy or halo; R6 and R18 are each independently selected from hydrogen, - alkyl or C C4 alkoxy; X represents -OR14 or -NHOH wherein R14 is hydrogen or Cj-C4 alkyl; Z represents C or SO; and m is 1, 2 or 3; and pharmaceutically acceptable salts thereof.
24. Use according to claim 23 wherein R6 is methyl
25. Use according to claim 23 wherein the compound is selected from: (2E,4E)-5-(4rbenzenesulfonylaminophenyl)-4-methylpenta-2,4-dienoic acid ethyl ester; (2E,4E)-5-(4-benzenesulfonylaminophenyl)-4-methylpenta-2,4-dienoic acid; and (2E,4E)-5-(4-benzenesulfonylaminophenyl)-4-methylpenta-2,4-dienoic acid hydroxamide.
26. Use according to any one of the preceding claims, wherein the disorder is a cancer, cardiac hypertrophy, a hematological disorder or a genetic-related metabolic disorder.
27. Use according to claim 26, wherein the cancer is leukemia, lung cancer, colon cancer, CNS cancer, melanoma, ovarian cancer, cervical cancer, renal cancer, prostate cancer and breast cancer.
28. A pharmaceutical composition comprising a pharmaceutically acceptable carried or diluent and, as active ingredient, a compound as defined in any one of claims 1 to 25 and which further comprises another chemotherapeutic or antineoplastic agent.
29. A composition according to claim 28, which further comprises a DNA methylation inhibitor.
30. Products containing a compound as defined in any one of claims 1 to 25 and another chemotherapeutic or antineoplastic agent as a combined preparation for simultaneous, separate or sequential use in treating a cancer.
31. Products according to claim 30, wherein the said agent is a DNA methylation inhibitor.
32. A method of treating a histone deacetylase-mediated disorder, which method comprises the step of administering to a subject having a said disorder a therapeutically effective amount of a compound as defined in any one of claims 1 to 25.
33. A method according to claim 32, comprising the further step of administering another chemotherapeutic or antineoplastic agent to said subject suffering from a cancer.
34. A method according to claim 33, wherein the said agent is a DNA methylation inhibitor.
35. A compound of formula (I) as defined in claim 1 wherein: when the symbol z^z represents a double bond, R7 represents hydrogen, halo, Cj- o alkyl, C6 or C10 aryl, pyrazinyl, pyrimidinyl, pyridazinyl, furanyl, thienyl, imidazolyl, pyrazolidinyl, oxadiazolyl, oxazyl, isoxazyl, thiadiazolyl, thiazolyl, 1,2,3- triazolyl, tetrazolyl, pyrazolyl, OR9, SR9 or NR9R10 wherein R9 and R10 are as defined above and R8 is absent; and W represents -C(R")=N-, -N=C(Rn)-, -C(Rn)(R12)-NR13-, -NR13-C(RU)(R12)-, -CO-NR11-, -NRπ-CO-, -SO2-NRu-, -NRu-SO2-, -C(Ru)(R12)-O-, -O-C(Ru)(R12)-, -C(Rn)(R12)-S-, -S-C(RU)(R12)-, -CO-, -NR11-, -SO-, -SO2-, O, S or -[C(Rn)R12]p- wherein R", R12 and R13 each independently represents hydrogen, - alkyl, C6 or C10 aryl or a 5- to 10- membered heterocyclic group alkyl and p is an integer of from 1 to 4; and pharmaceutically acceptable salts thereof, with the proviso that that compound is not 6-(4-(4-chlorobenzenesulfonyl amino)phenyl)hex-5-enoic acid or 7-(4-(4-chlorobenzenesulfonylamino)phenyl)hept-6- enoic acid.
36. A compound according to claim 35 and additionally as defined in any one of claims 2 to 9.
37. A compound according to claim 35, having the formula (II):
Figure imgf000068_0001
wherein R1, R2, R6, R18, W and X are as defined above and m is 1, 2 or 3; and pharmaceutically acceptable salts thereof.
38. A compound according to claim 37 wherein R1 and R2 independently represent hydrogen, CrC6 alkyl, Ct-C6 alkoxy, halo or ( - alkoxy )carbonyl; R6 and R18 are each independently selected from hydrogen, - alkyl or - alkoxy; W represents -CH=N-, N=CH-, -CONH-, -NHCO-, -SO2NH-, -NHSO2-, -OCH2-, -CH2O-, -CH2S- or -SCH2-; and X represents-NHOH, -CF3 or -OR14 wherein R14 is hydrogen or CrC4 alkyl.
39. A compound according to claim 37, wherein the compound is of formula (III):
Figure imgf000068_0002
wherein R1 represents hydrogen, - alkyl, -C4 alkoxy or halo; R6 and R18 are each independently selected from hydrogen, CrC4 alkyl or Cα-C4 alkoxy; X represents -OR14 or -NHOH wherein R14 is hydrogen or C C4 alkyl; Z represents C or SO; and m is 1, 2 or 3; and pharmaceutically acceptable salts thereof
40. A compound according to claim 39 and additionally as defined in claim 12 or claim 13.
41. A compound according to claim 35, having the formula (IV): 68
Figure imgf000069_0001
wherein R1, R2, R6, R18, W and X are as defined above, the symbol represents a single bond or a double bond and n is 1, 2 or 3; and pharmaceutically acceptable salts thereof.
42. A compound according to claim 41 wherein R1 and R2 each independently represent hydrogen, C,-C6 alkyl, - alkoxy, halo or ( - alkoxy )carbonyl; R6 and R18 are each independently selected from hydrogen, C,-C4 alkyl or - alkoxy; W represents -CH=N-, -N=CH-, CONH-, -NHCO-, -SO2NH-, -NHSO2-, -OCH2-, -CH2O-, -CH2S- or -SCH2-; and X represents -NHOH, CF3 or -OR14 wherein R14 is hydrogen or CrC4 alkyl.
43. A compound according to claim 41 and additionally as defined in any one of claims 16 to 18.
44. A compound of formula (VI) as defined in claim 19.
45. A compound according to claim 44 wherein m is 1, 2 or 3 and R is hydrogen
46. A compound according to claim 44 or claim 45 and additionally as defined in any one of claims 20 to 25.
PCT/GB2004/003155 2003-07-18 2004-07-19 Pharmaceutical agents WO2005011661A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
GB0316889A GB0316889D0 (en) 2003-07-18 2003-07-18 Pharmaceutical agents
GB0316889.5 2003-07-18

Publications (1)

Publication Number Publication Date
WO2005011661A1 true WO2005011661A1 (en) 2005-02-10

Family

ID=27764103

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/GB2004/003155 WO2005011661A1 (en) 2003-07-18 2004-07-19 Pharmaceutical agents

Country Status (2)

Country Link
GB (1) GB0316889D0 (en)
WO (1) WO2005011661A1 (en)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009079375A1 (en) 2007-12-14 2009-06-25 Georgetown University Histone deacetylase inhibitors
US7732475B2 (en) 2005-07-14 2010-06-08 Takeda San Diego, Inc. Histone deacetylase inhibitors
WO2014014900A1 (en) * 2012-07-16 2014-01-23 Chdi Foundation, Inc. Histone deacetylase inhibitors and compositions and methods of use thereof
US9765054B2 (en) 2011-01-24 2017-09-19 Chdi Foundation, Inc. Histone deacetylase inhibitors and compositions and methods of use thereof
US10478431B2 (en) 2014-11-26 2019-11-19 The J. David Gladstone Institutes Methods for treating a cytomegalovirus infection
WO2020186101A1 (en) 2019-03-12 2020-09-17 The Broad Institute, Inc. Detection means, compositions and methods for modulating synovial sarcoma cells

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0471259A1 (en) * 1990-08-16 1992-02-19 Bayer Ag Phenylsulfonamid substituted pyridinealken- and aminooxyalkan-carboxylic-acid derivatives
US5286736A (en) * 1990-11-22 1994-02-15 Dr. Karl Thomae Gmbh Pyridyl compounds and pharmaceutical compositions containing these compounds

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0471259A1 (en) * 1990-08-16 1992-02-19 Bayer Ag Phenylsulfonamid substituted pyridinealken- and aminooxyalkan-carboxylic-acid derivatives
US5286736A (en) * 1990-11-22 1994-02-15 Dr. Karl Thomae Gmbh Pyridyl compounds and pharmaceutical compositions containing these compounds

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
DATABASE CAPLUS CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; PRABHAKAR, Y. ET AL.: "QSAR study of the role of hydrophobicity in the activity of HMGR inhibitors", XP002301563, Database accession no. 1989:417158 *
DATABASE CAPLUS CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; TAKEUCHI, K. ET AL.: "Development of dual-acting agents for thromboxane receptor antagonism and thromboxane synthase inhibition I. Synthesis, structure-activity relationship, and evaluation of substituted omega-phenyl-(3-pyridyl)alkenoic acids", XP002301564, Database accession no. 1995:270973 *
DATABASE CAPLUS CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; XP002301565, Database accession no. 1994:298479 *
DATABASE CAPLUS CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; XP002301566, retrieved from STN Database accession no. 1992:214360 *

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7732475B2 (en) 2005-07-14 2010-06-08 Takeda San Diego, Inc. Histone deacetylase inhibitors
US7741494B2 (en) 2005-07-14 2010-06-22 Takeda San Diego, Inc. Histone deacetylase inhibitors
WO2009079375A1 (en) 2007-12-14 2009-06-25 Georgetown University Histone deacetylase inhibitors
US8293513B2 (en) 2007-12-14 2012-10-23 Georgetown University Histone deacetylase inhibitors
US9765054B2 (en) 2011-01-24 2017-09-19 Chdi Foundation, Inc. Histone deacetylase inhibitors and compositions and methods of use thereof
WO2014014900A1 (en) * 2012-07-16 2014-01-23 Chdi Foundation, Inc. Histone deacetylase inhibitors and compositions and methods of use thereof
US9505736B2 (en) 2012-07-16 2016-11-29 Chdi Foundation, Inc. Histone deacetylase inhibitors and compositions and methods of use thereof
US9855267B2 (en) 2012-07-16 2018-01-02 Chdi Foundation, Inc. Histone deacetylase inhibitors and compositions and methods of use thereof
US10478431B2 (en) 2014-11-26 2019-11-19 The J. David Gladstone Institutes Methods for treating a cytomegalovirus infection
WO2020186101A1 (en) 2019-03-12 2020-09-17 The Broad Institute, Inc. Detection means, compositions and methods for modulating synovial sarcoma cells

Also Published As

Publication number Publication date
GB0316889D0 (en) 2003-08-20

Similar Documents

Publication Publication Date Title
AU2017253937B2 (en) Compounds and compositions for treating conditions associated with NLRP activity
JP4405602B2 (en) Histone deacetylase inhibitor
AU2004274184B2 (en) Thiazole derivatives as cannabinoid receptor modulators
CA2552279C (en) Zn2+-chelating motif-tethered short -chain fatty acids as a novel class of histone deacetylase inhibitors
US7173060B2 (en) Oxime derivatives and their use as pharmaceutically active agents
US7683185B2 (en) Histone deacetylase inhibitors
AU2013296187B2 (en) Enzyme-activating compounds and compositions
JP2001521504A (en) Metalloproteinase inhibitors, pharmaceutical compositions containing them and their use as medicaments
EP1666469A1 (en) Plasminogen activator inhibitor-1 inhibitor
Thaler et al. Synthesis and biological evaluation of N-hydroxyphenylacrylamides and N-hydroxypyridin-2-ylacrylamides as novel histone deacetylase inhibitors
WO2008068170A1 (en) Hdac inhibitors
EP1317411A1 (en) Derivatives of naphthalene with comt inhibiting activity
WO2005011661A1 (en) Pharmaceutical agents
KR20120041070A (en) Aryloxyphenoxyacryl-based compounds having hif-1 inhibition activity, preparation method thereof and pharmaceutical composition containing the same as an active ingredient
AU2004276128B2 (en) Amide-type carboxamide derivatives
JP2005527518A (en) Novel chalcone derivatives and their use
CZ223697A3 (en) Substituted phenyl derivatives as endothelin antagonists
KR20020093127A (en) Peptide deformylase inhibitors
US5026861A (en) Substituted 1,5-dihydro-4-(N-methylhydroxylamino)-2H-pyrrol-2-ones
EP0994107A1 (en) 5-Arylisoxazol-4-yl-substituted 2-amino carboxylic acid compounds
EP2912015B1 (en) Mixed inhibitors of aminopeptidase n and neprylisin
KR101900574B1 (en) Novel N-hydroxybenzamide and Use Thereof
US11351151B2 (en) Compound having anticancer activity and preparation method and application
US20040267015A1 (en) Peptide deformylase inhibitors
JPH07503002A (en) Nitroquinolone derivatives as NMDA antagonists

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 BW 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 NA 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 NA 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 PL 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
122 Ep: pct application non-entry in european phase